Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Journal of Zhejiang University. Medical sciences ; (6): 1-11, 2023.
Article in English | WPRIM | ID: wpr-982057

ABSTRACT

RNA therapeutics inhibit the expression of specific proteins/RNAs by targeting complementary sequences of corresponding genes, or synthesize proteins encoded by the desired genes to treat genetic diseases. RNA-based therapeutics are categorized as oligonucleotide drugs (antisense oligonucleotides, small interfering RNA, RNA aptamers), and mRNA drugs. The antisense oligonucleotides and small interfering RNA for treatment of genetic diseases have been approved by the FDA in the United State, while RNA aptamers and mRNA drugs are still in clinical trials. Chemical modifications are applied to RNA drugs, such as pseudouridine modification of mRNA, to reduce immunogenicity and improve the efficacy. The secure and effective delivery systems like lipid-based nanoparticles, extracellular vesicles, and virus-like particles are under development to address stability, specificity, and safety issues of RNA drugs. This article provides an overview of the specific molecular mechanisms of 11 RNA drugs currently used for treating genetic diseases, and discusses the research progress of chemical modifications and delivery systems of RNA drugs.

2.
Singapore medical journal ; : 17-26, 2023.
Article in English | WPRIM | ID: wpr-969661

ABSTRACT

Inherited ocular diseases comprise a heterogeneous group of rare and complex diseases, including inherited retinal diseases (IRDs) and inherited optic neuropathies. Recent success in adeno-associated virus-based gene therapy, voretigene neparvovec (Luxturna®) for RPE65-related IRDs, has heralded rapid evolution in gene therapy platform technologies and strategies, from gene augmentation to RNA editing, as well as gene agnostic approaches such as optogenetics. This review discusses the fundamentals underlying the mode of inheritance, natural history studies and clinical trial outcomes, as well as current and emerging therapies covering gene therapy strategies, cell-based therapies and bionic vision.


Subject(s)
Humans , Eye Diseases/therapy
3.
Acta Pharmaceutica Sinica B ; (6): 903-915, 2023.
Article in English | WPRIM | ID: wpr-971765

ABSTRACT

We summarize the most important advances in RNA delivery and nanomedicine. We describe lipid nanoparticle-based RNA therapeutics and the impacts on the development of novel drugs. The fundamental properties of the key RNA members are described. We introduced recent advances in the nanoparticles to deliver RNA to defined targets, with a focus on lipid nanoparticles (LNPs). We review recent advances in biomedical therapy based on RNA drug delivery and state-of-the-art RNA application platforms, including the treatment of different types of cancer. This review presents an overview of current LNPs based RNA therapies in cancer treatment and provides deep insight into the development of future nanomedicines sophisticatedly combining the unparalleled functions of RNA therapeutics and nanotechnology.

4.
Acta Pharmaceutica Sinica ; (12): 1301-1311, 2022.
Article in Chinese | WPRIM | ID: wpr-924765

ABSTRACT

As one of the most serious hereditary neuromuscular disease, spinal muscular atrophy (SMA) is caused by the loss or mutation of survival motor neuron 1 (SMN1) gene. It leads to a decrease in the level of SMN protein and a consequent loss of alpha neurons and progressive muscle atrophy resulting in the progressive muscle weakness, the significant disability and the shortened lifespan. Up till now, only three drugs have been approved for SMA, including the gene therapy drug onasemnogene abeparvovec. The antisense oligonucleotide drug nusinersen and and the small molecule chemical drug risdiplam were briefly introduced. Some representative samples of the small molecule chemical drugs and antisense oligonucleotide drugs targeting SMN2 in the clinical trial or preclinical research phases were also reviewed.

