51
|
Epidermal or dermal specific knockout of PHD-2 enhances wound healing and minimizes ischemic injury. PLoS One 2014; 9:e93373. [PMID: 24695462 PMCID: PMC3973687 DOI: 10.1371/journal.pone.0093373] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 03/05/2014] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Hypoxia-inducible factor (HIF)-1α, part of the heterodimeric transcription factor that mediates the cellular response to hypoxia, is critical for the expression of multiple angiogenic growth factors, cell motility, and the recruitment of endothelial progenitor cells. Inhibition of the oxygen-dependent negative regulator of HIF-1α, prolyl hydroxylase domain-2 (PHD-2), leads to increased HIF-1α and mimics various cellular and physiological responses to hypoxia. The roles of PHD-2 in the epidermis and dermis have not been clearly defined in wound healing. METHODS Epidermal and dermal specific PHD-2 knockout (KO) mice were developed in a C57BL/6J (wild type) background by crossing homozygous floxed PHD-2 mice with heterozygous K14-Cre mice and heterozygous Col1A2-Cre-ER mice to get homozygous floxed PHD-2/heterozygous K14-Cre and homozygous floxed PHD-2/heterozygous floxed Col1A2-Cre-ER mice, respectively. Ten to twelve-week-old PHD-2 KO and wild type (WT) mice were subjected to wounding and ischemic pedicle flap model. The amount of healing was grossly quantified with ImageJ software. Western blot and qRT-PCR was run on protein and RNA from primary cells cultured in vitro. RESULTS qRT-PCR demonstrated a significant decrease of PHD-2 in keratinocytes and fibroblasts derived from tissue specific KO mice relative to control mice (*p<0.05). Western blot analysis showed a significant increase in HIF-1α and VEGF protein levels in PHD-2 KO mice relative to control mice (*p<0.05). PHD-2 KO mice showed significantly accelerated wound closure relative to WT (*p<0.05). When ischemia was analyzed at day nine post-surgery in a flap model, the PHD-2 tissue specific knockout mice showed significantly more viable flaps than WT (*p<0.05). CONCLUSIONS PHD-2 plays a significant role in the rates of wound healing and response to ischemic insult in mice. Further exploration shows PHD-2 KO increases cellular levels of HIF-1α and this increase leads to the transcription of downstream angiogenic factors such as VEGF.
Collapse
|
52
|
Lijkwan MA, Hellingman AA, Bos EJ, van der Bogt KEA, Huang M, Kooreman NG, de Vries MR, Peters HAB, Robbins RC, Hamming JF, Quax PHA, Wu JC. Short hairpin RNA gene silencing of prolyl hydroxylase-2 with a minicircle vector improves neovascularization of hindlimb ischemia. Hum Gene Ther 2014; 25:41-9. [PMID: 24090375 DOI: 10.1089/hum.2013.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In this study, we target the hypoxia inducible factor-1 alpha (HIF-1-alpha) pathway by short hairpin RNA interference therapy targeting prolyl hydroxylase-2 (shPHD2). We use the minicircle (MC) vector technology as an alternative for conventional nonviral plasmid (PL) vectors in order to improve neovascularization after unilateral hindlimb ischemia in a murine model. Gene expression and transfection efficiency of MC and PL, both in vitro and in vivo, were assessed using bioluminescence imaging (BLI) and firefly luciferase (Luc) reporter gene. C57Bl6 mice underwent unilateral electrocoagulation of the femoral artery and gastrocnemic muscle injection with MC-shPHD2, PL-shPHD2, or phosphate-buffered saline (PBS) as control. Blood flow recovery was monitored using laser Doppler perfusion imaging, and collaterals were visualized by immunohistochemistry and angiography. MC-Luc showed a 4.6-fold higher in vitro BLI signal compared with PL-Luc. BLI signals in vivo were 4.3×10(5)±3.3×10(5) (MC-Luc) versus 0.4×10(5)±0.3×10(5) (PL-Luc) at day 28 (p=0.016). Compared with PL-shPHD2 or PBS, MC-shPHD2 significantly improved blood flow recovery, up to 50% from day 3 until day 14 after ischemia induction. MC-shPHD2 significantly increased collateral density and capillary density, as monitored by alpha-smooth muscle actin expression and CD31(+) expression, respectively. Angiography data confirmed the histological findings. Significant downregulation of PHD2 mRNA levels by MC-shPHD2 was confirmed by quantitative polymerase chain reaction. Finally, Western blot analysis confirmed significantly higher levels of HIF-1-alpha protein by MC-shPHD2, compared with PL-shPHD2 and PBS. This study provides initial evidence of a new potential therapeutic approach for peripheral artery disease. The combination of HIF-1-alpha pathway targeting by shPHD2 with the robust nonviral MC plasmid improved postischemic neovascularization, making this approach a promising potential treatment option for critical limb ischemia.
Collapse
Affiliation(s)
- Maarten A Lijkwan
- 1 Department of Medicine and Radiology, Stanford University School of Medicine , Stanford, CA 94305
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Raynaud CM, Ahmad FS, Allouba M, Abou-Saleh H, Lui KO, Yacoub M. Reprogramming for cardiac regeneration. Glob Cardiol Sci Pract 2014; 2014:309-29. [PMID: 25763379 PMCID: PMC4352683 DOI: 10.5339/gcsp.2014.44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/18/2014] [Indexed: 01/10/2023] Open
Abstract
Treatment of cardiovascular diseases remains challenging considering the limited regeneration capacity of the heart muscle. Developments of reprogramming strategies to create in vitro and in vivo cardiomyocytes have been the focus point of a considerable amount of research in the past decades. The choice of cells to employ, the state-of-the-art methods for different reprogramming strategies, and their promises and future challenges before clinical entry, are all discussed here.
