151
|
Chen LX, Lin L, Wang HJ, Wei XL, Fu X, Zhang JY, Yu CL. Suppression of early experimental osteoarthritis by in vivo delivery of the adenoviral vector-mediated NF-kappaBp65-specific siRNA. Osteoarthritis Cartilage 2008; 16:174-84. [PMID: 17686636 DOI: 10.1016/j.joca.2007.06.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Accepted: 06/05/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study was to use adenoviral vector-mediated nuclear factor-kappaBp65 (NF-kappaBp65)-specific siRNA (Ad-siRNA(NF-kappaBp65)) to suppress the progression of early osteoarthritis (OA) in rat model, and therefore to explore a new gene therapy for OA. METHODS Reverse transcription polymerase chain reaction was performed to confirm the silencing effect of Ad-siRNA(NF-kappaBp65) in cultured rat chondrocytes. Transection of the medial collateral ligament plus partial medial meniscectomy was operated in the knee of rats to establish OA model. Histological analysis was made to assess the morphological change of cartilage and synovium, and enzyme-linked immunosorbent assay was made to measure the expression of cytokines, such as interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha), in synovial fluid. The silencing effect of Ad-siRNA(NF-kappaBp65) on NF-kappaBp65 in cartilage and synovium of knee was measured with Western blot and the activation of NF-kappaB was measured with electrophoretic mobility shift assays. RESULTS Ad-siRNA(NF-kappaBp65) can inhibit the activation of NF-kappaB and the expression of NF-kappaBp65 in cartilage and synovium of the knee, restrain the induction of IL-1beta and TNF-alpha in synovial fluid, alleviate the inflammation of synovium and reduce the degradation of cartilage in early phase of experimental OA. CONCLUSIONS Ad-siRNA(NF-kappaBp65) can suppress the progression of the early experimental OA which suggests that Ad-siRNA(NF-kappaBp65) has potential to be a useful preventive and therapeutic agent for OA.
Collapse
Affiliation(s)
- L X Chen
- Institute of Sports Medicine, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing 100083, PR China
| | | | | | | | | | | | | |
Collapse
|
152
|
Dieckhoff B, Petersen B, Kues WA, Kurth R, Niemann H, Denner J. Knockdown of porcine endogenous retrovirus (PERV) expression by PERV-specific shRNA in transgenic pigs. Xenotransplantation 2008; 15:36-45. [DOI: 10.1111/j.1399-3089.2008.00442.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
153
|
Competition potency of siRNA is specified by the 5′-half sequence of the guide strand. Biochem Biophys Res Commun 2008; 367:78-83. [DOI: 10.1016/j.bbrc.2007.12.099] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 12/11/2007] [Indexed: 12/24/2022]
|
154
|
ATHANASIADOU S, HUNTLEY JF. Emerging technologies and their applications in interactions between nutrition and immunity to gastrointestinal parasites in sheep. Parasite Immunol 2008; 30:101-11. [DOI: 10.1111/j.1365-3024.2008.00993.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
155
|
Sano M, Li H, Nakanishi M, Rossi JJ. Expression of Long Anti-HIV-1 Hairpin RNAs for the Generation of Multiple siRNAs: Advantages and Limitations. Mol Ther 2008; 16:170-7. [PMID: 17726454 DOI: 10.1038/sj.mt.6300298] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Promoter expressed long-hairpin RNAs (lhRNAs) that can be processed into multiple small interfering RNA (siRNAs) are being considered as effective agents for treating rapidly mutating viruses such as human immunodeficiency virus (HIV). In the present study, we have generated human U6 promoter-driven lhRNAs of 50, 53, and 80 base pairs (bp) targeting contiguous sequences within the tat and rev genes of HIV-1 and evaluated the efficacy of these lhRNAs as well as their processing in cells. By using multiple G:U mismatches in the stems, we have been able to readily incorporate the long-hairpin structures into a lentiviral vector transduction system. Here we show that such long hairpins can be stably and functionally expressed for a long term in HIV-1 susceptible T cells, where they provide potent inhibition of HIV replication against both non-mutant and mutant variants of HIV-1. Our studies provide strong support for the use of the G:U wobble pair containing lhRNAs to generate multiple siRNAs from a single transcript, but we also show that lhRNAs of 80 bp may be the upper size limit for effectively producing multiple, functional siRNAs.
Collapse
Affiliation(s)
- Masayuki Sano
- Division of Molecular Biology, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | | | | | | |
Collapse
|
156
|
Kavi HH, Fernandez H, Xie W, Birchler JA. Genetics and biochemistry of RNAi in Drosophila. Curr Top Microbiol Immunol 2008; 320:37-75. [PMID: 18268839 DOI: 10.1007/978-3-540-75157-1_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RNA interference (RNAi) is the technique employing double-stranded RNA to target the destruction of homologous messenger RNAs. It has gained wide usage in genetics. While having the potential for many practical applications, it is a reflection of a much broader spectrum of small RNA-mediated processes in the cell. The RNAi machinery was originally perceived as a defense mechanism against viruses and transposons. While this is certainly true, small RNAs have now been implicated in many other aspects of cell biology. Here we review the current knowledge of the biochemistry of RNAi in Drosophila and the involvement of small RNAs in RNAi, transposon silencing, virus defense, transgene silencing, pairing-sensitive silencing, telomere function, chromatin insulator activity, nucleolar stability, and heterochromatin formation. The discovery of the role of RNA molecules in the degradation of mRNA transcripts leading to decreased gene expression resulted in a paradigm shift in the field of molecular biology. Transgene silencing was first discovered in plant cells (Matzke et al. 1989; van der Krol et al. 1990; Napoli et al. 1990) and can occur on both the transcriptional and posttranscriptional levels, but both involve short RNA moieties in their mechanism. RNA interference (RNAi) is a type of gene silencing mechanism in which a double-stranded RNA (dsRNA) molecule directs the specific degradation of the corresponding mRNA (target RNA). The technique of RNAi was first discovered in Caenorhabditis elegans in 1994 (Guo and Kemphues 1994). Later the active component was found to be a dsRNA (Fire et al. 1998). In subsequent years, it has been found to occur in diverse eukaryotes
Collapse
Affiliation(s)
- Harsh H Kavi
- Division of Biological Sciences, University of Missouri, Tucker Hall, Columbia, MO 65211, USA
| | | | | | | |
Collapse
|
157
|
Abstract
During the past five years many patients suffering from congenital myasthenic syndromes (CMS) have been identified worldwide and novel causative genes and mutations have been discovered. The disease genes now include those encoding each subunit of the acetylcholine receptor (AChR), the ColQ part of acetylcholinesterase (AChE), choline acetyltransferase, Na(v)1.4, MuSK, and Dok-7. Moreover, emerging genotype-phenotype correlations are providing clues for targeted mutation analysis. This review focuses on the recent observations in selected CMS.
Collapse
Affiliation(s)
- Andrew G Engel
- Department of Neurology and Muscle Research Laboratory, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
158
|
|
159
|
Chen Y, Cheng G, Mahato RI. RNAi for treating hepatitis B viral infection. Pharm Res 2007; 25:72-86. [PMID: 18074201 PMCID: PMC2217617 DOI: 10.1007/s11095-007-9504-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 11/14/2007] [Indexed: 12/18/2022]
Abstract
Chronic hepatitis B virus (HBV) infection is one of the leading causes of liver cirrhosis and hepatocellular carcinoma (HCC). Current treatment strategies of HBV infection including the use of interferon (IFN)-α and nucleotide analogues such as lamivudine and adefovir have met with only partial success. Therefore, it is necessary to develop more effective antiviral therapies that can clear HBV infection with fewer side effects. RNA interference (RNAi), by which a small interfering RNA (siRNA) induces the gene silence at a post-transcriptional level, has the potential of treating HBV infection. The successful use of chemically synthesized siRNA, endogenous expression of small hairpin RNA (shRNA) or microRNA (miRNA) to silence the target gene make this technology towards a potentially rational therapeutics for HBV infection. However, several challenges including poor siRNA stability, inefficient cellular uptake, widespread biodistribution and non-specific effects need to be overcome. In this review, we discuss several strategies for improving the anti-HBV therapeutic efficacy of siRNAs, while avoiding their off-target effects and immunostimulation. There is an in-depth discussion on the (1) mechanisms of RNAi, (2) methods for siRNA/shRNA production, (3) barriers to RNAi-based therapies, and (4) delivery strategies of siRNA for treating HBV infection.