5.
JOURNAL OF RARE DISEASES ; (4): 199-205, 2022.
Article in English | WPRIM | ID: wpr-1005002

ABSTRACT

In recent years, antisense oligonucleotide (ASO) has been very active in the field of rare disease research and development, especially in Duchenne muscular dystrophy, where it made a major breakthrough. Duchenne muscular dystrophy (DMD) is a rare childhood myopathy caused by mutations in the dystrophin gene. Currently, the four ASO drugs approved internationally for DMD are all targeted at dystrophin, including eteplirsen, golodirsen, viltolarsen and casimersen. They all belong to phosphorodiamidate morpholino oligomers (PMO) antisense oligonucleotide drugs, so that their pharmacokinetic characteristics are similar. The drugs quickly spread to other tissues after intravenous administration. Because of the electrical neutrality of the PMO, they have a low binding rate to plasma proteins and are quickly metabolized by the kidney and excreted in the urine as archetypes. In addition, the likelihood of drug-drug interactions of ASO is low. Existing clinical studies have shown that they have certain clinical benefits and good tolerability, bringing new options for DMD treatment. This paper mainly discusses the pharmacological effects, pharmacokinetic characteristics, efficacy, and safety of ASO drugs for the treatment of DMD, hoping to provide scientific reference for the rational and safe clinical use of such drugs.

6.
Yonsei Medical Journal ; : 273-283, 2020.
Article in English | WPRIM | ID: wpr-816708

ABSTRACT

The reduction of survival motor neuron (SMN) protein causes spinal muscular atrophy (SMA), an autosomal recessive neuromuscular disease. Nusinersen is an antisense oligonucleotide, approved by the FDA, which specifically binds to the repressor within SMN2 exon 7 to enhance exon 7 inclusion and augment production of functional SMN protein. Nusinersen is the first new oligonucleotide-based drug targeting the central nervous system for the treatment of SMA. This review of nusinersen will discuss its action mechanism, cellular uptake, trafficking mechanisms, and administration approaches to cross the blood-brain barrier. Furthermore, nusinersen clinical trials will be assessed in terms of pharmacokinetics, tolerability and safety, the clinical outcomes of multiple intrathecal doses, and a discussion on the primary and secondary endpoints.

7.
Acta Pharmaceutica Sinica B ; (6): 734-745, 2020.
Article in English | WPRIM | ID: wpr-828846

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) is a transcriptional coactivator that binds to a diverse range of transcription factors. PPAR coactivator 1 (PGC-1) coactivators possess an extensive range of biological effects in different tissues, and play a key part in the regulation of the oxidative metabolism, consequently modulating the production of reactive oxygen species, autophagy, and mitochondrial biogenesis. Owing to these findings, a large body of studies, aiming to establish the role of PGC-1 in the neuromuscular system, has shown that PGC-1 could be a promising target for therapies targeting neuromuscular diseases. Among these, some evidence has shown that various signaling pathways linked to PGC-1 are deregulated in muscular dystrophy, leading to a reduced capacity for mitochondrial oxidative phosphorylation and increased reactive oxygen species (ROS) production. In the light of these results, any intervention aimed at activating PGC-1 could contribute towards ameliorating the progression of muscular dystrophies. PGC-1 is influenced by different patho-physiological/pharmacological stimuli. Natural products have been reported to display modulatory effects on PPAR activation with fewer side effects in comparison to synthetic drugs. Taken together, this review summarizes the current knowledge on Duchenne muscular dystrophy, focusing on the potential effects of natural compounds, acting as regulators of PGC-1.

8.
Acta Pharmaceutica Sinica B ; (6): 1347-1359, 2020.
Article in English | WPRIM | ID: wpr-828803

ABSTRACT

Gene therapy is rapidly emerging as a powerful therapeutic strategy for a wide range of neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Some early clinical trials have failed to achieve satisfactory therapeutic effects. Efforts to enhance effectiveness are now concentrating on three major fields: identification of new vectors, novel therapeutic targets, and reliable of delivery routes for transgenes. These approaches are being assessed closely in preclinical and clinical trials, which may ultimately provide powerful treatments for patients. Here, we discuss advances and challenges of gene therapy for neurodegenerative disorders, highlighting promising technologies, targets, and future prospects.

