1
|
Godinho BMDC, Gilbert JW, Haraszti RA, Coles AH, Biscans A, Roux L, Nikan M, Echeverria D, Hassler M, Khvorova A. Pharmacokinetic Profiling of Conjugated Therapeutic Oligonucleotides: A High-Throughput Method Based Upon Serial Blood Microsampling Coupled to Peptide Nucleic Acid Hybridization Assay. Nucleic Acid Ther 2017; 27:323-334. [PMID: 29022758 DOI: 10.1089/nat.2017.0690] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Therapeutic oligonucleotides, such as small interfering RNAs (siRNAs), hold great promise for the treatment of incurable genetically defined disorders by targeting cognate toxic gene products for degradation. To achieve meaningful tissue distribution and efficacy in vivo, siRNAs must be conjugated or formulated. Clear understanding of the pharmacokinetic (PK)/pharmacodynamic behavior of these compounds is necessary to optimize and characterize the performance of therapeutic oligonucleotides in vivo. In this study, we describe a simple and reproducible methodology for the evaluation of in vivo blood/plasma PK profiles and tissue distribution of oligonucleotides. The method is based on serial blood microsampling from the saphenous vein, coupled to peptide nucleic acid hybridization assay for quantification of guide strands. Performed with minimal number of animals, this method allowed unequivocal detection and sensitive quantification without the need for amplification, or further modification of the oligonucleotides. Using this methodology, we compared plasma clearances and tissue distribution profiles of two different hydrophobically modified siRNAs (hsiRNAs). Notably, cholesterol-hsiRNA presented slow plasma clearances and mainly accumulated in the liver, whereas, phosphocholine-docosahexaenoic acid-hsiRNA was rapidly cleared from the plasma and preferably accumulated in the kidney. These data suggest that the PK/biodistribution profiles of modified hsiRNAs are determined by the chemical nature of the conjugate. Importantly, the method described in this study constitutes a simple platform to conduct pilot assessments of the basic clearance and tissue distribution profiles, which can be broadly applied for evaluation of new chemical variants of siRNAs and micro-RNAs.
Collapse
Affiliation(s)
- Bruno M D C Godinho
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - James W Gilbert
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Reka A Haraszti
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Andrew H Coles
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Annabelle Biscans
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Loic Roux
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Mehran Nikan
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Dimas Echeverria
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Matthew Hassler
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Anastasia Khvorova
- 1 RNA Therapeutics Institute, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
2
|
Lu T, Lin Z, Ren J, Yao P, Wang X, Wang Z, Zhang Q. The Non-Specific Binding of Fluorescent-Labeled MiRNAs on Cell Surface by Hydrophobic Interaction. PLoS One 2016; 11:e0149751. [PMID: 26930565 PMCID: PMC4773022 DOI: 10.1371/journal.pone.0149751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/04/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND MicroRNAs are small noncoding RNAs about 22 nt long that play key roles in almost all biological processes and diseases. The fluorescent labeling and lipofection are two common methods for changing the levels and locating the position of cellular miRNAs. Despite many studies about the mechanism of DNA/RNA lipofection, little is known about the characteristics, mechanisms and specificity of lipofection of fluorescent-labeled miRNAs. METHODS AND RESULTS Therefore, miRNAs labeled with different fluorescent dyes were transfected into adherent and suspension cells using lipofection reagent. Then, the non-specific binding and its mechanism were investigated by flow cytometer and laser confocal microscopy. The results showed that miRNAs labeled with Cy5 (cyanine fluorescent dye) could firmly bind to the surface of adherent cells (Hela) and suspended cells (K562) even without lipofection reagent. The binding of miRNAs labeled with FAM (carboxyl fluorescein) to K562 cells was obvious, but it was not significant in Hela cells. After lipofectamine reagent was added, most of the fluorescently labeled miRNAs binding to the surface of Hela cells were transfected into intra-cell because of the high transfection efficiency, however, most of them were still binding to the surface of K562 cells. Moreover, the high-salt buffer which could destroy the electrostatic interactions did not affect the above-mentioned non-specific binding, but the organic solvent which could destroy the hydrophobic interactions eliminated it. CONCLUSIONS These results implied that the fluorescent-labeled miRNAs could non-specifically bind to the cell surface by hydrophobic interaction. It would lead to significant errors in the estimation of transfection efficiency only according to the cellular fluorescence intensity. Therefore, other methods to evaluate the transfection efficiency and more appropriate fluorescent dyes should be used according to the cell types for the accuracy of results.