Collapse
Affiliation(s)
| | | | - Mona Allouba
- Aswan Heart Center, Magdi Yacoub Foundation, Aswan, Egypt
| | - Haissam Abou-Saleh
- Qatar Cardiovascular Research Center, Qatar Foundation-Education City, Doha, Qatar
| | - Kathy O Lui
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, USA
| | | |
Collapse
|
54
|
Anderson CD, Urschitz J, Khemmani M, Owens JB, Moisyadi S, Shohet RV, Walton CB. Ultrasound directs a transposase system for durable hepatic gene delivery in mice. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2351-61. [PMID: 24035623 PMCID: PMC3838570 DOI: 10.1016/j.ultrasmedbio.2013.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/08/2013] [Accepted: 07/11/2013] [Indexed: 05/15/2023]
Abstract
Our aim was to evaluate the delivery of transposase-based vectors by ultrasound targeted microbubble destruction (UTMD) in mice. DNA vectors were attached to cationic lipid microbubbles (1-3 μm in diameter), injected intravenously and delivered to the liver by destruction of the carrier bubbles with ultrasound in burst mode at 1.0 MHz, 20-μs pulse duration, 10-Hz pulse repetition frequency and ∼1.3-MPa acoustic peak negative pressure. We evaluated the expression and genomic integration of conventional (pcDNA3) and piggyBac transposase-based (pmGENIE) reporter vectors. In vivo, we observed UTMD-mediated liver-specific expression of pmGENIE for an average of 24 d, compared with 4 d with pcDNA3. Reporter expression was located predominately near blood vessels initially, whereas expression after 3 d was more evenly distributed through the parenchyma of the liver. We confirmed random genomic integration for pmGENIE in vitro; however, integration events for pmGENIE in vivo were targeted to specific areas of chromosome 14. Our results suggest that a combination of UTMD and non-viral DNA transposase vectors can mediate weeks of hepatic-specific gene transfer in vivo, and analyses performed by non-restrictive linear amplification-mediated (nrLAM) polymerase chain reaction, cloning and sequencing identify an unexpected tropism for integration within a specific sequence on chromosome 14 in mice. UTMD delivery of transgenes may be useful for the treatment of hepatic gene deficiency disorders.
Collapse
Affiliation(s)
- Cynthia D Anderson
- Department of Cellular and Molecular Biology; John A. Burns School of Medicine, Honolulu, HI 96813
| | - Johann Urschitz
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI 96813
| | - Mark Khemmani
- Department of Medicine, John A. Burns School of Medicine, Honolulu, HI 96813
| | - Jesse B Owens
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI 96813
| | - Stefan Moisyadi
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, Honolulu, HI 96813
- Manoa Biosciences
| | - Ralph V Shohet
- Department of Medicine, John A. Burns School of Medicine, Honolulu, HI 96813
| | - Chad B Walton
- Department of Medicine, John A. Burns School of Medicine, Honolulu, HI 96813
| |
Collapse
|
55
|
In vivo electroporation of minicircle DNA as a novel method of vaccine delivery to enhance HIV-1-specific immune responses. J Virol 2013; 88:1924-34. [PMID: 24284319 DOI: 10.1128/jvi.02757-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
DNA vaccines offer advantage over conventional vaccines, as they are safer to use, easier to produce, and able to induce humoral as well cellular immune responses. Unfortunately, no DNA vaccines have been licensed for human use for the difficulties in developing an efficient and safe in vivo gene delivery system. In vivo electroporation (EP)-based DNA delivery has attracted great attention for its potency to enhance cellular uptake of DNA vaccines and function as an adjuvant. Minicircle DNA (a new form of DNA containing only a gene expression cassette and lacking a backbone of bacterial plasmid DNA) is a powerful candidate of gene delivery in terms of improving the levels and the duration of transgene expression in vivo. In this study, as a novel vaccine delivery system, we combined in vivo EP and the minicircle DNA carrying a codon-optimized HIV-1 gag gene (minicircle-gag) to evaluate the immunogenicity of this system. We found that minicircle-gag conferred persistent and high levels of gag expression in vitro and in vivo. The use of EP delivery further increased minicircle-based gene expression. Moreover, when delivered by EP, minicircle-gag vaccination elicited a 2- to 3-fold increase in cellular immune response and a 1.5- to 3-fold augmentation of humoral immune responses compared with those elicited by a pVAX1-gag positive control. Increased immunogenicity of EP-assisted minicircle-gag may benefit from increasing local antigen expression, upregulating inflammatory genes, and recruiting immune cells. Collectively, in vivo EP of minicircle DNA functions as a novel vaccine platform that can enhance efficacy and immunogenicity of DNA vaccines.
