1
|
Shiohama Y, Fujita R, Sonokawa M, Hisano M, Kotake Y, Krstic-Demonacos M, Demonacos C, Kashiwazaki G, Kitayama T, Fujii M. Elimination of Off-Target Effect by Chemical Modification of 5′-End of Small Interfering RNA. Nucleic Acid Ther 2022; 32:438-447. [DOI: 10.1089/nat.2021.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yasuo Shiohama
- Environmental and Biological Information Group, Tropical Biosphere Research Centre, University of the Ryukyus, Nishihara, Japan
| | - Ryosuke Fujita
- Department of Biological & Environmental Chemistry, School of Humanity Oriented Science and Technology, Kindai University, Iizuka, Japan
| | - Maika Sonokawa
- Department of Biological & Environmental Chemistry, School of Humanity Oriented Science and Technology, Kindai University, Iizuka, Japan
| | - Masaaki Hisano
- Department of Biological & Environmental Chemistry, School of Humanity Oriented Science and Technology, Kindai University, Iizuka, Japan
| | - Yojiro Kotake
- Department of Biological & Environmental Chemistry, School of Humanity Oriented Science and Technology, Kindai University, Iizuka, Japan
| | - Marija Krstic-Demonacos
- School of Science, Engineering and Environment, University of Salford, Salford, United Kingdom
| | - Constantinos Demonacos
- Division of Pharmacy and Optometry, Faculty of Biology Medicine and Health, School of Health Science, University of Manchester, Manchester, United Kingdom
| | - Gengo Kashiwazaki
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Takashi Kitayama
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Masayuki Fujii
- Department of Biological & Environmental Chemistry, School of Humanity Oriented Science and Technology, Kindai University, Iizuka, Japan
| |
Collapse
|
2
|
Yang C, Ma D, Lu L, Yang X, Xi Z. Synthesis of KUE-siRNA Conjugates for Prostate Cancer Cell-Targeted Gene Silencing. Chembiochem 2021; 22:2888-2895. [PMID: 34263529 DOI: 10.1002/cbic.202100243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Indexed: 11/06/2022]
Abstract
The delivery of siRNAs to selectively target cells poses a great challenge in RNAi-based cancer therapy. The lack of suitable cell-targeting methods seriously restricts the advance in delivering siRNAs to extrahepatic tissues. Based on prostate-specific membrane antigen (PSMA)-targeting ligands, we have synthesized a series of lysine-urea-glutamate (KUE)-siRNA conjugates and verified their effective cell uptake and gene silencing properties in prostate cancers. The results indicated that the KUE-siRNA conjugates could selectively enter PSMA+ LNCaP cells, eventually down-regulating STAT3 expression. Based on post-synthesis modification and receptor-mediated endocytosis, this strategy of constructing ligand-siRNA conjugates might provide a general method of siRNA delivery for cell-targeted gene silencing.
Collapse
Affiliation(s)
- Chao Yang
- Department of Chemical Biology, State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Nankai University, Tianjin, 300071, P. R. China
| | - Dejun Ma
- Department of Chemical Biology, State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Nankai University, Tianjin, 300071, P. R. China
| | - Liqing Lu
- Department of Chemical Biology, State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Nankai University, Tianjin, 300071, P. R. China
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, P. R. China
| | - Zhen Xi
- Department of Chemical Biology, State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
3
|
Kubo T, Nishimura Y, Sato Y, Yanagihara K, Seyama T. Sixteen Different Types of Lipid-Conjugated siRNAs Containing Saturated and Unsaturated Fatty Acids and Exhibiting Enhanced RNAi Potency. ACS Chem Biol 2021; 16:150-164. [PMID: 33346648 DOI: 10.1021/acschembio.0c00847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
SiRNAs are strong gene-silencing agents that function in a target sequence-specific manner. Although siRNAs might one day be used in therapy for intractable diseases such as cancers, a number of problems with siRNAs must first be overcome. In this study, we developed 16 different types of lipid-conjugated siRNAs (lipid-siRNAs) that could effectively inhibit the expression of target genes. We determined the hybridization properties, cellular uptake efficacies, and RNAi potencies of the resulting lipid-siRNAs. The lipid-siRNAs exhibited a mild interaction with Lipofectamine RNAiMAX (LFRNAi) as a transfection reagent, and a high membrane permeability was observed in all lipid-siRNAs-LFRNAi complexes; the conjugate siRNAs composed of 16-18 carbon chains as fatty acids showed an especially good cellular uptake efficacy. The in vitro RNAi effect of lipid-siRNAs targeted to a β-catenin gene exhibited a strong RNAi potency compared with those of unmodified siRNAs. In particular, the conjugate siRNAs composed of 16-18 carbon chains as fatty acids showed excellent RNAi potencies with prolonged effectivities. Interestingly, the RNAi potencies of conjugate siRNAs containing 18 carbon chains with a trans-form (elaidic acid and trans-vaccenic acid) were inferior to those of the carbon chains with a cis-form (oleic acid and cis-vaccenic acid). These lipid-siRNAs can solve the many problems hindering the clinical application of siRNAs.