Collapse
Affiliation(s)
- Ting Lu
- Division of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Zongwei Lin
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jianwei Ren
- Health Division of Guard Bureau, General Staff Department of Chinese PLA, Beijing, China
| | - Peng Yao
- Traditional Chinese Medicine Department, Jinan Firefighting Hospital, Jinan, China
| | - Xiaowei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Zhe Wang
- Division of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Qunye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
3
|
Ahlemeyer B, Vogt JF, Michel V, Hahn-Kohlberger P, Baumgart-Vogt E. Microporation is an efficient method for siRNA-induced knockdown of PEX5 in HepG2 cells: evaluation of the transfection efficiency, the PEX5 mRNA and protein levels and induction of peroxisomal deficiency. Histochem Cell Biol 2014; 142:577-91. [PMID: 25224142 DOI: 10.1007/s00418-014-1254-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2014] [Indexed: 11/26/2022]
Abstract
The pathomechanism of peroxisomal biogenesis disorders (PBDs), a group of inherited autosomal recessive diseases with mutations of peroxin (PEX) genes, is not yet fully understood. Therefore, several knockout models, e.g., the PEX5 knockout mouse, have been generated exhibiting a complete loss of peroxisomal function. In this study, we wanted to knockdown PEX5 using the siRNA technology (1) to mimic milder forms of PBDs in which the mutated peroxin has some residual function and (2) to analyze the cellular consequences of a reduction of the PEX5 protein without adaption during the development as it is the case in a knockout animal. First, we tried to optimize the transfection of the hepatoma cell line HepG2 with PEX5 siRNA using different commercially available liposomal and non-liposomal transfection reagents (Lipofectamine(®) 2000, FuGENE 6, HiPerFect(®), INTERFERin™, RiboJuice™) as well as microporation using the Neon™ Transfection system. Microporation was found to be superior to the transfection reagents with respect to the transfection efficiency (100 vs. 0-70%), to the reduction of PEX5 mRNA (by 90 vs. 0-50%) and PEX5 protein levels (by 70 vs. 0-50%). Interestingly, we detected that a part of the cleaved PEX5 mRNA still existed as 3' fragment (15%) 24 h after microporation. Using microporation, we further analyzed whether the reduced PEX5 protein level impaired peroxisomal function. We indeed detected a reduced targeting of SKL-tagged proteins into peroxisomes as well as an increased oxidative stress as found in PBD patients and respective knockout mouse models. Knockdown of the PEX5 protein and functional consequences were at a maximum 48 h after microporation. Thereafter, the PEX5 protein was resynthesized, which may allow the temporal analysis of the loss as well as the reconstitution of peroxisomes in the future. In conclusion, we propose microporation as an efficient and reproducible method to transfect HepG2 cells with PEX5 siRNA. We succeeded to transiently knockdown PEX5 mRNA and its protein level leading to functional consequences similar as observed in peroxisome deficiencies.
Collapse
Affiliation(s)
- Barbara Ahlemeyer
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, 35385, Giessen, Germany,
| | | | | | | | | |
Collapse
|
4
|
Han J, Wang QW, Wang SQ. Fluorescent tag is not a reliable marker for small RNA transfection in the presence of serum. J Biosci 2014; 38:471-8. [PMID: 23938380 DOI: 10.1007/s12038-013-9336-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemically synthetic siRNA and miRNA have become powerful tools to study gene function in the past decade. Fluorescent dyes covalently attached to the 5' or 3' ends of synthetic small RNAs are widely used for fluorescently imaging and detection of these RNAs. However, the reliability of fluorescent tags as small RNA markers in different conditions has not attracted enough attention. We used Cy3-labelled small RNAs to explore the reliability of fluorescent tags as small RNA markers in cell cultures involving serum. A strong Cy3-fluorescence signal was observed in the cytoplasm of the cells transfected with Cy3-miR24 in the culture medium containing fetal bovine serum (FBS), but qRT-PCR results showed that little miR24 were detected in these cells. Further study demonstrated that small RNAs were degraded in the presence of FBS, suggesting that it was Cy3-RNA fragments, rather than the original Cy3-miR24, diffused into cells. These phenomena disappeared when FBS was replaced by boiled-FBS, further supporting that the Cy3-fluorescence we observed in cells in the presence of FBS could not represent the presence of intact small RNAs. These findings addressed that fluorescent tags are not reliable for small RNA transfection in the presence of serum in culture.