Collapse
|
56
|
Keeney M, Ong SG, Padilla A, Yao Z, Goodman S, Wu JC, Yang F. Development of poly(β-amino ester)-based biodegradable nanoparticles for nonviral delivery of minicircle DNA. ACS NANO 2013; 7:7241-50. [PMID: 23837668 PMCID: PMC3789527 DOI: 10.1021/nn402657d] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Gene therapy provides a powerful tool for regulating cellular processes and tissue repair. Minicircle (MC) DNA are supercoiled DNA molecules free of bacterial plasmid backbone elements and have been reported to enhance prolonged gene expression compared to conventional plasmids. Despite the great promise of MC DNA for gene therapy, methods for safe and efficient MC DNA delivery remain lacking. To overcome this bottleneck, here we report the development of a poly(β-amino ester) (PBAE)-based, biodegradable nanoparticulate platform for efficient delivery of MC DNA driven by a Ubc promoter in vitro and in vivo. By synthesizing and screening a small library of 18 PBAE polymers with different backbone and end-group chemistry, we identified lead cationic PBAE structures that can complex with minicircle DNA to form nanoparticles, and delivery efficiency can be further modulated by tuning PBAE chemistry. Using human embryonic kidney 293 cells and mouse embryonic fibroblasts as model cell types, we identified a few PBAE polymers that allow efficient MC delivery at levels that are comparable or even surpassing Lipofectamine 2000. The biodegradable nature of PBAE-based nanoparticles facilitates in vivo applications and clinical translation. When injected via intraperitoneal route in vivo, MC alone resulted in high transgene expression, and a lead PBAE/MC nanoparticle formulation achieved a further 2-fold increase in protein expression compared to MC alone. Together, our results highlight the promise of PBAE-based nanoparticles as promising nonviral gene carriers for MC delivery, which may provide a valuable tool for broad applications of MC DNA-based gene therapy.
Collapse
Affiliation(s)
- Michael Keeney
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Sang-Ging Ong
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Amanda Padilla
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Zhenyu Yao
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Stuart Goodman
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence to Fan Yang, PhD. And Joseph C Wu, MD, PhD. ; ; Phone: 650-725-7128
| | - Fan Yang
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Correspondence to Fan Yang, PhD. And Joseph C Wu, MD, PhD. ; ; Phone: 650-725-7128
| |
Collapse
|
57
|
Hyun J, Grova M, Nejadnik H, Lo D, Morrison S, Montoro D, Chung M, Zimmermann A, Walmsley GG, Lee M, Daldrup-Link H, Wan DC, Longaker MT. Enhancing in vivo survival of adipose-derived stromal cells through Bcl-2 overexpression using a minicircle vector. Stem Cells Transl Med 2013; 2:690-702. [PMID: 23934910 DOI: 10.5966/sctm.2013-0035] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Tissue regeneration using progenitor cell-based therapy has the potential to aid in the healing of a diverse range of pathologies, ranging from short-gut syndrome to spinal cord lesions. However, there are numerous hurdles to be overcome prior to the widespread application of these cells in the clinical setting. One of the primary barriers to effective stem cell therapy is the hostile environment that progenitor cells encounter in the clinical injury wound setting. In order to promote cellular survival, stem cell differentiation, and participation in tissue regeneration, relevant cells and delivery scaffolds must be paired with strategies to prevent cell death to ensure that these cells can survive to form de novo tissue. The Bcl-2 protein is a prosurvival member of a family of proteins that regulate the mitochondrial pathway of apoptosis. Using several strategies to overexpress the Bcl-2 protein, we demonstrated a decrease in the mediators of apoptosis in vitro and in vivo. This was shown through the use of two different clinical tissue repair models. Cells overexpressing Bcl-2 not only survived within the wound environment at a statistically significantly higher rate than control cells, but also increased tissue regeneration. Finally, we used a nonintegrating minicircle technology to achieve this in a potentially clinically applicable strategy for stem cell therapy.
Collapse
Affiliation(s)
- Jeong Hyun
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Cam C, Segura T. Matrix-based gene delivery for tissue repair. Curr Opin Biotechnol 2013; 24:855-63. [PMID: 23680305 DOI: 10.1016/j.copbio.2013.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 12/28/2022]
Abstract
Scaffolds for tissue repair must provide structural and biochemical cues to initiate the complex cascade of events that lead to proper tissue formation. Incorporating genes into these scaffolds is an attractive alternative to protein delivery since gene delivery can be tunable to any DNA sequence and genes utilize the cells' machinery to continuously produce therapeutic proteins, leading to longer lasting transgene expression and activation of autocrine and paracrine signaling that are not activated with bulk protein delivery. In this review, we discuss the importance of scaffold design and the impact of its design parameters (e.g. material, architecture, vector incorporation, biochemical cue presentation) on transgene expression and tissue repair.
Collapse
Affiliation(s)
- Cynthia Cam
- Department of Bioengineering, University of California, Los Angeles, United States
| | | |
Collapse
|
59
|
Gao L, Xie L, Long X, Wang Z, He CY, Chen ZY, Zhang L, Nan X, Lei H, Liu X, Liu G, Lu J, Qiu B. Efficacy of MRI visible iron oxide nanoparticles in delivering minicircle DNA into liver via intrabiliary infusion. Biomaterials 2013; 34:3688-96. [DOI: 10.1016/j.biomaterials.2013.01.094] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 01/26/2013] [Indexed: 11/15/2022]
|
60
|
Madeira C, Rodrigues CAV, Reis MSC, Ferreira FFCG, Correia RESM, Diogo MM, Cabral JMS. Nonviral Gene Delivery to Neural Stem Cells with Minicircles by Microporation. Biomacromolecules 2013; 14:1379-87. [DOI: 10.1021/bm400015b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Catarina Madeira
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Carlos A. V. Rodrigues
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Mónica S. C. Reis
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Filipa F. C. G. Ferreira
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Raquel E. S. M. Correia
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Maria M. Diogo
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Technical University of Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| |
Collapse
|
61
|
Sharma N, Cai Y, Bak RO, Jakobsen MR, Schrøder LD, Mikkelsen JG. Efficient sleeping beauty DNA transposition from DNA minicircles. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e74. [PMID: 23443502 PMCID: PMC3586802 DOI: 10.1038/mtna.2013.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
DNA transposon-based vectors have emerged as new potential delivery tools in therapeutic gene transfer. Such vectors are now showing promise in hematopoietic stem cells and primary human T cells, and clinical trials with transposon-engineered cells are on the way. However, the use of plasmid DNA as a carrier of the vector raises safety concerns due to the undesirable administration of bacterial sequences. To optimize vectors based on the Sleeping Beauty (SB) DNA transposon for clinical use, we examine here SB transposition from DNA minicircles (MCs) devoid of the bacterial plasmid backbone. Potent DNA transposition, directed by the hyperactive SB100X transposase, is demonstrated from MC donors, and the stable transfection rate is significantly enhanced by expressing the SB100X transposase from MCs. The stable transfection rate is inversely related to the size of circular donor, suggesting that a MC-based SB transposition system benefits primarily from an increased cellular uptake and/or enhanced expression which can be observed with DNA MCs. DNA transposon and transposase MCs are easily produced, are favorable in size, do not carry irrelevant DNA, and are robust substrates for DNA transposition. In accordance, DNA MCs should become a standard source of DNA transposons not only in therapeutic settings but also in the daily use of the SB system.
