1
|
Connerty P, Moles E, de Bock CE, Jayatilleke N, Smith JL, Meshinchi S, Mayoh C, Kavallaris M, Lock RB. Development of siRNA-Loaded Lipid Nanoparticles Targeting Long Non-Coding RNA LINC01257 as a Novel and Safe Therapeutic Approach for t(8;21) Pediatric Acute Myeloid Leukemia. Pharmaceutics 2021; 13:pharmaceutics13101681. [PMID: 34683974 PMCID: PMC8539450 DOI: 10.3390/pharmaceutics13101681] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Standard of care therapies for children with acute myeloid leukemia (AML) cause potent off-target toxicity to healthy cells, highlighting the need to develop new therapeutic approaches that are safe and specific for leukemia cells. Long non-coding RNAs (lncRNAs) are an emerging and highly attractive therapeutic target in the treatment of cancer due to their oncogenic functions and selective expression in cancer cells. However, lncRNAs have historically been considered ‘undruggable’ targets because they do not encode for a protein product. Here, we describe the development of a new siRNA-loaded lipid nanoparticle for the therapeutic silencing of the novel oncogenic lncRNA LINC01257. Transcriptomic analysis of children with AML identified LINC01257 as specifically expressed in t(8;21) AML and absent in healthy patients. Using NxGen microfluidic technology, we efficiently and reproducibly packaged anti-LINC01257 siRNA (LNP-si-LINC01257) into lipid nanoparticles based on the FDA-approved Patisiran (Onpattro®) formulation. LNP-si-LINC01257 size and ζ-potential were determined by dynamic light scattering using a Malvern Zetasizer Ultra. LNP-si-LINC01257 internalization and siRNA delivery were verified by fluorescence microscopy and flow cytometry analysis. lncRNA knockdown was determined by RT-qPCR and cell viability was characterized by flow cytometry-based apoptosis assay. LNP-siRNA production yielded a mean LNP size of ~65 nm with PDI ≤ 0.22 along with a >85% siRNA encapsulation rate. LNP-siRNAs were efficiently taken up by Kasumi-1 cells (>95% of cells) and LNP-si-LINC01257 treatment was able to successfully ablate LINC01257 expression which was accompanied by a significant 55% reduction in total cell count following 48 h of treatment. In contrast, healthy peripheral blood mononuclear cells (PBMCs), which do not express LINC01257, were unaffected by LNP-si-LINC01257 treatment despite comparable levels of LNP-siRNA uptake. This is the first report demonstrating the use of LNP-assisted RNA interference modalities for the silencing of cancer-driving lncRNAs as a therapeutically viable and non-toxic approach in the management of AML.
Collapse
Affiliation(s)
- Patrick Connerty
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ernest Moles
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Charles E. de Bock
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Nisitha Jayatilleke
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
| | - Jenny L. Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (J.L.S.); (S.M.)
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA 98109, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (J.L.S.); (S.M.)
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA 98109, USA
| | - Chelsea Mayoh
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Richard B. Lock
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia; (P.C.); (E.M.); (C.E.d.B.); (N.J.); (C.M.); (M.K.)
- School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
- Correspondence: ; Tel.: +61-(02)-7209-6765
| |
Collapse
|
2
|
Bartman CM, Stelzig KE, Linden DR, Prakash YS, Chiarella SE. Passive siRNA transfection method for gene knockdown in air-liquid interface airway epithelial cell cultures. Am J Physiol Lung Cell Mol Physiol 2021; 321:L280-L286. [PMID: 34037474 DOI: 10.1152/ajplung.00122.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Differentiation of human bronchial epithelial cells (HBEs) in air-liquid interface (ALI) cultures recapitulates organotypic modeling of the in vivo environment. Although ALI cultures are invaluable for studying the respiratory epithelial barrier, loss-of-function studies are limited by potentially cytotoxic reagents in classical transfection methods, the length of the differentiation protocol, and the number of primary epithelial cell passages. Here, we present the efficacy and use of a simple method for small interfering RNA (siRNA) transfection of normal HBEs (NHBEs) in ALI cultures that does not require potentially cytotoxic transfection reagents and does not detrimentally alter the physiology or morphology of NHBEs during the differentiation process. This transfection protocol introduces a reproducible and efficient method for loss-of-function studies in HBE ALI cultures that can be leveraged for modeling the respiratory system and airway diseases.
