1
|
Castañeda-Lopez ME, Garza-Veloz I, Lopez-Hernandez Y, Barbosa-Cisneros OY, Martinez-Fierro ML. Anti-Inflammatory Effects of Modified Adenoviral Vectors for Gene Therapy: A View through Animal Models Tested. Immunol Invest 2016; 45:450-70. [PMID: 27245510 DOI: 10.3109/08820139.2016.1168831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The central dogma of gene therapy relies on the application of novel therapeutic genes to treat or prevent diseases. The main types of vectors used for gene transfer are adenovirus, retrovirus, lentivirus, liposome, and adeno-associated virus vectors. Gene therapy has emerged as a promising alternative for the treatment of inflammatory diseases. The main targets are cytokines, co-stimulatory molecules, and different types of cells from hematological and mesenchymal sources. In this review, we focus on molecules with anti-inflammatory effects used for in vivo gene therapy mediated by adenoviral gene transfer in the treatment of immune-mediated inflammatory diseases, with particular emphasis on autoinflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- M E Castañeda-Lopez
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - I Garza-Veloz
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - Y Lopez-Hernandez
- c CONACyT Research Fellow, Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud , Universidad Autonoma de Zacatecas , Mexico
| | - O Y Barbosa-Cisneros
- d Laboratory of Cell and Molecular Biology, Unidad Academica de Ciencias Quimicas de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - M L Martinez-Fierro
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| |
Collapse
|
2
|
Abstract
This introductory chapter provides a short review of the ideas and practical approaches that have led to the present and perceived future development of prenatal gene therapy. It summarizes the advantages and the potential adverse effects of this novel preventive and therapeutic approach to the management of prenatal diseases. It also provides guidance to the range of conditions to which prenatal gene therapy may be applied and to the technical approaches, vectors, and societal/ethical considerations for this newly emerging field of Fetal Medicine.
Collapse
|
3
|
Davey MG, Zoltick PW, Todorow CA, Limberis MP, Ruchelli ED, Hedrick HL, Flake AW. Jaagsiekte sheep retrovirus pseudotyped lentiviral vector-mediated gene transfer to fetal ovine lung. Gene Ther 2011; 19:201-9. [PMID: 21654824 DOI: 10.1038/gt.2011.83] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Viral vector-mediated gene transfer to the postnatal respiratory epithelium has, in general, been of low efficiency due to physical and immunological barriers, non-apical location of cellular receptors critical for viral uptake and limited transduction of resident stem/progenitor cells. These obstacles may be overcome using a prenatal strategy. In this study, HIV-1-based lentiviral vectors (LVs) pseudotyped with the envelope glycoproteins of Jaagsiekte sheep retrovirus (JSRV-LV), baculovirus GP64 (GP64-LV), Ebola Zaire-LV or vesicular stomatitis virus (VSVg-LV) and the adeno-associated virus-2/6.2 (AAV2/6.2) were compared for in utero transfer of a green fluorescent protein (GFP) reporter gene to ovine lung epithelium between days 65 and 78 of gestation. GFP expression was examined on day 85 or 136 of gestation (term is ∼145 days). The percentage of the respiratory epithelial cells expressing GFP in fetal sheep that received the JSRV-LV (3.18 × 10(8)-6.85 × 10(9) viral particles per fetus) was 24.6±0.9% at 3 weeks postinjection (day 85) and 29.9±4.8% at 10 weeks postinjection (day 136). Expression was limited to the surface epithelium lining fetal airways <100 μm internal diameter. Fetal airways were amenable to VSVg-LV transduction, although the percentage of epithelial expression was low (6.6±0.6%) at 1 week postinjection. GP64-LV, Ebola Zaire-LV and AAV2/6.2 failed to transduce the fetal ovine lung under these conditions. These data demonstrate that prenatal lung gene transfer with LV engineered to target apical surface receptors can provide sustained and high levels of transgene expression and support the therapeutic potential of prenatal gene transfer for the treatment of congenital lung diseases.