9.
Acta Pharmaceutica Sinica ; (12): 1251-1256, 2019.
Article in Chinese | WPRIM | ID: wpr-780223

ABSTRACT

Foxo-1 plays an important role in development of muscle atrophy, serving as a potential target for therapeutic treatment of the disease. In this study, the Foxo-1 mRNA was targeted by a Foxo-1 specific RNA oligonucleotide modified by 2'-O-methyl and with a butanol tag at the 3'-end. To understand the in vivo significance of new modified RNA oligos, efficacy, pharmacokinetic and safety profiles of the new modified RNA oligonucleotide targeting Foxo-1 were evaluated in mice. All experimental protocols were approved by the Animal Ethics Committee of Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention. The results showed that different doses of the RNA oligonucleotide can reduce the expression of Foxo-1 in mice by two routes of administration, leading to an increase in skeletal muscle mass of the mice. The results of pharmacokinetic evaluation showed that the plasma disappearance curve for the RNA oligonucleotide could be described by a two-compartmental model. The results of safety evaluation showed that no obvious adverse effects on renal and hepatic functions, nor on hematological parameters by intravenous or oral administration of the RNA oligo with a maximum dose of 30 mg·kg-1. Histopathology also did not reveal any significant changes in the morphology of the organs studied. In conclusion, the new modified RNA oligo is safe and effective in mice, providing experimental evidence supporting the significance for its clinical application.

10.
Arq. neuropsiquiatr ; 76(4): 265-272, Apr. 2018. tab, graf
Article in English | LILACS | ID: biblio-888378

ABSTRACT

ABSTRACT Spinal muscular atrophy (SMA) is a severe and clinically-heterogeneous motor neuron disease caused, in most cases, by a homozygous mutation in the SMN1 gene. Regarding the age of onset and motor involvement, at least four distinct clinical phenotypes have been recognized. This clinical variability is, in part, related to the SMN2 copy number. By now, only supportive therapies have been available. However, promising specific therapies are currently being developed based on different mechanisms to increase the level of SMN protein; in particular, intrathecal antisense oligonucleotides that prevent the skipping of exon 7 during SMN2 transcription, and intravenous SMN1 insertion using viral vector. These therapeutic perspectives open a new era in the natural history of the disease. In this review, we intend to discuss the most recent and promising therapeutic strategies, with special consideration to the pathogenesis of the disease and the mechanisms of action of such therapies.


RESUMO A atrofia muscular espinhal (AME) é uma grave doença dos neurônios motores, de grande variabilidade clínica e causada na maioria dos casos por mutação em homozigose no gene SMN1. Pelo menos quatro fenótipos clínicos distintos são reconhecidos com base na idade de início e no grau de envolvimento motor. Tal variabilidade clínica é em parte relacionada com o número de cópias do gene SMN2. Até recentemente, apenas terapias de suporte estavam disponíveis. Atualmente, terapias especificas estão sendo desenvolvidas com base em diferentes mecanismos para aumentar o nível de proteína SMN; em particular oligonucleotídeos antissenso por via intratecal e inserção de cópia do gene SMN1, via endovenosa, usando vetor viral. Nesta revisão, objetivamos discutir as mais recentes e promissoras estratégias terapêuticas, com consideração especial aos aspectos patogênicos da doença e aos mecanismos de ação de tais terapias.


Subject(s)
Humans , Oligonucleotides/administration & dosage , Muscular Atrophy, Spinal/therapy , Genetic Therapy/methods , DNA, Antisense/administration & dosage , Survival of Motor Neuron 1 Protein/administration & dosage , Phenotype , Injections, Spinal , Mutation
11.
Acta Medicinae Universitatis Scientiae et Technologiae Huazhong ; (6): 571-574, 2017.
Article in Chinese | WPRIM | ID: wpr-666908