Collapse
Affiliation(s)
- Colleen M Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kimberly E Stelzig
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - David R Linden
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
3
|
N-[4-( N,N,N-Trimethylammonium)Benzyl]Chitosan Chloride as a Gene Carrier: The Influence of Polyplex Composition and Cell Type. MATERIALS 2021; 14:ma14092467. [PMID: 34068680 PMCID: PMC8126137 DOI: 10.3390/ma14092467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/24/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022]
Abstract
Polyplex-based gene delivery systems are promising substitutes for viral vectors because of their high versatility and lack of disadvantages commonly encountered with viruses. In this work, we studied the DNA polyplexes with N-[4-(N,N,N-trimethylammonium)benzyl]chitosan chloride (TMAB-CS) of various compositions in different cell types. Investigations of the interaction of TMAB-CS with DNA by different physical methods revealed that the molecular weight and the degree of substitution do not dramatically influence the hydrodynamic properties of polyplexes. Highly substituted TMAB-CS samples had a high affinity for DNA. The transfection protocol was optimized in HEK293T cells and achieved the highest efficiency of 30-35%. TMAB-CS was dramatically less effective in nonadherent K562 cells (around 1% transfected cells), but it was more effective and less toxic than polyarginine.
Collapse
|
4
|
Remant KC, Thapa B, Valencia-Serna J, Domun SS, Dimitroff C, Jiang X, Uludağ H. Cholesterol grafted cationic lipopolymers: Potential siRNA carriers for selective chronic myeloid leukemia therapy. J Biomed Mater Res A 2019; 108:565-580. [PMID: 31714657 DOI: 10.1002/jbm.a.36837] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/21/2019] [Accepted: 09/04/2019] [Indexed: 01/22/2023]
Abstract
Synthetic siRNA technology has emerged as a promising approach for molecular therapy of cancer but, despite its potential for post-transcriptional gene silencing, there is an urgent need to develop efficient delivery systems particularly for difficult-to-transfect, anchorage-independent cells. In this study, we designed highly hydrophobic cationic lipopolymers by grafting cholesterol (Chol) onto low-molecular weight (0.6, 1.2, and 2.0 kDa) polyethylenimines (PEIs) to enable specific siRNA therapy to chronic myeloid leukemia (CML) cells. The siRNA binding by PEI-Chol led to nano-sized (100-200 nm diameter) polyplexes with enhanced ζ-potential (+20 to +35 mV) and ability to protect the loaded siRNA completely in fresh serum. The siRNA delivery to CML (K562) cells was proportional to degree of substitution and, unexpectedly, inversely proportional to molecular size of the polymeric backbone. Chol grafting with as little as ~1.0 Chol/PEI on 0.6 and 1.2 kDa PEIs enabled silencing of the reporter Green Fluorescent Protein gene as well as the endogenous BCR-Abl oncogene in K562 cells. The PEI-Chol mediated delivery of siRNAs specific for BCR-Abl and KSP genes significantly arrested the growth the cells which was significantly reflected in colony formation potency of K562 cells. BCR-Able siRNA mediated therapeutic efficacy was also observed in significantly increased caspase activity and apoptosis of K562 cells. Thus, Chol-grafted low-molecular weight PEIs appear to be unique siRNA carriers to realize the molecular therapy in CML cells.