Collapse
Affiliation(s)
- M G Davey
- The Children's Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
4
|
Saada J, Oudrhiri N, Bonnard A, de Lagausie P, Aissaoui A, Hauchecorne M, Oury JF, Aigrain Y, Peuchmaur M, Lehn JM, Lehn P, Luton D. Combining keratinocyte growth factor transfection into the airways and tracheal occlusion in a fetal sheep model of congenital diaphragmatic hernia. J Gene Med 2010; 12:413-22. [DOI: 10.1002/jgm.1451] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
5
|
Endo M, Henriques-Coelho T, Zoltick PW, Stitelman DH, Peranteau WH, Radu A, Flake AW. The developmental stage determines the distribution and duration of gene expression after early intra-amniotic gene transfer using lentiviral vectors. Gene Ther 2009; 17:61-71. [PMID: 19727133 DOI: 10.1038/gt.2009.115] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene transfer after intra-amniotic injection has, in general, been of low efficiency and limited to epithelial cells in the skin, pulmonary and gastrointestinal system. We have recently shown that early gestational administration results in a more efficient gene transfer to developmentally accessible stem cell populations in the skin and eye. In this study we present a comprehensive analysis of patterns of tissue expression seen after early intra-amniotic gene transfer (IAGT) using lentiviral vectors. To assess the influence of developmental stage on tissue expression, injections were administered from the late head fold/early somite stage (E8) to E18. In early gestation (E8-10), green fluorescent protein (GFP) expression was observed in multiple organs, derived from all three germ layers. Remarkably, GFP expression was observed in tissues derived from mesoderm and neural ectoderm at E8, whereas expression was limited to only epithelial cells of ectoderm- and endoderm-derived organs after E11. The amount and duration of gene expression was much higher after IAGT at early gestational time points. The observed temporal patterns of gene expression correspond to the predicted developmental accessibility of organ-specific cell populations. This model may be useful for the analyses of mechanisms of genetic and/or developmental disease and for the development of prenatal gene therapy for specific disorders.
Collapse
Affiliation(s)
- M Endo
- The Children's Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Genetically modified adenoviral vector with the protein transduction domain of Tat improves gene transfer to CAR-deficient cells. Biosci Rep 2009; 29:103-9. [PMID: 18721127 PMCID: PMC2630516 DOI: 10.1042/bsr20080023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The transduction efficiency of Ad (adenovirus) depends, to some extent, on the expression level of CAR (coxsackievirus and Ad receptor) of a target cell. The low level of CAR on the cell surface is a potential barrier to efficient gene transfer. To overcome this problem, PTD.AdeGFP (where eGFP is enhanced green fluorescent protein) was constructed by modifying the HI loop of Ad5 (Ad type 5) fibre with the Tat (trans-activating) PTD (protein transduction domain) derived from HIV. The present study showed that PTD.AdeGFP significantly improved gene transfer to multiple cell types deficient in expression of CAR. The improvement in gene transfer was not the result of charge-directed binding between the virus and the cell surface. Although PTD.AdeGFP formed aggregates, it infected target cells in a manner different from AdeGFP aggregates precipitated by calcium phosphate. In addition, PTD.AdeGFP was able to transduce target cells in a dynamin-independent pathway. The results provide some new clues as to how PTD.AdeGFP infects target cells. This new vector would be valuable in gene-function analysis and for gene therapy in cancer.
Collapse
|
7
|
Lentiviral transduction of the murine lung provides efficient pseudotype and developmental stage-dependent cell-specific transgene expression. Gene Ther 2008; 15:1167-75. [PMID: 18432275 DOI: 10.1038/gt.2008.74] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene transfer for cystic fibrosis (CF) airway disease has been hampered by the lung's innate refractivity to pathogen infection. We hypothesized that early intervention with an integrating gene transfer vector capable of transducing the lung via the lumen may be a successful therapeutic approach. An HIV-based lentiviral vector pseudotyped with the baculovirus gp64 envelope was applied to the fetal, neonatal or adult airways. Fetal intra-amniotic administration resulted in transduction of approximately 14% of airway epithelial cells, including both ciliated and non-ciliated epithelia of the upper, mid and lower airways; there was negligible alveolar or nasal transduction. Following neonatal intra-nasal administration we observed significant transduction of the airway epithelium (approximately 11%), although mainly in the distal lung, and substantial alveolar transduction. This expression was still detectable at 1 year after application. In the adult, the majority of transduction was restricted to the alveoli. In contrast, vesicular stomatitis virus glycoprotein pseudotyped virus transduced only alveoli after adult and neonatal application and no transduction was observed after fetal administration. Repeat administration did not increase transduction levels of the conducting airway epithelia. These data demonstrate that application at early developmental stages in conjunction with an appropriately pseudotyped virus provides efficient, high-level transgene expression in the murine lung. This may provide a modality for treatment for lung disease in CF.