ABSTRACT

Objective To explore the effects of miR-21 antisense oligonucleotides(ASO)and cisplatin combined on the growth of subcutaneous xenotransplanted osteosarcomas in nude mice.Methods Xenotransplanted osteosarcoma models were established in nude mice by injection of human osteosarcoma cells MG-63.Inoculated mice were randomly divided into four groups(blank control group,cisplatin group,anti-miR-21 group,anti-miR-21+cisplatin group).The rats were treated by injec-tion of anti-miR-21 into the tumors in anti-miR-21 group,by injection of cisplatin through tail veins in cisplatin group or by in-jection both of them in anti-miR-21+ cisplatin group.The volume and weight of tumors were measured and the inhibition rates of tumors were calculated on a daily basis.Meanwhile,MG-63 cells were divided into the same four groups as mentioned be-fore.The cell proliferation was detected by MTT assay and the cell apoptosis by Annexin V analysis.Results The inhibition of tumor growth(volume and weight)was obvious in the anti-miR-21+cisplatin group compared to the control groups.In vitro cell growth was significantly inhibited and cell apoptosis increased obviously in the anti-miR-21 + cisplatin group compared to the control groups(P <0.05).Conclusion MiR-21 ASO combined with cisplatin can inhibit cell proliferation,promote cell apopto-sis in vitro ,and obviously suppress tumor growth in vivo.They may be used to treat osteosarcoma.

12.
China Oncology ; (12): 167-172, 2015.
Article in Chinese | WPRIM | ID: wpr-465415

ABSTRACT

Background and purpose:As the development of the completion of the human genome project (HGP), the research focus is turning to the gene function research. At present, the domestic experimental research on the apoptosis of K562 cells induced by antisense olignonucleotides is rare. This study was aimed to investigate the effect of human chronic myelogenou leukemia (CML) bcr-abl fusion gene antisense oligonucletides on autophagy and apoptosis of CMLK562 cells in vitro. Methods:By liposome as the carrier, K562 cells were transfected with the bcr-abl gene antisense olignonucleotides. Hoechst staining method was used to observe the apoptosis inducing effect of different concentrations of oligonucleotides, the expressions of LC3-Ⅱ, autophagy-related protein, were determined by the Western blot method, the cell cycles were determined by lfow cytometry (FCM), and JEM-4000EX electron microscope technology was used to detect the apoptosis morphological changes. The apoptosis was detected by DNA agarose gel electrophoresis. Results:Hoechst staining results showed that the bcr-abl gene antisense oligonucletides signiifcantly promoted the apoptosis of K562 cells in a certain concentration dependent manner. Western blot showed that the expression level of LC3-Ⅱwas obviously higher in bcr-abl gene antisense oligonucletides transfected group than the control group, showing a promoting effect on cell autophagy. FCM test results showed that bcr-abl gene antisense oligonucleotides transfected K562 cells showed obvious cell cycle arrest, visible obvious apoptosis morphology under the electron microscope, and DNA Ladder showed obvious apoptosis fragments. Conclusion:The bcr-abl gene antisense olignonucleotides can signiifcantly induce the cell apoptosis of K562. This study provides a new method for CML therapy.

13.
Chinese Pharmacological Bulletin ; (12): 619-622, 2015.
Article in Chinese | WPRIM | ID: wpr-464380

ABSTRACT

Thromboembolic diseases are major health problems worldwide,and remain the leading cause of mortality and disabil-ity at present.Bleeding is the most important complication of an-tithrombotic therapy for thromboembolism,therefore research and development of new antithrombotic drugs with lowered bleeding risk is a significant medical need.The data that elevated plasma levels of FXI are associated with thromboembolic diseases,se-vere FXI deficiency reduced incidence of DVT and ischemic stroke,and FXI deficiency or inhibition in animals shows protec-tive effects against thrombus formation supporting FXI as a novel antithrombotic target with lowered bleeding risk.This paper re-views the progress on FXI as a novel antithrombotic target and the inhibitors target FXI.

