Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 453
Filtrar
2.
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
3.
Brain ; 144(10): 2985-2993, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-33993268

RESUMO

Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in mouse models of the disease. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analysed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.


Assuntos
Glicogênio Sintase/administração & dosagem , Doença de Lafora/tratamento farmacológico , Doença de Lafora/patologia , Oligorribonucleotídeos Antissenso/administração & dosagem , Animais , Feminino , Injeções Intraventriculares , Doença de Lafora/genética , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética
4.
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
5.
RNA ; 26(11): 1731-1742, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32759389

RESUMO

The measurement of RNA abundance derived from massively parallel sequencing experiments is an essential technique. Methods that reduce ribosomal RNA levels are usually required prior to sequencing library construction because ribosomal RNA typically comprises the vast majority of a total RNA sample. For some experiments, ribosomal RNA depletion is favored over poly(A) selection because it offers a more inclusive representation of the transcriptome. However, methods to deplete ribosomal RNA are generally proprietary, complex, inefficient, applicable to only specific species, or compatible with only a narrow range of RNA input levels. Here, we describe Ribo-Pop (ribosomal RNA depletion for popular use), a simple workflow and antisense oligo design strategy that we demonstrate works over a wide input range and can be easily adapted to any organism with a sequenced genome. We provide a computational pipeline for probe selection, a streamlined 20-min protocol, and ready-to-use oligo sequences for several organisms. We anticipate that our simple and generalizable "open source" design strategy would enable virtually any laboratory to pursue full transcriptome sequencing in their organism of interest with minimal time and resource investment.


Assuntos
Biologia Computacional/métodos , Oligorribonucleotídeos Antissenso/genética , RNA Ribossômico/análise , Sequência de Bases , Biologia Computacional/economia , Análise Custo-Benefício , Sequenciamento de Nucleotídeos em Larga Escala , Sondas de Oligonucleotídeos/genética , RNA Ribossômico/antagonistas & inibidores , Análise de Sequência de RNA/métodos , Fluxo de Trabalho
6.
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
7.
PLoS One ; 15(7): e0236195, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32678841

RESUMO

During infection, viruses enter susceptible host cells in order to replicate their components for production of new virions. In the process of infection, the gene expression of infected cells undergoes changes because of the production of viral components and due to the host response from detection of viral products. In the advent of RNA sequencing, the discovery of new genes and their functions in the host response generates new avenues for interventions in the host-pathogen interaction. We have identified a novel gene, Heatr9, as a virus and cytokine inducible viral responsive gene. We confirm Heatr9's expression in vitro and in vivo during virus infection and correlate it with viral burden. Heatr9 is induced by influenza virus and RSV. Heatr9 knockdown during viral infection was shown to affect chemokine expression. Our studies identify Heatr9 as a novel inflammatory and virus infection induced gene that can regulate the induction of specific cytokines.


Assuntos
Citocinas/metabolismo , Orthomyxoviridae/fisiologia , Proteínas de Ligação a RNA/metabolismo , Células A549 , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/virologia , Animais , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Citocinas/genética , Feminino , Loci Gênicos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oligorribonucleotídeos Antissenso/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Vírus Sinciciais Respiratórios/fisiologia , Regulação para Cima
8.
Yakugaku Zasshi ; 140(5): 687-700, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32378673

RESUMO

Recent advances in high-throughput technologies have revealed that 75% of the human genome is transcribed to RNA, whereas only 3% of transcripts are translated into proteins. Consequently, many long non-coding RNAs (lncRNAs) have been identified, which has improved our understanding of the complexity of biological processes. LncRNAs comprise multiple classes of RNA transcripts that regulate the transcription, stability and translation of protein-coding genes in a genome. Natural antisense transcripts (NATs) form one such class, and the GENCODE v30 catalog contains 16193 lncRNA loci, of which 5611 are antisense loci. This review outlines our emerging understanding of lncRNAs, with a particular focus on how lncRNAs regulate gene expression using interferon-α1 (IFN-α1) mRNA and its antisense partner IFN-α1 antisense (as)RNA as an example. We have identified and characterized the asRNA that determines post-transcriptional IFN-α1 mRNA levels. IFN-α1 asRNA stabilizes IFN-α1 mRNA by cytoplasmic sense-antisense duplex formation, which may enhance the accessibility of an RNA stabilizer protein or decrease the affinity of an RNA decay factor for the RNA. IFN-α1 asRNA can also act as competing molecules in the competing endogenous (ce)RNA network with other members of the IFNA multigene family mRNAs/asRNAs, and other cellular mRNA transcripts. Furthermore, antisense oligoribonucleotides representing functional domains of IFN-α1 asRNA inhibit influenza virus proliferation in the respiratory tract of virus-infected animals. Thus, these findings support, at least in part, the rationale that dissecting the activity of NAT on gene expression regulation promises to reveal previously unanticipated biology, with potential to provide new therapeutic approaches to diseases.