Collapse
Affiliation(s)
- K C Remant
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Juliana Valencia-Serna
- Department of Biomedical Engineering, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Suraj S Domun
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Cailean Dimitroff
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyan Jiang
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hasan Uludağ
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.,Department of Biomedical Engineering, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Zhang Z, Zhang HY, Zhang Y, Li H. Inactivation of the Ras/MAPK/PPARγ signaling axis alleviates diabetic mellitus-induced erectile dysfunction through suppression of corpus cavernosal endothelial cell apoptosis by inhibiting HMGCS2 expression. Endocrine 2019; 63:615-631. [PMID: 30460485 DOI: 10.1007/s12020-018-1810-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/30/2018] [Indexed: 01/30/2023]
Abstract
PURPOSE Diabetic mellitus-induced erectile dysfunction (DMED) represents a significant complication associated with diabetes mellitus (DM) that greatly affects human life quality. Various reports have highlighted the involvement of mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) in the regulation of mitochondrial fatty acid oxidation, which has also been linked with DM. Through bioinformatics analysis, HMGCS2 was determined to be a novel target among DM patients suffering from erectile dysfunction (ED), and enriched in the Ras/ERK/PPAR signaling axis. Owing to the fact that the key mechanism HMGCS2 involved in DM remains largely unknown, we set out to investigate the role of the Ras/MAPK/PPARγ signaling axis and HMGCS2 in the corpus cavernosal endothelial cells (CCECs) of rats with DMED. METHODS Firstly, bioinformatics analysis was used to screen out differentially expressed genes in DMED. Then, to investigate the influence of the Ras/MAPK/PPARγ signaling axis and HMGCS2 on DMED, a rat model of DMED was established and injected with Simvastatin and si-Hmgcs2. The individual expression patterns of Ras, MAPK, PPARγ and HMGCS2 were determined by RT-qPCR, immunohistochemistry and western blot analysis methods. Afterwards, to investigate the mechanism of Ras/MAPK/PPARγ signaling axis and HMGCS2, CCECs were isolated from DMED rats and transfected with agonists and inhibitors of the Ras/MAPK/PPARγ signaling axis and siRNA of HMGCS2, with their respective functions in apoptosis and impairment of CCECs evaluated using TUNEL staining and flow cytometry. RESULTS Microarray analysis and KEGG pathway enrichment analysis revealed that Ras/ERK/PPAR signaling axis mediated HMGCS2 in DMED. Among the DMED rats, the Ras/MAPK/PPAR signaling axis was also activated while the expression of HMGCS2 was upregulated. The activation of Ras was determined to be capable of upregulating ERK expression which resulted in the inhibition of the transcription of PPARγ and subsequent upregulation of HMGCS2 expression. The inhibited activation of the Ras/ERK/PPAR signaling axis and silencing HMGCS2 were observed to provide an alleviatory effect on the injury of DMED while acting to inhibit the apoptosis of CCECs. CONCLUSION Collectively, the key findings suggested that suppression of the Ras/MAPK/PPARγ signaling axis could downregulate expression of HMGCS2, so as to alleviate DMED. This study defines the potential treatment for DMED through inhibition of the Ras/MAPK/PPARγ signaling axis and silencing HMGCS2.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Urology, China-Japan Union Hospital of Jilin University, 130000, Changchun, P.R. China
| | - Hai-Yan Zhang
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, 130000, Changchun, P.R. China
| | - Ying Zhang
- Department of Pathology, China-Japan Union Hospital of Jilin University, 130000, Changchun, P.R. China
| | - Hai Li
- Department of Urology, China-Japan Union Hospital of Jilin University, 130000, Changchun, P.R. China.
| |
Collapse
|
6
|
Song X, Tang T, Li C, Liu X, Zhou L. CBX8 and CD96 Are Important Prognostic Biomarkers of Colorectal Cancer. Med Sci Monit 2018; 24:7820-7827. [PMID: 30383736 PMCID: PMC6225733 DOI: 10.12659/msm.908656] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies worldwide, with high morbidity and mortality rates. The purpose of this study was to identify potential biomarkers in the progression of CRC. MATERIAL AND METHODS Gene and isoform expression datasets of CRC was downloaded from The Cancer Genome Atlas (TCGA). EBSeq of R was used for the normalization of gene and isoform expression, as well as the identification of differential expression genes (DEGs) and isoforms (DEIs) of CRC samples compared with normal samples. The enriched functions of DEGs and DEIs were obtained based on the Database for Annotation, Visualization and Integrated Discovery (DAVID). An independent dataset, GSE38832, was downloaded from the Gene Expression Omnibus (GEO) database for survival analysis of genes with sustained decreased/increased expression values at both gene and isoform levels with the development of CRC. RESULTS A total of 2301 genes and 4241 isoforms were found to be significantly differentially expressed in stage I-IV CRC samples. They are closely associated with muscle or cell system activity. Sixteen genes were screened out with sustained decreased/increased expression values at both gene and isoform levels with the development of CRC. Aberrant CBX8 and CD96 expressions were found to be significantly associated with CRC survival. CONCLUSIONS Through combined analysis of gene and isoform expression profiles, we identified several potential biomarkers that may play an important role in the development of CRC and could be helpful in its early diagnosis and treatment.