Collapse
|
8
|
Abstract
Gene therapy uses the intracellular delivery of genetic material for the treatment of disease. A wide range of diseases - including cancer, vascular and neurodegenerative disorders and inherited genetic diseases - are being considered as targets for this therapy in adults. There are particular reasons why fetal application might prove better than application in the adult for treatment, or even prevention of early-onset genetic disorders such as cystic fibrosis and Duchenne muscular dystrophy. Research shows that gene transfer to the developing fetus targets rapidly expanding populations of stem cells, which are inaccessible after birth, and indicates that the use of integrating vector systems results in permanent gene transfer. In animal models of congenital disease such as haemophilia, studies show that the functionally immature fetal immune system does not respond to the product of the introduced gene, and therefore immune tolerance can be induced. This means that treatment could be repeated after birth, if that was necessary to continue to correct the disease. For clinicians and parents, fetal gene therapy would give a third choice following prenatal diagnosis of inherited disease, where termination of pregnancy or acceptance of an affected child are currently the only options. Application of this therapy in the fetus must be safe, reliable and cost-effective. Recent developments in the understanding of genetic disease, vector design, and minimally invasive delivery techniques have brought fetal gene therapy closer to clinical practice. However more research needs to be done in before it can be introduced as a therapy.
Collapse
Affiliation(s)
- Anna L David
- Department of Obstetrics & Gynaecology, Royal Free & University College London Medical School, 86-96 Chenies Mews, London, WC1E 6HX, UK.
| | | |
Collapse
|
9
|
David AL, Weisz B, Gregory L, Themis M, Cook T, Roubliova X, Deprest J, Coutelle C, Rodeck CH, Peebles DM. Ultrasound-guided injection and occlusion of the trachea in fetal sheep. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2006; 28:82-8. [PMID: 16795134 DOI: 10.1002/uog.2815] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
OBJECTIVES To access the fetal sheep trachea by ultrasound-guided transthoracic injection in order to deliver gene therapy vectors or occlude the trachea with a detachable balloon. METHODS Fetal sheep were operated on at a mean gestational age of 102 (range, 81-116) days (term = 145 days). Under ultrasound guidance, either a 20-G spinal (for vector delivery) or a 16-G Kellett (for placement of an occlusive balloon) needle was inserted via the fetal thorax into the fetal trachea. RESULTS Using the 20-G spinal needle the trachea was accessed successfully in 33/36 fetuses, with 97% survival. Failure to inject was related to fetal position and gestational age. Blood vessel damage causing significant morbidity occurred in two fetuses (6%). Tracheal occlusion was achieved by puncturing the trachea with the 16-G needle and advancing an endoluminal balloon in three out of five attempts in a mean time of 17 (range, 16-19) min, with 100% survival. In one case, the balloon became sited within the accessory lobe bronchus and was not inflated. At postmortem examination 21 days later, all balloons remained inflated and occluded the trachea, and the lung-to-body weight ratio and airways morphometric indices were consistent with relative pulmonary hyperplasia in the obstructed lungs. CONCLUSIONS Ultrasound-guided transthoracic tracheal puncture is a reliable technique in fetal sheep, with low morbidity and mortality. Using this technique, a detachable endotracheal balloon can be placed to provoke pulmonary growth. Advances in needle design and balloon size may improve the success rate.