Collapse
Affiliation(s)
| | | | | | - Kazuyoshi Yanagihara
- Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | | |
Collapse
|
4
|
Cheng L, Ma D, Lu L, Ouyang D, Xi Z. Building Customizable Multisite‐Targeting c‐Myc shRNA Array into Branch‐PCR‐Constructed DNA Nanovectors for Enhanced Tumor Cell Suppression. ChemistrySelect 2020. [DOI: 10.1002/slct.202002609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Longhuai Cheng
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Dejun Ma
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Liqing Lu
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Di Ouyang
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Zhen Xi
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| |
Collapse
|
5
|
Dellafiore M, Aviñó A, Alagia A, Montserrat JM, Iribarren AM, Eritja R. siRNA Modified with 2′-Deoxy-2′-C
-methylpyrimidine Nucleosides. Chembiochem 2018; 19:1409-1413. [DOI: 10.1002/cbic.201800077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Indexed: 01/23/2023]
Affiliation(s)
- María Dellafiore
- INGEBI (CONICET); Vuelta de Obligado 2490 -1428 Buenos Aires Argentina
| | - Anna Aviñó
- Institute for Advanced Chemistry of Catalonia (IQAC); Spanish Council for Scientific Research (CSIC); Jordi Girona 18-26 08034 Barcelona Spain
- NetworkingCenter on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Av. Monforte de Lemos 3-5. Pabellón 11. Planta 0 Madrid 28029 Spain
| | - Adele Alagia
- Institute for Advanced Chemistry of Catalonia (IQAC); Spanish Council for Scientific Research (CSIC); Jordi Girona 18-26 08034 Barcelona Spain
- NetworkingCenter on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Av. Monforte de Lemos 3-5. Pabellón 11. Planta 0 Madrid 28029 Spain
| | - Javier M. Montserrat
- Instituto de Ciencias; Universidad Nacional de General Sarmiento; J. M. Gutiérrez 1150 Los Polvorines Prov. Buenos Aires B1613GSX Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Godoy Cruz 2290 Buenos Aires C1425FQB Argentina
| | - Adolfo M. Iribarren
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET); Godoy Cruz 2290 Buenos Aires C1425FQB Argentina
- Laboratorio de Biotransformaciones; Universidad Nacional de Quilmes; Roque Saenz Peña352 1876 Bernal Prov Buenos Aires Argentina
| | - Ramon Eritja
- Institute for Advanced Chemistry of Catalonia (IQAC); Spanish Council for Scientific Research (CSIC); Jordi Girona 18-26 08034 Barcelona Spain
- NetworkingCenter on Bioengineering; Biomaterials and Nanomedicine (CIBER-BBN); Av. Monforte de Lemos 3-5. Pabellón 11. Planta 0 Madrid 28029 Spain
| |
Collapse
|
6
|
Kwon OS, Kwon SJ, Kim JS, Lee G, Maeng HJ, Lee J, Hwang GS, Cha HJ, Chun KH. Designing Tyrosinase siRNAs by Multiple Prediction Algorithms and Evaluation of Their Anti-Melanogenic Effects. Biomol Ther (Seoul) 2018; 26:282-289. [PMID: 29223142 PMCID: PMC5933895 DOI: 10.4062/biomolther.2017.115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 11/05/2022] Open
Abstract
Melanin is a pigment produced from tyrosine in melanocytes. Although melanin has a protective role against UVB radiation-induced damage, it is also associated with the development of melanoma and darker skin tone. Tyrosinase is a key enzyme in melanin synthesis, which regulates the rate-limiting step during conversion of tyrosine into DOPA and dopaquinone. To develop effective RNA interference therapeutics, we designed a melanin siRNA pool by applying multiple prediction programs to reduce human tyrosinase levels. First, 272 siRNAs passed the target accessibility evaluation using the RNAxs program. Then we selected 34 siRNA sequences with ΔG ≥-34.6 kcal/mol, i-Score value ≥65, and siRNA scales score ≤30. siRNAs were designed as 19-bp RNA duplexes with an asymmetric 3' overhang at the 3' end of the antisense strand. We tested if these siRNAs effectively reduced tyrosinase gene expression using qRT-PCR and found that 17 siRNA sequences were more effective than commercially available siRNA. Three siRNAs further tested showed an effective visual color change in MNT-1 human cells without cytotoxic effects, indicating these sequences are anti-melanogenic. Our study revealed that human tyrosinase siRNAs could be efficiently designed using multiple prediction algorithms.