Collapse
Affiliation(s)
- Yong Chen
- Huai-An 4th People’s Hospital, Jiangsu, China
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 19 S Manassas Street, Memphis, Tennessee 38103 USA
| | - Guofeng Cheng
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 19 S Manassas Street, Memphis, Tennessee 38103 USA
| | - Ram I. Mahato
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 19 S Manassas Street, Memphis, Tennessee 38103 USA
| |
Collapse
|
160
|
Gantier MP, Baugh JA, Donnelly SC. Nuclear transcription of long hairpin RNA triggers innate immune responses. J Interferon Cytokine Res 2007; 27:789-97. [PMID: 17892400 DOI: 10.1089/jir.2006.0152] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RNA interference (RNAi) is one of the most promising tools for deciphering the human genome and has great therapeutic potential. However, its high target specificity limits its efficiency for therapeutic protection from viruses with high rates of genetic mutation. This limitation may be overcome by the expression of long hairpin RNAs (lhRNAs). Indeed, lhRNAs have been shown recently to have increased efficacy over short interfering RNAs (siRNAs) as protective antiviral agents. Here, we investigate the expression of lhRNAs and demonstrate unintended effects. We show that overexpressed lhRNAs are exported to the cytoplasm. As a consequence, we detect activation of innate immune signaling pathways by lhRNAs. With growing concerns about the complexity of cytoplasmic detection of dsRNAs by the innate immune machinery, this work highlights the need for closer scrutiny when using lhRNAs as potential antiviral agents.
Collapse
Affiliation(s)
- Michael P Gantier
- School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, and St. Vincent's University Hospital, Dublin, Ireland
| | | | | |
Collapse
|
161
|
Cahill AL, Moore JM, Sabar FI, Harkins AB. Variability in RNA interference in neuroendocrine PC12 cell lines stably transfected with an shRNA plasmid. J Neurosci Methods 2007; 166:236-40. [PMID: 17767962 DOI: 10.1016/j.jneumeth.2007.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Accepted: 07/18/2007] [Indexed: 10/23/2022]
Abstract
RNA interference (RNAi) has quickly become a very powerful technique for specifically suppressing or knocking down the expression of any desired gene. Many fields of research, including neuroscience, have benefitted from RNAi methods. It has been well documented that different small interfering RNAs (siRNAs) and small hairpin RNAs (shRNAs) vary greatly in terms of their effectiveness, and much attention has been focused on guidelines and algorithms for the selection of effective siRNAs. However, it has not been widely appreciated that a single shRNA-expressing plasmid can also produce widely varying levels of knockdown in different stably transfected cell lines derived from the same transfection. Here we report that knockdown of three distinct target proteins varies from minimal to almost complete in independent, stably transfected PC12 cell lines. This variability in knockdown among cell lines emphasizes the importance of characterizing a number of cell lines when attempting to establish stable knockdown cell lines, but also offers the possibility of studying the effects of graded levels of protein expression.
Collapse
Affiliation(s)
- Anne L Cahill
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
162
|
Loss of the SdhB, but Not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Mol Cell Biol 2007; 28:718-31. [PMID: 17967865 DOI: 10.1128/mcb.01338-07] [Citation(s) in RCA: 334] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mitochondrial complex II is a tumor suppressor comprised of four subunits (SdhA, SdhB, SdhC, and SdhD). Mutations in any of these should disrupt complex II enzymatic activity, yet defects in SdhA produce bioenergetic deficiency while defects in SdhB, SdhC, or SdhD induce tumor formation. The mechanisms underlying these differences are not known. We show that the inhibition of distal subunits of complex II, either pharmacologically or via RNA interference of SdhB, increases normoxic reactive oxygen species (ROS) production, increases hypoxia-inducible factor alpha (HIF-alpha) stabilization in an ROS-dependent manner, and increases growth rates in vitro and in vivo without affecting hypoxia-mediated activation of HIF-alpha. Proximal pharmacologic inhibition or RNA interference of complex II at SdhA, however, does not increase normoxic ROS production or HIF-alpha stabilization and results in decreased growth rates in vitro and in vivo. Furthermore, the enhanced growth rates resulting from SdhB suppression are inhibited by the suppression of HIF-1alpha and/or HIF-2alpha, indicating that the mechanism of SdhB-induced tumor formation relies upon ROS production and subsequent HIF-alpha activation. Therefore, differences in ROS production, HIF proliferation, and cell proliferation contribute to the differences in tumor phenotype in cells lacking SdhB as opposed to those lacking SdhA.
Collapse
|
163
|
Abstract
RNA interference (RNAi) is an evolutionally conserved gene silencing mechanism present in a variety of eukaryotic species. RNAi uses short double-stranded RNA (dsRNA) to trigger degradation or translation repression of homologous RNA targets in a sequence-specific manner. This system can be induced effectively in vitro and in vivo by direct application of small interfering RNAs (siRNAs), or by expression of short hairpin RNA (shRNA) with non-viral and viral vectors. To date, RNAi has been extensively used as a novel and effective tool for functional genomic studies, and has displayed great potential in treating human diseases, including human genetic and acquired disorders such as cancer and viral infections. In the present review, we focus on the recent development in the use of RNAi in the prevention and treatment of viral infections. The mechanisms, strategies, hurdles and prospects of employing RNAi in the pharmaceutical industry are also discussed.
Collapse
Affiliation(s)
- Yan Ma
- Stanley Ho Centre for Emerging Infectious Diseases, and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | | | | |
Collapse
|
164
|
Sun P, Gao J, Liu YL, Wei LW, Wu LP, Liu ZY. RNA interference (RNAi)-mediated vascular endothelial growth factor-C (VEGF-C) reduction interferes with lymphangiogenesis and enhances epirubicin sensitivity of breast cancer cells. Mol Cell Biochem 2007; 308:161-8. [PMID: 17938864 DOI: 10.1007/s11010-007-9624-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Accepted: 10/03/2007] [Indexed: 01/02/2023]
Abstract
It has been reported that over-expression of vascular endothelial growth factor-C (VEGF-C) in tumors leads to increased lymphangiogenesis and resistance to chemotherapy. Therefore, we hypothesized that VEGF-C would be a good molecular target for cancer gene therapy. In this study, we silenced the expression of VEGF-C with the highly specific post-transcriptional suppression of RNA interference (RNAi) in human breast cancer MCF-7 cell line. The expression of VEGF-C was examined by reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA), and the effect of plasmid on human lymphatic endothelial cells (HLECs) in vitro was analyzed by migration and 3-(4, 5-dimethylt-hiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. The sensitivity to anticancer agents was evaluated by MTT and apoptosis assay, and apoptosis-related genes bcl-2/bax ratio was determined by Western Blotting. Results showed that of three siRNA-expressing vectors, P-1/siRNA most significantly suppressed the expression of VEGF-C mRNA and protein (38.1% of control and 117.8 +/- 24.2 pg/ml, respectively) and interfered with proliferation and migration of HLECs in vitro. Moreover, transfection of VEGF-C/siRNA combined with Epirubicin markedly decreased breast cancer cells viability, reaching up to 38.5%, and increased apoptosis rate from 13.1% to 38.9%, as determined by decrease of bcl-2/bax ratio. In summary, VEGF-C would be a good molecular target, and a combination of Epirubicin and RNAi targeting VEGF-C could be an effective means for suppressing lymphatic metastasis and enhancing chemosensitivity of human breast cancer cells.
Collapse
Affiliation(s)
- Ping Sun
- Department of Anatomy, School of Medicine, Shandong University, No. 44, WenHua Xi Road, JiNan, Shandong 250012, PR China.
| | | | | | | | | | | |
Collapse
|
165
|
Vickers TA, Lima WF, Nichols JG, Crooke ST. Reduced levels of Ago2 expression result in increased siRNA competition in mammalian cells. Nucleic Acids Res 2007; 35:6598-610. [PMID: 17905815 PMCID: PMC2095815 DOI: 10.1093/nar/gkm663] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Administration of small interfering RNAs (siRNAs) leads to degradation of specific mRNAs utilizing the cellular RNA interference (RNAi) machinery. It has been demonstrated that co-administration of siRNAs may lead to attenuation of activity of one of the siRNAs. Utilizing antisense and siRNA-mediated RNA-induced silencing complex (RISC) gene reduction we show that siRNA competition is correlated with differences in the cellular expression levels of Ago2, while levels of other RISC proteins have no effect on competition. We also show that under certain conditions siRNA competition rather than reduction of cellular RISC levels may be responsible for apparent reduction in siRNA activity. Furthermore, exploiting siRNA competition, we show that the RISC pathway loads and results in detectable cleavage of the target RNA in ∼2 h after transfection. The RISC pathway is also capable of being reloaded even in the absence of new protein synthesis. RISC reloading and subsequent induction of detectable cleavage of a new target RNA, requires about 9–12 h following the initial transfection.