Collapse
Affiliation(s)
- Jing Han
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | | | | |
Collapse
|
5
|
Kamiya Y, Ito A, Ito H, Urushihara M, Takai J, Fujii T, Liang X, Kashida H, Asanuma H. Selective labeling of mature RISC using a siRNA carrying fluorophore–quencher pair. Chem Sci 2013. [DOI: 10.1039/c3sc51197a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
6
|
Simmons O, Maples PB, Senzer N, Nemunaitis J. Ewing's Sarcoma: Development of RNA Interference-Based Therapy for Advanced Disease. ISRN ONCOLOGY 2012; 2012:247657. [PMID: 22523703 PMCID: PMC3317005 DOI: 10.5402/2012/247657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 12/04/2011] [Indexed: 12/12/2022]
Abstract
Ewing's sarcoma tumors are associated with chromosomal translocation between the EWS gene and the ETS transcription factor gene. These unique target sequences provide opportunity for RNA interference(i)-based therapy. A summary of RNAi mechanism and therapeutically designed products including siRNA, shRNA and bi-shRNA are described. Comparison is made between each of these approaches. Systemic RNAi-based therapy, however, requires protected delivery to the Ewing's sarcoma tumor site for activity. Delivery systems which have been most effective in preclinical and clinical testing are reviewed, followed by preclinical assessment of various silencing strategies with demonstration of effectiveness to EWS/FLI-1 target sequences. It is concluded that RNAi-based therapeutics may have testable and achievable activity in management of Ewing's sarcoma.
Collapse
Affiliation(s)
| | | | - Neil Senzer
- Gradalis, Inc., Dallas, TX 75201, USA
- Mary Crowley Cancer Research Centers, Dallas, TX 75201, USA
- Texas Oncology, PA, Dallas, TX 75251, USA
- Medical City Dallas Hospital, Dallas, TX 75230, USA
| | - John Nemunaitis
- Gradalis, Inc., Dallas, TX 75201, USA
- Mary Crowley Cancer Research Centers, Dallas, TX 75201, USA
- Texas Oncology, PA, Dallas, TX 75251, USA
- Medical City Dallas Hospital, Dallas, TX 75230, USA
| |
Collapse
|
7
|
Nemunaitis J, Rao DD, Liu SH, Brunicardi FC. Personalized cancer approach: using RNA interference technology. World J Surg 2011; 35:1700-14. [PMID: 21557010 DOI: 10.1007/s00268-011-1100-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Normal cellular survival is dependent on the cooperative expression of genes' signaling through a broad array of DNA patterns. Cancer, however, has an Achilles' heel. Its altered cellular survival is dependent on a limited subset of signals through mutated DNA, possibly as few as three. Identification and control of these signals through the use of RNA interference (RNAi) technology may provide a unique clinical opportunity for the management of cancer that employs genomic-proteomic profiling to provide a molecular characterization of the cancer, leading to targeted therapy customized to an individual cancer signal. Such an approach has been described as "personalized therapy." The present review identifies unique developing technology that employs RNAi as a method to target, and therefore block, signaling from mutated DNA and describes a clinical pathway toward its development in cancer therapy.
Collapse
Affiliation(s)
- John Nemunaitis
- Mary Crowley Cancer Research Centers, 1700 Pacific Avenue, Suite 1100, Dallas, TX, USA.
| | | | | | | |
Collapse
|
8
|
Broegger T, Jacobsen JCB, Secher Dam V, Boedtkjer DMB, Kold-Petersen H, Pedersen FS, Aalkjaer C, Matchkov VV. Bestrophin is important for the rhythmic but not the tonic contraction in rat mesenteric small arteries. Cardiovasc Res 2011; 91:685-93. [PMID: 21498420 DOI: 10.1093/cvr/cvr111] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS We have previously characterized a cGMP-dependent Ca(2+)-activated Cl(-) current in vascular smooth muscle cells (SMCs) and have shown its dependence on bestrophin-3 expression. We hypothesize that this current is important for synchronization of SMCs in the vascular wall. In the present study, we aimed to test this hypothesis by transfecting rat mesenteric small arteries in vivo with siRNA specifically targeting bestrophin-3. METHODS AND RESULTS The arteries were tested 3 days after transfection in vitro for isometric force development and for intracellular Ca(2+) in SMCs. Bestrophin-3 expression was significantly reduced compared with arteries transfected with mutated siRNA. mRNA levels for bestrophin-1 and -2 were also significantly reduced by bestrophin-3 down-regulation. This is suggested to be secondary to specific bestrophin-3 down-regulation since siRNAs targeting different exons of the bestrophin-3 gene had identical effects on bestrophin-1 and -2 expression. The transfection affected neither the maximal contractile response nor the sensitivity to norepinephrine and arginine-vasopressin. The amplitude of agonist-induced vasomotion was significantly reduced in arteries down-regulated for bestrophins compared with controls, and asynchronous Ca(2+) waves appeared in the SMCs. The average frequency of vasomotion was not different. 8Br-cGMP restored vasomotion in arteries where the endothelium was removed, but oscillation amplitude was still significantly less in bestrophin-down-regulated arteries. Thus, vasomotion properties were consistent with those previously characterized for rat mesenteric small arteries. Data from our mathematical model are consistent with the experimental results. CONCLUSION This study demonstrates the importance of bestrophins for synchronization of SMCs and strongly supports our hypothesis for generation of vasomotion.