Collapse
Affiliation(s)
- Ok-Seon Kwon
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Soo-Jung Kwon
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Jin Sang Kim
- Leaders Cosmetics Co., Ltd., Anseong 17599, Republic of Korea
| | - Gunbong Lee
- Leaders Cosmetics Co., Ltd., Anseong 17599, Republic of Korea
| | - Han-Joo Maeng
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwi Seo Hwang
- Laboratory of Cell Differentiation Research, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Hyuk-Jin Cha
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
7
|
Morihiro K, Kasahara Y, Obika S. Biological applications of xeno nucleic acids. MOLECULAR BIOSYSTEMS 2017; 13:235-245. [PMID: 27827481 DOI: 10.1039/c6mb00538a] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Xeno nucleic acids (XNAs) are a group of chemically modified nucleic acid analogues that have been applied to various biological technologies such as antisense oligonucleotides, siRNAs and aptamers.
Collapse
Affiliation(s)
- Kunihiko Morihiro
- National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan and Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Yuuya Kasahara
- National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan and Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Satoshi Obika
- National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan and Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
8
|
Greco CT, Andrechak JC, Epps TH, Sullivan MO. Anionic Polymer and Quantum Dot Excipients to Facilitate siRNA Release and Self-Reporting of Disassembly in Stimuli-Responsive Nanocarrier Formulations. Biomacromolecules 2017; 18:1814-1824. [PMID: 28441861 PMCID: PMC5672795 DOI: 10.1021/acs.biomac.7b00265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The incorporation of anionic excipients into polyplexes is a promising strategy for modulating siRNA binding versus release and integrating diagnostic capabilities; however, specific design criteria and structure-function relationships are needed to facilitate the development of nanocarrier-based theranostics. Herein, we incorporated poly(acrylic acid) (PAA) and quantum dot (QD) excipients into photolabile siRNA polyplexes to increase gene silencing efficiencies by up to 100% and enable self-reporting of nanocarrier disassembly. Our systematic approach identified the functional relationships between gene silencing and key parameters such as excipient loading fractions and molecular weights that facilitated the establishment of design rules for optimization of nanocarrier efficacy. For example, we found that PAA molecular weights ∼10-20× greater than that of the coencapsulated siRNA exhibited the most efficient release and silencing. Furthermore, siRNA release assays and RNAi modeling allowed us to generate a PAA "heat map" that predicted gene silencing a priori as a function of PAA molecular weight and loading fraction. QDs further promoted selective siRNA release and provided visual as well as Förster resonance energy transfer (FRET)-based monitoring of the dynamic changes in nanostructure in situ. Moreover, even with the addition of anionic components, our formulations exhibited substantially improved stability and shelf life relative to typical formulations, with complete stability after a week of storage and full activity in the presence of serum. Taken together, this study enabled synergistic improvements in siRNA release and diagnostic capabilities, along with the development of mechanistic insights that are critical for advancing the translation of nucleic acid theranostics into the clinic.
Collapse
Affiliation(s)
- Chad T Greco
- Department of Chemical and Biomolecular Engineering and §Department of Materials Science and Engineering, University of Delaware , Newark, Delaware 19716, United States
| | - Jason C Andrechak
- Department of Chemical and Biomolecular Engineering and §Department of Materials Science and Engineering, University of Delaware , Newark, Delaware 19716, United States
| | - Thomas H Epps
- Department of Chemical and Biomolecular Engineering and §Department of Materials Science and Engineering, University of Delaware , Newark, Delaware 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering and §Department of Materials Science and Engineering, University of Delaware , Newark, Delaware 19716, United States
| |
Collapse
|
9
|
Paces J, Nic M, Novotny T, Svoboda P. Literature review of baseline information to support the risk assessment of RNAi‐based GM plants. ACTA ACUST UNITED AC 2017. [PMCID: PMC7163844 DOI: 10.2903/sp.efsa.2017.en-1246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jan Paces
- Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic (IMG)
| | | | | | - Petr Svoboda
- Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic (IMG)
| |
Collapse
|
10
|
Selvam C, Mutisya D, Prakash S, Ranganna K, Thilagavathi R. Therapeutic potential of chemically modified siRNA: Recent trends. Chem Biol Drug Des 2017; 90:665-678. [PMID: 28378934 DOI: 10.1111/cbdd.12993] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/17/2022]
Abstract
Small interfering RNAs (siRNAs) are one of the valuable tools to investigate the functions of genes and are also used for gene silencing. It has a wide scope in drug discovery through in vivo target validation. siRNA therapeutics are not optimal drug-like molecules due to poor bioavailability and immunogenic and off-target effects. To overcome the challenges associated with siRNA therapeutics, identification of appropriate chemical modifications that improves the stability, specificity and potency of siRNA is essential. This review focuses on the various chemical modifications and their implications in siRNA therapy.