Collapse
Affiliation(s)
- Timothy A Vickers
- Isis Pharmaceuticals. 1896 Rutherford Road, Carlsbad, CA 92008, USA.
| | | | | | | |
Collapse
|
166
|
Wolvetang EJ, Pera MF, Zuckerman KS. Gap junction mediated transport of shRNA between human embryonic stem cells. Biochem Biophys Res Commun 2007; 363:610-5. [PMID: 17900528 DOI: 10.1016/j.bbrc.2007.09.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 09/06/2007] [Indexed: 11/30/2022]
Abstract
Gap junction intracellular communication (GJIC) allows the direct transport of small molecules between adjacent cells. We hypothesized that siRNAs in one hESC could inhibit target RNA expression in another hESC via GJIC. We co-cultured green fluorescent protein (GFP)-expressing ENVY hESC with non-GFP-expressing hESC, which had been transduced to stably express shRNA directed against GFP. We discovered that the GFP shRNA expressing hESC inhibited GFP expression in the adjacent GFP-expressing hESC in a dose-dependent manner. This downregulation of GFP expression in ENVY cells was not observed when the co-cultured cells had been transduced with a non-functional GFP shRNA that was mutated at two nucleotides or when the cells were incubated with the GJIC inhibitor, alpha-glycyrrhetinic acid (alpha-GA). We conclude that 21-23 bp double-stranded shRNA/siRNA oligonucleotides are able to move through gap junctions between hESCs and thus can affect gene expression in neighbouring hESC. This novel intercellular gene expression regulatory mechanism may offer new approaches to manipulation of hESC.
Collapse
Affiliation(s)
- Ernst J Wolvetang
- The Australian Stem Cell Centre and Department of Anatomy and Cell Biology, Monash University, Building 75 STRIP, Wellington Road, Clayton, Vic. 3800, Australia.
| | | | | |
Collapse
|
167
|
Han Y, Qin W, Huang G. Knockdown of RCAS1 expression by RNA interference recovers T cell growth and proliferation. Cancer Lett 2007; 257:182-90. [PMID: 17825484 DOI: 10.1016/j.canlet.2007.07.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2007] [Revised: 05/28/2007] [Accepted: 07/16/2007] [Indexed: 10/22/2022]
Abstract
Receptor binding cancer antigen expressed on SiSo cells (RCAS1), a tumor-associated antigen, was expressed in various malignant tissues. It is involved in the tumor immune escape. Here, we reported the evidence that knockdown of RCAS1 expression by RNA interference can recovers T cell growth and proliferation. We designed a small hairpin RNA to knockdown RCAS1 expression in MCF-7 cells effectively. Adding RCAS1 protein resulted in a reduced T cell growth rate, an increased T cell apoptosis ratio, the higher activity of Caspase-3 proteases, and decreased IFN-gamma secretion. The suppression of RCAS1 expression effectively recover T cell proliferation, reduce apoptosis and partially reverse the T cell function of IFN-gamma secretion.
Collapse
Affiliation(s)
- Yuan Han
- Department of Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 145 Middle Shandong Road, Shanghai 200001, People's Republic of China
| | | | | |
Collapse
|
168
|
Wang Q, Peng Z, Xiao S, Geng S, Yuan J, Li Z. RNAi-mediated inhibition of COL1A1 and COL3A1 in human skin fibroblasts. Exp Dermatol 2007; 16:611-7. [PMID: 17576241 DOI: 10.1111/j.1600-0625.2007.00574.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Types I and III collagens are the major collagens comprising skin connective tissue. Defects in these collagens lead to diseases of dermal connective tissue and fibre hyperplasia. RNA interference (RNAi) provides a powerful tool to inhibit specific gene expression. In this study, we generated small interfering RNAs (siRNA) expression cassettes (SECs) by polymerase chain reaction (PCR) as a method to quickly screen the efficacy of siRNAs. We then cloned the most efficient SECs into vectors, using a rapid and novel method intrinsic to the design of the SEC, and transfected human skin fibroblasts (HSF) to generate stable lines. We show that the transfection of SECs into HSFs resulted in specific and effective repression of COL1A1 and COL3A1 expression (5.00% and 6.48% of control levels) provided a rapid method for testing candidate siRNA sequences. We report the use of vector-based RNAi to establish stable HSF cell lines with persistent knockdown over at least 30 days (25.21% and 22.12% of control levels). These stably modified HSF cell lines may be used for the study of other types of collagen or proteins of the extracellular matrix (ECM).
Collapse
Affiliation(s)
- Qiong Wang
- Department of Dermatology, Second Hospital of Xi'an Jiaotong University, Shaanxi, China
| | | | | | | | | | | |
Collapse
|
169
|
Pulukuri SMK, Rao JS. Small interfering RNA directed reversal of urokinase plasminogen activator demethylation inhibits prostate tumor growth and metastasis. Cancer Res 2007; 67:6637-46. [PMID: 17638874 PMCID: PMC2390768 DOI: 10.1158/0008-5472.can-07-0751] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent studies have shown that small interfering RNA (siRNA) silences genes at the transcriptional level in human cells. However, the therapeutic potential of siRNA-mediated transcriptional gene silencing remains unclear. Here, we show that siRNA targeted to the urokinase plasminogen activator (uPA) promoter induced epigenetic transcriptional silencing in human prostate cancer cells. This silencing resulted in a dramatic reduction of tumor cell invasion and angiogenesis in vitro. Furthermore, the results from a bioluminescence tumor/metastasis model showed that the silencing of uPA significantly inhibits prostate tumor growth and the incidence of lung metastasis. Our findings represent a potentially powerful new approach to not only epigenetic silencing of metastasis or growth-promoting genes as a cancer therapy, but also as a means to shed light on how aberrant de novo methylation during cancer progression might be targeted to specific sequences.
Collapse
Affiliation(s)
- Sai Murali Krishna Pulukuri
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL 61605, USA
| | | |
Collapse
|
170
|
Abstract
MicroRNAs (miRNAs) are emerging as potent regulators of many biological processes, including cellular differentiation and disease. Recently, miRNA has been directly involved in innate immunity and transduction signalling by Toll-like receptors and the ensuing cytokine response. In this review, we present an overview of what is currently known of the involvement of miRNA and RNA interference components in the fine-tuning of innate immune responses.
Collapse
Affiliation(s)
- Michael P Gantier
- Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
171
|
Qiu Z, Huang C, Sun J, Qiu W, Zhang J, Li H, Jiang T, Huang K, Cao J. RNA interference-mediated signal transducers and activators of transcription 3 gene silencing inhibits invasion and metastasis of human pancreatic cancer cells. Cancer Sci 2007; 98:1099-106. [PMID: 17459060 PMCID: PMC11158338 DOI: 10.1111/j.1349-7006.2007.00485.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Signal transducers and activators of transcription-3 (STAT3), a central cytoplasmic transcription factor, is frequently overexpressed and constitutively activated by tyrosine during malignant transformation. The overexpression and phosphorylation of STAT3 in pancreatic cancer has been described only recently, but the roles and mechanism still remain unclear. In this study, we elucidate the significance of the STAT3 signaling pathway in metastatic potentials of pancreatic cancer. We stably silence the expression of the STAT3 and p-STAT3 by using RNA interference (RNAi) in the pancreatic cancer cell line SW1990, and then reduce its invasion capacity in vitro and metastasis capacity in vivo compared to parental cells or cells tansfected with a control vector. Furthermore, silencing SW1990 cells with the STAT3 gene by RNAi also led to a decrease of matrix metalloproteinases-2 (MMP-2) and vascular endothelial growth factor (VEGF) at the mRNA and protein level. Collectively, these studies suggest that activation of the STAT3 signaling pathway plays an important role in the progression of pancreatic cancer, and that silence of the STAT3 gene with RNAi may be a useful anti-invasive therapeutic option in pancreatic cancer.
Collapse
Affiliation(s)
- Zhengjun Qiu
- Department of General Surgery, Affiliated First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Réjiba S, Wack S, Aprahamian M, Hajri A. K-ras oncogene silencing strategy reduces tumor growth and enhances gemcitabine chemotherapy efficacy for pancreatic cancer treatment. Cancer Sci 2007; 98:1128-36. [PMID: 17489984 PMCID: PMC11159785 DOI: 10.1111/j.1349-7006.2007.00506.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pancreatic adenocarcinoma remains a fatal disease characterized by rapid tumor progression, high metastatic potential and profound chemoresistance. Gemcitabine is the current standard chemotherapy for advanced pancreatic cancer, but it is still far from optimal and novel therapeutic strategies are needed urgently. Mutations in the k-ras gene have been found in more than 90% of pancreatic cancers and are believed to play a key role in this malignancy. Thus, the goal of this study was to investigate the impact of k-ras oncogene silencing on pancreatic tumor growth. Additionally, we examined whether combining k-ras small interfering RNA (siRNA) with gemcitabine has therapeutic potential for pancreatic cancer. The treatment of tumor cell cultures with the corresponding k-ras siRNA resulted in a significant inhibition of k-ras endogenous expression and cell proliferation. In vivo, tumor xenografts were significantly reduced with k-ras siRNA(GAT) delivered by electroporation. Moreover, combined treatment with pSsik-ras(GAT) plus gemcitabine resulted in strong growth inhibition of orthotopic pancreatic tumors. Survival rate was significantly prolonged and the mean tumor volume was dramatically reduced in mice receiving the combined treatment compared with single agents. Collectively, these findings show that targeting mutant k-ras through specific siRNA might be effective for k-ras oncogene silencing and tumor growth inhibition. The improvement of gemcitabine-based chemotherapy suggests that this strategy might be used therapeutically against human pancreatic cancer to potentiate the effects of conventional therapy.