Collapse
Affiliation(s)
- A L David
- Department of Obstetrics and Gynaecology, Royal Free and University College Medical School, University College London, London, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Rinaldi M, Signori E, Rosati P, Cannelli G, Parrella P, Iannace E, Monego G, Ciafrè SA, Farace MG, Iurescia S, Fioretti D, Rasi G, Fazio VM. Feasibilty of in utero DNA vaccination following naked gene transfer into pig fetal muscle: Transgene expression, immunity and safety. Vaccine 2006; 24:4586-91. [PMID: 16154671 DOI: 10.1016/j.vaccine.2005.08.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The high toll of death among first-week infants is due to infections occurring at the end of pregnancy, during birth or by breastfeeding. This problem significantly concerns industrialized countries also. To prevent the typical "first-week infections", a vaccine would be protective as early as at the birth. In utero DNA immunization has demonstrated the effectiveness in inducing specific immunity in newborns. We have already published results of a 2-year follow-up showing long-term safety, protective antibody titers at birth and long-term immune memory, following intramuscular in utero anti-HBV DNA immunization in 90-days pig fetuses. We have now analyzed further parameters of short-term safety. Two different reporter genes were injected in the thigh muscles of 90-days fetuses. At 8 days following DNA injection, we found high-level of transgenes expression in all injected fetuses. A step gradient of expression from the area of injection was observed with both reporter genes. CMV promoter/enhancer produced higher levels of expression compared to SV40 promoter/enhancer. Moreover, no evidence of local or systemic flogistic alterations or fetal malformations, mortality or haemorrhage following intramuscular injection were observed. A single anti-HBV s-antigen DNA immunization in 90-days fetuses supported protective antibody levels in all immunized newborns, lasting at least up to 4 months after birth. Our report further sustains safety and efficacy of intramuscular in utero naked gene transfer and immunization. This approach may support therapeutic or prophylactic procedure in many early life-threatening pathologic conditions.
Collapse
Affiliation(s)
- Monica Rinaldi
- Institute of Neurobiology and Molecular Medicine, CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Buckley SMK, Waddington SN, Jezzard S, Lawrence L, Schneider H, Holder MV, Themis M, Coutelle C. Factors influencing adenovirus-mediated airway transduction in fetal mice. Mol Ther 2005; 12:484-92. [PMID: 16099411 DOI: 10.1016/j.ymthe.2005.02.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 02/15/2005] [Accepted: 02/22/2005] [Indexed: 11/30/2022] Open
Abstract
Intra-amniotic injection of adenovirus allows transduction of the fetal airways following natural fetal breathing movements. This administration method is promising for use in gene therapy for cystic fibrosis and other diseases for which the main target for exogenous gene expression is the lung. Here we have investigated factors that may affect the efficacy of gene transfer to the murine fetal lung. We examined marker compound distribution and transgene expression (from a first-generation adenoviral vector) at different stages of development. This demonstrated that fetal breathing movements at 15-16 days of gestation are of sufficient intensity to carry marker/vector into the fetal lungs. These movements can be significantly stimulated by the combination of intra-amniotic theophylline administration and postoperative exposure of the dam to elevated CO(2) levels. However, the most important factor for efficient and consistent pulmonary transgene delivery is the dose of adenoviral vector used, as both the degree of transduction and the percentage of lungs transduced increases with escalating viral dose.
Collapse
Affiliation(s)
- S M K Buckley
- Gene Therapy Research Group, Department of Cell and Molecular Biology, SAF Building, Imperial College, South Kensington, London SW7 2AZ, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Shen JS, Meng XL, Maeda H, Ohashi T, Eto Y. Widespread gene transduction to the central nervous system by adenovirus in utero: implication for prenatal gene therapy to brain involvement of lysosomal storage disease. J Gene Med 2005; 6:1206-15. [PMID: 15459963 DOI: 10.1002/jgm.630] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In some lysosomal storage diseases, considerable alterations of the central nervous system (CNS) occur prior to birth and neurodegeneration progresses rapidly soon after birth causing early death in patients. No effective treatment is available after birth. Treatment may need to be initiated before birth to prevent the onset or progression of neurological changes and thereby irreversible brain damage. The aim of this study is to investigate the feasibility and effectiveness of brain-directed prenatal gene therapy for lysosomal storage diseases. METHODS Recombinant adenovirus encoding the lacZ gene was injected into the lateral ventricles of mouse embryos and the pattern of gene transduction to the CNS was investigated. In the therapeutic experiment, adenovirus expressing beta-glucuronidase was injected into the cerebral ventricles of the embryos of mucopolysaccharidosis VII mice and the therapeutic effects on the brain were evaluated. RESULTS Injection of adenoviral vectors to the cerebral ventricles of mouse embryos led to widespread gene transduction throughout the brain and the spinal cord and transgene expression persisted over 10 months in those surviving the procedure. The prenatal transduction of the therapeutic gene to the brain of the mucopolysaccharidosis VII mouse efficiently prevented lysosomal storage in most brain cells before birth until 4 months after birth. CONCLUSIONS Brain-directed in utero gene therapy through an intra-ventricular route would be an effective strategy to treat some lysosomal storage diseases with early and severe CNS alterations.