Collapse
Affiliation(s)
- Chelliah Selvam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Daniel Mutisya
- Department of Science and Mathematics, Albany State University, Albany, GA, USA
| | - Sandhya Prakash
- Department of Biotechnology, Faculty of Engineering, Karpagam University, Coimbatore, India
| | - Kasturi Ranganna
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Ramasamy Thilagavathi
- Department of Biotechnology, Faculty of Engineering, Karpagam University, Coimbatore, India
| |
Collapse
|
11
|
Scherman D, Rousseau A, Bigey P, Escriou V. Genetic pharmacology: progresses in siRNA delivery and therapeutic applications. Gene Ther 2017; 24:151-156. [PMID: 28121307 DOI: 10.1038/gt.2017.6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/03/2017] [Indexed: 12/16/2022]
Abstract
In the RNA interference process, the catalytic degradation of an endogenous mRNA results from the Watson-Crick complementary recognition by either a small silencing synthetic double-stranded ribonucleotide (siRNA) or by a small hairpin RNA (shRNA) produced in the cell by transcription from a DNA template. This interference process ideally results in an exquisitely specific mRNA suppression. The present review is dedicated to siRNAs. It describes the mechanism of RNA silencing and the main siRNA delivery techniques, with a focus on siRNA self-complexing to cationic lipids to form nanoparticles, which are called lipoplexes. The addition to lipoplexes of an anionic polymer leads to the ternary formulation APIRL (Anionic-Polymer-Interfering-RNA-Lipoplexes) with increased in vivo stability and biological efficacy. In terms of clinical development, the review focuses on therapeutic applications by intravenous delivery to the liver and inflammatory joints, and to localized siRNA delivery to the ocular sphere.
Collapse
Affiliation(s)
- D Scherman
- CNRS, UTCBS UMR 8258, Paris, France.,Université Paris Descartes, Faculty of Pharmacy, Sorbonne-Paris-Cité, UTCBS, Paris, France.,Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,INSERM, UTCBS U 1022, F-75006 Paris, France, 4,avenue de l'Observatoire, Paris, France
| | - A Rousseau
- CNRS, UTCBS UMR 8258, Paris, France.,Université Paris Descartes, Faculty of Pharmacy, Sorbonne-Paris-Cité, UTCBS, Paris, France.,Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,INSERM, UTCBS U 1022, F-75006 Paris, France, 4,avenue de l'Observatoire, Paris, France
| | - P Bigey
- CNRS, UTCBS UMR 8258, Paris, France.,Université Paris Descartes, Faculty of Pharmacy, Sorbonne-Paris-Cité, UTCBS, Paris, France.,Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,INSERM, UTCBS U 1022, F-75006 Paris, France, 4,avenue de l'Observatoire, Paris, France
| | - V Escriou
- CNRS, UTCBS UMR 8258, Paris, France.,Université Paris Descartes, Faculty of Pharmacy, Sorbonne-Paris-Cité, UTCBS, Paris, France.,Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France.,INSERM, UTCBS U 1022, F-75006 Paris, France, 4,avenue de l'Observatoire, Paris, France
| |
Collapse
|
12
|
Bradford BJ, Cooper CA, Tizard ML, Doran TJ, Hinton TM. RNA interference-based technology: what role in animal agriculture? ANIMAL PRODUCTION SCIENCE 2017. [DOI: 10.1071/an15437] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal agriculture faces a broad array of challenges, ranging from disease threats to adverse environmental conditions, while attempting to increase productivity using fewer resources. RNA interference (RNAi) is a biological phenomenon with the potential to provide novel solutions to some of these challenges. Discovered just 20 years ago, the mechanisms underlying RNAi are now well described in plants and animals. Intracellular double-stranded RNA triggers a conserved response that leads to cleavage and degradation of complementary mRNA strands, thereby preventing production of the corresponding protein product. RNAi can be naturally induced by expression of endogenous microRNA, which are critical in the regulation of protein synthesis, providing a mechanism for rapid adaptation of physiological function. This endogenous pathway can be co-opted for targeted RNAi either through delivery of exogenous small interfering RNA (siRNA) into target cells or by transgenic expression of short hairpin RNA (shRNA). Potentially valuable RNAi targets for livestock include endogenous genes such as developmental regulators, transcripts involved in adaptations to new physiological states, immune response mediators, and also exogenous genes such as those encoded by viruses. RNAi approaches have shown promise in cell culture and rodent models as well as some livestock studies, but technical and market barriers still need to be addressed before commercial applications of RNAi in animal agriculture can be realised. Key challenges for exogenous delivery of siRNA include appropriate formulation for physical delivery, internal transport and eventual cellular uptake of the siRNA; additionally, rigorous safety and residue studies in target species will be necessary for siRNA delivery nanoparticles currently under evaluation. However, genomic incorporation of shRNA can overcome these issues, but optimal promoters to drive shRNA expression are needed, and genetic engineering may attract more resistance from consumers than the use of exogenous siRNA. Despite these hurdles, the convergence of greater understanding of RNAi mechanisms, detailed descriptions of regulatory processes in animal development and disease, and breakthroughs in synthetic chemistry and genome engineering has created exciting possibilities for using RNAi to enhance the sustainability of animal agriculture.
