Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Recept Signal Transduct Res ; 41(5): 504-510, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32998602

RESUMO

BACKGROUND: Cell division control (CDC) 42 has been involved in the regulation of diverse cancers. Macrophage recruitment plays an important role in the pathogenesis and development of tumor. However, it remains unclear whether CDC42 contributes to macrophage recruitment and lung tumorigenesis in vivo. METHODS: Small interference RNA (siRNA) was used to knock down CDC42 in the Lewis lung carcinoma (LLC)1. The invasion capability of CDC42 knockdown LLC1 cells was evaluated. LLC1 cells with CDC42 targeted small hairpin RNA (shRNA) were inoculated into C57BL/6 mice to establish the tumor-bearing animal model Tumor size and metastasis related proteins were measured. In addition, the invasion of macrophages in the tumor site as well as macrophage chemokine were also determined in the model. RESULTS: The capacity of invasion and metastasis of LLC1 cells significantly decreased when CDC42 was knocked down. When inoculated with CDC42 knockdown LLC1 cells in vivo, the tumor size and metastasis related proteins levels both decreased. The invasion capacity of macrophages and the associated macrophage chemokine were also significantly down-regulated. CONCLUSION: Our data suggest that the inhibition of CDC42 expression in lung cancer cells can significantly prevent the pathogenesis and development of tumor in an allograft tumor model in vivo, which might provide a novel therapeutic target and potential strategy for lung cancer treatment in the future.


Assuntos
Carcinogênese/patologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Modelos Animais de Doenças , Macrófagos/imunologia , RNA Interferente Pequeno/genética , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Animais , Apoptose , Carcinogênese/imunologia , Carcinogênese/metabolismo , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína cdc42 de Ligação ao GTP/genética
2.
J Control Release ; 342: 228-240, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35016917

RESUMO

RNA interference (RNAi) is being extensively investigated as a potential therapeutic strategy for cancer treatment. However, RNAi-based therapeutics have not yet been used to treat cancer because of their instability and the difficulty of microRNA (miRNA) delivery. Plasmonic nanoparticle-based RNAi nanotherapeutics have been developed for accurate and sensitive diagnosis and a strong therapeutic effect on cancers by leveraging their ease-of-use and specific properties such as photothermal conversion. In this review, recent strategies and advances in plasmonic nanoparticle-based miRNA delivery are briefly presented to facilitate the detection and treatment of several cancers. The challenges and potential opportunities afforded by the RNAi-based theragnosis field are discussed. We expect that the RNAi-integrated plasmonic nanotherapeutics discussed in this review can provide insights for the early diagnosis and effective treatment of cancer.


Assuntos
MicroRNAs , Nanopartículas , Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , MicroRNAs/genética , MicroRNAs/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/terapia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico
3.
Int J Radiat Biol ; 97(11): 1569-1577, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34402389

RESUMO

PURPOSE: Ovarian cancer is one of the most common gynecological cancers in women with a low 5-year survival rate. Evaluation of hyaluronic acid-binding protein 1 (HABP1) level can provide important information for the diagnosis and treatment of ovarian cancer. In this study, we designed a novel HABP1 probe based on 99mTc-radiolabeled small-interference RNA (siRNA) for detecting HABP1 expression noninvasively in vivo, thereby providing a new method for its diagnosis and treatment. METHODS: A specific siHABP1 was selected because of its targetability and silencing effect. A negative control siRNA (NCsiRNA) with no homology with the human genome was used. SiHABP1 and NCsiRNA were radiolabeled with 99mTc using the bifunctional chelating agent hydrazinonicotinamide (HYNIC). The radiochemical purity and in vitro stability of the probe were determined by HPLC. The binding activity was measured by western blotting (WB) and RT-PCR. The HABP1-overexpressing human ovarian cancer cell line HO-8910 was used for cell uptake experiments, which were performed with or without transfection and measured with a gamma counter. HO8910-bearing mice were imaged at 1, 4, and 10 h, and biodistribution analysis was performed at 1, 4, 6, and 10 h after injection of 99mTc-HYNIC-siRNA. RESULTS: 99mTc-HYNIC-siHABP1 had high radiochemical purity and good in vitro stability, and showed the same binding capacity and silencing effect as siHABP1. SPECT imaging showed that tumors were clearly visualized at 10 h after injection of 99mTc-HYNIC-siHABP1 but not after 99mTc-HYNIC-NCsiRNA, implying specific binding. The biodistribution results were consistent with those of SPECT imaging. CONCLUSIONS: We showed that 99mTc-HYNIC-siHABP1 is a feasible probe for the noninvasive visualization of HABP1 expression in ovarian cancer.