Assuntos
Regulação da Expressão Gênica/genética , RNA Antissenso/fisiologia , RNA não Traduzido/fisiologia , Animais , Genoma Humano/genética , Humanos , Interferon-alfa/química , Interferon-alfa/genética , Família Multigênica , Oligorribonucleotídeos Antissenso/fisiologia , Orthomyxoviridae/fisiologia , Estabilidade de RNA , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Sistema Respiratório/virologia , Transcrição Gênica/genética , Replicação Viral
10.
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
11.
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
12.
Curr Gene Ther ; 19(3): 172-196, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31566126

RESUMO

Long-term research on various types of RNAs has led to further understanding of diverse mechanisms, which eventually resulted in the rapid development of RNA-based therapeutics as powerful tools in clinical disease treatment. Some of the developing RNA drugs obey the antisense mechanisms including antisense oligonucleotides, small interfering RNAs, microRNAs, small activating RNAs, and ribozymes. These types of RNAs could be utilized to inhibit/activate gene expression or change splicing to provide functional proteins. In the meantime, some others based on different mechanisms like modified messenger RNAs could replace the dysfunctional endogenous genes to manage some genetic diseases, and aptamers with special three-dimensional structures could bind to specific targets in a high-affinity manner. In addition, the recent most popular CRISPR-Cas technology, consisting of a crucial single guide RNA, could edit DNA directly to generate therapeutic effects. The desired results from recent clinical trials indicated the great potential of RNA-based drugs in the treatment of various diseases, but further studies on improving delivery materials and RNA modifications are required for the novel RNA-based drugs to translate to the clinic. This review focused on the advances and clinical studies of current RNA-based therapeutics, analyzed their challenges and prospects.


Assuntos
Ensaios Clínicos como Assunto , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Terapia Genética , Aptâmeros de Nucleotídeos/genética , Sistemas CRISPR-Cas , Humanos , MicroRNAs/genética , Oligorribonucleotídeos Antissenso/genética , RNA Catalítico , RNA Interferente Pequeno/genética
13.
Genes (Basel) ; 10(9)2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31547430

RESUMO

Amyloid beta-peptide is produced by the cleavage of amyloid precursor protein by two secretases, a ß-secretase, beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and a γ-secretase. It has been hypothesised that partial inhibition of BACE1 in individuals with a high risk of developing Alzheimer's disease may be beneficial in preventing cognitive decline. In this study, we report the development of a novel antisense oligonucleotide (AO) that could efficiently downregulate the BACE1 transcript and partially inhibit BACE1 protein. We designed and synthesised a range of 2'-OMethyl-modified antisense oligonucleotides with a phosphorothioate backbone across various exons of the BACE1 transcript, of which AO2, targeting exon 2, efficiently downregulated BACE1 RNA expression by 90%. The sequence of AO2 was later synthesised with a phosphorodiamidate morpholino chemistry, which was found to be not as efficient at downregulating BACE1 expression as the 2'-OMethyl antisense oligonucleotides with a phosphorothioate backbone variant. AO2 also reduced BACE1 protein levels by 45%. In line with our results, we firmly believe that AO2 could be used as a potential preventative therapeutic strategy for Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Ácido Aspártico Endopeptidases/genética , Regulação para Baixo , Oligorribonucleotídeos Antissenso/genética , RNA Mensageiro/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Células HEK293 , Humanos , Oligorribonucleotídeos Antissenso/metabolismo , RNA Mensageiro/genética , Terapêutica com RNAi/métodos
14.
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
15.
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
16.
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
17.
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
18.
Clin Transl Sci ; 12(2): 98-112, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30706991

RESUMO

The therapeutic pathways that modulate transcription mechanisms currently include gene knockdown and splicing modulation. However, additional mechanisms may come into play as more understanding of molecular biology and disease etiology emerge. Building on advances in chemistry and delivery technology, oligonucleotide therapeutics is emerging as an established, validated class of drugs that can modulate a multitude of genetic targets. These targets include over 10,000 proteins in the human genome that have hitherto been considered undruggable by small molecules and protein therapeutics. The approval of five oligonucleotides within the last 2 years elicited unprecedented excitement in the field. However, there are remaining challenges to overcome and significant room for future innovation to fully realize the potential of oligonucleotide therapeutics. In this review, we focus on the translational strategies encompassing preclinical evaluation and clinical development in the context of approved oligonucleotide therapeutics. Translational approaches with respect to pharmacology, pharmacokinetics, cardiac safety evaluation, and dose selection that are specific to this class of drugs are reviewed with examples. The mechanism of action, chemical evolution, and intracellular delivery of oligonucleotide therapies are only briefly reviewed to provide a general background for this class of drugs.


Assuntos
Terapia Genética/métodos , RNA Mensageiro/genética , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Ensaios Clínicos como Assunto , Aprovação de Drogas , Sistemas de Liberação de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Oligorribonucleotídeos Antissenso/administração & dosagem , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/farmacocinética , Interferência de RNA , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/agonistas , RNA Mensageiro/antagonistas & inibidores , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacocinética , Transcrição Gênica/efeitos dos fármacos
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...