Collapse
Affiliation(s)
- Nynne Sharma
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
62
|
Gaspar VM, Cruz C, Queiroz JA, Pichon C, Correia IJ, Sousa F. Sensitive Detection of Peptide–Minicircle DNA Interactions by Surface Plasmon Resonance. Anal Chem 2013; 85:2304-11. [DOI: 10.1021/ac303288x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Vítor M. Gaspar
- CICS-UBI - Centro de Investigação
em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Carla Cruz
- CICS-UBI - Centro de Investigação
em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - João A. Queiroz
- CICS-UBI - Centro de Investigação
em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Chantal Pichon
- Centre de Biophysique Moléculaire
CNRS UPR4301, INSERM and University of Orléans, F-45071 Orléans cedex 2, France
| | - Ilídio J. Correia
- CICS-UBI - Centro de Investigação
em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Fani Sousa
- CICS-UBI - Centro de Investigação
em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
63
|
Wan Q, Xie L, Gao L, Wang Z, Nan X, Lei H, Long X, Chen ZY, He CY, Liu G, Liu X, Qiu B. Self-assembled magnetic theranostic nanoparticles for highly sensitive MRI of minicircle DNA delivery. NANOSCALE 2013; 5:744-752. [PMID: 23224057 DOI: 10.1039/c2nr32438e] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
As a versatile gene vector, minicircle DNA (mcDNA) has a great potential for gene therapy. However, some serious challenges remain, such as to effectively deliver mcDNA into targeted cells/tissues and to non-invasively monitor the delivery of the mcDNA. Superparamagnetic iron oxide (SPIO) nanoparticles have been extensively used for both drug/gene delivery and diagnosis. In this study, an MRI visible gene delivery system was developed with a core of SPIO nanocrystals and a shell of biodegradable stearic acid-modified low molecular weight polyethyleneimine (Stearic-LWPEI) via self-assembly. The Stearic-LWPEI-SPIO nanoparticles possess a controlled clustering structure, narrow size distribution and ultrasensitive imaging capacity. Furthermore, the nanoparticle can effectively bind with mcDNA and protect it from enzymatic degradation. In conclusion, the nanoparticle shows synergistic advantages in the effective transfection of mcDNA and non-invasive MRI of gene delivery.
Collapse
Affiliation(s)
- Qian Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Watcharanurak K, Nishikawa M, Takahashi Y, Takakura Y. Controlling the kinetics of interferon transgene expression for improved gene therapy. J Drug Target 2012; 20:764-9. [PMID: 22994266 DOI: 10.3109/1061186x.2012.716848] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
65
|
Jang ES, Park KS. Lipofectamine-2000 Assisted Magnetofection to Fibroblast Cells Using Polyethyleneimine-Fe 3O 4@SiO 2Nanoparticles. B KOREAN CHEM SOC 2012. [DOI: 10.5012/bkcs.2012.33.8.2567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
66
|
Mohsin S, Khan M, Toko H, Bailey B, Cottage CT, Wallach K, Nag D, Lee A, Siddiqi S, Lan F, Fischer KM, Gude N, Quijada P, Avitabile D, Truffa S, Collins B, Dembitsky W, Wu JC, Sussman MA. Human cardiac progenitor cells engineered with Pim-I kinase enhance myocardial repair. J Am Coll Cardiol 2012; 60:1278-87. [PMID: 22841153 DOI: 10.1016/j.jacc.2012.04.047] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 03/09/2012] [Accepted: 04/02/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The goal of this study was to demonstrate the enhancement of human cardiac progenitor cell (hCPC) reparative and regenerative potential by genetic modification for the treatment of myocardial infarction. BACKGROUND Regenerative potential of stem cells to repair acute infarction is limited. Improved hCPC survival, proliferation, and differentiation into functional myocardium will increase efficacy and advance translational implementation of cardiac regeneration. METHODS hCPCs isolated from the myocardium of heart failure patients undergoing left ventricular assist device implantation were engineered to express green fluorescent protein (hCPCe) or Pim-1-GFP (hCPCeP). Functional tests of hCPC regenerative potential were performed with immunocompromised mice by using intramyocardial adoptive transfer injection after infarction. Myocardial structure and function were monitored by echocardiographic and hemodynamic assessment for 20 weeks after delivery. hCPCe and hCPCeP expressing luciferase were observed by using bioluminescence imaging to noninvasively track persistence. RESULTS hCPCeP exhibited augmentation of reparative potential relative to hCPCe control cells, as shown by significantly increased proliferation coupled with amelioration of infarction injury and increased hemodynamic performance at 20 weeks post-transplantation. Concurrent with enhanced cardiac structure and function, hCPCeP demonstrated increased cellular engraftment and differentiation with improved vasculature and reduced infarct size. Enhanced persistence of hCPCeP versus hCPCe was revealed by bioluminescence imaging at up to 8 weeks post-delivery. CONCLUSIONS Genetic engineering of hCPCs with Pim-1 enhanced repair of damaged myocardium. Ex vivo gene delivery to modify stem cells has emerged as a viable option addressing current limitations in the field. This study demonstrates that efficacy of hCPCs from the failing myocardium can be safely and significantly enhanced through expression of Pim-1 kinase, setting the stage for use of engineered cells in pre-clinical settings.