Collapse
Affiliation(s)
- Soukaina Réjiba
- Institut de Recherche sur les Cancers de l'Appareil Digestif (IRCAD), INSERM Unit 701, Tumor Biology and Gene Therapy Department, Strasbourg 67091, France
| | | | | | | |
Collapse
|
173
|
Allen D, Kenna PF, Palfi A, McMahon HP, Millington-Ward S, O'Reilly M, Humphries P, Farrar GJ. Development of strategies for conditional RNA interference. J Gene Med 2007; 9:287-98. [PMID: 17397092 DOI: 10.1002/jgm.1018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND RNA interference (RNAi) represents a powerful tool with which to undertake sequence-dependent suppression of gene expression. Synthesized double-stranded RNA (dsRNA) or dsRNA generated endogenously from plasmid or viral vectors can be used for RNAi. For the latter, polymerase III promoters which drive ubiquitous expression in all tissues have typically been adopted. Given that dsRNA molecules must contain few 5' and 3' over-hanging bases to maintain potency, employing polymerase II promoters to drive tissue-specific expression of RNAi may be problematic due to potential inclusion of nucleotides 5' and 3' of siRNA sequences. METHODS To circumvent this, polymerase II promoters in combination with cis-acting hammerhead ribozymes and short-hairpin RNA sequences have been explored as a means to generate potent dsRNA molecules in tissues defined by the promoter in use. RESULTS The novel constructs evaluated in this study produced functional siRNA which suppressed the enhanced green fluorescent protein (eGFP) both in vitro and in vivo (in mice). Additionally, the constructs did not appear to elicit a significant type-1 interferon response compared to traditional H1-transcribed shRNA. CONCLUSIONS Given the potential 'off-target' effects of dsRNAs, it would be preferable in many cases to limit expression of dsRNA to the tissue of interest and moreover would significantly augment the resolution of RNAi technologies. Notably, the system under evaluation in this study could readily be adapted to achieve this objective.
Collapse
Affiliation(s)
- Danny Allen
- Department of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Schellander K, Hoelker M, Tesfaye D. Selective degradation of transcripts in mammalian oocytes and embryos. Theriogenology 2007; 68 Suppl 1:S107-15. [PMID: 17573104 DOI: 10.1016/j.theriogenology.2007.05.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During the last decade several gene expression analysis studies have been carried out to investigate the transcriptional profile of bovine embryos in response to various culture and treatments conditions. Despite this fact, the function of a large number of genes in mammalian embryogenesis has not yet been investigated or is not known. The conventional gene-knockout experiments have been used extensively to study the function of genes in mammalian embryogenesis. However, these studies are relatively slow and cannot keep pace with the rapid accumulation of new sequence information produced by various genome projects. For this, the posttranscriptional gene silencing (PTGS) by double-stranded RNA (dsRNA), or RNA interference (RNAi), has emerged as a new tool for studying gene function in an increasing number of organisms. The present review will focus on recent developments in the use of RNAi for selective degradation of transcripts in mammalian embryos to elucidate their function in early development.
Collapse
Affiliation(s)
- K Schellander
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Endenicher Allee 15, 53115 Bonn, Germany.
| | | | | |
Collapse
|
175
|
Nakaya N, Hemish J, Krasnov P, Kim SY, Stasiv Y, Michurina T, Herman D, Davidoff MS, Middendorff R, Enikolopov G. noxin, a novel stress-induced gene involved in cell cycle and apoptosis. Mol Cell Biol 2007; 27:5430-44. [PMID: 17515607 PMCID: PMC1952090 DOI: 10.1128/mcb.00551-06] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We describe a novel stress-induced gene, noxin, and a knockout mouse line with an inactivated noxin gene. The noxin gene does not have sequelogs in the genome and encodes a highly serine-rich protein with predicted phosphorylation sites for ATM, Akt, and DNA-dependent protein kinase kinases; nuclear localization signals; and a Zn finger domain. noxin mRNA and protein levels are under tight control by the cell cycle. noxin, identified as a nitric oxide-inducible gene, is strongly induced by a wide range of stress signals: gamma- and UV irradiation, hydrogen peroxide, adriamycin, and cytokines. This induction is dependent on p53. Noxin accumulates in the nucleus in response to stress and, when ectopically expressed, Noxin arrests the cell cycle at G1; although it also induces p53, the cell cycle arrest function of Noxin is independent of p53 activity. noxin knockout mice are viable and fertile; however, they have an enlarged heart, several altered hematopoietic parameters, and a decreased number of spermatids. Importantly, loss or downregulation of Noxin leads to increased cell death. Our results suggest that Noxin may be a component of the cell defense system: it is activated by various stress stimuli, helps cells to withdraw from cycling, and opposes apoptosis.
Collapse
Affiliation(s)
- Naoki Nakaya
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Xu D, Lin JS, Ren JH, Chen Q, Yao JJ, He XX. Effect of nephroblastoma overexpressed gene on biological behaviors of rat hepatic stellate cells. Shijie Huaren Xiaohua Zazhi 2007; 15:1602-1608. [DOI: 10.11569/wcjd.v15.i14.1602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct recombinant plasmids that can express small interfering RNA (siRNA) targeting nephroblastoma overexpressed gene (NOV) and investigate its effect on the biological behaviors of hepatic stellate cells (HSCs).
METHODS: Hairpin siRNA templates targeting NOV gene were synthesized and cloned into plasmid vector psiRNA-H1neo. Three vector-derived siRNAs (denoted psiRNA1, 2 and 3) and one mocking pconsiRNA (as control) were constructed. The recombinant NOV siRNA plasmids were constructed and identified using restrictive enzyme analysis and DNA sequencing, and then transfected into HSCs by lipofectamine. HSCs were divided into group psiRNA1, psiRNA2, psiRNA3 and pconsiRNA, transfected with the corresponding recombinant plasmids. Blank group consisted of HSCs contaning no plasmids. The expression of NOV and α-smooth muscle actin (α-SMA) were detected by semi-quantitative reverser transcription-polymerase chain reaction (RT-PCR) and Western blot. The mRNA expression of type Ⅰ collagen and type Ⅲ collagen were detected by semi-quantitative RT-PCR. Cell proliferation was assayed by MTT method and cell apoptosis by flow cytometry.
RESULTS: Restrictive enzyme analysis and DNA sequencing revealed the successful construction of siRNA expression plasmids. Compared with negative control group, extrogenous recombinant plasmid psiRNA2 reduced the mRNA levels of NOV (decreased rate: 73.0% vs 23.2%, P < 0.05), type Ⅰ collagen (decreased rate: 59.8% vs 17.0%, P < 0.05), type Ⅲ collagen (decreased rate: 37.1% vs 6.6%, P < 0.05), and inhibit the expression of α-SMA at mRNA level (decreased rate: 51.4% vs 15.1%, P < 0.05) and protein level. Compared with non-transfection group, extrogenous recombinant plasmid psiRNA2 significantly decreased the proliferating activity of HSC (24 h: 0.172 ± 0.005 vs 0.318 ± 0.018, P < 0.05; 48 h: 0.296 ± 0.004 vs 0.472 ± 0.029, P < 0.05; 72 h: 0.432 ± 0.024 vs 0.672 ± 0.050, P < 0.05). No obvious changes were found in psiRNA1 group and psiRNA3 group (all P > 0.05).
CONCLUSION: NOV can increase the secretion of extracellular matrix (ECM) and promote the proliferation and activation of HSCs. NOV may be a novel target for gene therapy of liver fibrosis.
Collapse
|
177
|
WU C, LU Y. Inclusion of high molecular weight dextran in calcium phosphate-mediated transfection significantly improves gene transfer efficiency. Cell Mol Biol (Noisy-le-grand) 2007; 53:67-74. [PMID: 17531163 PMCID: PMC2830788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Accepted: 10/27/2006] [Indexed: 05/15/2023]
Abstract
Calcium phosphate-based mammalian cell transfection is a widely used gene transfer technology. To facilitate the efficiency of this gene transfer method, several polysaccharide compounds were tested and evaluated for their effectiveness in enhancing DNA transfection. Using a HIV-1-derived lentivirus vector plasmid as a gene transfer indicator, we demonstrated that the addition of high molecular weight dextran-500 at 0.6-1.2% in the 2x Hepes buffered saline (HBS) increased transfection efficiency by over 50% (as reflected by the number of GFP-positive cells) and increased the titer of resulting lentivirus vector particles even more (up to 4-fold). This enhancement of transfection efficiency was further increased when higher molecular weight dextran formulations were used in place of dextran-500, and also when dextran was used in combination with polybrene, another polycationic chemical compound. Examination of transfected cells showed that dextran had no apparent adverse effect on cell viability and growth. Our data represent the first report showing that dextran can be used to enhance calcium phosphate-mediated gene transfer; this may be useful in applications for the generation of high-titer virus vector stocks using transient transfection technology.