Collapse
Affiliation(s)
- Torbjoern Broegger
- Department of Physiology and Biophysics, Aarhus University, Ole Worms Alle bygn.4, 1163, Aarhus C 8000, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Siddiqui A, Patwardhan GA, Liu YY, Nazzal S. Mixed backbone antisense glucosylceramide synthase oligonucleotide (MBO-asGCS) loaded solid lipid nanoparticles: in vitro characterization and reversal of multidrug resistance in NCI/ADR-RES cells. Int J Pharm 2010; 400:251-9. [PMID: 20816930 DOI: 10.1016/j.ijpharm.2010.08.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 08/04/2010] [Accepted: 08/27/2010] [Indexed: 01/11/2023]
Abstract
In this study, solid lipid nanoparticles (SLN) loaded with MBO-asGCS oligonucleotide were prepared, characterized and evaluated for cytotoxicity against NCI/ADR-RES human ovary cancer cells. Two types of cetyltrimethyl ammonium bromide (CTAB) stabilized SLN, with or without ceramide VI, were prepared by mixed homogenization/ultrasonication technique. Complexes were characterized for size, zeta-potential, and stability in biorelevant media and against DNaseI activity. Binding and release studies were further confirmed by gel electrophoresis. Cytotoxicity of the SLN against NCI/ADR-RES cells was evaluated by quantizing ATP. SLN with ceramide VI had lower particle size (74.6 nm) with improved stability in RPMI media when compared to reference SLN without ceramide VI (167.16 nm). Both SLN however had similar cytotoxicity profile with an optimum binding at CTAB to MBO-asGCS ratio of 6:1. Blank SLN, and free MBO-asGCS in the presence and absence of free doxorubicin had insignificant effect on the viability of NCI/ADR-RES cells. However, when cells were concurrently treated with MBO-asGCS loaded SLN and free doxorubicin, cell viability significantly decreased to approximately 12%. These results suggested that SLN enhanced internalization and uptake of MBO-asGCS oligonucleotide, which led to the downregulation of GCS and subsequently reversing the resistance of the cells to doxorubicin.
Collapse
Affiliation(s)
- Akhtar Siddiqui
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, United States
| | | | | | | |
Collapse
|
10
|
QIP, a protein that converts duplex siRNA into single strands, is required for meiotic silencing by unpaired DNA. Genetics 2010; 186:119-26. [PMID: 20551436 DOI: 10.1534/genetics.110.118273] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RNA interference (RNAi) depends on the production of small RNA to regulate gene expression in eukaryotes. Two RNAi systems exist to control repetitive selfish elements in Neurospora crassa. Quelling targets transgenes during vegetative growth, whereas meiotic silencing by unpaired DNA (MSUD) silences unpaired genes during meiosis. The two mechanisms require common RNAi proteins, such as RNA-directed RNA polymerases, Dicers, and Argonaute slicers. We have previously demonstrated that, while Quelling depends on the redundant dicer activity of DCL-1 and DCL-2, only DCL-1 is required for MSUD. Here, we show that QDE-2-interacting protein (QIP), an exonuclease that is important for the production of single-stranded siRNA during Quelling, is also required for MSUD. QIP is crucial for sexual development and is shown to colocalize with other MSUD proteins in the perinuclear region.
Collapse
|
11
|
Abstract
With the recent discovery of small interfering RNA (siRNA), to silence the expression of genes in vitro and in vivo, there has been a need to deliver these molecules to the cell nucleus. Forming a lipid/nucleic acid complex has become a solution and is explored here. Certain methods and ideas are used, such as: the positive/negative electrostatic interaction with a cationic lipid and an anionic RNA molecule, the size of the lipid vesicle aiding the uptake target tissues, targeted lipoplexes which can increase efficiency, and the protection of the siRNA molecule from the natural defenses of the immune system. Many lipid formulations exist and can be experimented with to achieve varying results depending on the application.