Assuntos
Neoplasias Ovarianas , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Proteínas Mitocondriais , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Compostos Radiofarmacêuticos , Distribuição Tecidual
4.
G3 (Bethesda) ; 11(4)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33617633

RESUMO

Plant long non-coding RNAs (lncRNAs) function in diverse biological processes, and lncRNA expression is under epigenetic regulation, including by cytosine DNA methylation. However, it remains unclear whether 5-methylcytosine (5mC) plays a similar role in different sequence contexts (CG, CHG, and CHH). In this study, we characterized and compared the profiles of genome-wide lncRNA profiles (including long intergenic non-coding RNAs [lincRNAs] and long noncoding natural antisense transcripts [lncNATs]) of a null mutant of the rice DNA methyltransferase 1, OsMET1-2 (designated OsMET1-2-/-) and its isogenic wild type (OsMET1-2+/+). The En/Spm transposable element (TE) family, which was heavily methylated in OsMET1-2+/+, was transcriptionally de-repressed in OsMET1-2-/- due to genome-wide erasure of CG methylation, and this led to abundant production of specific lncRNAs. In addition, RdDM-mediated CHH hypermethylation was increased in the 5'-upstream genomic regions of lncRNAs in OsMET1-2-/-. The positive correlation between the expression of lincRNAs and that of their proximal protein-coding genes was also analyzed. Our study shows that CG methylation negatively regulates the TE-related expression of lncRNA and demonstrates that CHH methylation is also involved in the regulation of lncRNA expression.


Assuntos
Oryza , RNA Longo não Codificante , Metilação de DNA , Epigênese Genética , Regulação da Expressão Gênica de Plantas , Genoma de Planta , Metiltransferases/genética , Mutação , Oryza/genética , RNA Longo não Codificante/genética
5.
Am J Transl Res ; 12(11): 7420-7429, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33312378

RESUMO

Fibroblasts migrating to peritoneum injuries play an important role in the development of postoperative peritoneal adhesions due to the excessive synthesis and deposition of extracellular matrix (ECM). This effect is mainly induced by the transforming growth factor-ß (TGF-ß). Studies indicate that elevated TGF-ß1 levels and TGF-ß1/Smad signaling are both implicated in the formation of peritoneal adhesions. To confirm the effect of TGF-ß1/Smad signaling interference in regulating excessive ECM synthesis, a total of four different R-Smad-targeting small interference RNA (siRNA) duplexes (Smad2-500, Smad2-956, Smad3-378, Smad3-1385) were tested in this study using a TGF-ß1-stimulated adhesion tissue fibroblasts (ATFs) cell model. The in vitro assessments show that all proposed siRNAs are capable of significantly downregulating the mRNA and protein levels of Smad2 and Smad3 in ATFs. They also inhibit the phosphorylation of both Smads, which confirms their effect in blocking the TGF-ß1/Smad signaling pathway. Moreover, the siRNA duplexes can appreciably lower the elevated levels of fibronectin and collagen 3 alpha 1 (COL3A1) in TGF-ß1-stimulated ATFs, and the Smad3-378 siRNA can actually restore both molecules (fibronectin and COL3A1) to normal levels. Therefore, the Smad3-378 siRNA is suitable for both adhesion prevention and wound healing. Overall, our results indicate that postoperative adhesion prophylaxis may be achieved by temporarily blocking TGF-ß1/Smad signaling transduction.