Collapse
Affiliation(s)
- Sadia Mohsin
- SDSU Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
A simple and rapid nonviral approach to efficiently transfect primary tissue–derived cells using polyethylenimine. Nat Protoc 2012; 7:935-45. [DOI: 10.1038/nprot.2012.038] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
68
|
Lee JM, Kim BS, Lee H, Im GI. In vivo tracking of mesechymal stem cells using fluorescent nanoparticles in an osteochondral repair model. Mol Ther 2012; 20:1434-42. [PMID: 22491215 DOI: 10.1038/mt.2012.60] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We devised and tested an in vivo system to monitor the migration of mesenchymal stem cells (MSCs) within the marrow cavity. In vitro studies confirmed that platelet-derived growth factor (PDGF)-AA had the most potent chemotactic effect of the tested factors, and possessed the greatest number of receptors in MSCs. MSCs were labeled with fluorescent nanoparticles and injected into the marrow cavity of nude rats through osteochondral defects created in the distal femur. The defects were sealed with HCF (heparin-conjugated fibrin) or PDGF-AA-loaded HCF. In the HCF-only group, the nanoparticle-labeled MSCs dispersed outside the marrow cavity within 3 days after injection. In the PDGF-AA-loaded HCF group, the labeled cells moved time-dependently for 14 days toward the osteochondral defect. HCF-PDGF in low dose (LD; 8.5 ng/µl) was more effective than HCF-PDGF in high dose (HD; 17 ng/µl) in recruiting the MSCs to the osteochondral defect. After 21 days, the defects treated with PDGF and transforming growth factor (TGF)-β1-loaded HCF showed excellent cartilage repair compared with other groups. Further studies confirmed that this in vivo osteochondral MSCs tracking system (IOMTS) worked for other chemoattractants (chemokine (C-C motif) ligand 2 (CCL2) and PDGF-BB). IOMTS can provide a useful tool to examine the effect of growth factors or chemokines on endogenous cartilage repair.
Collapse
Affiliation(s)
- Jong Min Lee
- Department of Orthopaedics, Dongguk University Ilsan Hospital, Goyang, Korea
| | | | | | | |
Collapse
|
69
|
Levi B, Hyun JS, Nelson ER, Li S, Montoro DT, Wan DC, Jia FJ, Glotzbach JC, James AW, Lee M, Huang M, Quarto N, Gurtner GC, Wu JC, Longaker MT. Nonintegrating knockdown and customized scaffold design enhances human adipose-derived stem cells in skeletal repair. Stem Cells 2012; 29:2018-29. [PMID: 21997852 DOI: 10.1002/stem.757] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An urgent need exists in clinical medicine for suitable alternatives to available techniques for bone tissue repair. Human adipose-derived stem cells (hASCs) represent a readily available, autogenous cell source with well-documented in vivo osteogenic potential. In this article, we manipulated Noggin expression levels in hASCs using lentiviral and nonintegrating minicircle short hairpin ribonucleic acid (shRNA) methodologies in vitro and in vivo to enhance hASC osteogenesis. Human ASCs with Noggin knockdown showed significantly increased bone morphogenetic protein (BMP) signaling and osteogenic differentiation both in vitro and in vivo, and when placed onto a BMP-releasing scaffold embedded with lentiviral Noggin shRNA particles, hASCs more rapidly healed mouse calvarial defects. This study therefore suggests that genetic targeting of hASCs combined with custom scaffold design can optimize hASCs for skeletal regenerative medicine.
Collapse
Affiliation(s)
- Benjamin Levi
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, California 94305-5148, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Hsu CYM, Uludağ H. Nucleic-acid based gene therapeutics: delivery challenges and modular design of nonviral gene carriers and expression cassettes to overcome intracellular barriers for sustained targeted expression. J Drug Target 2012; 20:301-28. [PMID: 22303844 DOI: 10.3109/1061186x.2012.655247] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The delivery of nucleic acid molecules into cells to alter physiological functions at the genetic level is a powerful approach to treat a wide range of inherited and acquired disorders. Biocompatible materials such as cationic polymers, lipids, and peptides are being explored as safer alternatives to viral gene carriers. However, the comparatively low efficiency of nonviral carriers currently hampers their translation into clinical settings. Controlling the size and stability of carrier/nucleic acid complexes is one of the primary hurdles as the physicochemical properties of the complexes can define the uptake pathways, which dictate intracellular routing, endosomal processing, and nucleocytoplasmic transport. In addition to nuclear import, subnuclear trafficking, posttranscriptional events, and immune responses can further limit transfection efficiency. Chemical moieties, reactive linkers or signal peptide have been conjugated to carriers to prevent aggregation, induce membrane destabilization and localize to subcellular compartments. Genetic elements can be inserted into the expression cassette to facilitate nuclear targeting, delimit expression to targeted tissue, and modulate transgene expression. The modular option afforded by both gene carriers and expression cassettes provides a two-tier multicomponent delivery system that can be optimized for targeted gene delivery in a variety of settings.