Collapse
Affiliation(s)
- C. WU
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii 96822, USA
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, Hawaii 96822, USA
| | - Y. LU
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii 96822, USA
- Dr. Yuanan Lu, Environmental Health Laboratory, Department of Public Health Sciences, 1960 East West Road, Biomed D104, University of Hawaii, Honolulu, Hawaii 96822, USA. Fax (808) 956–2702;
| |
Collapse
|
178
|
Li D, Chen XQ, Li WJ, Yang YH, Wang JZ, Yu ACH. Cytoglobin Up-regulated by Hydrogen Peroxide Plays a Protective Role in Oxidative Stress. Neurochem Res 2007; 32:1375-80. [PMID: 17476593 DOI: 10.1007/s11064-007-9317-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 02/14/2007] [Indexed: 11/24/2022]
Abstract
Cytoglobin (Cygb) is a recently discovered intracellular respiratory globin, which exists in all types of cells. It has been suggested that Cygb has a role in protecting cells against oxidative stress. In the present study we have tested this hypothesis. The N2a neuroblastoma cells were exposed to various kinds of insults, including hydrogen peroxide (H(2)O(2)), hypoxia, kainic acid, high extracellular CaCl(2), high osmolarity, UV irradiation and heat shock. Among them, only H(2)O(2)-treatment induced a significant up-regulation of cytoglobin mRNA level. We stably transfected N2a cells with Cygb-siRNA vectors and successfully knocked down Cygb. The Cygb-siRNA could exacerbate cell death upon H(2)O(2)-treatment, as demonstrated by MTT cell viability assay. Thus, Cygb in neuronal cells might be specifically induced under oxidative stress to protect them from death.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathophysiology and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
179
|
Li GQ, Xu WZ, Wang JX, Deng WW, Li D, Gu HX. Combination of small interfering RNA and lamivudine on inhibition of human B virus replication in HepG2.2.15 cells. World J Gastroenterol 2007; 13:2324-7. [PMID: 17511031 PMCID: PMC4147141 DOI: 10.3748/wjg.v13.i16.2324] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the inhibition of hepatitis B virus (HBV) replication and expression by combination of siRNA and lamivudine in HepG2.2.15 cells.
METHODS: Recombinant plasmid psil-HBV was constructed and transfected into HepG2.2.15 cells. The transfected cells were cultured in lamivudine-containing medium (0.05 μmol/L) and harvested at 48, 72 and 96 h. The concentration of HBeAg and HBsAg was determined using ELISA. HBV DNA replication was examined by real-time PCR and the level of HBV mRNA was measured by RT-PCR.
RESULTS: In HepG2.2.15 cells treated with combination of siRNA and lamivudine, the secretion of HBeAg and HBsAg into the supernatant was found to be inhibited by 91.80% and 82.40% (2.89 ± 0.48 vs 11.73 ± 0.38, P < 0.05; 4.59 ± 0.57 vs 16.25 ± 0.48, P < 0.05) at 96 h, respectively; the number of HBV DNA copies within culture medium was also significantly decreased at 96 h (1.04 ± 0.26 vs 8.35 ± 0.33, P < 0.05). Moreover, mRNA concentration in HepG2.2.15 cells treated with combination of siRNA and lamivudine was obviously lower compared to those treated either with siRNA or lamivudine (19.44 ± 0.17 vs 33.27 ± 0.21 or 79.9 ± 0.13, P < 0.05).
CONCLUSION: Combination of siRNA and lamivudine is more effective in inhibiting HBV replication as compared to the single use of siRNA or lamivudine in HepG2.2.15 cells.
Collapse
Affiliation(s)
- Gui-Qiu Li
- Department of Microbiology, Harbin Medical University, Harbin 150081, and Department of Anesthesiology, Second Affiliated Hospital, Jiamusi University, Heilongjiang Province, China
| | | | | | | | | | | |
Collapse
|
180
|
Liu L, Li P, Jiang L, Dong C, Wang L, Che Y, Zhao H, Dong S, Li Q. Using interferon-alpha to block expression of cellular ribosome subunit S24 variant 2 in human fibroblasts inhibits translation of the poliovirus genome. Viral Immunol 2007; 20:142-9. [PMID: 17425428 DOI: 10.1089/vim.2006.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Studies about the proteins induced by interferon (IFN-)-alpha stimulation have provided some data on their mechanism of antiviral effect. These proteins were confirmed to contribute to antiviral functions. In this study, IFN-alpha stimulation of human fibroblasts was shown to induce the inhibition of S24 variant 2 (a structural component of the ribosomal small subunit) at the mRNA and protein levels, implying a possible antiviral mechanism for IFN-alpha in human fibroblasts. The delay of poliovirus replication by IFN-alpha was partially compensated for by S24 variant 2 expressed in pcDNA vector-transfected cells, and the interference RNA of S24 variant 2 was able to induce mimetically, to some extent, this poliovirus replication delay. These observations revealed that S24 variant 2 could be involved in the antiviral effects of IFN-alpha in human fibroblasts.
Collapse
Affiliation(s)
- Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Tsang SY, Moore JC, Van Huizen R, W.Y.Chan C, Li RA. Ectopic expression of systemic RNA interference defective protein in embryonic stem cells. Biochem Biophys Res Commun 2007; 357:480-6. [PMID: 17434453 PMCID: PMC2464293 DOI: 10.1016/j.bbrc.2007.03.187] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2007] [Accepted: 03/28/2007] [Indexed: 11/15/2022]
Abstract
RNA interference (RNAi), a post-transcriptional gene silencing mechanism originally described in Caenorhabditis elegans, involves sequence-specific mRNA degradation mediated by double-stranded RNAs (dsRNAs). Passive dsRNA uptake has been uniquely observed in C. elegans due to the expression of systemic RNA interference defective-1 (SID-1). Here we investigated the ability of ectopic SID-1 expression to enable passive cellular uptake of short interfering RNA (siRNA) or double stranded RNA (dsRNA) in pluripotent mouse embryonic stem cells (mESCs). When SID-1-GFP and the Firefly luciferase reporter gene (luc(Fir)) were co-expressed in mESCs, luc(Fir) activity could be suppressed by simple incubation with dsRNAs/siRNAs that were designed to specifically target luc(Fir). By contrast, suppression was not observed in mESCs expressing luc(Fir) and GFP alone or when either GFP- or SID-1-GFP-expressing cells were incubated with control dsRNAs/siRNAs (non-silencing or Renilla luciferase-specific). These results may lead to high-throughput experimental strategies for studying ESC differentiation and novel approaches to genetically inhibit or eliminate the tumorigenicity of ESCs.
Collapse
Affiliation(s)
- Suk Ying Tsang
- Stem Cell Program, University of California, Davis, CA, USA
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA, USA
- Corresponding author: Ronald Li, University, of California 2425, Stockton Blvd., Rm 650, Sacramento CA 95817, Tel. 916 453-2225, Fax. 916 453-2238, E-mail:
| | - Jennifer C. Moore
- Stem Cell Program, University of California, Davis, CA, USA
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA, USA
| | - Rika Van Huizen
- Stem Cell Program, University of California, Davis, CA, USA
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA, USA
| | - Camie W.Y.Chan
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA, USA
- Institute of Pediatric Regenerative Medicine, Shriners Hospital for Children of North America, Sacramento, CA, USA
| | - Ronald A. Li
- Stem Cell Program, University of California, Davis, CA, USA
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA, USA
- Institute of Pediatric Regenerative Medicine, Shriners Hospital for Children of North America, Sacramento, CA, USA
| |
Collapse
|
182
|
Cheng JC, Horwitz EM, Karsten SL, Shoemaker L, Kornblum HI, Malik P, Sakamoto KM. Report on the Workshop “New Technologies in Stem Cell Research,” Society for Pediatric Research, San Francisco, California, April 29, 2006. Stem Cells 2007; 25:1070-88. [PMID: 17255523 DOI: 10.1634/stemcells.2006-0397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Jerry C Cheng
- Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories and Mattel Children's Hospital, Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|
183
|
Kumar LD, Clarke AR. Gene manipulation through the use of small interfering RNA (siRNA): from in vitro to in vivo applications. Adv Drug Deliv Rev 2007; 59:87-100. [PMID: 17434644 DOI: 10.1016/j.addr.2007.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 03/04/2007] [Indexed: 12/19/2022]
Abstract
The conventional approach to investigate genotype-phenotype relationships has been the generation of gene targeted murine strains. However, the emergence of RNAi technologies has opened the possibility of much more rapid (and indeed more cost effective) genetic manipulation in vivo at the level of the transcriptome. Successful application of RNAi in vivo depends on intracellular targeted delivery of siRNA/shRNA molecules for efficient knockdown of the desired gene. In this review, we discuss the rationale and different strategies of using siRNA/shRNA for accomplishing the silencing of targeted genes in a spatial and /or temporally regulated manner. We also summarise the steps involved in extending these approaches to in vivo applications, with a specific focus upon the development of silencing in the mouse.