14.
China Oncology ; (12): 516-521, 2015.
Article in Chinese | WPRIM | ID: wpr-459820

ABSTRACT

Background and purpose:MiR-224 is overexpressed in hepatocellular carcinoma, and participate in invasion and metastasis of cancer. The aim of this study was to investigate the effects of miR-224 antisense oligonucleotide (ASO) on the proliferation and apoptosis of Hep3B cells.Methods:After transfection with miR-224 ASO, and detecting the miR-224 mRNA expression of Hep3B cells by real-time quantitative PCR; the miR-224 expression in Hep3B cells was measured and cell proliferation was analyzed by MTT assay and the colony formation experimentin vitro andin vivo. The cell apoptosis was analyzed by flow cytometry.Results:Compared with the control group, miR-224 ASO significantly reduced the miR-224 mRNA expression in the Hep3B cell(P<0.05), MTT assay results showed that Hep3B cells survived rate decreased greatly after transfection with miR-224 ASO. Clone formation assay revealed that the colony formation rate in miR-224 ASO group was significantly lower than that in the control group.In vivo study further confirmed that miR-224 ASO could inhibit the proliferation of Hep3B cells,and miR-224 ASO group grew substantially slow compared with the negative control. Flow cytometry indicated that miR-224 ASO group promoted apoptosis significantly.Conclusion:miR-224 was overexpressed in Hep3B cells. Reducing the expression of miR-224 can effectively inhibit the growth of Hep3B cells and promote apoptosis. miR-224 may become a new target for the regulation of gene expression in hepatocellular carcinoma.

15.
Tianjin Medical Journal ; (12): 203-207, 2014.
Article in Chinese | WPRIM | ID: wpr-473479

ABSTRACT

Objective To investigate the expression of miR-30d in breast cancer tissues and demonstrate the regula-tive effects of miR-30d ASO on the invasion and migration of breast cancer cells in vitro. Methods The expression levels of miR-30d in 108 breast cancer tissues and their adjacent tissues were detected by real-time quantitative PCR method. Af-ter transfection with miR-30d ASO, the biological effects of miR-30d on in breast cancer cells was measured by transwell as-say and wound healing assay. The expressions of matrix metalloproteinase (MMP)-2 and MMP-9 were analyzed by Western blot assay. Results The expression level of miR-30d was found to be over-expressed in breast cancer tissues(P<0.05). Compared with control group and nonsense interference group, the miR-30d expression was significantly decreased in breast cancer cells(transfection with miR-30d ASO). Results of transwell and wound healing assay showed that the invasion and mi-gration ability decreased significantly after transfection with miR-30d ASO, and expressions of MMP-2 and MMP-9 were down-regulated (P<0.05). Conclusion miR-30d was over-expressed in human breast cancer. The invasion and migration of breast cancer cells can be effectively inhibited by decreasing the expression of miR-30d. miR-30d may become a new tar-get for the regulation of invasion and migration in breast cancer.

16.
Journal of Leukemia & Lymphoma ; (12): 428-430,435, 2013.
Article in Chinese | WPRIM | ID: wpr-601264

ABSTRACT

Objective To explore the inhibitory effect of anti-miRNA-17 oligonucleotide on leukemic K562 cells.Methods K562 cells were transfected with anti-miRNA-17 oligonucleotide,cell viability was analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetra-zoliunbromide (MTT) assay.Apoptosis was detected by flow cytometry,expression of miRNA-17 in K562 cells was measured by real-time PCR.Results MTT results showed transfection of antisense nucleic acid significantly decreased cell proliferation activity,after 24,48,72 h they were respectively 0.8719±0.001,0.7102±0.002,0.5507±0.001,the difference being statistically significant (P < 0.05) when compared to the random control (t =182.575,269.77,660.4) or control group (t =537.98,571.20,1230.51).FCM test results showed that after 48 h of transfecting antisense nucleic acid apoptosis rate was (20.14 ± 0.01) %,and was statistically significant compared to the randomized control or blank control group (t =2347.6,2568.2,P < 0.01).Fluorescence real-time quantitative PCR confirmed antisense nucleic acid significantly decreased the relative expression level of miR-17 in K562 cells (0.07).The difference was statistically significant compared to the random group or blank group (1,1.01) (t =148.63,147.04,P < 0.05).Conclusion Targeted inhibition of miR-17 with oligonucleotide can suppress K562 cell growth and induce apoptosis.