Collapse
Affiliation(s)
- Charlie Yu Ming Hsu
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Cananda
| | | |
Collapse
|
71
|
Van Overstraeten-Schlögel N, Shim YH, Tevel V, Piel G, Piette J, Dubois P, Raes M. Assessment of new biocompatible poly(N-(morpholino)ethyl methacrylate)-based copolymers by transfection of immortalized keratinocytes. Drug Deliv 2012; 19:112-22. [PMID: 22239537 DOI: 10.3109/10717544.2011.649219] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Skin carcinomas are among the most commonly diagnosed tumors in the world. In this study, we investigated the transfection of immortalized keratinocytes, used as an in vitro model for skin carcinoma, using the antisense technology and poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA)-based copolymers. In order to improve the transfection efficiency of the classic PDMAEMA polymers, copolymers were synthesized including a poly(N-morpholino)ethylmethacrylate) (PMEMA) moiety for an improved proton-sponge effect, intended to favour the release of the oligonucleotide from the acidic endosome. These copolymers were synthesized either statistically (with alternating PDMAEMA and PMEMA fragments) or in blocks (one PDMAEMA block followed by one PMEMA block). MTT assays were performed using the PDMAEMA-PMEMA copolymers and revealed no significant cytotoxicity of these polymers at an N/P ratio of 7.3. Using fluorescent oligonucleotides and analyzing transfection efficiency by flow cytometry, we noticed no significant differences between the two kinds of copolymers. However copolymers with a higher DMAEMA content and a higher Mn were also those displaying the highest vectorization efficiency. Confocal microscopy showed that these copolymers induced a fine granular distribution of the transfected antisense oligonucleotides inside the cells. We also assessed the functionality of the transfected antisense oligonucleotide by transfecting immortalized GFP expressing keratinocytes with a GFP antisense oligonucleotide using these copolymers. A significant silencing was achieved with a PDMAEMA-PMEMA in block copolymer (Mn=41,000, 89 % PDMAEMA). Together, these results suggest that PDMAEMA-PMEMA copolymers combining low toxicity, vectorization and proton sponge properties, can be efficiently used to transfect immortalized keratinocytes and so open new perspectives in the therapy of skin carcinomas as well as of other skin diseases of genetic or immunological origin.
Collapse
|
72
|
Xu BL, Yuan L, Wu JX, Xu N, Fang WJ, Zhao P, Huang WL. Mc-hES, a novel plasmid carrying human endostatin gene, inhibits nasopharyngeal carcinoma growth. Cancer Gene Ther 2011; 19:110-7. [PMID: 22158523 DOI: 10.1038/cgt.2011.72] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Conventional plasmids for gene therapy produce low-level and short-term gene expression. Here, we first created minicircle carrying endostatin (mc-hES) for measurement of transfection efficiency. Compared with pcDNA-hES, MC-mediated endostatin gene transfer in vitro resulted in seven-fold greater endostatin expression levels in transfected cells and inhibited the growth of Human umbilical vein endothelial cells (HUVEC) more efficiently. HUVEC cell migration and tube-formation assays suggested that MC-mediated endostatin gene has significant anti-migration and anti-tube-formation capacity than that in pcDNA-hES. In vivo experiments showed that after transfection, mc-hES inhibited the growth of nasopharyngeal carcinoma xenografts. The tumor inhibition rates of mc-hES and pcDNA-hES were 60.8% and 26.9%, respectively (P<0.05). MC-mediated intratumoral endostatin expression in vivo was 2.2-17.9 times higher than pcDNA-hES in xenografted mice and lasted for 20 days. Our results suggest that minicircle DNA vectors might be a promising vector for biotherapy and should be further investigated.
Collapse
Affiliation(s)
- B-L Xu
- The Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, PRC
| | | | | | | | | | | | | |
Collapse
|
73
|
Vandermeulen G, Marie C, Scherman D, Préat V. New generation of plasmid backbones devoid of antibiotic resistance marker for gene therapy trials. Mol Ther 2011; 19:1942-9. [PMID: 21878901 PMCID: PMC3222533 DOI: 10.1038/mt.2011.182] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 08/03/2011] [Indexed: 12/29/2022] Open
Abstract
Since it has been established that the injection of plasmid DNA can lead to an efficient expression of a specific protein in vivo, nonviral gene therapy approaches have been considerably improved, allowing clinical trials. However, the use of antibiotic resistance genes as selection markers for plasmid production raises safety concerns which are often pointed out by the regulatory authorities. Indeed, a horizontal gene transfer to patient's bacteria cannot be excluded, and residual antibiotic in the final product could provoke allergic reactions in sensitive individuals. A new generation of plasmid backbones devoid of antibiotic resistance marker has emerged to increase the safety profile of nonviral gene therapy trials. This article reviews the existing strategies for plasmid maintenance and, in particular, those that do not require the use of antibiotic resistance genes. They are based either on the complementation of auxotrophic strain, toxin-antitoxin systems, operator-repressor titration, RNA markers, or on the overexpression of a growth essential gene. Minicircles that allow removing of the antibiotic resistance gene from the initial vector will also be discussed. Furthermore, reported use of antibiotic-free plasmids in preclinical or clinical studies will be listed to provide a comprehensive view of these innovative technologies.