Collapse
|
13
|
Nothisen M, Bagilet J, Behr JP, Remy JS, Kotera M. Structure Tuning of Cationic Oligospermine-siRNA Conjugates for Carrier-Free Gene Silencing. Mol Pharm 2016; 13:2718-28. [PMID: 27398779 DOI: 10.1021/acs.molpharmaceut.6b00309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oligospermine-siRNA conjugates are able to induce efficient luciferase gene silencing upon carrier-free transfection. These conjugates are readily accessible by a versatile automated chemistry that we developed using a DMT-spermine phosphoramidite reagent. In this article, we used this chemistry to study a wide range of structural modifications of the oligospermine-siRNA conjugates, i.e., variation of conjugate positions and introduction of chemical modifications to increase nuclease resistance. At first we examined gene silencing activity of a series of siRNA-tris(spermine) conjugates with and without chemical modifications in standard carrier assisted conditions. The three spermine units attached at one of the two ends of the sense strand or at the 3'-end of the antisense strand are compatible with gene silencing activity whereas attachment of spermine units at the 5'-end of the antisense strand abolished the activity. 2'-O-Methylated nucleotides introduced in the sense strand are compatible while not in the antisense strand. Thiophosphate links could be used without activity loss at the 3'-end of both strands and at the 5'-end of the sense strand to conjugate oligospermine. Consequently a series of oligospermine-siRNA conjugates containing 15 to 45 spermines units in various configurations were chosen, prepared, and examined in carrier-free conditions. Attachment of 30 spermine units singly at the 5'-end of the sense strand provides the most potent carrier-free siRNA. Longevity of luciferase gene silencing was studied using oligospermine-siRNA conjugates. Five day long efficiency with more than 80% gene expression knockdown was observed upon transfection without vector. Oligospermine-siRNA conjugates targeting cell-constitutive natural lamin A/C gene were prepared. Efficient gene silencing was observed upon carrier-free transfection of siRNA conjugates containing 20 or 30 spermine residues grafted at the 5'-end of the sense strand.
Collapse
Affiliation(s)
- Marc Nothisen
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jérémy Bagilet
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jean-Paul Behr
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jean-Serge Remy
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Mitsuharu Kotera
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| |
Collapse
|
14
|
Bienk K, Hvam ML, Pakula MM, Dagnæs-Hansen F, Wengel J, Malle BM, Kragh-Hansen U, Cameron J, Bukrinski JT, Howard KA. An albumin-mediated cholesterol design-based strategy for tuning siRNA pharmacokinetics and gene silencing. J Control Release 2016; 232:143-51. [DOI: 10.1016/j.jconrel.2016.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/30/2016] [Accepted: 04/06/2016] [Indexed: 11/30/2022]
|
15
|
Lozac'h F, Christensen J, Faller T, van de Kerkhof E, Krauser J, Garnier M, Litherland K, Catoire A, Natt F, Hunziker J, Swart P. ADME studies of [5-(3)H]-2'-O-methyluridine nucleoside in mice: a building block in siRNA therapeutics. Pharmacol Res Perspect 2016; 4:e00209. [PMID: 26977299 PMCID: PMC4777266 DOI: 10.1002/prp2.209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022] Open
Abstract
The chemical modification 2′‐O‐methyl of nucleosides is often used to increase siRNA stability towards nuclease activities. However, the metabolic fate of modified nucleosides remains unclear. Therefore, the aim of this study was to determine the mass balance, pharmacokinetic, and absorption, distribution, metabolism, and excretion (ADME)‐properties of tritium‐labeled 2′‐O‐methyluridine, following a single intravenous dose to male CD‐1 mice. The single intravenous administration of [5‐3H]‐2′‐O‐methyluridine was well tolerated in mice. Radioactivity was rapidly and widely distributed throughout the body and remained detectable in all tissues investigated throughout the observation period of 48 h. After an initial rapid decline, blood concentrations of total radiolabeled components declined at a much slower rate. [3H]‐2′‐O‐Methyluridine represented a minor component of the radioactivity in plasma (5.89% of [3H]‐AUC0‐48 h). Three [3H]‐2′‐O‐methyluridine metabolites namely uridine (M1), cytidine (M2), and uracil (M3) were the major circulating components representing 32.8%, 8.11%, and 23.6% of radioactivity area under the curve, respectively. The highest concentrations of total radiolabeled components and exposures were observed in kidney, spleen, pineal body, and lymph nodes. The mass balance, which is the sum of external recovery of radioactivity in excreta and remaining radioactivity in carcass and cage wash, was complete. Renal excretion accounted for about 52.7% of the dose with direct renal excretion of the parent in combination with metabolism to the endogenous compounds cytidine, uracil, cytosine, and cytidine.