Collapse
Affiliation(s)
- Lekha Dinesh Kumar
- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
184
|
Bhagavati S, Song X, Siddiqui MAQ. RNAi inhibition of Pax3/7 expression leads to markedly decreased expression of muscle determination genes. Mol Cell Biochem 2007; 302:257-62. [PMID: 17396232 DOI: 10.1007/s11010-007-9444-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Accepted: 03/02/2007] [Indexed: 10/23/2022]
Abstract
Skeletal muscle regeneration by cell transplantation for the treatment of muscle diseases requires the identification and isolation of well-defined, early skeletal muscle progenitor cells. It is known that myogenesis is governed by the sequential and compound activation of the muscle determination genes, the myogenic regulatory factors (MRFs). Recently it has been proposed that the transcription factors Pax3 and Pax7 trigger the expression of the MRFs and thereby specify a novel population of cells destined to enter the myogenic program. We directly tested this hypothesis using RNA interference methodology to reduce the levels of Pax3 and Pax7 RNA in mouse embryoid bodies developing in vitro. We found that decreasing the levels of Pax3/Pax7 RNA leads to a marked and selective decrease in Myf5, MyoD and Desmin expression. Pax3 and Pax7 expressing cells from developing embryos may thus serve as the earliest known skeletal muscle progenitor cells potentially useful for cell transplantation studies.
Collapse
Affiliation(s)
- Satyakam Bhagavati
- Department of Neurology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA.
| | | | | |
Collapse
|
185
|
Luo Q, Kang Q, Song WX, Luu HH, Luo X, An N, Luo J, Deng ZL, Jiang W, Yin H, Chen J, Sharff KA, Tang N, Bennett E, Haydon RC, He TC. Selection and validation of optimal siRNA target sites for RNAi-mediated gene silencing. Gene 2007; 395:160-9. [PMID: 17449199 DOI: 10.1016/j.gene.2007.02.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Revised: 02/13/2007] [Accepted: 02/15/2007] [Indexed: 11/23/2022]
Abstract
RNA interference (RNAi)-mediated gene silencing has become a valuable tool for functional studies, reverse genomics, and drug discoveries. One major challenge of using RNAi is to identify the most effective short interfering RNAs (siRNAs) sites of a given gene. Although several published bioinformatic prediction models have proven useful, the process to select and validate optimal siRNA sites for a given gene remains empirical and laborious. Here, we developed a fluorescence-based selection system using a retroviral vector backbone, namely pSOS, which was based on the premise that candidate siRNAs would knockdown the chimeric transcript between GFP and target gene. The expression of siRNA was driven by the opposing convergent H1 and U6 promoters. This configuration simplifies the cloning of duplex siRNA oligonucleotide cassettes. We demonstrated that GFP signal reduction was closely correlated with siRNA knockdown efficiency of human beta-catenin, as well as with the inhibition of beta-catenin/Tcf4 signaling activity. The pSOS should not only facilitate the selection and validation of candidate siRNA sites, but also provide efficient delivery tools of siRNAs via viral vectors in mammalian cells. Thus, the pSOS system represents an efficient and user-friendly strategy to select and validate siRNA target sites.
Collapse
Affiliation(s)
- Qing Luo
- The Children's Hospital, and the Key Laboratory of Diagnostic Medicine designated by the Ministry of Education, Chongqing University of Medical Sciences, Chongqing 400016, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Jian R, Cheng X, Jiang J, Deng S, Hu F, Zhang J. A cDNA-based random RNA interference library for functional genetic screens in embryonic stem cells. Stem Cells 2007; 25:1904-12. [PMID: 17379769 DOI: 10.1634/stemcells.2006-0448] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To facilitate high-throughput functional genetic screens in embryonic stem cells, a simple and efficient system to construct cDNA-based random RNA interference (RNAi) library was developed in the study. Previous studies have demonstrated that sequence-specific gene silencing could be induced by long double-stranded RNA (dsRNA) in mouse embryos, mouse oocytes, embryonic stem cells, and other mammalian cells. Based on these findings, a dsRNA-expressing RNAi vector system was designed. This study provided evidence that the vector design could induce efficient knockdown of expression of both exogenous egfp gene and endogenous MTM1 gene in mouse embryonic stem cells. A random RNAi library was established by cloning enzyme-digested cDNA of mouse embryonic stem (ES) cells into the BamHI site of the convergent dual promoter RNAi vector. Sequencing of 20 randomly selected clones from the library showed that 17 contained inserts and that all of them were unique sequences. A functional genetic screen of genes involving in self-renewal and differentiation with the random RNAi library identified ubiquitin. The ubiquitin knockdown ES cell line generated 20%-30% of undifferentiated colonies in the absence of leukemia inhibitor factor, whereas parental ES cells and control vector pDCont transfectants produced less than 5% of colonies of undifferentiated cells, suggesting that ubiquitin plays a role in ES cell differentiation. The random RNAi library provides a useful tool for investigation of molecular mechanisms of cellular development and differentiation.
Collapse
Affiliation(s)
- Rui Jian
- Laboratory of Infection Immunity, Department of Microbiology, Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | |
Collapse
|
187
|
Li L, Yang L, Scudiero DA, Miller SA, Yu ZX, Stukenberg PT, Shoemaker RH, Kotin RM. Development of recombinant adeno-associated virus vectors carrying small interfering RNA (shHec1)-mediated depletion of kinetochore Hec1 protein in tumor cells. Gene Ther 2007; 14:814-27. [PMID: 17330085 DOI: 10.1038/sj.gt.3302933] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transcript depletion using small interfering RNA (siRNA) technology represents a potentially valuable technique for the treatment of cancer. However, delivering therapeutic quantities of siRNA into solid tumors by chemical transfection is not feasible, whereas viral vectors efficiently transduce many human tumor cell lines. Yet producing sufficient quantities of viral vectors that elicit acute and selective cytotoxicity remains a major obstacle for preclinical and clinical trials. Using the invertebrate Spodoptera frugiperda (Sf9) cell line, we were able to produce high titer stocks of cytotoxic recombinant adeno-associated virus (rAAV) that express short hairpin RNA (shRNA) and that efficiently deplete Hec1 (highly expressed in cancer 1), or Kntc2 (kinetochore-associated protein 2), a kinetochore protein directly involved in kinetochore microtubule interactions, chromosome congression and spindle checkpoint signaling. Depletion of Hec1 protein results in persistent spindle checkpoint activation followed by cell death. Because Hec1 expression and activity are only present in mitotic cells, non-dividing cells were not affected by rAAV treatment. On the basis of the results of screening 56 human tumor cell lines with three different serotype vectors, we used a tumor xenograft model to test the effects in vivo. The effects of the shHec1 vector were evident in sectioned and stained tumors. The experiments with rAAV-shRNA vectors demonstrate the utility of producing vectors in invertebrate cells to obtain sufficient concentrations and quantities for solid tumor therapy. This addresses an important requirement for cancer gene therapy, to produce cytotoxic vectors in sufficient quantities and concentrations to enable quantitative transduction and selective killing of solid tumor cells.
Collapse
Affiliation(s)
- L Li
- Laboratory of Biochemical Genetics, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Abstract
Double stranded short interfering RNAs (siRNAs) mediate gene silencing in a sequence specific manner. By virtue of their specific gene silencing activity and owing to the recent discoveries on their plasmid and virus driven expression, siRNAs are being widely adopted in research and therapeutics. Efforts were made to optimize the siRNA expression system for the application in therapy. One major obstacle in developing RNA interference (RNAi) therapy is the delivery of siRNAs to the target cells. Combination of novel molecular targeting technologies, such as recombinant protein technology and ribosome display technology, will enable to deliver gene silencing agents to target cells specifically and efficiently.
Collapse
Affiliation(s)
- Atsushi Inoue
- National Institute of Advanced Industrial Science and Technology, Tsukuba Science City, Japan
| | | | | |
Collapse
|
189
|
Abstract
RNA interference (RNAi) is a naturally occurring cellular defense mechanism against viral infections and transposon invasion. Short double-stranded RNA molecules, so-called small-interfering (si)RNAs, bind their complementary mRNA leading to the mRNA's degradation. During the past few years, RNAi has become a valuable tool for transient as well as stable repression of gene expression rendering the time-consuming production of knockout animals superfluous. In this chapter the usability of the RNAi technology in cancer research will be described, focusing on the application of large-scale screens for identification of new components in cancer-relevant signal pathways (e.g., p53, RAS). The screens are especially helpful in the detection of potential anticancer drug targets or siRNAs with therapeutic potential.
Collapse
Affiliation(s)
- Uta Fuchs
- Dr. von Haunersches Kinderspital, Ludwig Maximilians Universität München, München, Germany
| | | |
Collapse
|
190
|
Abstract
Silencing of gene expression by RNA interference (RNAi) has become a powerful tool for functional genomics in mammalian cells. Furthermore, RNAi holds promise as a simple, fast and cost-effective approach to studying mammalian gene function in vivo and as a novel therapeutic approach. This review provides an overview of the progress of RNAi in vivo, with emphasis on systemic/local siRNA delivery, viral shRNA vectors, shRNA vector transgenic mice and conditional systems to control shRNA vectors. Taken together, the data from 80 in vivo studies show that RNAi is a useful tool that offers new opportunities for functional genomics in mice.