Collapse
Affiliation(s)
- Jeffrey Hughes
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
12
|
Rao DD, Vorhies JS, Senzer N, Nemunaitis J. siRNA vs. shRNA: similarities and differences. Adv Drug Deliv Rev 2009; 61:746-59. [PMID: 19389436 DOI: 10.1016/j.addr.2009.04.004] [Citation(s) in RCA: 419] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 04/13/2009] [Indexed: 12/11/2022]
Abstract
RNA interference (RNAi) is a natural process through which expression of a targeted gene can be knocked down with high specificity and selectivity. Using available technology and bioinformatics investigators will soon be able to identify relevant bio molecular tumor network hubs as potential key targets for knockdown approaches. Methods of mediating the RNAi effect involve small interfering RNA (siRNA), short hairpin RNA (shRNA) and bi-functional shRNA. The simplicity of siRNA manufacturing and transient nature of the effect per dose are optimally suited for certain medical disorders (i.e. viral injections). However, using the endogenous processing machinery, optimized shRNA constructs allow for high potency and sustainable effects using low copy numbers resulting in less off-target effects, particularly if embedded in a miRNA scaffold. Bi-functional design may further enhance potency and safety of RNAi-based therapeutics. Remaining challenges include tumor selective delivery vehicles and more complete evaluation of the scope and scale of off-target effects. This review will compare siRNA, shRNA and bi-functional shRNA.
Collapse
|
13
|
Affiliation(s)
- Jens Kurreck
- Institut für Industrielle Genetik, Universität Stuttgart, Allmandring 31, 70569 Stuttgart (Deutschland), Fax: (+49) 711‐685 66973 http://www.uni‐stuttgart.de/iig/institut/staff/kurreck/index.html
| |
Collapse
|
14
|
Abstract
An efficient mechanism for the sequence-specific inhibition of gene expression is RNA interference. In this process, double-stranded RNA molecules induce cleavage of a selected target RNA (see picture). This technique has in recent years developed into a standard method of molecular biology. Successful applications in animal models have already led to the initiation of RNAi-based clinical trials as a new therapeutic option.Only ten years ago Andrew Fire and Craig Mello were able to show that double-stranded RNA molecules could inhibit the expression of homologous genes in eukaryotes. This process, termed RNA interference, has developed into a standard method of molecular biology. This Review provides an overview of the molecular processes involved, with a particular focus on the posttranscriptional inhibition of gene expression in mammalian cells, the possible applications in research, and the results of the first clinical studies.
Collapse
Affiliation(s)
- Jens Kurreck
- Institute of Industrial Genetics, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany.
| |
Collapse
|
15
|
|
16
|
Matchkov VV, Larsen P, Bouzinova EV, Rojek A, Boedtkjer DMB, Golubinskaya V, Pedersen FS, Aalkjaer C, Nilsson H. Bestrophin-3 (vitelliform macular dystrophy 2-like 3 protein) is essential for the cGMP-dependent calcium-activated chloride conductance in vascular smooth muscle cells. Circ Res 2008; 103:864-72. [PMID: 18776041 DOI: 10.1161/circresaha.108.178517] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the biophysical fingerprints (ion selectivity, voltage-dependence, kinetics, etc) of Ca(2+)-activated Cl(-) currents are well established, their molecular identity is still controversial. Several molecular candidates have been suggested; however, none of them has been fully accepted. We have recently characterized a cGMP-dependent Ca(2+)-activated Cl(-) current with unique characteristics in smooth muscle cells. This novel current has been shown to coexist with a "classic" (cGMP-independent) Ca(2+)-activated Cl(-) current and to have characteristics distinct from those previously known for Ca(2+)-activated Cl(-) currents. Here, we suggest that a bestrophin, a product of the Best gene family, is responsible for the cGMP-dependent Ca(2+)-activated Cl(-) current based on similarities between the membrane currents produced by heterologous expressions of bestrophins and the cGMP-dependent Ca(2+)-activated Cl(-) current. This is supported by similarities in the distribution pattern of the cGMP-dependent Ca(2+)-activated Cl(-) current and bestrophin-3 (the product of Best-3 gene) expression in different smooth muscle. Furthermore, downregulation of Best-3 gene expression with small interfering RNA both in cultured cells and in vascular smooth muscle cells in vivo was associated with a significant reduction of the cGMP-dependent Ca(2+)-activated Cl(-) current, whereas the magnitude of the classic Ca(2+)-activated Cl(-) current was not affected. The majority of previous suggestions that bestrophins are a new Cl(-) channel family were based on heterologous expression in cell culture studies. Our present results demonstrate that at least 1 family member, bestrophin-3, is essential for a well-defined endogenous Ca(2+)-activated Cl(-) current in smooth muscles in the intact vascular wall.