6.
J Insect Physiol ; 111: 47-52, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30393170

RESUMO

RNA interference (RNAi) is a powerful tool for artificially manipulating gene expression in diverse organisms. In the honey bee, Apis mellifera, both long double stranded RNA (dsRNA) and small interference RNA (siRNA) have been successfully used to reduce targeted gene expression and induce specific phenotypes. However, whether dsRNA and siRNA have different effects and efficiencies in gene silencing has never been investigated in honey bees. Thus, we tested the effect of dsRNA and siRNA on the tyramine receptor 1 (tyr1), which encodes a receptor of neurotransmitter tyramine, in honey bee brains at mRNA and protein levels over time. We found that both dsRNA and siRNA achieved successful gene knockdown. The siRNA mixes affected tyr1 gene expression faster than dsRNA, and the duration of the knockdown between dsRNA and siRNA varied. We also found that the turnover rate of TYR1 protein was relatively fast, which is consistent with its role as a neurotransmitter receptor. Our study reveals the different efficiencies of dsRNA and siRNA in honey bee brains. We show that consideration of the gene regions targeted by RNAi, prior screening for RNAi molecules and combing siRNAs are important strategies to enhance RNAi efficiency.


Assuntos
Abelhas/genética , Proteínas de Insetos/genética , Interferência de RNA , RNA de Cadeia Dupla/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores de Amina Biogênica/genética , Animais , Abelhas/metabolismo , Encéfalo/metabolismo , Proteínas de Insetos/metabolismo , Receptores de Amina Biogênica/metabolismo
7.
J Gastrointest Cancer ; 48(2): 156-163, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27629422

RESUMO

PURPOSE: Overexpression of HMGA2, known as small non-histone chromosomal protein, is associated with progression of various tumors, including colorectal cancers. The aim of this study was to investigate the effect of a specific HMGA2 siRNA on apoptosis and cell cycle of HCT-116 (colorectal carcinoma) cells. METHODS: The cells were transfected with siRNAs using a transfection reagent. The cytotoxic effects of HMGA2 siRNA on colorectal carcinoma cells were determined using MTT assay. Relative HMGA2 mRNA and protein levels were measured by QRT-PCR and western blotting, respectively. Apoptosis was measured by a TUNEL test based on labeling of DNA strand breaks. We also evaluated caspase-3, caspase-8, and caspase-9 expression by QRTPCR to determine which pathway is involved in apoptosis. Cell cycle was assessed by FACS and cell cycle analysis using PI DNA staining. RESULTS: HMGA2 siRNA significantly reduced both mRNA and protein expression levels 48 h after transfection and dose-dependent manner in colorectal carcinoma cells. We also showed that the silencing of HMGA2 led to the induction of apoptosis through intrinsic pathway and cell cycle arrest in G2/M phases of interphase in HCT-116 cells in vitro. CONCLUSIONS: These results propose that HMGA2 might play an important role in the progression of colorectal carcinoma and might be a potential therapeutic target for trigger apoptosis and cell cycle arrest in colorectal carcinoma.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Proliferação de Células , Progressão da Doença , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HCT116 , Humanos , Marcação In Situ das Extremidades Cortadas , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
8.
Mol Imaging Biol ; 18(3): 353-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26452556

RESUMO

PURPOSE: Noninvasive quantification of chemokine receptor 4 (CXCR4) expression could serve as a prognostic indicator and may be of value for the design of personalized therapies and posttreatment monitoring. The objective of the present study was to assess the use of (99m)Tc-radiolabeled small-interference RNA (siRNA) targeting CXCR4 to detect CXCR4 expression in vivo. PROCEDURES: CXCR4 siRNAs were radiolabeled with (99m)Tc using the bifunctional chelator hydrazinonicotinamide (HYNIC), and the labeling efficiency, specific activity and radiochemical purity were determined. The stability of the probe in serum was assessed by measuring its radiochemical purity and inhibitory activity by RT-PCR and western blotting. Biodistribution studies and static imaging were performed in MDA-MB-231 tumor-bearing mice. RESULTS: Radiochemical purity remained highly stable in PBS and fresh human serum at room temperature and at 37 °C. Radiolabeled siRNA1 showed strong inhibitory effects similar to those of unlabeled siRNA1 on both CXCR4 messenger RNA (mRNA) and protein in vitro. The excretion of the probe occurred mainly through the liver and kidneys. Tumors were clearly visualized at 1-10 h after injection of the probe, but not after injection of the control probe. CONCLUSIONS: (99m)Tc-labeled CXCR4 siRNA1 shows tumor-specific accumulation and could be a promising strategy for the visualization of CXCR4 expression in human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Hidrazinas/metabolismo , Ácidos Nicotínicos/metabolismo , RNA Interferente Pequeno/metabolismo , Compostos Radiofarmacêuticos/química , Receptores CXCR4/metabolismo , Tecnécio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Endogâmicos BALB C , Imagem Molecular , Oligonucleotídeos Antissenso/metabolismo , Receptores CXCR4/genética , Distribuição Tecidual
9.
Adv Pharm Bull ; 6(2): 171-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27478778