Collapse
Affiliation(s)
- Frederic Lozac'h
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Jesper Christensen
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Thomas Faller
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Esther van de Kerkhof
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Joel Krauser
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Maxime Garnier
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Karine Litherland
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Alexandre Catoire
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Francois Natt
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Jurg Hunziker
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| | - Piet Swart
- Novartis Institutes for Biomedical Research, Drug Metabolism and Pharmacokinetics Novartis Pharma AG Fabrikstrasse 14, 1.17 CH-4002 Basel Switzerland
| |
Collapse
|
16
|
siRNAmod: A database of experimentally validated chemically modified siRNAs. Sci Rep 2016; 6:20031. [PMID: 26818131 PMCID: PMC4730238 DOI: 10.1038/srep20031] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/21/2015] [Indexed: 11/21/2022] Open
Abstract
Small interfering RNA (siRNA) technology has vast potential for functional genomics and development of therapeutics. However, it faces many obstacles predominantly instability of siRNAs due to nuclease digestion and subsequently biologically short half-life. Chemical modifications in siRNAs provide means to overcome these shortcomings and improve their stability and potency. Despite enormous utility bioinformatics resource of these chemically modified siRNAs (cm-siRNAs) is lacking. Therefore, we have developed siRNAmod, a specialized databank for chemically modified siRNAs. Currently, our repository contains a total of 4894 chemically modified-siRNA sequences, comprising 128 unique chemical modifications on different positions with various permutations and combinations. It incorporates important information on siRNA sequence, chemical modification, their number and respective position, structure, simplified molecular input line entry system canonical (SMILES), efficacy of modified siRNA, target gene, cell line, experimental methods, reference etc. It is developed and hosted using Linux Apache MySQL PHP (LAMP) software bundle. Standard user-friendly browse, search facility and analysis tools are also integrated. It would assist in understanding the effect of chemical modifications and further development of stable and efficacious siRNAs for research as well as therapeutics. siRNAmod is freely available at: http://crdd.osdd.net/servers/sirnamod.
Collapse
|
17
|
Leo L, Yu W, Baas PW. Using siRNA to study microtubule-related proteins in cultured neurons. Methods Cell Biol 2016; 131:163-76. [DOI: 10.1016/bs.mcb.2015.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Nawrot B, Michalak O, Mikołajczyk B, Stec WJ. Acyclic analogs of nucleosides based on tris(hydroxymethyl)phosphine oxide: synthesis and incorporation into short DNA oligomers. HETEROCYCL COMMUN 2015. [DOI: 10.1515/hc-2015-0173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractTris-(hydroxymethyl)phosphine oxide (THPO) to a certain extent resembles a part of 2′-deoxyribofuranose, although it exists in an acyclic form only and the oxygen atom at the THPO phosphorus center provides additional hydration site or acceptor of hydrogen bonds. After proper protection of hydroxyl groups, THPO was functionalized with nucleobases and converted into phosphoramidite monomers suitable for incorporation into growing oligonucleotide chains within the solid phase synthesis protocol. The resultant THPO-DNA analogs show reduced affinity to complementary DNA strands, and are resistant towards snake venom and calf spleen exonucleases.
Collapse
Affiliation(s)
- Barbara Nawrot
- 1Department of Bioorganic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Olga Michalak
- 2Pharmaceutical Research Institute, 8 Rydygiera Street, 01-793 Warsaw, Poland
| | - Barbara Mikołajczyk
- 3Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| | - Wojciech J. Stec
- 3Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
19
|
Kong LL, Zhuang XM, Yang HY, Yuan M, Xu L, Li H. Inhibition of P-glycoprotein Gene Expression and Function Enhances Triptolide-induced Hepatotoxicity in Mice. Sci Rep 2015; 5:11747. [PMID: 26134275 PMCID: PMC4488747 DOI: 10.1038/srep11747] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/04/2015] [Indexed: 11/29/2022] Open
Abstract
Triptolide (TP) is the major active principle of Tripterygium wilfordii Hook f. and very effective in treatment of autoimmune diseases. However, TP induced hepatotoxicity limited its clinical applications. Our previous study found that TP was a substrate of P-glycoprotein and its hepatobiliary clearance was markedly affected by P-gp modulation in sandwich-cultured rat hepatocytes. In this study, small interfering RNA (siRNA) and specific inhibitor tariquidar were used to investigate the impact of P-gp down regulation on TP-induced hepatotoxicity. The results showed that when the function of P-gp was inhibited by mdr1a-1 siRNA or tariquidar, the systemic and hepatic exposures of TP were significantly increased. The aggravated hepatotoxicity was evidenced with the remarkably lifted levels of serum biomarkers (ALT and AST) and pathological changes in liver. The other toxicological indicators (MDA, SOD and Bcl-2/Bax) were also significantly changed by P-gp inhibition. The data analysis showed that the increase of TP exposure in mice was quantitatively correlated to the enhanced hepatotoxicity, and the hepatic exposure was more relevant to the toxicity. P-gp mediated clearance played a significant role in TP detoxification. The risk of herb-drug interaction likely occurs when TP is concomitant with P-gp inhibitors or substrates in clinic.