Collapse
Affiliation(s)
- Vladimir V Matchkov
- Water and Salt Centre, Institute of Physiology and Biophysics, University of Aarhus, 8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kurreck J. The Role of Backbone Modifications in Oligonucleotide-Based Strategies. THERAPEUTIC OLIGONUCLEOTIDES 2008. [DOI: 10.1039/9781847558275-00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Jens Kurreck
- Institute for Chemistry and Biochemistry, Free University Berlin Thielallee 63 14195 Berlin Germany and Institute of Industrial Genetics, University of Stuttgart Allmandring 31 70569 Stuttgart Germany
| |
Collapse
|
18
|
Järve A, Müller J, Kim IH, Rohr K, MacLean C, Fricker G, Massing U, Eberle F, Dalpke A, Fischer R, Trendelenburg MF, Helm M. Surveillance of siRNA integrity by FRET imaging. Nucleic Acids Res 2007; 35:e124. [PMID: 17890733 PMCID: PMC2094076 DOI: 10.1093/nar/gkm694] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Techniques for investigation of exogenous small interfering RNA (siRNA) after penetration of the cell are of substantial interest to the development of efficient transfection methods as well as to potential medical formulations of siRNA. A FRET-based visualization method including the commonplace dye labels fluorescein and tetramethylrhodamin (TMR) on opposing strands of siRNA was found compatible with RNA interference (RNAi). Investigation of spectral properties of three labelled siRNAs with differential FRET efficiencies in the cuvette, including pH dependence and FRET efficiency in lipophilic environments, identified the ratio of red and green fluorescence (R/G-ratio) as a sensitive parameter, which reliably identifies samples containing >90% un-degraded siRNA. Spectral imaging of siRNAs microinjected into cells showed emission spectra indistinguishable from those measured in the cuvette. These were used to establish a calibration curve for assessing the degradation state of siRNA in volume elements inside cells. An algorithm, applied to fluorescence images recorded in standard green and red fluorescence channels, produces R/G-ratio images of high spatial resolution, identifying volume elements in the cell with high populations of intact siRNA with high fidelity. To demonstrate the usefulness of this technique, the movement of intact siRNA molecules are observed after introduction into the cytosol by microinjection, standard transfection and lipofection with liposomes.
Collapse
Affiliation(s)
- Anne Järve
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Julius Müller
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Il-Han Kim
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Karl Rohr
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Caroline MacLean
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Gert Fricker
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Ulrich Massing
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Florian Eberle
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Alexander Dalpke
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Roger Fischer
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Michael F. Trendelenburg
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
| | - Mark Helm
- Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Bioinformatics and Functional Genomics, IPMB, University of Heidelberg, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany, Department of Pharmaceutical Technology und Pharmacology, IPMB, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany, Tumor Biology Center, Department of Clinical Research, Breisacher Str. 117, D-79106 Freiburg, Germany, Department of Hygiene and Medical Microbiology, University of Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany and IPMB Master Program Molecular Biotechnology
- *To whom correspondence should be addressed. +49 6221 544879+49 6221 546430
| |
Collapse
|
19
|
Krützfeldt J, Kuwajima S, Braich R, Rajeev KG, Pena J, Tuschl T, Manoharan M, Stoffel M. Specificity, duplex degradation and subcellular localization of antagomirs. Nucleic Acids Res 2007; 35:2885-92. [PMID: 17439965 PMCID: PMC1888827 DOI: 10.1093/nar/gkm024] [Citation(s) in RCA: 347] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
MicroRNAs (miRNAs) are an abundant class of 20–23-nt long regulators of gene expression. The study of miRNA function in mice and potential therapeutic approaches largely depend on modified oligonucleotides. We recently demonstrated silencing miRNA function in mice using chemically modified and cholesterol-conjugated RNAs termed ‘antagomirs’. Here, we further characterize the properties and function of antagomirs in mice. We demonstrate that antagomirs harbor optimized phosphorothioate modifications, require >19-nt length for highest efficiency and can discriminate between single nucleotide mismatches of the targeted miRNA. Degradation of different chemically protected miRNA/antagomir duplexes in mouse livers and localization of antagomirs in a cytosolic compartment that is distinct from processing (P)-bodies indicates a degradation mechanism independent of the RNA interference (RNAi) pathway. Finally, we show that antagomirs, although incapable of silencing miRNAs in the central nervous system (CNS) when injected systemically, efficiently target miRNAs when injected locally into the mouse cortex. Our data further validate the effectiveness of antagomirs in vivo and should facilitate future studies to silence miRNAs for functional analysis and in clinically relevant settings.