Collapse
Affiliation(s)
- Xin Song
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Tao Tang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Chaofeng Li
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Xin Liu
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China (mainland)
| | - Lei Zhou
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China (mainland)
| |
Collapse
|
7
|
Hazzan T, Guhl S, Artuc M, Franke K, Worm M, Zuberbier T, Babina M. An efficient method for gene knock-down by RNA interference in human skin mast cells. Exp Dermatol 2017; 26:1136-1139. [PMID: 28418623 DOI: 10.1111/exd.13358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2017] [Indexed: 12/12/2022]
Abstract
Mast cells (MCs) from human skin have been notoriously resistant to gene manipulation, and a method to knock-down gene expression in in situ differentiated MCs is highly desired. The Dharmacon Accell® transfection system proved successful on several "difficult-to-transfect" cells. In the present work, we therefore tested this method on skin-derived MCs using different siRNA entities. The siRNA was readily taken up, followed by pronounced, specific reduction of gene and protein expression. Hence, we present the first efficient technique for the manipulation of gene expression in primary skin MCs ex vivo, which combines high transfection rates with retained cell viability.
Collapse
Affiliation(s)
- Tarek Hazzan
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Guhl
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Metin Artuc
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kristin Franke
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Margitta Worm
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Zuberbier
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Magda Babina
- Department of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Wang LL, Burdick JA. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies. Adv Healthc Mater 2017; 6:10.1002/adhm.201601041. [PMID: 27976524 PMCID: PMC5226889 DOI: 10.1002/adhm.201601041] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Indexed: 12/20/2022]
Abstract
It has been nearly two decades since RNA-interference (RNAi) was first reported. While there are no approved clinical uses, several phase II and III clinical trials suggest the great promise of RNAi therapeutics. One challenge for RNAi therapies is the controlled localization and sustained presentation to target tissues, to both overcome systemic toxicity concerns and to enhance in vivo efficacy. One approach that is emerging to address these limitations is the entrapment of RNAi molecules within hydrogels for local and sustained release. In these systems, nucleic acids are either delivered as siRNA conjugates or within nanoparticles. A plethora of hydrogels has been implemented using these approaches, including both traditional hydrogels that have already been developed for other applications and new hydrogels developed specifically for RNAi delivery. These hydrogels have been applied to various applications in vivo, including cancer, bone regeneration, inflammation and cardiac repair. This review will examine the design and implementation of such hydrogel RNAi systems and will cover the most recent applications of these systems.
Collapse
Affiliation(s)
- Leo L. Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
9
|
Gherardini L, Bardi G, Gennaro M, Pizzorusso T. Novel siRNA delivery strategy: a new "strand" in CNS translational medicine? Cell Mol Life Sci 2014; 71:1-20. [PMID: 23508806 PMCID: PMC11113879 DOI: 10.1007/s00018-013-1310-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 12/12/2022]
Abstract
RNA interference has been envisaged as a powerful tool for molecular and clinical investigation with a great potential for clinical applications. In recent years, increased understanding of cancer biology and stem cell biology has dramatically accelerated the development of technology for cell and gene therapy in these areas. This paper is a review of the most recent report of innovative use of siRNA to benefit several central nervous system diseases. Furthermore, a description is made of innovative strategies of delivery into the brain by means of viral and non-viral vectors with high potential for translation into clinical use. Problems are also highlighted that might hamper the transition from bench to bed, analyzing the lack of reliable preclinical models with predictive validity and the lack of effective delivery systems, which are able to overcome biological barriers and specifically reach the brain site of action.
Collapse
Affiliation(s)
| | - Giuseppe Bardi
- Center for MicroBioRobotics @SSSA, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | | | - Tommaso Pizzorusso
- Institute of Neuroscience, CNR, Via Moruzzi, 1 56124 Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Florence, Italy
| |
Collapse
|
10
|
Valencia-Serna J, Gul-Uludağ H, Mahdipoor P, Jiang X, Uludağ H. Investigating siRNA delivery to chronic myeloid leukemia K562 cells with lipophilic polymers for therapeutic BCR-ABL down-regulation. J Control Release 2013; 172:495-503. [DOI: 10.1016/j.jconrel.2013.05.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/09/2013] [Accepted: 05/20/2013] [Indexed: 11/30/2022]
|
11
|
Discovery of siRNA lipid nanoparticles to transfect suspension leukemia cells and provide in vivo delivery capability. Mol Ther 2013; 22:359-370. [PMID: 24002693 DOI: 10.1038/mt.2013.210] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/19/2013] [Indexed: 12/15/2022] Open
Abstract
As a powerful research tool, siRNA's therapeutic and target validation utility with leukemia cells and long-term gene knockdown is severely restricted by the lack of omnipotent, safe, stable, and convenient delivery. Here, we detail our discovery of siRNA-containing lipid nanoparticles (LNPs) able to effectively transfect several leukemia and difficult-to-transfect adherent cell lines also providing in vivo delivery to mouse spleen and bone marrow tissues through tail-vein administration. We disclose a series of novel structurally related lipids accounting for the superior transfection ability, and reveal a correlation between expression of Caveolins and successful transfection. These LNPs, bearing low toxicity and long stability of >6 months, are ideal for continuous long-term dosing. Our discovery represents the first effective siRNA-containing LNPs for leukemia cells, which not only enables high-throughput siRNA screening with leukemia cells and difficult-to-transfect adherent cells but also paves the way for the development of therapeutic siRNA for leukemia treatment.