Collapse
Affiliation(s)
- Gaëlle Vandermeulen
- Université catholique de Louvain, Louvain Drug Research Institute, Pharmaceutics and Drug Delivery, Brussels, Belgium
| | | | | | | |
Collapse
|
74
|
de Almeida PE, van Rappard JRM, Wu JC. In vivo bioluminescence for tracking cell fate and function. Am J Physiol Heart Circ Physiol 2011; 301:H663-71. [PMID: 21666118 DOI: 10.1152/ajpheart.00337.2011] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tracking the fate and function of cells in vivo is paramount for the development of rational therapies for cardiac injury. Bioluminescence imaging (BLI) provides a means for monitoring physiological processes in real time, ranging from cell survival to gene expression to complex molecular processes. In mice and rats, BLI provides unmatched sensitivity because of the absence of endogenous luciferase expression in mammalian cells and the low background luminescence emanating from animals. In the field of stem cell therapy, BLI provides an unprecedented means to monitor the biology of these cells in vivo, giving researchers a greater understanding of their survival, migration, immunogenicity, and potential tumorigenicity in a living animal. In addition to longitudinal monitoring of cell survival, BLI is a useful tool for semiquantitative measurements of gene expression in vivo, allowing a better optimization of drug and gene therapies. Overall, this technology not only enables rapid, reproducible, and quantitative monitoring of physiological processes in vivo but also can measure the influences of therapeutic interventions on the outcome of cardiac injuries.
Collapse
Affiliation(s)
- Patricia E de Almeida
- Department of Medicine, Stanford University School of Medicine, Stanford, California 94305-5454, USA
| | | | | |
Collapse
|
75
|
Zuo Y, Wu J, Xu Z, Yang S, Yan H, Tan L, Meng X, Ying X, Liu R, Kang T, Huang W. Minicircle-oriP-IFNγ: a novel targeted gene therapeutic system for EBV positive human nasopharyngeal carcinoma. PLoS One 2011; 6:e19407. [PMID: 21573215 PMCID: PMC3088667 DOI: 10.1371/journal.pone.0019407] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 03/31/2011] [Indexed: 12/15/2022] Open
Abstract
Background Nonviral vectors are attractively used for gene therapy owing to their distinctive advantages. Our previous study has demonstrated that transfer of human IFNγ gene into nasopharyngeal carcinoma (NPC) by using a novel nonviral vector, minicircle (mc), under the control of cytomegalovirus (CMV) promoter was effective to inhibit tumor growth. However, therapies based on CMV promoter cannot express the targeted genes in cancer tissues. Previous studies indicated that the development of human NPC was closely associated with Epstein-Barr virus (EBV) and demonstrated the transcriptional enhancer function of oriP when bound by EBV protein. Therefore, the present study is to explore the targeted gene expression and the anti-tumor effect of a novel tumor-specific gene therapeutic system (mc-oriP-IFNγ) in which the transgene expression was under the transcriptional regulation of oriP promoter. Methodology/Principal Findings Dual-luciferase reporter assay and ELISA were used to assess the expression of luciferase and IFNγ. WST assay was used to assess the cell proliferation. RT-PCR was used to detect the mRNA level of EBNA1. RNAi was used to knockdown the expression of EBNA1. NPC xenograft models in nude mice were used to investigate the targeted antitumor efficacy of mc-oriP-IFNγ. Immunohistochemistry was used to detect the expression and the activity of the IFNγ in tumor sections. Our results demonstrated that mc-oriP vectors mediated comparable gene expression and anti-proliferative effect in the EBV-positive NPC cell line C666-1 compared to mc-CMV vectors. Furthermore, mc-oriP vectors exhibited much lower killing effects on EBV-negative cell lines compared to mc-CMV vectors. The targeted expression of mc-oriP vectors was inhibited by EBNA1-siRNA in C666-1. This selective expression was corroborated in EBV-positive and -negative tumor models. Conclusions/Significance This study demonstrates the feasibility of mc-oriP-IFNγ as a safe and highly effective targeted gene therapeutic system for the treatment of EBV positive NPC.
Collapse
Affiliation(s)
- Yufang Zuo
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jiangxue Wu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zumin Xu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Department of Radiation Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Shiping Yang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haijiao Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Li Tan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiangqi Meng
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiaofang Ying
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ranyi Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Institute of Microbiology, Chinese Academy of Science, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
76
|
Narsinh K, Narsinh KH, Wu JC. Derivation of human induced pluripotent stem cells for cardiovascular disease modeling. Circ Res 2011; 108:1146-56. [PMID: 21527744 PMCID: PMC3098466 DOI: 10.1161/circresaha.111.240374] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 03/17/2011] [Indexed: 01/29/2023]
Abstract
The successful derivation of human induced pluripotent stem cells (hiPSCs) by dedifferentiation of somatic cells offers significant potential to overcome obstacles in the field of cardiovascular disease. hiPSC derivatives offer incredible potential for new disease models and regenerative medicine therapies. However, many questions remain regarding the optimal starting materials and methods to enable safe, efficient derivation of hiPSCs suitable for clinical applications. Initial reprogramming experiments were carried out using lentiviral or retroviral gene delivery methods. More recently, various nonviral methods that avoid permanent and random transgene insertion have emerged as alternatives. These include transient DNA transfection using plasmids or minicircles, protein transduction, or RNA transfection. In addition, several small molecules have been found to significantly augment hiPSC derivation efficiency, allowing the use of a fewer number of genes during pluripotency induction. We review these various methods for the derivation of hiPSCs, focusing on their ultimate clinical applicability, with an emphasis on their potential for use as cardiovascular therapies and disease-modeling platforms.