Collapse
Affiliation(s)
- Jan Krützfeldt
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Satoru Kuwajima
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Ravi Braich
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Kallanthottathil G. Rajeev
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - John Pena
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Thomas Tuschl
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Muthiah Manoharan
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - Markus Stoffel
- Laboratory of Metabolic Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA, Alnylam Pharmaceuticals Inc., 300 3rd Street, Cambridge, MA 02142 USA and Howard Hughes Medical Institute, Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
- *To whom correspondence should be addressed. +41 44 633 4560+41 44 633 1051
| |
Collapse
|
20
|
Bajaj BG, Verma SC, Lan K, Cotter MA, Woodman ZL, Robertson ES. KSHV encoded LANA upregulates Pim-1 and is a substrate for its kinase activity. Virology 2006; 351:18-28. [PMID: 16647097 DOI: 10.1016/j.virol.2006.03.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 02/17/2006] [Accepted: 03/24/2006] [Indexed: 11/18/2022]
Abstract
Pim kinases are proto-oncogenes that are upregulated in a number of B cell cancers, including Epstein-Barr Virus (EBV) associated Burkitt's lymphoma. They have also been shown to be upregulated in Kaposi sarcoma-associated herpes virus (KSHV) infected primary B cells. Most cells in KSHV-associated tumors are latently infected and express only a small subset of viral genes, with KSHV latency associated nuclear antigen (LANA) being constitutively expressed. LANA regulates the transcription of a large number of cellular and viral genes. Here, we show that LANA upregulates transcription from the Pim-1 promoter (pPim-1) and map this activation to a region in the promoter located within the sequence (-681 to +37). We show that LANA expressing cells can proliferate faster and are better protected from drug induced apoptosis. Since transition through cell cycle check points and anti-apoptosis are functions associated with Pim-1, it is likely that higher Pim-1 expression in cells expressing LANA is responsible, at least in part, for this effect. A Pim-1 phosphorylation site was also identified within the amino-terminal domain of LANA. Using in vitro kinase assays, we confirmed that LANA was indeed a Pim-1 substrate, and the failure of Pim-1 to phosphorylate LANA mutated at SS205/6RR identified this site as the specific serine residues phosphorylated by Pim-1. This report provides valuable insight into yet another cellular signaling pathway subverted by KSHV LANA and suggests a contribution to KSHV related oncogenesis.
Collapse
Affiliation(s)
- Bharat G Bajaj
- Department of Microbiology and the Tumor Virology Program, Abramson Comprehensive Cancer Center, University of Pennsylvania Medical School, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
21
|
Zhang Y, Cristofaro P, Silbermann R, Pusch O, Boden D, Konkin T, Hovanesian V, Monfils PR, Resnick M, Moss SF, Ramratnam B. Engineering mucosal RNA interference in vivo. Mol Ther 2006; 14:336-42. [PMID: 16766229 DOI: 10.1016/j.ymthe.2006.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 04/04/2006] [Accepted: 04/04/2006] [Indexed: 01/22/2023] Open
Abstract
Mucosal surfaces serve as a gateway to disease. Here, we demonstrate that RNA interference can be used to manipulate mucosal gene expression in vivo. Using a murine model, we show that direct application of liposome-complexed siRNA mediates gene-specific silencing in cervicovaginal and rectal mucosa. A single vaginal or rectal administration of siRNA targeting hematopoietic or somatic cell gene products reduced corresponding mRNA levels by up to 90%. Using a murine model of inflammatory bowel disease, we found that the rectal application of siRNA targeting TNF-alpha led to relative mucosal resistance to experimental colitis. Liposomal siRNA formulations proved nontoxic, did not elicit a nonspecific interferon response, and provide a means for genetic engineering of mucosal surfaces in vivo.
Collapse
Affiliation(s)
- Yingjie Zhang
- Laboratory of Retrovirology, Division of Infectious Diseases, Department of Medicine, Brown Medical School, 55 Claverick Street, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shiu PKT, Zickler D, Raju NB, Ruprich-Robert G, Metzenberg RL. SAD-2 is required for meiotic silencing by unpaired DNA and perinuclear localization of SAD-1 RNA-directed RNA polymerase. Proc Natl Acad Sci U S A 2006; 103:2243-8. [PMID: 16461906 PMCID: PMC1413707 DOI: 10.1073/pnas.0508896103] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A gene unpaired during the meiotic homolog pairing stage in Neurospora generates a sequence-specific signal that silences the expression of all copies of that gene. This process is called Meiotic Silencing by Unpaired DNA (MSUD). Previously, we have shown that SAD-1, an RNA-directed RNA polymerase (RdRP), is required for MSUD. We isolated a second gene involved in this process, sad-2. Mutated Sad-2 (RIP) alleles, like those of Sad-1, are dominant and suppress MSUD. Crosses homozygous for Sad-2 are blocked at meiotic prophase. SAD-2 colocalizes with SAD-1 in the perinuclear region, where small interfering RNAs have been shown to reside in mammalian cells. A functional sad-2(+) gene is necessary for SAD-1 localization, but the converse is not true. The data suggest that SAD-2 may function to recruit SAD-1 to the perinuclear region, and that the proper localization of SAD-1 is important for its activity.