Collapse
Affiliation(s)
- Ling-Lei Kong
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xiao-Mei Zhuang
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hai-Ying Yang
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Mei Yuan
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liang Xu
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hua Li
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
20
|
Marimani MD, Ely A, Buff MCR, Bernhardt S, Engels JW, Scherman D, Escriou V, Arbuthnot P. Inhibition of replication of hepatitis B virus in transgenic mice following administration of hepatotropic lipoplexes containing guanidinopropyl-modified siRNAs. J Control Release 2015; 209:198-206. [PMID: 25937322 DOI: 10.1016/j.jconrel.2015.04.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Abstract
Chronic infection with hepatitis B virus (HBV) occurs commonly and complications that arise from persistence of the virus are associated with high mortality. Available licensed drugs have modest curative efficacy and advancing new therapeutic strategies to eliminate the virus is therefore a priority. HBV is susceptible to inactivation by exogenous gene silencers that harness RNA interference (RNAi) and the approach has therapeutic potential. To advance RNAi-based treatment for HBV infection, use in vivo of hepatotropic lipoplexes containing siRNAs with guanidinopropyl (GP) modifications is reported here. Lipoplexes contained polyglutamate, which has previously been shown to facilitate formulation and improve efficiency of the non-viral vectors. GP moieties were included in a previously described anti-HBV siRNA that effectively targeted the conserved viral X sequence. Particles had physical properties that were suitable for use in vivo: average diameter was approximately 50-200 nm and surface charge (zeta potential) was +65 mV. Efficient hepatotropic delivery of labeled siRNA was observed following systemic intravenous injection of the particles into HBV transgenic mice. Good inhibition of markers of viral replication was observed without evidence of toxicity. Efficacy of the GP-modified siRNAs was significantly more durable and formulations made up with chemically modified siRNAs were less immunostimulatory. An RNAi-mediated mechanism was confirmed by demonstrating that HBV mRNA cleavage occurred in vivo at the intended target site. Collectively these data indicate that GP-modified siRNAs formulated in anionic polymer-containing lipoplexes are effective silencers of HBV replication in vivo and have therapeutic potential.
Collapse
Affiliation(s)
- Musa D Marimani
- Wits/SA MRC Antiviral Gene Therapy Research Unit, School of Pathology, Health Sciences Faculty, University of the Witwatersrand, Johannesburg, Private Bag 3, Wits 2050, South Africa
| | - Abdullah Ely
- Wits/SA MRC Antiviral Gene Therapy Research Unit, School of Pathology, Health Sciences Faculty, University of the Witwatersrand, Johannesburg, Private Bag 3, Wits 2050, South Africa
| | - Maximilian C R Buff
- Goethe-University, Institute of Organic Chemistry & Chemical Biology, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Stefan Bernhardt
- Goethe-University, Institute of Organic Chemistry & Chemical Biology, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Joachim W Engels
- Goethe-University, Institute of Organic Chemistry & Chemical Biology, Max-von-Laue-Str. 7, 60438 Frankfurt am Main, Germany
| | - Daniel Scherman
- UTCBS, CNRS UMR8258, INSERM U1022, Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Virginie Escriou
- UTCBS, CNRS UMR8258, INSERM U1022, Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Patrick Arbuthnot
- Wits/SA MRC Antiviral Gene Therapy Research Unit, School of Pathology, Health Sciences Faculty, University of the Witwatersrand, Johannesburg, Private Bag 3, Wits 2050, South Africa.
| |
Collapse
|
21
|
Fan M, Zhang Y, Huang Z, Liu J, Guo X, Zhang H, Luo H. Optimizations of siRNA design for the activation of gene transcription by targeting the TATA-box motif. PLoS One 2014; 9:e108253. [PMID: 25250958 PMCID: PMC4176967 DOI: 10.1371/journal.pone.0108253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/20/2014] [Indexed: 12/24/2022] Open
Abstract
Small interfering RNAs (siRNAs) are widely used to repress gene expression by targeting mRNAs. Some reports reveal that siRNAs can also activate or inhibit gene expression through targeting the gene promoters. Our group has found that microRNAs (miRNAs) could activate gene transcription via interaction with the TATA-box motif in gene promoters. To investigate whether siRNA targeting the same region could upregulate the promoter activity, we test the activating efficiency of siRNAs targeting the TATA-box motif of 16 genes and perform a systematic analysis to identify the common features of the functional siRNAs for effective activation of gene promoters. Further, we try various modifications to improve the activating efficiency of siRNAs and find that it is quite useful to design the promoter-targeting activating siRNA by following several rules such as (a) complementary to the TATA-box-centered region; (b) UA usage at the first two bases of the antisense strand; (c) twenty-three nucleotides (nts) in length; (d) 2'-O-Methyl (2'-OMe) modification at the 3' terminus of the antisense strand; (e) avoiding mismatches at the 3' end of the antisense strand. The optimized activating siRNAs potently enhance the expression of interleukin-2 (IL-2) gene in human and mouse primary CD4+ T cells with a long-time effect. Taken together, our study provides a guideline for rational design the promoter-targeting siRNA to sequence-specifically enhance gene expression.