Collapse
Affiliation(s)
- R Kühn
- Institute for Developmental Genetics, GSF, National Research Center for Environment and Health, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| | | | | |
Collapse
|
191
|
Lin DI, Barbash O, Kumar KS, Weber JD, Harper JW, Klein-Szanto AJP, Rustgi A, Fuchs SY, Diehl JA. Phosphorylation-dependent ubiquitination of cyclin D1 by the SCF(FBX4-alphaB crystallin) complex. Mol Cell 2006; 24:355-66. [PMID: 17081987 PMCID: PMC1702390 DOI: 10.1016/j.molcel.2006.09.007] [Citation(s) in RCA: 287] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Revised: 07/11/2006] [Accepted: 09/12/2006] [Indexed: 11/19/2022]
Abstract
Growth factor-dependent accumulation of the cyclin D1 proto-oncogene is balanced by its rapid phosphorylation-dependent proteolysis. Degradation is triggered by threonine 286 phosphorylation, which promotes its ubiquitination by an unknown E3 ligase. We demonstrate that Thr286-phosphorylated cyclin D1 is recognized by a Skp1-Cul1-F box (SCF) ubiquitin ligase where FBX4 and alphaB crystallin govern substrate specificity. Overexpression of FBX4 and alphaB crystallin triggered cyclin D1 ubiquitination and increased cyclin D1 turnover. Impairment of SCF(FBX4-alphaB crystallin) function attenuated cyclin D1 ubiquitination, promoting cyclin D1 overexpression and accelerated cell-cycle progression. Purified SCF(FBX4-alphaB crystallin) catalyzed polyubiquitination of cyclin D1 in vitro. Consistent with a putative role for a cyclin D1 E3 ligase in tumorigenesis, FBX4 and alphaB crystallin expression was reduced in tumor-derived cell lines and a subset of primary human cancers that overexpress cyclin D1. We conclude that SCF(FBX4-alphaB crystallin) is an E3 ubiquitin ligase that promotes ubiquitin-dependent degradation of Thr286-phosphorylated cyclin D1.
Collapse
Affiliation(s)
- Douglas I. Lin
- The Leonard and Madlyn Abramson Family Cancer Research
Institute and Cancer Center
- Department of Cancer Biology, University of Pennsylvania,
Philadelphia, Pennsylvania 19104, USA
| | - Olena Barbash
- The Leonard and Madlyn Abramson Family Cancer Research
Institute and Cancer Center
| | - K.G. Suresh Kumar
- Department of Animal Biology, University of Pennsylvania,
Philadelphia, PA
| | - Jason D. Weber
- Department of Medicine Washington University School of
Medicine, St Louis MO
| | - J. Wade Harper
- Department of Pathology, Harvard Medical School, Boston,
Massachusetts 02115, USA
| | | | - Anil Rustgi
- The Leonard and Madlyn Abramson Family Cancer Research
Institute and Cancer Center
- Department of Medicine University of Pennsylvania, USA
| | - Serge Y. Fuchs
- Department of Animal Biology, University of Pennsylvania,
Philadelphia, PA
| | - J. Alan Diehl
- The Leonard and Madlyn Abramson Family Cancer Research
Institute and Cancer Center
- Department of Cancer Biology, University of Pennsylvania,
Philadelphia, Pennsylvania 19104, USA
- To whom correspondence should be directed:
| |
Collapse
|
192
|
Hiraoka-Kanie M, Miyagishi M, Yamashita JK. Differentiation stage-specific analysis of gene function with inducible short hair-pin RNA in differentiating embryonic stem cells. Biochem Biophys Res Commun 2006; 351:669-74. [PMID: 17084387 DOI: 10.1016/j.bbrc.2006.10.108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Accepted: 10/17/2006] [Indexed: 11/30/2022]
Abstract
Cell differentiation is regulated by spatial and temporal coordination of gene expressions. Previously, we have established an embryonic stem (ES) cell differentiation system that can trace early cardiovascular developmental process in vitro. Here we show that tetracycline-induced short hair-pin RNA (shRNA) expression in differentiating ES cells successfully suppressed stage-specific genes for differentiation and modified cell fates. We established ES cell lines carrying shRNA gene driven by tRNA(val) promoter with tetracycline operator sequences (tet-ON system). When expression of vascular endothelial growth factor receptor-2 (VEGFR2) gene, a vascular progenitor and mesoderm marker and an essential gene for endothelial cell (EC) differentiation, was suppressed by shRNA in early ES cell differentiation, appearance of VEGFR2(+) mesoderm cells was substantially reduced. Suppression of VEGFR2 expression at mesoderm stage almost completely inhibited EC differentiation from VEGFR2(+) mesoderm cells. This novel experimental system, thus, can selectively determine stage-specific roles of genes in differentiation in vitro.
Collapse
Affiliation(s)
- Mina Hiraoka-Kanie
- Laboratory of Stem Cell Differentiation, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
193
|
Effect of Survivin-siRNA-mediated gene silencing on Survivin expression in osteosarcoma cell line MG-63. Chin J Cancer Res 2006. [DOI: 10.1007/s11670-006-0286-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
194
|
Stein J, Shah R, Steiner D, Dey A. RNAi-mediated silencing of prohormone convertase (PC) 5/6 expression leads to impairment in processing of cocaine- and amphetamine-regulated transcript (CART) precursor. Biochem J 2006; 400:209-15. [PMID: 16800814 PMCID: PMC1635452 DOI: 10.1042/bj20060506] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Understanding the functions of the widely expressed PCs (prohormone/proprotein convertases), including PC5/6, furin and PACE4 (paired basic amino acid cleaving enzyme 4), in animal models is difficult since individual knockouts of these PCs in mice exhibit early embryonic lethality. To investigate the roles of PC5/6 in processing pro-CART (pro-cocaine- and amphetamine-regulated transcript), an important anorexigenic peptide precursor, we have generated GH3 cells silenced for PC5/6 expression by RNAi (RNA interference). We show, following transient knockdown of PC5/6 in these neuroendocrine cells, that generation of the two bioactive forms, CART I (amino acids 42-89/55-102) and CART II (amino acids 49-89/62-102), from pro-CART is impaired due to a lack particularly of the A isoform of PC5/6. The results indicate that PC5/6A shares specificities primarily with PC2 (PC5/6A<PC2) in cleaving the pairs of basic residues, KR (40, 41 //53, 54/) and KK (47, 48//60, 61/), within the pro-CART isoforms [see Dey, Zhu, Carroll, Turck, Stein and Steiner (2003) J. Biol. Chem. 278, 15007-15014]. We do not find any significant role of PC5/6A in processing the RXXR (29-32/) site for production of intermediate CART (amino acids 33-102) from long pro-CART. The findings taken altogether indicate that PC5/6 participates in normal processing of pro-CART.
Collapse
Affiliation(s)
- Jeffrey Stein
- *Department of Biochemistry and Molecular Biology, University of Chicago, 5841 South Maryland Avenue, MC-1027, Chicago, IL 60637, U.S.A
| | - Rohan Shah
- *Department of Biochemistry and Molecular Biology, University of Chicago, 5841 South Maryland Avenue, MC-1027, Chicago, IL 60637, U.S.A
| | - Donald F. Steiner
- *Department of Biochemistry and Molecular Biology, University of Chicago, 5841 South Maryland Avenue, MC-1027, Chicago, IL 60637, U.S.A
- †Howard Hughes Medical Institute, University of Chicago, 5841 South Maryland Avenue, MC-1028, Chicago, IL 60637, U.S.A
| | - Arunangsu Dey
- *Department of Biochemistry and Molecular Biology, University of Chicago, 5841 South Maryland Avenue, MC-1027, Chicago, IL 60637, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
195
|
Tagami T, Barichello JM, Kikuchi H, Ishida T, Kiwada H. The gene-silencing effect of siRNA in cationic lipoplexes is enhanced by incorporating pDNA in the complex. Int J Pharm 2006; 333:62-9. [PMID: 17097247 DOI: 10.1016/j.ijpharm.2006.09.057] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2006] [Revised: 09/22/2006] [Accepted: 09/26/2006] [Indexed: 01/13/2023]
Abstract
Efficient delivery is a key issue in translating interference RNA technology into a feasible therapy. The efficiency of carrier systems used for this technology is commonly tested by co-transfection, i.e. simultaneous transfection with an exogenous gene and with the siRNA. Two approaches can be distinguished: (1) with the two transfectants in the same carrier complex (siRNA/pDNA/carrier) and (2) with the two transfectants in different carrier complexes (pDNA/carrier and siRNA/carrier). The process to prepare the nucleic acid(s)-carrier complexes and the transfection procedure may affect the effectiveness of the gene-silencing process. In this study, two preparation methods were compared, namely the co-preparation of an siRNA/pDNA/liposome lipoplex (Method I) and the separate preparation of an siRNA/liposome lipoplex and a pDNA/liposome lipoplex (Method II). siRNA in the lipoplex produced by Method I showed a stronger gene-silencing effect than that in the lipoplexes prepared by Method II. There was no significant difference between the two methods in the amount of siRNA delivered to cells. Cellular entry and intracellular trafficking of siRNA/pDNA/liposome lipoplex is likely to differ from those of the separate lipoplexes. When in Method II non-transcriptional pDNA was included in the complex with siRNA, the gene-silencing effect was significantly enhanced. If and to what extent the experimental design is suitable to quantify RNA interference remains to be demonstrated.