Collapse
Affiliation(s)
- Patrick K. T. Shiu
- *Division of Biological Sciences, University of Missouri, Columbia, MO 65211
- To whom correspondence may be addressed. E-mail:
or
| | - Denise Zickler
- Institut de Génétique et Microbiologie, Université Paris-Sud, 91405 Orsay Cedex, France
| | - Namboori B. Raju
- Department of Biological Sciences, Stanford University, Stanford, CA 94305; and
| | | | - Robert L. Metzenberg
- Department of Biology, California State University, Northridge, CA 91330
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
23
|
Leng Q, Scaria P, Zhu J, Ambulos N, Campbell P, Mixson AJ. Highly branched HK peptides are effective carriers of siRNA. J Gene Med 2005; 7:977-86. [PMID: 15772938 DOI: 10.1002/jgm.748] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Both viral and nonviral carriers have been used to carry small interfering RNA molecules (siRNA) to their cytosolic mRNA target. To date, few peptide carriers have been developed that have proved effective for siRNA delivery. Our previous branched carriers composed of histidine and lysine were useful for transfection of plasmids. In this study, we determined if these and more highly branched HK polymers were effective carriers of siRNA. METHODS Several branched polymers were synthesized on a Ranin Voyager synthesizer. These polymers were then screened for their ability to transfer siRNA into SVR-bag4 cells, MDA-MB-435 cells, and C6 cells. After one polymer, H3K8b, was identified as an effective carrier of siRNA, additional polymers were synthesized to determine the essential domains for siRNA transport. The size/zeta-potential of HK : siRNA complexes were measured with the N4 submicron particle size analyzer and the Delsa 440 SX zeta-potential analyzer, respectively. Toxicity of the highly branched polymers in complex with siRNA was investigated by flow cytometry. RESULTS In an endothelial cell line (SVR-bag4) that stably expressed beta-galactosidase (beta-gal), an siRNA in complex with the H3K8b polymer inhibited beta-gal expression by more than 80%. In contrast, the polymer H2K4b, which was an effective carrier of plasmids, was not an efficient carrier of siRNA. The size and surface charge did not distinguish effective from ineffective HK carriers of siRNA. By modifying H3K8b, we then determined what properties of H3K8b augmented siRNA delivery. The histidine-rich domain and the length of the terminal arms of H3K8 were important for siRNA delivery. The modestly more effective analog of H3K8b containing an integrin ligand, H3K8b(+RGD), was able to inhibit markedly intracellular beta-gal expression. Furthermore, we determined that H3K8b(+RGD) in complex with a luciferase-targeting siRNA inhibited luciferase expression in MDA-MB-435 cells. At its optimal concentration for inhibiting its target, H3K8b(+RGD) : siRNA complex had minimal toxicity. In contrast, carriers of siRNA such as Oligofectamine and Lipofectamine 2000 were significantly more toxic. CONCLUSIONS Both the degree of complexity and the sequence specificity are important factors to be considered for developing the HK carrier of siRNA. In particular, we found that certain branched HK polymers (H3K8b, H3K8b(+RGD), and similar structural analogs) with eight terminal branches and a histidine-rich domain were effective carriers of siRNA.
Collapse
Affiliation(s)
- Qixin Leng
- Department of Pathology, University of Maryland Baltimore, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
24
|
Schubert S, Grünweller A, Erdmann VA, Kurreck J. Local RNA target structure influences siRNA efficacy: systematic analysis of intentionally designed binding regions. J Mol Biol 2005; 348:883-93. [PMID: 15843020 DOI: 10.1016/j.jmb.2005.03.011] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 03/01/2005] [Accepted: 03/02/2005] [Indexed: 11/26/2022]
Abstract
Contradictory reports in the literature have emphasised either the sequence of small interfering RNAs (siRNA) or the structure of their target molecules to be the major determinant of the efficiency of RNA interference (RNAi) approaches. In the present study, we analyse systematically the contributions of these parameters to siRNA activity by using deliberately designed mRNA constructs. The siRNA target sites were included in well-defined structural elements rendering them either highly accessible or completely involved in stable base-pairing. Furthermore, complementary sequence elements and various hairpins with different stem lengths and designs were used as target sites. Only one of the strands of the siRNA duplex was found to be capable of silencing via its respective target site, indicating that thermodynamic characteristics intrinsic to the siRNA strands are a basic determinant of siRNA activity. A significant obstruction of gene silencing by the same siRNA, however, was observed to be caused by structural features of the substrate RNA. Bioinformatic analysis of the mRNA structures suggests a direct correlation between the extent of gene-knockdown and the local free energy in the target region. Our findings indicate that, although a favourable siRNA sequence is a necessary prerequisite for efficient RNAi, complex target structures may limit the applicability even of carefully chosen siRNAs.
Collapse
Affiliation(s)
- Steffen Schubert
- Free University Berlin, Institute for Chemistry (Biochemistry), Thielallee 63, 14195 Berlin, Germany
| | | | | | | |
Collapse
|