Collapse
Affiliation(s)
- Miaomiao Fan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yijun Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhuoqiong Huang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuemin Guo
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (HZ); (HL)
| | - Haihua Luo
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (HZ); (HL)
| |
Collapse
|
22
|
Fiszer A, Krzyzosiak WJ. Oligonucleotide-based strategies to combat polyglutamine diseases. Nucleic Acids Res 2014; 42:6787-810. [PMID: 24848018 PMCID: PMC4066792 DOI: 10.1093/nar/gku385] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Considerable advances have been recently made in understanding the molecular aspects of pathogenesis and in developing therapeutic approaches for polyglutamine (polyQ) diseases. Studies on pathogenic mechanisms have extended our knowledge of mutant protein toxicity, confirmed the toxicity of mutant transcript and identified other toxic RNA and protein entities. One very promising therapeutic strategy is targeting the causative gene expression with oligonucleotide (ON) based tools. This straightforward approach aimed at halting the early steps in the cascade of pathogenic events has been widely tested for Huntington's disease and spinocerebellar ataxia type 3. In this review, we gather information on the use of antisense oligonucleotides and RNA interference triggers for the experimental treatment of polyQ diseases in cellular and animal models. We present studies testing non-allele-selective and allele-selective gene silencing strategies. The latter include targeting SNP variants associated with mutations or targeting the pathologically expanded CAG repeat directly. We compare gene silencing effectors of various types in a number of aspects, including their design, efficiency in cell culture experiments and pre-clinical testing. We discuss advantages, current limitations and perspectives of various ON-based strategies used to treat polyQ diseases.
Collapse
Affiliation(s)
- Agnieszka Fiszer
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
23
|
Polonyi C, Alshiekh A, Sarsam LA, Clausén M, Elmroth SKC. Cisplatin-induced duplex dissociation of complementary and destabilized short GG-containing duplex RNAs. Dalton Trans 2014; 43:11941-9. [DOI: 10.1039/c4dt00213j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The reactivity of a series of small size RNAs towards mono-aquated cisplatin was monitored using UV/vis spectroscopy. Subtle changes of local melting behavior were found to influence metal binding kinetics, with an increase in reactivity following central destabilization.
Collapse
Affiliation(s)
- Christopher Polonyi
- Biochemistry and Structural Biology
- KILU
- Lund University
- SE-221 00 Lund, Sweden
| | - Alak Alshiekh
- Biochemistry and Structural Biology
- KILU
- Lund University
- SE-221 00 Lund, Sweden
| | - Lamya A. Sarsam
- Biochemistry and Structural Biology
- KILU
- Lund University
- SE-221 00 Lund, Sweden
| | - Maria Clausén
- Biochemistry and Structural Biology
- KILU
- Lund University
- SE-221 00 Lund, Sweden
| | - Sofi K. C. Elmroth
- Biochemistry and Structural Biology
- KILU
- Lund University
- SE-221 00 Lund, Sweden
| |
Collapse
|
24
|
Tam YYC, Chen S, Cullis PR. Advances in Lipid Nanoparticles for siRNA Delivery. Pharmaceutics 2013; 5:498-507. [PMID: 24300520 PMCID: PMC3836621 DOI: 10.3390/pharmaceutics5030498] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 01/21/2023] Open
Abstract
Technological advances in both siRNA (small interfering RNA) and whole genome sequencing have demonstrated great potential in translating genetic information into siRNA-based drugs to halt the synthesis of most disease-causing proteins. Despite its powerful promises as a drug, siRNA requires a sophisticated delivery vehicle because of its rapid degradation in the circulation, inefficient accumulation in target tissues and inability to cross cell membranes to access the cytoplasm where it functions. Lipid nanoparticle (LNP) containing ionizable amino lipids is the leading delivery technology for siRNA, with five products in clinical trials and more in the pipeline. Here, we focus on the technological advances behind these potent systems for siRNA-mediated gene silencing.
Collapse
Affiliation(s)
- Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C. V6T 1Z3, Canada.
| | | | | |
Collapse
|
25
|
Feng XY, Zhao YM, Wang WJ, Ge LH. Msx1regulates proliferation and differentiation of mouse dental mesenchymal cells in culture. Eur J Oral Sci 2013; 121:412-20. [PMID: 24028588 DOI: 10.1111/eos.12078] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2013] [Indexed: 11/26/2022]
Affiliation(s)
- Xiao-yu Feng
- Department of Pediatric Dentistry; Peking University School and Hospital of Stomatology; Beijing; China
| | - Yu-ming Zhao
- Department of Pediatric Dentistry; Peking University School and Hospital of Stomatology; Beijing; China
| | - Wen-jun Wang
- Department of Pediatric Dentistry; Peking University School and Hospital of Stomatology; Beijing; China
| | - Li-hong Ge
- Department of Pediatric Dentistry; Peking University School and Hospital of Stomatology; Beijing; China
| |
Collapse
|
26
|
Polonyi C, Elmroth SKC. Time dependence of cisplatin-induced duplex dissociation of 15-mer RNAs and mature miR-146a. Dalton Trans 2013; 42:14959-62. [DOI: 10.1039/c3dt51788h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|