17.
Chinese Journal of Experimental Ophthalmology ; (12): 749-753, 2013.
Article in Chinese | WPRIM | ID: wpr-636190

ABSTRACT

Background Platelet-derived growth facto(PDGF) affectthe proliferation of human lenepithelial cell(LECs),and human LECexpresPDGF-α recepto(PDGFR-α) throughoutheilifetime.The binding of activated PDGF-α receptowith PDGF promotethe synthesiof DNA.Othestudiedemonstrated thasilencing of PDGFR-α by antisense oligodeoxynucleotide(ASODN) inhibitthe growth of RPE cellin proliferative vitreoretinopathy (PVR),buwhethethitechnique ifeasible foLECiunclear.Objective Thistudy wato investigate the effecof the knockdown of the PDGFR-α on the proliferation of human LECin vitro,and to offean experimental basifothe gene therapy of posteriocapsule opacification.MethodHuman LECstrain SRA01/ 04 wacultured in α-MEM containing fetal bovine serum.The cellwere incubated in 6-well platea5 × 104 cells/ well and transfection of ASODN-containing liposome waperformed.The cellwere divided into the blank control group (with blank liposome),PDGFR-α missense oligodeoxynucleotide(MSODN) group (with PDGFR-α MSODN + liposome),0.5 μmol/L PDGFR-α ASODN group (with 0.5 μmol/L PDGFR-α ASODN+liposome) and 1.0 μmol/L PDGFR-α ASODN group (with 1.0 μ mol/L PDGFR-α ASODN+liposome).The morphology of LECwaexamined undean inverse microscope 24 houraftetransfection.The expression of PDGFR-α mRNin the cellwadetected by reverse transcription-PC(RT-PCR).The rate of proliferation (A490) of the cellwaassayed using Mtand the inhibitory rate of PDGFR-α ASODN on proliferation wameasured.The percentage of LECin G1 phase waanalyzed by flow cytometer.ResultThe LECgrew well and exhibited polygonal shape in the blank control group and PDGFR-α MSODN group 24 houraftetransfection.Buin the 0.5 μmol/L and 1.0 μmol/L PDGFR-α ASODN groups,the cellappeared round in shape and the numberof cellwere obviously decreased.The expression of PDGFR-α mRNdetected by RT-Pcdemonstrated highelevel in the blank control group and PDGFR-α MSODN group;however,the PDGFR-α mRNexpression waobviously lowein the 0.5 μmol/L and 1.0 μmol/L PDGFR-α ASODN groups.The A490 value wa0.661 ± 0.036,0.655 ± 0.016,0.529 ± 0.030 and 0.441 ± 0.039 in the blank control group,PDGFR-α MSODN group,0.5 μmol/L PDGFR-α ASODN group and 1.0 μmol/L PDGFR-α ASODN group,respectively,showing significandecline in the 0.5 μmol/L PDGFR-α ASODN group and 1.0 μ mol/L PDGFR-α ASODN group in comparison with the blank control group (F=34.08,P<0.01).The percentageof LECin G1 phase were (47.73±1.18)%,(49.48±1.09)%,(53.31±1.30)% and (59.98±0.95) % in the blank control group,PDGFR-α MSODN group,0.5 μmol/L PDGFR-α ASODN group and 1.0 μmol/L PDGFR-α ASODN group,showing significandifference among them (F =68.41,P<0.01),and thain the 0.5 μmol/L PDGFR-α ASODN group o1.0 μmol/L PDGFR-α ASODN group showed significantly increase in comparison with the blank control group (P<0.05).ConclusionPDGFR-α silencing could inhibithe proliferation of human LECin vitro.