Collapse
Affiliation(s)
- Tatsuaki Tagami
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Health Biosciences, The University of Tokushima, 1-78-1 Sho-machi, Tokushima 770-8505, Japan
| | | | | | | | | |
Collapse
|
196
|
Zhao ZF, Yang H, Han DW, Zhao LF, Zhang GY, Zhang Y, Liu MS. Inhibition of hepatitis B virus expression and replication by RNA interference in HepG2.2.15. World J Gastroenterol 2006; 12:6046-9. [PMID: 17009407 PMCID: PMC4124416 DOI: 10.3748/wjg.v12.i37.6046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the inhibition of hepatitis B virus replication and expression by transfecting vector-based small interference RNA (siRNA) pGenesil-HBV X targeting HBV X gene region into HepG2.2.15 cells.
METHODS: pGenesil-HBV X was constructed and transfected into HepG2.2.15 cells via lipofection. HBV antigen secretion was determined 24, 48, and 72 h after transfection by time-resolved immunofluorometric assays (TRFIA). HBV replication was examined by fluorescence quantitative PCR, and the expression of cytoplasmic viral proteins was determined by immunohistochemistry.
RESULTS: The secretion of HBsAg and HBeAg into the supernatant was found to be inhibited by 28.5% and 32.2% (P < 0.01), and by 38.67% (P < 0.05) and 42.86% (P < 0.01) at 48 h and 72 h after pGenesil-HBV X transfection, respectively. Immunohistochemical staining for cytoplasmic HBsAg showed a similar decline in HepG2.2.15 cells 48 h after transfection. The number of HBV genomes within culture supernatants was also significantly decreased 48 h and 72 h post-transfection as quantified by fluorescence PCR (P < 0.05).
CONCLUSION: In HepG2.2.15 cells, HBV replication and expression is inhibited by vector-based siRNA pGenesil-HBV X targeting the HBV X coding region.
Collapse
Affiliation(s)
- Zhong-Fu Zhao
- Institute of Hepatology, Changzhi Medical College, Changzhi 046000, Shanxi Province, China.
| | | | | | | | | | | | | |
Collapse
|
197
|
Streicher KL, Yang ZQ, Draghici S, Ethier SP. Transforming function of the LSM1 oncogene in human breast cancers with the 8p11-12 amplicon. Oncogene 2006; 26:2104-14. [PMID: 17001308 PMCID: PMC2435249 DOI: 10.1038/sj.onc.1210002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Amplification of the 8p11-12 region occurs in 15-20% of breast cancers, but the driving oncogene at this locus has yet to be definitively identified. We mapped the 8p11-12 amplicon in breast cancer cell lines and primary human breast cancers and identified the candidate oncogene human Sm-like protein (hLsm1, LSM1) based on increases in copy number and expression level relative to human mammary epithelial cells. To examine the oncogenic role of LSM1, we overexpressed this gene in MCF10A mammary epithelial cells and inhibited its production in the SUM44 breast cancer cell line, which has a natural amplification and overexpression of LSM1. Our data confirmed that LSM1 is an oncogene from the 8p11-12 amplicon by showing that hLsm1 overexpression induced growth factor-independent proliferation and soft agar colony formation in MCF10A cells, and hLsm1 inhibition in SUM44 cells dramatically reduced soft agar growth. Little is known about hLsm1 function other than its involvement in mRNA degradation; therefore, we used expression microarray analysis to investigate how hLsm1 affects cell transformation in MCF10A and SUM44 cells. We identified numerous genes altered following hLsm1 overexpression common to SUM44 breast cancer cells that play important roles in cell cycle regulation, cell proliferation and other cancer-promoting processes. Future work will continue to characterize these important changes to achieve a more complete understanding of the mechanism of hLsm1's effect on cancer progression.
Collapse
Affiliation(s)
- K L Streicher
- Breast Cancer Program, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
198
|
Oeggerli M, Schraml P, Ruiz C, Bloch M, Novotny H, Mirlacher M, Sauter G, Simon R. E2F3 is the main target gene of the 6p22 amplicon with high specificity for human bladder cancer. Oncogene 2006; 25:6538-43. [PMID: 16953223 DOI: 10.1038/sj.onc.1209946] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Amplification of 6p22 occurs in about 10-20% of bladder cancers and is associated with enhanced tumour cell proliferation. Candidate target genes for the 6p22 amplicon include E2F3 and the adjacent gene NM_017774. To clarify which gene is representing the main target, we compared the prevalence of the amplification and the functional role of both genes. Amplification of E2F3 and NM_017774 was analysed by fluorescence in situ hybridization on a bladder cancer tissue microarray composed of 2317 cancer samples. Both genes showed amplification in 104 of 893 (11.6%) interpretable tumours and were exclusively found co-amplified. Additional gene expression analysis by real-time polymerase chain reaction in 12 tumour-derived cell lines revealed that amplification of 6p22 was always associated with co-overexpression of E2F3 and NM_017774. Furthermore, RNA interference was used to study the influence of reduced gene expression on cell growth. In tumour cells with and without the 6p22 amplicon, knockdown of E2F3 always lead to unequivocal reduction of proliferation, whereas knockdown of NM_017774 was only capable to slow down cell proliferation in non-amplified cells. Our findings point out that E2F3 but not NM_017774 is driving enhanced proliferation of 6p22 amplified tumour cells. We conclude that E2F3 must be responsible for the growth advantage of 6p22 amplified bladder cancer cells.
Collapse
Affiliation(s)
- M Oeggerli
- Department of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Siu YT, Chin KT, Siu KL, Yee Wai Choy E, Jeang KT, Jin DY. TORC1 and TORC2 coactivators are required for tax activation of the human T-cell leukemia virus type 1 long terminal repeats. J Virol 2006; 80:7052-9. [PMID: 16809310 PMCID: PMC1489057 DOI: 10.1128/jvi.00103-06] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) Tax protein activates viral transcription from the long terminal repeats (LTR). Mechanisms through which Tax activates LTR have been established, but coactivators of this process remain to be identified and characterized. Here we show that all three members of the TORC family of transcriptional regulators are coactivators of Tax for LTR-driven expression. TORC coactivation requires CREB, but not ATF4 or other bZIP factors. Tax physically interacts with TORC1, TORC2, and TORC3 (TORC1/2/3), and the depletion of TORC1/2/3 inhibited Tax activity. TORC coactivation can be further enhanced by transcriptional coactivator p300. In addition, coactivators in the p300 family are required for full activity of Tax independently of TORC1/2/3. Thus, both TORC and p300 families of coactivators are essential for optimal activation of HTLV-1 transcription by Tax.
Collapse
Affiliation(s)
- Yeung-Tung Siu
- Department of Biochemistry, The University of Hong Kong, 3/F Laboratory Block, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong
| | | | | | | | | | | |
Collapse
|
200
|
Strat A, Gao L, Utsuki T, Cheng B, Nuthalapaty S, Mathis JM, Odaka Y, Giordano T. Specific and nontoxic silencing in mammalian cells with expressed long dsRNAs. Nucleic Acids Res 2006; 34:3803-10. [PMID: 16916791 PMCID: PMC1540741 DOI: 10.1093/nar/gkl532] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A number of groups have developed libraries of siRNAs to identify genes through functional genomics. While these studies have validated the approach of making functional RNAi libraries to understand fundamental cellular mechanisms, they require information and knowledge of existing sequences since the RNAi sequences are generated synthetically. An alternative strategy would be to create an RNAi library from cDNA. Unfortunately, the complexity of such a library of siRNAs would make screening difficult. To reduce the complexity, longer dsRNAs could be used; however, concerns of induction of the interferon response and off-target effects of long dsRNAs have prevented their use. As a first step in creating such libraries, long dsRNA was expressed in mammalian cells. The 250 nt dsRNAs were capable of efficiently silencing a luciferase reporter gene that was stably transfected in MDA-MB-231 cells without inducing the interferon response or off-target effects any more than reported for siRNAs. In addition, a long dsRNA expressed in the same cell line was capable of silencing endogenous c-met expression and inhibited cell migration, whereas the dsRNA against luciferase had no effect on c-met or cell migration. The studies suggest that large dsRNA libraries are feasible and that functional selection of genes will be possible.
Collapse
Affiliation(s)
- Aurel Strat
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Lu Gao
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Tada Utsuki
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Bing Cheng
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Sam Nuthalapaty
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - J. Mike Mathis
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Yoshinobu Odaka
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences CenterShreveport, LA, USA
| | - Tony Giordano
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences CenterShreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences CenterShreveport, LA, USA
- To whom correspondence should be addressed. Tel: +1 318 675 7791; Fax: +1 318 675 5180;
| |
Collapse
|