RESUMO

PURPOSE: HMGI-C (High Mobility Group protein Isoform I-C) protein is a member of the high-mobility group AT-hook (HMGA) family of small non-histone chromosomal protein that can modulate transcription of an ample number of genes. Genome-wide studies revealed up regulation of the HMGI-C gene in many human cancers. We suggested that HMGI-C might play a critical role in the progression and migration of various tumors. However, the exact role of HMGI-C in breast adenocarcinoma has not been cleared. METHODS: The cells were transfected with siRNAs using transfection reagent. Relative HMGI-C mRNA and protein levels were measured by quantitative real-time PCR and Western blotting, respectively. The cytotoxic effects of HMGI-C siRNA, Paclitaxel alone and combination on breast adenocarcinoma cells were determined using MTT assay. The migration after treatment by HMGI-C siRNA, Paclitaxel alone and combination were detected by wound-healing respectively. RESULTS: HMGI-C siRNA significantly reduced both mRNA and protein expression levels in a 48 hours after transfection and dose dependent manner. We observed that the knockdown of HMGI-C led to the significant reduced cell viability and inhibited cells migration in MDA-MB-468 cells in vitro. CONCLUSION: These results propose that HMGI-C silencing and Paclitaxel treatment alone can inhibit the proliferation and migration significantly, furthermore, synergic effect of HMGI-C siRNA and Paclitaxel showed higher inhibition compared to mono treatment. Taken together, HMGI-C could be used as a promising therapeutic agent in the treatment of human breast adenocarcinoma. Therefore HMGI-C siRNA may be an effective adjuvant in human breast adenocarcinoma.

10.
Cell Cycle ; 15(19): 2585-2592, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27245202

RESUMO

PURPOSE: The HMGI-C (high mobility group protein isoform I-C) protein is a member of the high-mobility group AT-hook (HMGA) family of small non-histone chromosomal proteins that can modulate transcription of an ample number of genes. Genome-wide studies reveal upregulation of the HMGI-C gene in many human cancers, which suggests that HMGI-C might play a critical role in the progression of various tumors. However, the exact role of HMGI-C in breast adenocarcinoma has not been made clear. METHODS: HMGI-C mRNA expression in breast cancer samples and marginal normal tissues was characterized using qRT-PCR. The cytotoxic effects of HMGI-C siRNA on breast adenocarcinoma cells were determined using MTT assay. Relative HMGI-C mRNA and protein levels were measured by quantitative real-time PCR and western blotting, respectively. Apoptosis detection was done using TUNEL and Annexin-V/PI assays, P53, caspase 3, 9, 8 and Bcl2 proteins evaluated by protein gel blot and miR34a, Let-7a genes investigates by QRT-PCR assay. Cell cycle was analyzed by flow cytometry assay using propidium iodide DNA staining. RESULTS: An overexpression of HMGA2 was revealed with highly statistically significant differences between breast cancer samples and marginal normal tissues (P < 0.0001). HMGI-C siRNA significantly reduced both mRNA and protein expression levels in a 48-hour period after transfection and in a dose-dependent manner. We observed that the knockdown of HMGI-C led to the significant induction of apoptosis via mitochondrial pathway by inducing miR34a and cell cycle arrest in MDA-MB-468 cells in vitro. CONCLUSIONS: These results propose that HMGI-C might play a critical role in the progression of breast adenocarcinoma. Here we introduced HMGI-C as a potential therapeutic target for trigger apoptosis and cell cycle arrest in human breast adenocarcinoma. Therefore HMGI-C siRNA may be an effective adjuvant in human breast adenocarcinoma.


Assuntos
Adenocarcinoma/patologia , Apoptose , Neoplasias da Mama/patologia , Caspase 9/metabolismo , Proteína HMGA2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adenocarcinoma/genética , Apoptose/genética , Neoplasias da Mama/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Proteína HMGA2/genética , Humanos , MicroRNAs/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA