51
|
Rijnkels JM, Whiteley LO, Beijersbergen van Henegouwen GMJ. Time and Dose-related Ultraviolet B Damage in Viable Pig Skin Explants Held in a Newly Developed Organ Culture System¶. Photochem Photobiol 2007. [DOI: 10.1562/0031-8655(2001)0730499tadrub2.0.co2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
52
|
Fuller DH, Loudon P, Schmaljohn C. Preclinical and clinical progress of particle-mediated DNA vaccines for infectious diseases. Methods 2007; 40:86-97. [PMID: 16997717 DOI: 10.1016/j.ymeth.2006.05.022] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2006] [Accepted: 05/10/2006] [Indexed: 11/23/2022] Open
Abstract
This review provides an overview of studies employing particle-mediated epidermal delivery (PMED) or the gene gun to administer DNA vaccines for infectious diseases in preclinical studies employing large animal models and in human clinical trials. It reviews the immunogenicity and protective efficacy of PMED DNA vaccines in nonhuman primates and swine and studies that have directly compared the effectiveness of PMED in these large animal models to existing licensed vaccines and intramuscular or intradermal delivery of DNA vaccines with a needle. Various clinical trials employing PMED have been completed and an overview of the immunogenicity, safety, and tolerability of this approach in humans is described. Finally, efforts currently in progress for commercial development of particle-mediated DNA vaccines are discussed.
Collapse
Affiliation(s)
- Deborah H Fuller
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, School of Medicine, 260 Kappa Drive, PA 15238, USA.
| | | | | |
Collapse
|
53
|
Scheerlinck JPY, Greenwood DLV. Particulate delivery systems for animal vaccines. Methods 2007; 40:118-24. [PMID: 16997719 DOI: 10.1016/j.ymeth.2006.05.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Accepted: 05/05/2006] [Indexed: 11/28/2022] Open
Abstract
The requirements for veterinary vaccines are different to those of human vaccines. Indeed, while more side effects can be tolerated in animals than in humans; there are stricter requirements in terms of cost, ease of delivery (including to wildlife), and a need to develop vaccines in species for which relatively little is known in terms of molecular immunology. By their nature particulate vaccine delivery systems are well suited to address these challenges. Here, we review particulate vaccine delivery systems, ranging from cm-sized long-distance ballistic devices to nano-bead technology for veterinary species and wildlife.
Collapse
|
54
|
Chauhan A, Tikoo A, Kapur AK, Singh M. The taming of the cell penetrating domain of the HIV Tat: myths and realities. J Control Release 2006; 117:148-62. [PMID: 17196289 PMCID: PMC1859861 DOI: 10.1016/j.jconrel.2006.10.031] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 10/20/2006] [Indexed: 01/08/2023]
Abstract
Protein transduction with cell penetrating peptides over the past several years has been shown to be an effective way of delivering proteins in vitro and now several reports have also shown valuable in vivo applications in correcting disease states. An impressive bioinspired phenomenon of crossing biological barriers came from HIV transactivator Tat protein. Specifically, the protein transduction domain of HIV Tat has been shown to be a potent pleiotropic peptide in protein delivery. Various approaches such as molecular modeling, arginine guanidinium head group structural strategy, multimerization of PTD sequence and phage display system have been applied for taming of the PTD. This has resulted in identification of PTD variants which are efficient in cell membrane penetration and cytoplasmic delivery. In spite of these state of the art technologies, the dilemma of low protein transduction efficiency and target specific delivery of PTD fusion proteins remains unsolved. Moreover, some misconceptions about PTD of Tat in the literature require considerations. We have assembled critical information on secretory, plasma membrane penetration and transcellular properties of Tat and PTD using molecular analysis and available experimental evidences.
Collapse
Affiliation(s)
- Ashok Chauhan
- Department of Neurology, Richard Johnson Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, 509 Pathology, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|
55
|
Andreadis ST. Gene-modified tissue-engineered skin: the next generation of skin substitutes. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2006; 103:241-74. [PMID: 17195466 DOI: 10.1007/10_023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tissue engineering combines the principles of cell biology, engineering and materials science to develop three-dimensional tissues to replace or restore tissue function. Tissue engineered skin is one of most advanced tissue constructs, yet it lacks several important functions including those provided by hair follicles, sebaceous glands, sweat glands and dendritic cells. Although the complexity of skin may be difficult to recapitulate entirely, new or improved functions can be provided by genetic modification of the cells that make up the tissues. Gene therapy can also be used in wound healing to promote tissue regeneration or prevent healing abnormalities such as formation of scars and keloids. Finally, gene-enhanced skin substitutes have great potential as cell-based devices to deliver therapeutics locally or systemically. Although significant progress has been made in the development of gene transfer technologies, several challenges have to be met before clinical application of genetically modified skin tissue. Engineering challenges include methods for improved efficiency and targeted gene delivery; efficient gene transfer to the stem cells that constantly regenerate the dynamic epidermal tissue; and development of novel biomaterials for controlled gene delivery. In addition, advances in regulatable vectors to achieve spatially and temporally controlled gene expression by physiological or exogenous signals may facilitate pharmacological administration of therapeutics through genetically engineered skin. Gene modified skin substitutes are also employed as biological models to understand tissue development or disease progression in a realistic three-dimensional context. In summary, gene therapy has the potential to generate the next generation of skin substitutes with enhanced capacity for treatment of burns, chronic wounds and even systemic diseases.
Collapse
Affiliation(s)
- Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical & Biological Engineering, University at Buffalo, The State University of New York (SUNY), Amherst, NY 14260, USA.
| |
Collapse
|
56
|
Branski LK, Herndon DN, Jeschke MG. Gentherapie mit Wachstumsfaktoren – ein neuer therapeutischer Ansatz für akute und chronische Wunden? Visc Med 2006. [DOI: 10.1159/000094885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
57
|
Gene technology and tissue engineering. MINIM INVASIV THER 2006; 11:93-9. [PMID: 16754057 DOI: 10.1080/136457002320174159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The interest in gene therapy to treat human diseases has increased with the advances in recombinant DNA technology and the improved physical, chemical and biological methods of delivering genes to mammalian cells. Areas of therapeutic interest for gene therapy relevant for tissue engineering are, for example, in the treatment of wounds, skin diseases, nerve, bone, and muscle diseases. The transfer of a gene into a cell can lead to the addition or modification of a function and may be an attractive alternative to the pharmacological use of proteins. The complementation of defective functions could also be an effective treatment for inherited skin diseases with a gene defect. The two major challenges facing gene technology in tissue engineering are the problem of identifying appropriate genes that are effective in tissue repair, and the reliable expression of the therapeutic gene at clinically beneficial levels. This review discusses principles and methods of delivering genes encoding growth factors into cells, together with their respective advantages and disadvantages.
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW Peanut allergy is among the most serious, life-threatening food sensitivities, and recent studies indicate increasing prevalence, particularly among children. Our objective is to highlight recent advances in the immunology and treatment of peanut allergy. RECENT FINDINGS Peanut sensitization may be both a Th1- and Th2-driven process, and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) may play a role in regulating the response intensity. Preliminary work shows that the food matrix is important in the immune response to peanut and that purified peanut allergens may have little intrinsic stimulatory capacity. Studies characterizing peanut allergens have revealed Ara h 1 and Ara h 2 as the most potent allergens, but Ara h 3 may be more allergenic than previously thought. There appears to be a relationship between the diversity of IgE-binding patterns and the severity of clinical symptoms. Multiple novel approaches to treatment are being investigated, which include traditional Chinese medicine, various forms of modified immunotherapy and the use of adjuvants in modified immunotherapy. SUMMARY By understanding the immunologic response to peanut and the roles of the major peanut allergens, it may be possible to predict those at risk for severe reactions, prevent peanut sensitization and effectively treat those already sensitized.
Collapse
Affiliation(s)
- Kricia Palmer
- Duke University Medical Center, Pediatric Allergy and Immunology, Durham, New Carolina 27710, USA
| | | |
Collapse
|
59
|
Birchall J, Coulman S, Anstey A, Gateley C, Sweetland H, Gershonowitz A, Neville L, Levin G. Cutaneous gene expression of plasmid DNA in excised human skin following delivery via microchannels created by radio frequency ablation. Int J Pharm 2006; 312:15-23. [PMID: 16469457 DOI: 10.1016/j.ijpharm.2005.12.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 12/05/2005] [Accepted: 12/05/2005] [Indexed: 01/09/2023]
Abstract
The skin is a valuable organ for the development and exploitation of gene medicines. Delivering genes to skin is restricted however by the physico-chemical properties of DNA and the stratum corneum (SC) barrier. In this study, we demonstrate the utility of an innovative technology that creates transient microconduits in human skin, allowing DNA delivery and resultant gene expression within the epidermis and dermis layers. The radio frequency (RF)-generated microchannels were of sufficient morphology and depth to permit the epidermal delivery of 100 nm diameter nanoparticles. Model fluorescent nanoparticles were used to confirm the capacity of the channels for augmenting diffusion of macromolecules through the SC. An ex vivo human organ culture model was used to establish the gene expression efficiency of a beta-galactosidase reporter plasmid DNA applied to ViaDerm treated skin. Skin treated with ViaDerm using 50 microm electrode arrays promoted intense levels of gene expression in the viable epidermis. The intensity and extent of gene expression was superior when ViaDerm was used following a prior surface application of the DNA formulation. In conclusion, the RF-microchannel generator (ViaDerm) creates microchannels amenable for delivery of nanoparticles and gene therapy vectors to the viable region of skin.
Collapse
Affiliation(s)
- James Birchall
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3XF, and Gwent Healthcare NHS Trust, Royal Gwent Hospital, South Wales, UK.
| | | | | | | | | | | | | | | |
Collapse
|
60
|
López E, del Pozo V, Miguel T, Sastre B, Seoane C, Civantos E, Llanes E, Baeza ML, Palomino P, Cárdaba B, Gallardo S, Manzarbeitia F, Zubeldia JM, Lahoz C. Inhibition of chronic airway inflammation and remodeling by galectin-3 gene therapy in a murine model. THE JOURNAL OF IMMUNOLOGY 2006; 176:1943-50. [PMID: 16424226 DOI: 10.4049/jimmunol.176.3.1943] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously demonstrated that treatment of acute asthmatic rats with gene therapy using plasmid-encoding Galectin-3 (Gal-3) resulted in an improvement of cellular and functional respiratory parameters. The next question that we wanted to clarify was if in a chronic situation where the treated animal continues to inhale the Ag, does this procedure prevent the chronicity and the remodeling? Chronic inflammation was induced by intranasal administration of OVA over a period of 12 wk. In the treated group, the Gal-3 gene was introduced by intranasal instillation in 50 mul of plasmid-encoding Gal-3. Noninvasive airway responsiveness to methacholine was tested at different times. Cells were obtained by bronchoalveolar lavage and used for RNA extraction and cytometric studies. Eosinophils were counted in blood and bronchoalveolar lavage fluid. Real-time PCR was used to measure Gal-3 and cytokine mRNA expression in lung. Lungs were paraffined and histologic analyses were performed (H&E, periodic acid-Schiff, and Masson Trichrome stain). Our results showed that 12 wk after the first intranasal Ag instillation in chronically asthmatic mice, treatment with the Gal-3 gene led to an improvement in the eosinophil count and the normalization of hyperresponsiveness to methacholine. Concomitantly, this treatment resulted in an improvement in mucus secretion and subepithelial fibrosis in the chronically asthmatic mice, with a quantitatively measured reduction in lung collagen, a prominent feature of airway remodeling. Plasmid-encoding Gal-3 acts as a novel treatment for chronic asthma in mice producing nearly complete blockade of Ag responses with respect to eosinophil airway accumulation, airway hyperresponsiveness, and remodeling.
Collapse
Affiliation(s)
- Esther López
- Immunology Department, Fundación Jiménez Díaz Capio, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Jeschke MG, Sandmann G, Finnerty CC, Herndon DN, Pereira CT, Schubert T, Klein D. The structure and composition of liposomes can affect skin regeneration, morphology and growth factor expression in acute wounds. Gene Ther 2006; 12:1718-24. [PMID: 16034455 DOI: 10.1038/sj.gt.3302582] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Liposomal gene transfer is an effective therapeutic approach to improve dermal and epidermal regeneration. The purpose of the present study was to define whether the biological or chemical structure of a liposome influences cellular and biological regeneration in the skin, and to determine by which mechanisms possible changes occur. Rats were inflicted a full-excision acute wound and divided into three groups to receive weekly subcutaneous injections of DMRIE liposomes plus the Lac Z gene, or DOTAP/Chol liposomes plus the Lac Z gene, or saline. Planimetry, immunological assays, histological and immunohistochemical techniques were used to determine cellular responses after gene transfer, protein expression, dermal and epidermal regeneration. DOTAP/Chol increased IGF-I and KGF protein concentration and caused concomitant cellular responses, for example, by increasing IGFBP-3, P<0.05. DOTAP/Chol liposomes improved epidermal regeneration by exhibiting the most rapid area and linear wound re-epithelization compared to DMRIE or control, P<0.001. DOTAP/Chol and DMRIE exerted promitogenic and antiapoptotic effects on basal keratinocytes, P<0.05. Dermal regeneration was improved in DOTAP/Chol-treated animals by an increased collagen deposition and morphology, P<0.001. DOTAP/Chol liposomes increased vascular endothelial growth factor concentrations and thus neovascularization when compared with DMRIE and saline, P<0.001. In the present study, we showed that different liposomes have different effects on intracellular and biological responses based on its chemical and molecular structure. For gene transfer in acute wounds, the administration of DOTAP/Chol liposomes appears to be beneficial.
Collapse
Affiliation(s)
- M G Jeschke
- Galveston Burns Unit and Department of Surgery, Shriners Hospital for Children, University Texas Medical Branch, TX 77550, USA
| | | | | | | | | | | | | |
Collapse
|
62
|
Cardones AR, Leitner WW, Fang L, Murakami T, Kapoor V, Udey MC, Hwang ST. Genetic immunization with LYVE-1 cDNA yields function-blocking antibodies against native protein. Microvasc Res 2006; 71:32-9. [PMID: 16257423 DOI: 10.1016/j.mvr.2005.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Revised: 09/22/2005] [Accepted: 09/22/2005] [Indexed: 11/24/2022]
Abstract
LYVE-1 is a surface bound hyaluronic acid (HA) receptor that is preferentially expressed by lymphatic endothelial cells (LEC). cDNA encoding full-length human LYVE-1 was coated onto gold particles that were then delivered via helium-assisted jet propulsion (gene gun) into the skin of Balb/C mice. LYVE-1 antisera, but not control pre-immune sera, recognized LYVE-1-transfected 293T cells by flow cytometry. While 40-70% of cultured human dermal microvascular endothelial cells (HMEC) were positive for LYVE-1 staining, human lung microvascular endothelial cells (LMEC) were negative. LYVE-1 antisera was used to effectively separate HMEC into LYVE-1 (hi) and LYVE-1(lo) populations that were enriched or depleted, respectively, for podoplanin, another LEC marker. By immunohistochemistry, LYVE-1 antisera detected CD31(lo) podoplanin(hi) lymphatic channels in normal and psoriatic human skin as well as in human tonsil. LYVE-1 antisera also blocked binding of FITC-labeled HA to HMEC (but not LMEC), demonstrating that these antibodies recognized regions of LYVE-1 required for HA binding. In summary, gene gun-assisted delivery of cDNA encoding LYVE-1 into skin resulted in reliable production of antisera that specifically and functionally recognized native LYVE-1 protein.
Collapse
Affiliation(s)
- Adela R Cardones
- Dermatology, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 12N258, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
63
|
Yang L, Shirakata Y, Tamai K, Dai X, Hanakawa Y, Tokumaru S, Yahata Y, Tohyama M, Shiraishi K, Nagai H, Wang X, Murakami S, Sayama K, Kaneda Y, Hashimoto K. Microbubble-enhanced ultrasound for gene transfer into living skin equivalents. J Dermatol Sci 2005; 40:105-14. [PMID: 16111869 DOI: 10.1016/j.jdermsci.2005.07.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Revised: 07/01/2005] [Accepted: 07/11/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Gene transfer to skin is an attractive therapeutic approach because of the accessibility of the skin and the high rate of cure for many cutaneous diseases. However, safety concerns over viral vectors and the low efficiency of most non-viral gene transfer techniques have encumbered their clinical application for gene transfer. By contrast, efficient gene transfers into various cell types using microbubble-enhanced ultrasound has been reported. OBJECTIVES The purpose of this study was to investigate whether ultrasound with microbubble enhancement allowed effective transfer of foreign genes into living skin equivalents (LSEs). METHODS Microbubbles and plasmid DNA encoding green fluorescent protein (GFP) were added to the dermal-epidermal junctions of LSEs, which were then exposed to ultrasound. The LSEs were harvested at different time points to investigate transgene expression using confocal laser microscopy. Transfected LSEs were also transplanted onto nude mice, and the in vivo transgene expression was observed. RESULTS From days 2 to 7 after transfection, most GFP-positive cells continued to migrate upward from the basal layer, while other GFP-positive cells lagged behind or remained in the basal layer on days 5 and 7. Transfection resulted in 20-30% GFP-positive cells. Multiple transfections further increased the percentage of transfected cells and resulted in multi-layer transgene expression. Grafts from the transfected LSEs survived on nude mice and continued to express GFP up to 2 weeks post-transplantation. CONCLUSION Gene transfer into LSE using ultrasound with microbubble enhancement is an effective alternative to viral and non-viral methods.
Collapse
Affiliation(s)
- Lujun Yang
- Department of Dermatology, Ehime University School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Petrie NC, Vranckx JJ, Hoeller D, Yao F, Eriksson E. Gene delivery of PDGF for wound healing therapy. J Tissue Viability 2005; 15:16-21. [PMID: 16302501 DOI: 10.1016/s0965-206x(05)54002-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nicola C Petrie
- Laboratory of Wound Repair and Gene Transfer, Division of Plastic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | |
Collapse
|
65
|
Wolff JA, Budker V. The mechanism of naked DNA uptake and expression. ADVANCES IN GENETICS 2005; 54:3-20. [PMID: 16096005 DOI: 10.1016/s0065-2660(05)54001-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The administration of naked nucleic acids into animals is increasingly being used as a research tool to elucidate mechanisms of gene expression and the role of genes and their cognate proteins in the pathogenesis of disease in animal models (Herweijer and Wolff, 2003; Hodges and Scheule, 2003). It is also being used in several human clinical trials for genetic vaccines, Duchenne muscular dystrophy, peripheral limb ischemia, and cardiac ischemia (Davis et al., 1996; Romero et al., 2002; Tsurumi et al., 1997). Naked DNA is an attractive non-viral vector because of its inherent simplicity and because it can easily be produced in bacteria and manipulated using standard recombinant DNA techniques. It shows very little dissemination and transfection at distant sites following delivery and can be readministered multiple times into mammals (including primates) without inducing an antibody response against itself (i.e., no anti-DNA antibodies generated) (Jiao et al., 1992). Also, contrary to common belief, long-term foreign gene expression from naked plasmid DNA (pDNA) is possible even without chromosome integration if the target cell is postmitotic (as in muscle) or slowly mitotic (as in hepatocytes) and if an immune reaction against the foreign protein is not generated (Herweijer et al., 2001; Miao et al., 2000; Wolff et al., 1992; Zhang et al., 2004). With the advent of intravascular and electroporation techniques, its major restriction--poor expression levels--is no longer limiting and levels of foreign gene expression in vivo are approaching what can be achieved with viral vectors. Direct in vivo gene transfer with naked DNA was first demonstrated when efficient transfection of myofibers was observed following injection of mRNA or pDNA into skeletal muscle (Wolff et al., 1990). It was an unanticipated finding in that the use of naked nucleic acids was the control for experiments designed to assess the ability of cationic lipids to mediate expression in vivo. Subsequent studies also found foreign gene expression after direct injection in other tissues such as heart, thyroid, skin, and liver (Acsadi et al., 1991; Hengge et al., 1996; Kitsis and Leinwand, 1992; Li et al., 1997; Sikes and O'Malley 1994; Yang and Huang, 1996). However, the efficiency of gene transfer into skeletal muscle and these other tissues by direct injection is relatively low and variable, especially in larger animals such as nonhuman primates (Jiao et al., 1992). After our laboratory had developed novel transfection complexes of pDNA and amphipathic compounds and proteins, we sought to deliver them to hepatocytes in vivo via an intravascular route into the portal vein. Our control for these experiments was naked pDNA and we were once again surprised that this control group had the highest expression levels (Budker et al., 1996; Zhang et al., 1997). High levels of expression were achieved by the rapid injection of naked pDNA in relatively large volumes via the portal vein, the hepatic vein, and the bile duct in mice and rats. The procedure also proved effective in larger animals such as dogs and nonhuman primates (Eastman et al., 2002; Zhang et al., 1997). The next major advance was the demonstration that high levels of expression could also be achieved in hepatocytes in mice by the rapid injection of naked DNA in large volumes simply into the tail vein (Liu et al., 1999; Zhang et al., 1999). This hydrodynamic tail vein (HTV) procedure is proving to be a very useful research tool not only for gene expression studies, but also more recently for the delivery of small interfering RNA (siRNA) (Lewis et al., 2002; McCaffrey et al., 2002). The intravascular delivery of naked pDNA to muscle cells is also attractive particularly since many muscle groups would have to be targeted for intrinsic muscle disorders such as Duchenne muscular dystrophy. High levels of gene expression were first achieved by the rapid injection of naked DNA in large volumes via an artery route with both blood inflow and outflow blocked surgically (Budker et al., 1998; Zhang et al., 2001). Intravenous routes have also been shown to be effective (Hagstrom et al., 2004; Liang et al., 2004; Liu et al., 2001). For limb muscles, the ability to use a peripheral limb vein for injection and a proximal, external tourniquet to block blood flow renders the procedure to be clinically viable. This review concerns itself with the mechanism by which naked DNA is taken up by cells in vivo. A greater understanding of the mechanisms involved in the uptake and expression of naked DNA, and thus connections between postulated mechanisms and expression levels, is emphasized. Inquiries into the mechanism not only aid these practical efforts, but are also interesting on their own account with relevance to viral transduction and cellular processes. The delivery to hepatocytes is first discussed given the greater information available for this process, and then uptake by myofibers is discussed.
Collapse
Affiliation(s)
- Jon A Wolff
- Department of Pediatrics, Waisman Center, University of Wisconsin-Madison Madison, Wisconsin 53705, USA
| | | |
Collapse
|
66
|
Sawamura D, Abe R, Goto M, Akiyama M, Hemmi H, Akira S, Shimizu H. Direct injection of plasmid DNA into the skin induces dermatitis by activation of monocytes through toll-like receptor 9. J Gene Med 2005; 7:664-71. [PMID: 15655803 DOI: 10.1002/jgm.709] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Direct injection of naked DNA into skin can be efficiently used to transfer genes into keratinocytes in vivo. However, bacterial DNA is known to be a potent stimulus for vertebrate immune cells and immune systems. Towards the clinical use of this method, this study examined whether the application of plasmid DNA by direct injection induces any adverse skin effects. METHODS Several plasmid preparations were prepared and directly injected into rat and human skin. Migration, IL-6 production, and reactive oxygen production assays were performed to determine the type of the primary cells responsible for the reaction. Involvement of toll-like receptor (TLR) 9 was examined by experiments using TLR9-knockout mice. RESULTS Injection of several plasmid preparations into rat and human skin resulted in an inflammatory reaction at the treated site. Contamination by endotoxin in the plasmid preparation was shown to worsen this skin inflammation reaction. Immunohistochemical analysis showed that the infiltrating cells in the skin lesions were predominantly monocytes and neutrophils. Further experiments examining migration, IL-6 production, and reactive oxygen production indicated that the primary responsible cells were monocytes rather than neutrophils. Since it was recently shown that cytosine-guanosine dinucleotide (CpG) motif is critical for immune reaction induction in bacterial DNA and cellular responses were mediated by TLR9, we injected plasmids into the ear skin of TLR9-knockout mice. A decrease in ear swelling was noted in the knockout mice, compared to controls, suggesting that plasmid-DNA-induced dermatitis was mediated mostly by TLR9. CONCLUSIONS This study demonstrates that injection of plasmid DNA induces skin inflammation initiated by monocyte activation via TRL9. We should therefore attempt to counteract this dermatitis during the clinical use of the naked DNA injection method in skin.
Collapse
Affiliation(s)
- D Sawamura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | |
Collapse
|
67
|
Meykadeh N, Mirmohammadsadegh A, Wang Z, Basner-Tschakarjan E, Hengge UR. Topical application of plasmid DNA to mouse and human skin. J Mol Med (Berl) 2005; 83:897-903. [PMID: 15902389 DOI: 10.1007/s00109-005-0669-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Accepted: 03/03/2005] [Indexed: 10/25/2022]
Abstract
Gene expression following direct injection of naked plasmid DNA into the skin has been demonstrated in the past. Topical application of plasmid DNA represents an attractive route of gene delivery. If successful, it would have great prospects in skin gene therapy since it is painless and easy to apply. In this study, we analyzed the expression of plasmid DNA in vivo and in vitro following topical application of plasmid DNA in various liposomal spray formulations. Therefore, different concentrations of plasmid DNA expressing enhanced green fluorescent protein (pEGFP-N1) were sprayed onto mouse or human skin once daily for three consecutive days and compared with direct injection. Gene expression was assessed 24 h after the final topical application of various liposomal DNA formulations. The results showed that EGFP mRNA and protein were detectable by RT-PCR and Western blot, respectively. However, epicutaneously applied EGFP plasmid DNA did not lead to microscopically detectable EGFP protein, when assessed by confocal laser microscopy or fluorescence-activated cell sorting in contrast to about 4% of fluorescent keratinocytes following intradermal injection. In an in vivo mouse model, the application of pEGFP-N1 DNA led to the generation of GFP-specific antibodies. These results indicate that topical spray application of pEGFP-N1 liposomal DNA formulations is a suitable method for plasmid DNA delivery to the skin, yielding limited gene expression. This spray method may thus be useful for DNA vaccination. To increase its attractiveness for skin gene therapy, the improvement of topical formulations with enhanced DNA absorption is desirable.
Collapse
Affiliation(s)
- Nuschin Meykadeh
- Department of Dermatology, Heinrich-Heine-University of Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
68
|
Theopold C, Yao F, Eriksson E. Gene therapy in the treatment of lower extremity wounds. INT J LOW EXTR WOUND 2005; 3:69-79. [PMID: 15866792 DOI: 10.1177/1534734604265431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This article presents a brief overview of the etiology of chronic wounds of the lower extremities and their current medical and surgical treatment. Gene therapy as a potential tool for treating therapeutically challenging wounds is described in terms of the vectors employed in gene transfer, as well as the strategies used to promote wound healing. Results from animal model studies, as well as clinical trials, are presented.
Collapse
Affiliation(s)
- Christoph Theopold
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
69
|
Hengge UR, Bardenheuer W. Gene therapy and the skin. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2005; 131C:93-100. [PMID: 15468151 DOI: 10.1002/ajmg.c.30038] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Significant progress has been made during the past decade in corrective gene therapy of the skin. This includes advances in vector technology, targeted gene expression, gene replacement, gene correction, and the availability of appropriate animal models for a variety of candidate diseases. While non-viral integration of large genes such as essential basement membrane proteins has been mastered, new challenges such as the control of immune responses lie ahead of the research community. Among the first skin diseases, patients with junctional epidermolysis bullosa (JEB) and xeroderma pigmentosum (XP) will enter clinical trials.
Collapse
Affiliation(s)
- U R Hengge
- Department of Dermatology, University of Düsseldorf, Germany.
| | | |
Collapse
|
70
|
Meilander-Lin NJ, Cheung PJ, Wilson DL, Bellamkonda RV. Sustained in Vivo Gene Delivery from Agarose Hydrogel Prolongs Nonviral Gene Expression in Skin. ACTA ACUST UNITED AC 2005; 11:546-55. [PMID: 15869433 DOI: 10.1089/ten.2005.11.546] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prolonging gene expression in skin using safe, nonviral gene delivery techniques could impact skin regeneration and wound healing, decrease infection, and potentially improve the success of tissue-engineered skin. To this end, an injectable, agarose-based delivery system was tested and shown to prolong nonviral gene expression in the skin. DNA was compacted with polylysine to improve DNA stability in the presence of nucleases. Up to 25 microg of compacted luciferase plasmid with or without agarose hydrogel was injected intradermally in rodents. Bioluminescence imaging was used for longitudinal, noninvasive monitoring of gene expression in vivo for 35 days. Injections of DNA in solution produced gene expression for only 5-7 days, whereas the sustained release of compacted DNA from the agarose system prolonged expression, with more than 500 pg (20% of day 1 levels) of luciferase per site for at least 35 days. Southern blotting confirmed that the agarose system extended DNA retention, with significant plasmid present through day 7, as compared with DNA in solution, which had detectable DNA only on day 1. Histology revealed that agarose invoked a wound-healing response through day 14. Tissue-engineering and wound-healing applications may benefit from the agarose gene delivery system.
Collapse
Affiliation(s)
- Nancy J Meilander-Lin
- Biomaterials, Cell and Tissue Engineering Laboratory, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
71
|
Abstract
The skin is an attractive target for gene therapy because it is easily accessible and shows great potential as an ectopic site for protein delivery in vivo. Genetically modified epidermal cells can be used to engineer three-dimensional skin substitutes, which when transplanted can act as in vivo 'bioreactors' for delivery of therapeutic proteins locally or systemically. Although some gene transfer technologies have the potential to afford permanent genetic modification, differentiation and eventual loss of genetically modified cells from the epidermis results in temporary transgene expression. Therefore, to achieve stable long-term gene expression, it is critical to deliver genes to epidermal stem cells, which possess unlimited growth potential and self-renewal capacity. This review discusses the recent advances in epidermal stem cell isolation, gene transfer and engineering of skin substitutes. Recent efforts that employ gene therapy and tissue engineering for the treatment of genetic diseases, chronic wounds and systemic disorders, such as leptin deficiency or diabetes, are reviewed. Finally, the use of gene-modified tissue-engineered skin as a biological model for understanding tissue development, wound healing and epithelial carcinogenesis is also discussed.
Collapse
Affiliation(s)
- Stelios T Andreadis
- University at Buffalo, Bioengineering Laboratory, Department of Chemical and Biological Engineering, State University of New York, Amherst, NY 14260, USA.
| |
Collapse
|
72
|
Woodley DT, Keene DR, Atha T, Huang Y, Ram R, Kasahara N, Chen M. Intradermal injection of lentiviral vectors corrects regenerated human dystrophic epidermolysis bullosa skin tissue in vivo. Mol Ther 2005; 10:318-26. [PMID: 15294178 DOI: 10.1016/j.ymthe.2004.05.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 05/10/2004] [Indexed: 11/21/2022] Open
Abstract
Dystrophic epidermolysis bullosa (DEB) is a family of inherited mechanobullous disorders caused by mutations in the gene, COL7A1, that codes for type VII, (anchoring fibril), collagen, which is critical for epidermal-dermal adherence. Most gene therapy approaches have been ex vivo, involving cell culture and culture graft transplantation, which is logistically difficult. To develop a more simplified approach, we engineered a self-inactivating lentiviral vector expressing human type VII collagen and injected this vector intradermally into hairless, immunodeficient mice and into a human DEB composite skin equivalent grafted onto immunodeficient mice. In both situations, the vector transduced dermal cells, which in turn synthesized and exported type VII collagen into the extracellular space. Remarkably, the type VII collagen selectively adhered to and incorporated into the basement membrane zone (BMZ) between the dermis and the epidermis, where it formed anchoring fibril structures. In the case of the DEB skin equivalent, the newly expressed type VII collagen reversed the DEB phenotype characterized by poor epidermal-dermal adherence and anchoring fibril defects. A single lentiviral vector injection provided stable type VII collagen at the BMZ for at least 3 months. These data demonstrate efficient and long-term type VII collagen gene transfer in vivo using direct intradermal injection of an engineered lentiviral vector.
Collapse
Affiliation(s)
- David T Woodley
- Division of Dermatology, Department of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | | | | | | | | | | |
Collapse
|
73
|
Hengge UR, Bardenheuer W, Doroudi R, Mirmohammadsadegh A. Thérapie génique et peau. Ann Dermatol Venereol 2005; 132:154-63. [PMID: 15798569 DOI: 10.1016/s0151-9638(05)79230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- U-R Hengge
- Département de Dermatologie, Université Heinrich-Heine, Duesseldorf, Allemagne.
| | | | | | | |
Collapse
|
74
|
Dietz GPH, Bähr M. Delivery of bioactive molecules into the cell: the Trojan horse approach. Mol Cell Neurosci 2005; 27:85-131. [PMID: 15485768 DOI: 10.1016/j.mcn.2004.03.005] [Citation(s) in RCA: 358] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 02/17/2004] [Accepted: 03/16/2004] [Indexed: 01/12/2023] Open
Abstract
In recent years, vast amounts of data on the mechanisms of neural de- and regeneration have accumulated. However, only in disproportionally few cases has this led to efficient therapies for human patients. Part of the problem is to deliver cell death-averting genes or gene products across the blood-brain barrier (BBB) and cellular membranes. The discovery of Antennapedia (Antp)-mediated transduction of heterologous proteins into cells in 1992 and other "Trojan horse peptides" raised hopes that often-frustrating attempts to deliver proteins would now be history. The demonstration that proteins fused to the Tat protein transduction domain (PTD) are capable of crossing the BBB may revolutionize molecular research and neurobiological therapy. However, it was only recently that PTD-mediated delivery of proteins with therapeutic potential has been achieved in models of neural degeneration in nerve trauma and ischemia. Several groups have published the first positive results using protein transduction domains for the delivery of therapeutic proteins in relevant animal models of human neurological disorders. Here, we give an extensive review of peptide-mediated protein transduction from its early beginnings to new advances, discuss their application, with particular focus on a critical evaluation of the limitations of the method, as well as alternative approaches. Besides applications in neurobiology, a large number of reports using PTD in other systems are included as well. Because each protein requires an individual purification scheme that yields sufficient quantities of soluble, transducible material, the neurobiologist will benefit from the experiences of other researchers in the growing field of protein transduction.
Collapse
|
75
|
Abstract
Significant progress has been made in corrective gene therapy of the skin in the last decade. This includes advances in vector technology, targeted gene expression, gene replacement, gene correction, and the availability of appropriate animal models for a variety of candidate diseases. While non-viral integration of large genes such as essential basement membrane proteins has been mastered, new challenges such as the control of immune responses lie ahead of the research community until skin gene therapy will become clinical reality. Among the first skin diseases patients with junctional epidermolysis bullosa and xeroderma pigmentosum have entered clinical trials.
Collapse
Affiliation(s)
- Ulrich R Hengge
- Department of Dermatology, Heinrich-Heine-University, D-40225 Duesseldorf, Germany.
| |
Collapse
|
76
|
Bellocq NC, Kang DW, Wang X, Jensen GS, Pun SH, Schluep T, Zepeda ML, Davis ME. Synthetic Biocompatible Cyclodextrin-Based Constructs for Local Gene Delivery to Improve Cutaneous Wound Healing. Bioconjug Chem 2004; 15:1201-11. [PMID: 15546185 DOI: 10.1021/bc0498119] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The localized, sustained delivery of growth factors for wound healing therapy is actively being explored by gene transfer to the wound site. Biocompatible matrices such as bovine collagen have demonstrated usefulness in sustaining gene therapy vectors that express growth factors in local sites for tissue repair. Here, new synthetic biocompatible materials are prepared and shown to deliver a protein to cultured cells via the use of an adenoviral delivery vector. The synthetic construct consists of a linear, beta-cyclodextrin-containing polymer and an adamantane-based cross-linking polymer. When the two polymers are combined, they create an extended network by the formation of inclusion complexes between the cyclodextrins and adamantanes. The properties of the network are altered by controlling the polymer molecular weights and the number of adamantanes on the cross-linking polymer, and these modifications and others such as replacement of the beta-cyclodextrin (host) and adamantane (guest) with other cyclodextrins (hosts such as alpha, gamma, and substituted members) and inclusion complex forming molecules (guests) provide the ability to rationally design network characteristics. Fibroblasts exposed to these synthetic constructs show proliferation rates and migration patterns similar to those obtained with collagen. Gene delivery (green fluorescent protein) to fibroblasts via the inclusion of adenoviral vectors in the synthetic construct is equivalent to levels observed with collagen. These in vitro results suggest that the synthetic constructs are suitable for in vivo tissue repair applications.
Collapse
Affiliation(s)
- Nathalie C Bellocq
- Insert Therapeutics, Inc., 2585 Nina Street, Pasadena, California 91107, USA
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Settelen N, Roch O, Bock D, Rooke R, Braun S, Meyer O. Controlled plasmid gene transfer to murine renal carcinoma by hexadecylphosphocholine. J Control Release 2004; 94:237-44. [PMID: 14684287 DOI: 10.1016/j.jconrel.2003.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We report here that the anticancer drug hexadecylphosphocholine (HPC) can control plasmid DNA-mediated gene transfer to renal carcinoma following intratumoral administration. Significant improvement of gene expression levels could be achieved depending on HPC dose administered. Optimal concentration of HPC co-injected with plasmid DNA was found to be 0.2% (w/v) showing up to a 10-fold increase in reporter gene expression levels when compared to DNA administered alone. In vivo gene transfer activity of HPC was not affected by the nature of the diluent used, i.e. glucose-based or saline-based isotonic solutions. Although in vitro transfection activity of HPC formulations could not be evidenced, a liposome leakage assay revealed that HPC could significantly destabilize stable lipid membranes suggesting that a possible membrane permeation enhancer activity of HPC combined to the physical stress induced by the intratumor injection may facilitate plasmid DNA entry inside the cells resulting in increased gene expression. HPC/plasmid formulations represent new and attractive non-viral gene delivery systems with potential in cancer gene therapy and vaccination.
Collapse
Affiliation(s)
- Nathalie Settelen
- Non-viral Gene Therapy Laboratory, Transgene S.A., 11 rue de Molsheim, 67082 Strasbourg, France
| | | | | | | | | | | |
Collapse
|
78
|
Byrnes CK, Malone RW, Akhter N, Nass PH, Wetterwald A, Cecchini MG, Duncan MD, Harmon JW. Electroporation enhances transfection efficiency in murine cutaneous wounds. Wound Repair Regen 2004; 12:397-403. [PMID: 15260804 DOI: 10.1111/j.1067-1927.2004.012409.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Transfection of wounds with DNA-encoding growth factors has the potential to improve healing, but current means of nonviral gene delivery are inefficient. Repeated high doses of DNA, necessary to achieve reliable gene expression, are detrimental to healing. We assessed the ability of in vivo electroporation to enhance gene expression. Full-thickness cutaneous excisional wounds were created on the dorsum of female mice. A luciferase- encoding plasmid driven by a CMV promoter was injected at the wound border. Following plasmid administration, electroporative pulses were applied to injection sites. Pulse parameters were varied over a range of voltage, duration, and number. Animals were euthanized at intervals after transfection and the luciferase activity measured. Application of electric pulses consistently increased luciferase expression. The electroporative effect was most marked at a plasmid dose of 50 micro g, where an approximate tenfold increase was seen. Six 100- micro s-duration pulses of 1750 V/cm were found to be the most effective in increasing luciferase activity. High numbers of pulses tended to be less effective than smaller numbers. This optimal electroporation regimen had no detrimental effect on wound healing. We conclude that electroporation increases the efficiency of transgene expression and may have a role in gene therapy to enhance wound healing.
Collapse
Affiliation(s)
- Colman K Byrnes
- Section of Surgical Sciences, Johns Hopkins Medical Institutions, Johns Hopkins Bayview Medical Center, 4940 Eastern Avenue, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Ogura M, Sato S, Nakanishi K, Uenoyama M, Kiyozumi T, Saitoh D, Ikeda T, Ashida H, Obara M. In vivo targeted gene transfer in skin by the use of laser-induced stress waves. Lasers Surg Med 2004; 34:242-8. [PMID: 15022251 DOI: 10.1002/lsm.20024] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVES Much interest has been shown in the use of lasers for nonviral targeted gene transfer, since the spatial characteristics of laser light are quite well defined. The aim of this study was to demonstrate in vivo gene transfer by the use of laser-induced stress waves (LISWs). STUDY DESIGN/MATERIALS AND METHODS After reporter genes had been intradermally injected to rat skin in vivo, a laser target was placed on the gene-injected skin. LISWs were generated by the irradiation of an elastic laser target with 532-nm nanosecond laser pulses of a Q-switched Nd:YAG laser. RESULTS Levels of luciferase activities for the skin exposed to LISWs were two orders of magnitude higher than those for the skin injected with naked DNA. Expressions of enhanced green fluorescent protein (EGFP) and beta-galactosidase were observed only in the area that was exposed to LISWs, and in addition, epidermal cells were selectively transfected. No major side effects were observed, and luciferase activity levels as high as 10(5) RLU per mg of protein were sustained even 5 days after gene transfer. CONCLUSION Highly efficient and site-specific gene transfer can be achieved by applying a few pulses of nanosecond pulsed LISWs to rat skin in vivo.
Collapse
Affiliation(s)
- Makoto Ogura
- Department of Electronics and Electrical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Barfoed AM, Kristensen B, Dannemann-Jensen T, Viuff B, Bøtner A, Kamstrup S, Blixenkrone Møller M. Influence of routes and administration parameters on antibody response of pigs following DNA vaccination. Vaccine 2004; 22:1395-405. [PMID: 15063562 DOI: 10.1016/j.vaccine.2003.10.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2003] [Accepted: 10/27/2003] [Indexed: 11/26/2022]
Abstract
Using the nucleoprotein of porcine reproductive and respiratory syndrome virus as model antigen, we optimised parameters for gene gun vaccination of pigs, including firing pressure and vaccination site. As criteria for optimisation, we characterised particle penetration and local tissue damage by histology. For selected combinations, vaccination efficiency in terms of antibody response was studied. Gene gun vaccination on ear alone was as efficient as a multi-site (ear, thorax, inguinal area, tongue mucosa) gene gun approach, and more efficient than combined intramuscular (i.m.)/intradermal (i.d.) injection of plasmid DNA. This indicates, that the ear is an attractive site for gene gun vaccination of pigs.
Collapse
Affiliation(s)
- Annette Malene Barfoed
- Laboratory of Virology and Immunology, Department of Veterinary Microbiology, The Royal Veterinary and Agricultural University, Stigbøjlen 7, DK-1870 Frederiksberg C, Denmark
| | | | | | | | | | | | | |
Collapse
|
81
|
Basner-Tschakarjan E, Mirmohammadsadegh A, Baer A, Hengge UR. Uptake and trafficking of DNA in keratinocytes: evidence for DNA-binding proteins. Gene Ther 2004; 11:765-74. [PMID: 14724668 DOI: 10.1038/sj.gt.3302221] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The skin is an interesting organ for human gene therapy due to accessibility, immunologic potential and synthesis capabilities. In this study, we attempted to visualize and measure the uptake of naked FITC-labeled plasmid by FACS analysis detecting up to 15% internalization in a dose- and time-dependent manner. Cycloheximide treatment inhibited the uptake by >90%, suggesting a protein-mediated uptake. The inhibition of different internalization pathways demonstrated that blocking macropinocytosis (by amiloride and N,N-dimethylamylorid) reduced DNA uptake by >85%, while the inhibition of clathrin-coated pits (by chlorpromazine) and caveolae (by nystatin/filipin III) did not limit the uptake. Colocalization studies using confocal laser microscopy revealed a time-dependent accumulation of plasmid DNA in endosomes and lysosomes. When a green fluorescent protein (GFP) expression vector was used, specific GFP-RNA became detectable by reverse transcriptase-PCR, whereas measurable amounts of protein could not be identified in FACS experiments. To detect the potential DNA receptors on the keratinocyte surface, membrane proteins were extracted and subjected to South-Western blotting using digoxigenin-labeled calf thymus and lambda-phage DNA. Two DNA-binding proteins, ezrin and moesin, known as plasma membrane-actin linkers, were identified by one- and two-dimensional-South-Western blots and matrix-assisted laser desorption and ionization-mass spectrometry. Ezrin and moesin are functionally associated with a number of transmembrane receptors such as the EGF, CD44 or ICAM-1 receptor. Taken together, naked plasmid DNA seems to enter human keratinocytes through different pathways, mainly by macropinocytosis. Two DNA-binding proteins were identified that seemed to be involved in binding/trafficking of internalized DNA.
Collapse
Affiliation(s)
- E Basner-Tschakarjan
- Department of Dermatology, Heinrich Heine-University Düsseldorf, Düsseldorf, Germany
| | | | | | | |
Collapse
|
82
|
Siprashvili Z, Scholl FA, Oliver SF, Adams A, Contag CH, Wender PA, Khavari PA. Gene transfer via reversible plasmid condensation with cysteine-flanked, internally spaced arginine-rich peptides. Hum Gene Ther 2003; 14:1225-33. [PMID: 12952594 DOI: 10.1089/104303403767740768] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nonviral gene transfer offers biosafety, stability, and expense advantages over viruses; however, it has suffered from poor efficiency. Because arginine-rich peptides facilitate uptake of macromolecules such as proteins, liposomes, and iron nanoparticles, we explored their potential in enhancing plasmid DNA delivery. In their unmodified form, known protein transduction sequences, including hepta-arginine and Tat(47-57), failed to support effective gene delivery. However, by flanking a core of consecutive arginines with amino- and carboxy-terminal cysteines in vitro gene transfer was observed. Furthermore, interspersing arginines with glycine and histidine residues achieved reversible plasmid condensation and dramatically increased transfection levels in a variety of cell types. Unlike most available cationic homopolymers that function only in vitro, these new peptides also increased gene expression in both murine and human tissue in vivo. Thus, cysteine-flanked, internally spaced arginine-rich (CFIS-R) peptides represent a new approach to efficient nonviral plasmid delivery using rationally designed protein transduction domains.
Collapse
|
83
|
Ortiz-Urda S, Lin Q, Yant SR, Keene D, Kay MA, Khavari PA. Sustainable correction of junctional epidermolysis bullosa via transposon-mediated nonviral gene transfer. Gene Ther 2003; 10:1099-104. [PMID: 12808440 DOI: 10.1038/sj.gt.3301978] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2002] [Accepted: 12/06/2002] [Indexed: 01/13/2023]
Abstract
Sustainable correction of severe human genetic disorders of self-renewing tissues, such as the blistering skin disease junctional epidermolysis bullosa (JEB), is facilitated by stable genomic integration of therapeutic genes into somatic tissue stem cells. While integrating viral vectors can achieve this, they suffer from logistical and biosafety concerns. To circumvent these limitations, we used the Sleeping Beauty transposable element to integrate the LAMB3 cDNA into genomes of epidermal holoclones from six unrelated JEB patients. These cells regenerate human JEB skin that is normalized at the level of laminin 5 protein expression, hemidesmosome formation and blistering. Transposon-mediated gene delivery therefore affords an opportunity for stable gene delivery in JEB and other human diseases.
Collapse
Affiliation(s)
- S Ortiz-Urda
- Program in Epithelial Biology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
84
|
Gharwan H, Wightman L, Kircheis R, Wagner E, Zatloukal K. Nonviral gene transfer into fetal mouse livers (a comparison between the cationic polymer PEI and naked DNA). Gene Ther 2003; 10:810-7. [PMID: 12704421 DOI: 10.1038/sj.gt.3301954] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We investigated the efficacy and safety of the cationic polymer polyethylenimine (PEI) as a potential tool for intrauterine gene delivery into livers of fetal mice in the last trimester of pregnancy (E17.5). Using luciferase as a reporter gene, transferrin-conjugated and ligand-free PEI/DNA complexes (containing 3 microg DNA) with varying PEI-nitrogen/DNA-phosphate (N/P) ratios and different PEI forms, branched (800, 25 kDa) and linear (22 kDa), were compared with naked DNA. Transgene expression was measured 48 h after administration of PEI/DNA complexes or naked DNA. Highest luciferase activity (9.8 x 10(3) relative light units (RLU)/mg of tissue protein) was observed with ligand-free PEI22/DNA mixtures at N/P 6.0. In addition, this formulation was associated with very low toxicity as compared to the other PEI/DNA-injected groups. Using beta-galactosidase as a reporter gene, transfection of single, but also small, clusters of cells was demonstrated throughout the liver. Injection of 3 microg naked DNA resulted in an 11-fold lower transgene expression value (0.9 x 10(3) RLU/mg of tissue protein) as compared to PEI22/DNA complexes. However, the administration of higher concentrated naked DNA (9 microg) into fetal livers yielded expression levels of 3.2 x 10(4) RLU/mg of tissue protein, a more than three-fold increase compared to PEI22/DNA complexes. Furthermore, the gene transfer efficacy of concentrated naked DNA was approximately 40 times higher in fetuses than in adults (0.8 x 10(3) RLU/mg of tissue protein), indicating that fetal tissue is especially amenable to the uptake and expression of naked DNA.
Collapse
Affiliation(s)
- H Gharwan
- Institute of Pathology, University of Graz, Auenbruggerplatz, Graz, Styria, Austria
| | | | | | | | | |
Collapse
|
85
|
Ohyama M, Ota T, Aoki M, Tsunoda K, Harada R, Koyasu S, Nishikawa T, Amagai M. Suppression of the immune response against exogenous desmoglein 3 in desmoglein 3 knockout mice: an implication for gene therapy. J Invest Dermatol 2003; 120:610-5. [PMID: 12648224 DOI: 10.1046/j.1523-1747.2003.12090.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Gene therapies for recessive genetic diseases may provoke unwanted immune responses against the introduced gene product because patients, especially those with null mutation of a certain protein, have no tolerance for the protein of interest. This study used desmoglein 3 knockout (Dsg3-/-) mice as a disease model for a genetic defect in DSG3, to investigate whether nonviral gene therapy induces an immune response against Dsg3 and whether the reaction against Dsg3 can be prevented. When mouse Dsg3 cDNA was injected in the skin of Dsg3-/- mice, 50% of treated Dsg3-/- mice developed anti-Dsg3 IgG, which can bind native Dsg3 in vivo. To prevent this response, we used an anti-CD40L monoclonal antibody, MR1, which blocks the costimulatory interaction between CD40 and CD40L. To evaluate the effect of MR1, we grafted Dsg3+/+skin on Dsg3-/- mice, to mimic stable gene transfer of Dsg3. After skin grafting, all the recipient Dsg3-/- mice were treated with either MR1 (n=8) or control hamster IgG (n=8). All of the control IgG-treated mice developed circulating anti-Dsg3 IgG about 2 wk after grafting, and IgG deposition was observed on the surfaces of keratinocytes in the grafted Dsg3+/+skin. Such anti-Dsg3 IgG production was significantly prevented, however, when the recipient mice were treated with MR1. These findings suggested that gene therapies for recessive diseases may provoke an immune response against the transgene product, and that the CD40-CD40L interaction might be a reasonable target for effective prevention of such undesirable immune responses, leading, in turn, to a successful gene therapy.
Collapse
Affiliation(s)
- Manabu Ohyama
- Department of Dermatology , Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Ma H, Xu R, Cheng H, Kuo HS, During M, Fang RH. Gene transfer into human keloid tissue with adeno-associated virus vector. THE JOURNAL OF TRAUMA 2003; 54:569-73. [PMID: 12634540 DOI: 10.1097/01.ta.0000042016.45195.4c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Gene transfer is a new territory for clinicians. Intractable disorders might be approached in such a way. Adeno-associated virus (AAV) vector has been transfected successfully into a variety of tissues including skin. We evaluated the ability of this vector to transfer and cause expression of the reporter gene in human keloid tissue. METHODS Human keloid specimens were injected with an AAV vector encoding beta-galactosidase and incubated for 4 weeks after injection. The presence of mRNA and beta-galactosidase enzymatic activity were assayed by reverse-transcriptase polymerase chain reaction and the X-gal technique. RESULTS Gene expression shown by reverse-transcriptase polymerase chain reaction was observed in keloid tissue 4 weeks after injection, and so was the positive X-gal staining. CONCLUSION Our results showed that AAV vector could transduce human keloid tissue effectively. Replacement of the reporter gene with a functioning gene might be feasible for keloid treatment.
Collapse
Affiliation(s)
- Hsu Ma
- School of Medicine, NationalYang-Ming University and Department of Neurosurgery, Veterans General Hospital-Taipei, Taiwan.
| | | | | | | | | | | |
Collapse
|
87
|
Brus C, Santi P, Colombo P, Kissel T. Distribution and quantification of polyethylenimine oligodeoxynucleotide complexes in human skin after iontophoretic delivery using confocal scanning laser microscopy. J Control Release 2002; 84:171-81. [PMID: 12468220 DOI: 10.1016/s0168-3659(02)00295-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Iontophoresis may be a potentially useful technique for the delivery of oligonucleotides into the skin. To enhance intracellular uptake during iontophoresis, we investigated the dermal delivery of oligodeoxynucleotides (ODN) as a polyelectrolyte complex with polyethylenimine (PEI). Perpendicular cross-sectioning was performed to visualize and quantify the penetration properties of double labeled PEI/ODN complexes across full thickness human skin. Due to the net positive charge of the complexes, anodal iontophoresis was expected to enhance skin delivery by electrorepulsion compared to passive diffusion. Confocal laser scanning microscopy demonstrated that non-complexed ODN could penetrate the skin after 1 h of cathodal iontophoresis but not by passive diffusion or anodal iontophoresis. However, extensive degradation occurred as documented by a dramatic decrease of fluorescence intensity within viable skin tissue after 10 h. Anodal iontophoresis of the complexes led to a deep penetration of both the TAMRA-labeled ODN and the Oregon Green-labeled PEI. A constant increase in fluorescence indicated a protective effect of the polymer against nuclease degradation. Co-localization of red and green fluorescence was noted within numerous nuclei of epidermal keratinocytes. In contrast, passive diffusion of the complexes did not lead to successful uptake into keratinocytes and was limited to the stratum corneum. Complexation of ODN by PEI, therefore, seems to be a promising method to enhance both the transport of charged complexes into the skin and to facilitate intracellular uptake, which may potentially be useful for the local treatment of skin diseases using ODN.
Collapse
Affiliation(s)
- Carola Brus
- Department of Pharmaceutics and Biopharmacy, Philipps University of Margburg, Ketzerbach 63, 35037 Marburg, Germany
| | | | | | | |
Collapse
|
88
|
De Rose R, Tennent J, McWaters P, Chaplin PJ, Wood PR, Kimpton W, Cahill R, Scheerlinck JPY. Efficacy of DNA vaccination by different routes of immunisation in sheep. Vet Immunol Immunopathol 2002; 90:55-63. [PMID: 12406655 DOI: 10.1016/s0165-2427(02)00221-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
DNA vaccination, delivered through various routes, has been used extensively in laboratory animals. Few studies have focused on veterinary species and while results obtained in laboratory animals can often be extrapolated to veterinary species this is not always the case. In this study we have compared the effect of the route of immunisation with DNA on the induction of immune responses and protection of sheep to challenge with live Corynebacterium pseudotuberculosis. Intramuscular injection of plasmid DNA encoding an inactivated form of the phospholipase D (PLD) antigen linked to CTLA4-Ig resulted in the induction of a strong memory response and sterile immunity following challenge in 45% of the animals. In contrast, gene gun delivery or subcutaneous (SC) injection of the DNA vaccine induced comparatively poor responses and insignificant levels of protection. Thus, DNA vaccine efficacy in sheep is strongly influenced by the route of vaccination. Amongst intramuscular vaccinates, protected sheep had significantly elevated IgG2 responses compared to unprotected animals, while both subgroups had equivalent IgG1 levels. This suggests that the presence of IgG2 antibodies and hence a Th1-like response, induced by the DNA vaccine gave rise to protective immunity against C. pseudotuberculosis.
Collapse
Affiliation(s)
- Robert De Rose
- The Cooperative Research Centre for Vaccine Technology, CSIRO Livestock Industries, Private Bag No 24, Vic 3220, Geelong, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Sandgren S, Cheng F, Belting M. Nuclear targeting of macromolecular polyanions by an HIV-Tat derived peptide. Role for cell-surface proteoglycans. J Biol Chem 2002; 277:38877-83. [PMID: 12163493 DOI: 10.1074/jbc.m205395200] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
New therapies based on gene transfer and protein delivery require a better understanding of the basic mechanisms of macromolecular membrane transport. We have studied cellular uptake of macromolecular polyanions, i.e. DNA and glycosaminoglycans, and a polybasic HIV-Tat derived peptide (GRKKRRQRRRPPQC) using fluorescence assisted cell sorting and confocal fluorescence microscopy. The transactivator of HIV transcription (Tat) peptide stimulated cellular uptake of both DNA and heparan sulfate in a time-, concentration-, and temperature-dependent manner. Peptide-polyanion complexes accumulated in large, acidic, cytoplasmic vesicles formed de novo. This was followed by transfer of polyanion into the nuclear compartment and subsequent disappearance of the endolysosomal vesicles. In the absence of polyanion the Tat peptide displayed rapid accumulation in the nuclear compartment. However, in the presence of polyanion the peptide was almost exclusively retained in cytoplasmic vesicles. Cell-surface proteoglycans played a pivotal role in the uptake of complexes exhibiting a relatively high peptide to polyanion ratio, corresponding to a net positive charge of the complexes. Uptake of polyanions per se or complexes with a relatively low peptide to polyanion ratio was favored by proteoglycan deficiency in the recipient cells, indicating the existence of distinct transport mechanisms. Moreover, expression of full-length HIV-Tat as well as exogenous addition of HIV-Tat peptide resulted in cellular accumulation of endogenous proteoglycans. We conclude that an HIV-Tat derived peptide efficiently targets extraneous DNA and glycosaminoglycans to the nuclear compartment and that proteoglycans serve a regulatory role in these processes, which may have implications for directed gene and drug delivery in vivo.
Collapse
Affiliation(s)
- Staffan Sandgren
- Department of Cell and Molecular Biology, Section of Cell and Matrix Biology, Lund University, BMC, C13, S-221 84 Lund, Sweden
| | | | | |
Collapse
|
90
|
Ortiz-Urda S, Thyagarajan B, Keene DR, Lin Q, Fang M, Calos MP, Khavari PA. Stable nonviral genetic correction of inherited human skin disease. Nat Med 2002; 8:1166-70. [PMID: 12244305 DOI: 10.1038/nm766] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2002] [Accepted: 08/27/2002] [Indexed: 01/29/2023]
Abstract
Current gene-transfer technologies display limitations in achieving effective gene delivery. Among these limitations are difficulties in stably integrating large corrective sequences into the genomes of long-lived progenitor-cell populations. Current larger-capacity viral vectors suffer from biosafety concerns, whereas plasmid-based approaches have poor efficiency of stable gene transfer. These barriers hinder genetic correction of many severe inherited human diseases, such as the blistering skin disorder recessive dystrophic epidermolysis bullosa (RDEB), caused by mutations in the large COL7A1 gene. To circumvent these barriers, we used the phi C31 bacteriophage integrase, which stably integrates large DNA sequences containing a specific 285-base-pair attB sequence into genomic 'pseudo-attP sites'. phi C31 integrase-based gene transfer stably integrated the COL7A1 cDNA into genomes of primary epidermal progenitor cells from four unrelated RDEB patients. Skin regenerated using these cells displayed stable correction of hallmark RDEB disease features, including Type VII collagen protein expression, anchoring fibril formation and dermal-epidermal cohesion. These findings establish a practical approach to nonviral genetic correction of severe human genetic disorders requiring stable genomic integration of large DNA sequences.
Collapse
Affiliation(s)
- Susana Ortiz-Urda
- VA Palo Alto Healthcare System and the Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | |
Collapse
|
91
|
del Pozo V, Rojo M, Rubio ML, Cortegano I, Cárdaba B, Gallardo S, Ortega M, Civantos E, López E, Martín-Mosquero C, Peces-Barba G, Palomino P, González-Mangado N, Lahoz C. Gene therapy with galectin-3 inhibits bronchial obstruction and inflammation in antigen-challenged rats through interleukin-5 gene downregulation. Am J Respir Crit Care Med 2002; 166:732-7. [PMID: 12204873 DOI: 10.1164/rccm.2111031] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The pathophysiology of asthma involves an intricate network of molecular and cellular interactions. Elevated Th2 cytokines (interleukin [IL]-5 and IL-4) associated with eosinophilic inflammation characterize allergic diseases and provide potential targets for immunomodulation. Recent evidence has demonstrated that galectin-3 induces selective downregulation of IL-5 gene expression in several cell types (eosinophils, T cell lines, and antigen specific T cells). Accordingly, we sought to elucidate whether in vivo intratracheal instillation of plasmid DNA encoding galectin-3 would inhibit an experimental asthmatic reaction in a rat model with increased eosinophils and T cells in bronchoalveolar fluid and impaired pulmonary function. We found that instillation of galectin-3 gene in these rats led to normalization of the eosinophil and T cell count in bronchoalveolar lavage fluid and that there was a strong concomitant inhibition of IL-5 mRNA in the lungs. As a consequence, galectin-3-treated rats showed recovery of pulmonary functional parameters, such as pulmonary pressure and expiratory flows. These data emphasize the potential utility of galectin-3 as a novel therapeutic approach for treatment of allergic asthma.
Collapse
|
92
|
Sawamura D, Akiyama M, Shimizu H. Direct injection of naked DNA and cytokine transgene expression: implications for keratinocyte gene therapy. Clin Exp Dermatol 2002; 27:480-4. [PMID: 12372091 DOI: 10.1046/j.1365-2230.2002.01101.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intradermally injected DNA diffuses into the epidermis and can then enter keratinocytes and become expressed by these cells. Using this method, plasmids containing cytokine genes that have been introduced into keratinocytes can induce a level of cytokine expression sufficient to provide biological effects in the treated skin. Furthermore, transgenic cytokines released from the transduced keratinocytes can also enter the circulation and have downstream effects on other target organs. Thus far, naked DNA injection appears to be a safe, simple, and relatively efficient method that enables genes to be expressed in transplanted human skin on immunosuppressed animals. In humans, keratinocyte gene therapy using the cytokine gene DNA injection method has the potential to become a powerful therapeutic tool for dermatologists in the management of certain inflammatory and other dermatoses.
Collapse
Affiliation(s)
- D Sawamura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | |
Collapse
|
93
|
Zhang L, Nolan E, Kreitschitz S, Rabussay DP. Enhanced delivery of naked DNA to the skin by non-invasive in vivo electroporation. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1572:1-9. [PMID: 12204326 DOI: 10.1016/s0304-4165(02)00270-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
DNA delivery to skin may be useful for the treatment of skin diseases, DNA vaccinations, and other gene therapy applications requiring local or systemic distribution of a transgene product. However, the effective, consistent and patient-friendly transfection of skin cells remains a challenge. In a mouse model, we evaluated the effectiveness of intradermal injection of plasmid DNA followed by noninvasive in vivo electroporation (EP) as a method to improve transfection in skin. We achieved a several hundred-fold stimulation of gene expression by EP, sufficient to produce clinically relevant amounts of transgene product. We studied the effect of DNA dose and time after treatment as well as various EP pulse parameters on the efficiency of gene expression. EP under conditions of constant charge transfer revealed that the applied voltage was the main determinant for transgene expression efficiency while other pulse parameters had lesser effects. Patient-friendly, noninvasive meander electrodes which we designed for clinical applications proved equally effective and safe as plate electrodes. We also showed for the first time that noninvasive EP is effective in stimulating transfection and gene expression in human skin, particularly in the epidermis. Our findings demonstrate the applicability of EP-enhanced DNA delivery to skin for gene therapy, DNA immunization and other areas.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Research and Development, Genetronics, Inc., San Diego, CA 92121-1334, USA.
| | | | | | | |
Collapse
|
94
|
Abstract
Recent progress in molecular genetics has illuminated the basis for a wide variety of inherited and acquired diseases. Gene therapy offers an attractive therapeutic approach capitalizing upon these new mechanistic insights. The skin is a uniquely attractive tissue site for development of new genetic therapeutic approaches both for its accessibility as well as for the large number of diseases that are amenable in principle to cutaneous gene transfer. Amongst these opportunities are primary monogenic skin diseases, chronic wounds and systemic disorders characterized by low or absent levels of circulating polypeptides. For cutaneous gene therapy to be effective, however, significant progress is required in a number of domains. Recent advances in vector design, administration, immune modulation, and regulation of gene expression have brought the field much nearer to clinical utility.
Collapse
Affiliation(s)
- P A Khavari
- VA Palo Alto Healthcare System and the Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | | | | |
Collapse
|
95
|
Sawamura D, Yasukawa K, Kodama K, Yokota K, Sato-Matsumura KC, Toshihiro T, Shimizu H. The majority of keratinocytes incorporate intradermally injected plasmid DNA regardless of size but only a small proportion of cells can express the gene product. J Invest Dermatol 2002; 118:967-71. [PMID: 12060390 DOI: 10.1046/j.1523-1747.2002.01756.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The expression of intradermally injected DNA by keratinocytes is found mainly in the upper and middle layers of the epidermis. To investigate the mechanism of this selective expression, we observed the sequential changes in the distribution of interleukin-6-expressing keratinocytes after the introduction of the interleukin-6 gene. Transgene expression first occurred in basal keratinocytes and subsequently expanded to all epidermal layers and then remained in the upper layers. Semiquantitative analysis indicated that keratinocytes in the lower layers incorporated and lost DNA earlier than those in the upper layers. In order to examine the effect of the DNA size on the transgene expression, we constructed a plasmid containing a full-length 9 kb cDNA of type VII collagen and introduced it into keratinocytes. The expression pattern of type VII collagen in the epidermis was the same as those for smaller genes. This suggests that plasmid size has little or no effect on the expression pattern of the transfected gene. To trace the introduced plasmid, we intradermally injected a green fluorescence protein expression plasmid coupled with a rhodamine flag. Almost all keratinocytes in the injected areas showed rhodamine fluorescence. Furthermore, some cells also expressed green fluorescence protein. A lack of rhodamine fluorescence in the nucleus suggested an impairment of plasmid DNA transport from the cytoplasm to the nucleus. Collectively, our results show that the majority of keratinocytes take up the intradermally injected DNA regardless of its size, but that the transfer of DNA from the cytoplasm to the nucleus is limiting the transgene expression.
Collapse
Affiliation(s)
- Daisuke Sawamura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | |
Collapse
|
96
|
Baqué P, Pierrefite-Carle V, Gavelli A, Brossette N, Benchimol D, Bourgeon A, Staccini P, Saint-Paul MC, Rossi B. Naked DNA injection for liver metastases treatment in rats. Hepatology 2002; 35:1144-52. [PMID: 11981764 DOI: 10.1053/jhep.2002.32709] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
The cytosine deaminase (CD) gene converts the nontoxic prodrug, 5-fluorocytosine (5-FC), into 5-fluorouracil (5-FU). We previously showed that injection of CD-bearing cancer cells followed by 5-FC treatment can act as an autologous tumor vaccine in a syngenic liver metastasis model in rats. In the present work, we analyzed the antitumor efficiency of a direct intratumoral injection of a CD-expressing plasmid. In rats bearing microscopic or macroscopic metastases in right and left liver lobes, an injection of a CD-expressing plasmid was performed in the left lobe tumor, followed by 5-FC treatment of the animals. A significant regression of the DNA-injected tumor was observed in 5-FC-treated rats, both in microscopic (P =.007) or advanced (P <.0001) tumor models. Moreover, this treatment also induced a potent distant bystander effect on untreated controlateral liver tumors and extrahepatic metastases, resulting in an increased survival compared with control animals in both tumor models (P <.05). In conclusion, these data suggest that direct intratumoral injection of a CD-expressing plasmid, associated to 5-FC administration, can constitute a powerful and innocuous alternative treatment for unresectable liver metastases from colon carcinoma.
Collapse
Affiliation(s)
- Patrick Baqué
- Service de Chirurgie Abdominale et Thoracique, Hôpital l'Archet II, Nice cédex 3, France
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Rao KVN, He YX, Ramaswamy K. Suppression of cutaneous inflammation by intradermal gene delivery. Gene Ther 2002; 9:38-45. [PMID: 11850721 DOI: 10.1038/sj.gt.3301622] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2001] [Accepted: 09/28/2001] [Indexed: 11/08/2022]
Abstract
Biological effects of in vivo transfection of a potential anti-inflammatory gene, designated Sm16, cloned from the human parasite Schistosoma mansoni were analyzed in these studies. A single intradermal injection of a full-length cDNA of Sm16 resulted in the expression of Sm16 in the epidermis, dermis, skin migratory cells and skin-draining lymph nodes of mice for up to 7 days. Subsequently the anti-inflammatory effect of this gene expression was evaluated by inducing an inflammatory response in the skin of mice. These studies showed that Sm16 gene delivery resulted in a significant suppression of cutaneous inflammation as shown by a reduction in cutaneous edema, decrease in neutrophil infiltration, suppression of pro-inflammatory cytokine expression and down-regulation of ICAM-1 expression in the skin inflammatory site. Cells collected from the skin-draining lymph nodes showed reduced proliferation to mitogen. Multiple intradermal injection of Sm16 cDNA failed to induce any antibody response in mice for up to 8 weeks after initial injection. These findings suggest a potential for developing Sm16 gene delivery as a therapeutic agent for treating inflammatory skin disorders.
Collapse
Affiliation(s)
- K V N Rao
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL 61107, USA
| | | | | |
Collapse
|
98
|
Chesnoy S, Huang L. Enhanced cutaneous gene delivery following intradermal injection of naked DNA in a high ionic strength solution. Mol Ther 2002; 5:57-62. [PMID: 11786046 DOI: 10.1006/mthe.2001.0511] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intradermal injection of naked DNA results in gene transfer to skin cells, but the efficiency of this gene transfer method is relatively low and variable. We have systematically optimized several parameters to obtain reproducible, high-level gene transfer to the mouse skin. Older mice (approximately 7 weeks) showed a significant decrease in gene expression compared with younger mice (4-5 weeks old). The composition of the solvent vehicle (electrolyte versus nonelectrolyte) strongly affected gene expression in the skin. A higher level of gene expression was achieved when naked DNA was dissolved in isotonic phosphate buffered saline solution compared with isotonic dextrose solution. Finally, transfection efficiency in older mice was greatly improved by increasing the ionic strength of the solvent vehicle. The improved transfection efficiency was due to an enhanced DNA uptake by the skin cells. Gene transfer was most evident in the subdermal smooth muscle cells and epidermal cells. With the optimized conditions, gene transfer mediated by intradermal injection of naked DNA was comparable in efficiency to electroporation. However, cellular distributions of the gene transfer of the two methods were different.
Collapse
Affiliation(s)
- Sophie Chesnoy
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
99
|
Kircheis R, Wightman L, Wagner E. Design and gene delivery activity of modified polyethylenimines. Adv Drug Deliv Rev 2001; 53:341-58. [PMID: 11744176 DOI: 10.1016/s0169-409x(01)00202-2] [Citation(s) in RCA: 500] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The polycation polyethylenimine (PEI) has recently been widely employed for the design of DNA delivery vehicles. Gene delivery using PEI involves condensation of DNA into compact particles, uptake into the cells, release from the endosomal compartment into the cytoplasm, and uptake of the DNA into the nucleus. Particularly for in vivo gene delivery, optimal coordination and timing between DNA complexation for protection of the DNA from nucleases and the disassembly of the complexes is essential. For in vivo application, DNA complexes have to pass a variety of anatomical and physiological barriers, and an environment of biological fluids and extracellular matrix before reaching their targets. Furthermore, targeted gene delivery is seriously hampered by non-specific interactions with non-target cells. Strategies have been developed to protect transfection complexes from non-specific interactions and to increase target specificity and gene expression.
Collapse
Affiliation(s)
- R Kircheis
- Department of Cancer Vaccines and Gene Therapy, Boehringer Ingelheim Austria GmbH, Dr. Boehringer-Gasse 5-11, A-1121 Vienna, Austria.
| | | | | |
Collapse
|
100
|
Maruyama H, Ataka K, Higuchi N, Sakamoto F, Gejyo F, Miyazaki J. Skin-targeted gene transfer using in vivo electroporation. Gene Ther 2001; 8:1808-12. [PMID: 11803401 DOI: 10.1038/sj.gt.3301604] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2001] [Accepted: 08/31/2001] [Indexed: 11/09/2022]
Abstract
The skin is an important target for gene transfer because of its easy accessibility. Using plasmid DNA expressing rat erythropoietin (pCAGGS-Epo) as the vector, we previously demonstrated long-term Epo delivery in rats by muscle-targeted gene transfer using in vivo electroporation. Here we examined whether this electroporation approach could be applied to gene delivery in rat skin. To optimize gene transfer, we tested the efficiency of skin-targeted Epo gene transfer with three types of electrodes at three different electrode voltages. Each rat was injected intradermally with a total of 800 microg of pCAGGS-Epo, in the abdominal area. Plate-and-fork-type electrodes were effective for Epo delivery by skin-targeted gene transfer at low voltages (12 approximately 24 V). The vector-derived Epo mRNA was expressed only at the DNA injection site. The Epo gene was expressed in a dose-dependent manner, the expression persisted for 7 weeks, and hematocrit levels were increased for 11 weeks. Skin injection with pCAGGS-lacZ showed lacZ gene expression in the epidermis on day 1 after injection and in the subcutaneous muscle layer on day 7. Slight skin damage due to the gene transfer procedure was evident on day 1, but absent by day 7. These results demonstrate that skin-targeted pCAGGS-Epo transfer by in vivo electroporation at low voltage is a useful procedure for the short-term delivery of Epo.
Collapse
Affiliation(s)
- H Maruyama
- Department of Medicine (II), Niigata University School of Medicine, Niigata, Japan
| | | | | | | | | | | |
Collapse
|