18.
Academic Journal of Second Military Medical University ; (12): 852-856, 2013.
Article in Chinese | WPRIM | ID: wpr-839438

ABSTRACT

Objective To investigate the effect of cellular Fas-associated death domain-like interleukin-1β-converting enzyme-inhibitory protein (c-FLIP) antisense oligonucleotide (ASODN)-loaded nanoparticles (NP) on the human orbital rhabdomyosarcoma xenograft in nude mice, so as to assess the feasibility of nanoparticles as a gene vector. Methods The model of human orbital rhabdomyosarcoma xenograft was established in nude mice, and the tumors were injected with c-FLIP ASODN NP, c-FLIP ASODN or normal saline (NS). The tumor volume and histopathological changes of tumor were observed. Western blotting analysis and immunohistochemical analysis were used to examine the expression of c-FLIP in tumor tissues of each group. Apoptosis of tumor cells was detected using TUNEL method. Results The growth of human orbital rhabdomyosarcoma in nude mice was significantly inhibited in ASODN NP group compared with the other two groups. Western blotting analysis showed that c-FLIP protein expression in ASODN NP and ASODN groups was significantly decreased compared with NS group (P<0. 05). Immunohistochemical study showed that c-FLIP expressionwas found in the endochylema, and the c-FLIP positive cells in ASODN NP group was significantly less than those in the other two groups (P<0.05). Tumor cell apoptosis was observed in both ASODN NP and ASODN groups, with more found in the former, and only a few apoptotic cells were found in the NS group. Conclusion c-FLIP ASODN NP can effectively inhibit the growth of human orbital rhabdomyosarcoma xenograft in nude mice, indicating that nanoparticles may serve as a safe and effective vector for ASODN.

19.
China Oncology ; (12): 285-291, 2013.
Article in Chinese | WPRIM | ID: wpr-433490

ABSTRACT

10.3969/j.issn.1007-3969.2013.04.008

20.
China Oncology ; (12): 432-438, 2013.
Article in Chinese | WPRIM | ID: wpr-435565

ABSTRACT

Background and purpose: The miR-224 in a variety of malignant tumors is overexpression, however, its expression and function in colon cancer are not clear. The aim of this study was to investigate the expression of miR-301 in colon cancer tissues and demonstrate the regulative effects of miR-301 ASO on the proliferation and apoptosis of colon cancer cell in vitro and in vitro. Methods:The expression of miR-301 in 120 colon cancer tissues and their adjacent tissues was detected by real-time quantitative PCR method. After transfection with miR-301ASO, the biological effects of miR-301 in SW620 cells were measured by MTT assay, the colony formation experiment, flow cytometry and the in vivo experiment. Results: The expression level of miR-301 was found to be overexpressed in 63.33% (76/120) of the colon cancer cases (P<0.05). miR-301 expression in SW620 cells (transfection with miR-301 ASO, 0.09±0.01) was significantly less than control group (0.50±0.07, P=0.00). MTT assay results showed that SW620 cells survived rate at 24, 48 and 96 h decreased greatly after transfection with miR-301ASO (P=0.00). Clone formation assay revealed that miR-301 ASO group colony formation rate (5.33%±0.74%) was significantly lower than the control group (33.33%±8.38%, P=0.00). In vivo study further confirmed that miR-301ASO could inhibit the proliferation of SW620 cells (P<0.05), and miR-301ASO group grew substantially slow compared with the negative control group (P=0.00). Flow cytometry indicated that the apoptotic index in miR-301 ASO group (15.68±1.46) was significantly higher than the control group (3.36±0.88, P=0.02). In addition, the Bcl2 mRNA and protein were significantly decreased after reduce the expression of miR-301 (P=0.00, P=0.00). Conclusion:MiR-301 was overexpressed in human colon cancer. Reduce the expression of miR-301 can effectively inhibit the growth of colon cancer cells and promote apoptosis. MiR-301 may become a new target for the regulation of gene expression in colon cancer.

SELECTION OF CITATIONS
SEARCH DETAIL