Collapse
Affiliation(s)
- Kamileh Narsinh
- Department of Medicine, Stanford University School of Medicine
- Department of Radiology, Stanford University School of Medicine
- Department of Chemistry and Biochemistry, University of California, Los Angeles
| | - Kazim H. Narsinh
- Department of Medicine, Stanford University School of Medicine
- Department of Radiology, Stanford University School of Medicine
| | - Joseph C. Wu
- Department of Medicine, Stanford University School of Medicine
- Department of Radiology, Stanford University School of Medicine
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| |
Collapse
|
77
|
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5111, USA
| | | |
Collapse
|
78
|
Kay MA, He CY, Chen ZY. A robust system for production of minicircle DNA vectors. Nat Biotechnol 2010; 28:1287-9. [PMID: 21102455 PMCID: PMC4144359 DOI: 10.1038/nbt.1708] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 10/15/2010] [Indexed: 12/02/2022]
Abstract
Minicircle DNA vectors allow sustained transgene expression in quiescent cells and tissues. To improve minicircle production, we genetically modified Escherichia coli to construct a producer strain that stably expresses a set of inducible minicircle-assembly enzymes, ΦC31 integrase and I-SceI homing endonuclease. This bacterial strain produces purified minicircles in a time frame and quantity similar to those of routine plasmid DNA preparation, making it feasible to use minicircles in place of plasmids in mammalian transgene expression studies.
Collapse
Affiliation(s)
- Mark A Kay
- Correspondence should be address to MAK () or ZYC ()
| | - Cheng-Yi He
- Department of Pediatrics and Genetics
- The Center for Clinical Science Research, Room 2105, Stanford University, 269 Campus Drive, Stanford, California 94305-5164, USA
| | - Zhi-Ying Chen
- Correspondence should be address to MAK () or ZYC ()
| |
Collapse
|
79
|
Hu S, Huang M, Li Z, Jia F, Ghosh Z, Lijkwan MA, Fasanaro P, Sun N, Wang X, Martelli F, Robbins RC, Wu JC. MicroRNA-210 as a novel therapy for treatment of ischemic heart disease. Circulation 2010; 122:S124-31. [PMID: 20837903 PMCID: PMC2952325 DOI: 10.1161/circulationaha.109.928424] [Citation(s) in RCA: 365] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND MicroRNAs are involved in various critical functions, including the regulation of cellular differentiation, proliferation, angiogenesis, and apoptosis. We hypothesize that microRNA-210 can rescue cardiac function after myocardial infarction by upregulation of angiogenesis and inhibition of cellular apoptosis in the heart. METHODS AND RESULTS Using microRNA microarrays, we first showed that microRNA-210 was highly expressed in live mouse HL-1 cardiomyocytes compared with apoptotic cells after 48 hours of hypoxia exposure. We confirmed by polymerase chain reaction that microRNA-210 was robustly induced in these cells. Gain-of-function and loss-of-function approaches were used to investigate microRNA-210 therapeutic potential in vitro. After transduction, microRNA-210 can upregulate several angiogenic factors, inhibit caspase activity, and prevent cell apoptosis compared with control. Afterward, adult FVB mice underwent intramyocardial injections with minicircle vector carrying microRNA-210 precursor, minicircle carrying microRNA-scramble, or sham surgery. At 8 weeks, echocardiography showed a significant improvement of left ventricular fractional shortening in the minicircle vector carrying microRNA-210 precursor group compared with the minicircle carrying microRNA-scramble control. Histological analysis confirmed decreased cellular apoptosis and increased neovascularization. Finally, 2 potential targets of microRNA-210, Efna3 and Ptp1b, involved in angiogenesis and apoptosis were confirmed through additional experimental validation. CONCLUSIONS MicroRNA-210 can improve angiogenesis, inhibit apoptosis, and improve cardiac function in a murine model of myocardial infarction. It represents a potential novel therapeutic approach for treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Shijun Hu
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | - Mei Huang
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | - Zongjin Li
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | - Fangjun Jia
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | - Zhumur Ghosh
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | | | | | - Ning Sun
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| | - Xi Wang
- Department of Cardiothoracic Surgery, Stanford, CA, USA
| | | | | | - Joseph C. Wu
- Department of Medicine, Division of Cardiology, Stanford, CA, USA
- Department of Radiology, Molecular Imaging Program, Stanford, CA, USA
| |
Collapse
|
80
|
Abstract
Ocular gene therapy is becoming a well-established field. Viral gene therapies for the treatment of Leber's congentinal amaurosis (LCA) are in clinical trials, and many other gene therapy approaches are being rapidly developed for application to diverse ophthalmic pathologies. Of late, development of non-viral gene therapies has been an area of intense focus and one technology, polymer-compacted DNA nanoparticles, is especially promising. However, development of pharmaceutically and clinically viable therapeutics depends not only on having an effective and safe vector but also on a practical treatment strategy. Inherited retinal pathologies are caused by mutations in over 220 genes, some of which contain over 200 individual disease-causing mutations, which are individually very rare. This review will focus on both the progress and future of nanoparticles and also on what will be required to make them relevant ocular pharmaceutics.
Collapse
Affiliation(s)
- Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center
| | - Muna I. Naash
- Department of Cell Biology, University of Oklahoma Health Sciences Center
| |
Collapse
|