Collapse
|
12
|
Kim JS, Lee YK, Jeong HY, Kang SJ, Kim MW, Ryu SH, Kim HS, Kim KS, Kim DE, Park YS. Sendai F/HN viroplexes for efficient transfection of leukemic T cells. Yonsei Med J 2013; 54:1149-57. [PMID: 23918564 PMCID: PMC3743179 DOI: 10.3349/ymj.2013.54.5.1149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Most chemical transfection reagents are ineffective for the transfection of cells in suspension, such as leukemic cell and stem cell lineages. We developed two different types of viroplexes, cationic Sendai F/HN viroplexes (CSVs) and protamine sulfate-condensed cationic Sendai F/HN viroplexes (PCSVs) for the efficient transfection of T-leukemic cells. MATERIALS AND METHODS The viroplex systems were prepared by reconstitution of fusogenic Sendai F/HN proteins in DMKE (O,O'-dimyristyl-N-lysyl glutamate) cationic liposomes. The viroplexes were further optimized for plasmid DNA and siRNA delivery to suspension cells. The particle size and surface charge of the viroplexes were analyzed with a ζ-sizer. Transfection of plasmid DNA (pDNA) and small interfering RNA (siRNA) by CSVs or PCSV was evaluated by measurement of transgene expression, confocal microscopy, FACS, and RT-PCR. RESULTS The optimized CSVs and PCSVs exhibited enhanced gene and siRNA delivery in the tested suspension cell lines (Jurkat cells and CEM cells), compared with conventional cationic liposomes. In the case of pDNA transfection, the CSVs and PCSVs show at least 10-fold and 100-fold higher transgene expression compared with DMKE lipoplexes (or lipofectamine 2000), respectively. The CSVs showed more effective siRNA delivery to the suspension cells than cationic liposomes, as assessed by confocal microscopy, FACS, and RT-PCR. The effective transfection by the CSVs and PCSVs is presumably due to fusogenic activity of F/HN proteins resulting in facilitated internalization of pDNA and siRNA. CONCLUSION This study suggests that Sendai F/HN viroplexes can be widely applicable for the transfection of pDNA and siRNA to suspension cell lines.
Collapse
Affiliation(s)
- Jung Seok Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Yeon Kyung Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Seung Hyun Ryu
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| | - Hong Sung Kim
- Department of Biomedical Laboratory Science, Korea Nazarene University, Cheonan, Korea
| | - Keun Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon, Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Korea
| |
Collapse
|
13
|
Xie Q, Luo J, Zhu Z, Wang G, Wang J, Niu B. Nucleofection of a DNA vaccine into human monocyte-derived dendritic cells. Cell Immunol 2012; 276:135-43. [PMID: 22632899 DOI: 10.1016/j.cellimm.2012.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/19/2012] [Accepted: 04/30/2012] [Indexed: 11/27/2022]
Abstract
An efficient method for delivering DNA vaccines into dendritic cells is considered to be of paramount importance. Electroporation-based technology (nucleofection) has gained increasingly popularity, but few reports focused on the possible functional consequences related to this method. In this study, the nucleofection technique was used to transfer the recombinant plasmid into hMoDCs for phenotype expression analysis and immunopotency detection. The results showed that the nucleofection of increasing concentrations of plasmid DNA decreased the viability of the hMoDCs. The welfare of nucleofected hMoDCs depended on the dosage of the plasmid and the plasmid's retention time within the cells. Accompanied by the process of nucleofection, it would bring some non-specific changes. The methodology reported here is suggestive of a feasible system for DNA vaccine transfer into hMoDCs with the caution of certain undesired effect.
Collapse
Affiliation(s)
- Qiu Xie
- Department of Biotechnology, Capital Institute of Pediatrics, Beijing 100020, China
| | | | | | | | | | | |
Collapse
|