Collapse
Affiliation(s)
- Jin-Song Shen
- Department of Gene Therapy, Institute of DNA Medicine, The Jikei University School of Medicine, Minato-Ku, Tokyo 105-8461, Japan
| | | | | | | | | |
Collapse
|
13
|
Theiss AL, Simmons JG, Jobin C, Lund PK. Tumor necrosis factor (TNF) alpha increases collagen accumulation and proliferation in intestinal myofibroblasts via TNF receptor 2. J Biol Chem 2005; 280:36099-109. [PMID: 16141211 DOI: 10.1074/jbc.m505291200] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intestinal fibrosis is an incurable complication of Crohn's disease involving increased numbers of collagen-producing myofibroblasts. Tumor necrosis factor (TNF) alpha has defined proinflammatory roles in Crohn's disease but its role in fibrosis is unclear. We tested the hypothesis that TNFalpha increases collagen accumulation and proliferation in intestinal myofibroblasts and has additive effects in combination with insulin-like growth factor (IGF) I. The mechanisms, TNF receptor isoform, and downstream signaling pathways were examined. Intestinal myofibroblasts from wild-type (WT) mice or mice homozygous for disruption of genes encoding TNFR1 (TNFR1-/-), TNFR2 (TNFR2-/-), or both (TNFR1/2-/-), were treated with TNFalpha, IGF-I, or both. In WT cells, TNFalpha and IGF-I stimulated type I collagen accumulation and DNA synthesis in an additive manner. IGF-I, but not TNFalpha, stimulated type I collagen gene activation. TNFalpha, but not IGF-I, induced tissue inhibitor of metalloproteinase-1 (TIMP-1) expression and reduced matrix metalloproteinases-2 activity and collagen degradation. TNFalpha also activated ERK1/2. These responses to TNFalpha were absent in TNFR2-/- and TNFR1/2-/- myofibroblasts, whereas TNFR1-/- cells showed similar responses to WT. Inhibition of ERK1/2 diminished TNFalpha induced DNA synthesis in WT and TNFR1-/- cells. Differences in TNFalpha-induced STAT3/DNA binding activity and not NFkappaB and AP-1 transcriptional activation correlated with impaired collagen accumulation/TIMP-1 induction in TNFR2(-/-) cells. Constitutively active STAT3 rescued TIMP-1 expression in TNFR2-/- cells. We conclude that TNFalpha and IGF-I may additively contribute to fibrosis during intestinal inflammation. TNFR2 is a primary mediator of fibrogenic actions of TNFalpha acting through ERK1/2 to stimulate proliferation and through STAT3 to stimulate TIMP-1 and inhibit collagen degradation.
Collapse
Affiliation(s)
- Arianne L Theiss
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
14
|
Waddington SN, Kramer MG, Hernandez-Alcoceba R, Buckley SMK, Themis M, Coutelle C, Prieto J. In utero gene therapy: current challenges and perspectives. Mol Ther 2005; 11:661-76. [PMID: 15851005 DOI: 10.1016/j.ymthe.2005.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Over the past few years, considerable progress in prenatal diagnosis and surgery combined with improvements in vector design vindicate a reappraisal of the feasibility of in utero gene therapy for serious monogenetic diseases. As adult gene therapy gathers pace, several apparent obstacles to its application as a treatment may be overcome by pre- or early postnatal treatment. This review will examine the concepts and practice of prenatal vector administration. We aim to highlight the advantages of early therapeutic intervention focusing on diseases that could benefit greatly from a prenatal gene therapy approach. We will pay special attention to the strategies and vectors that are most likely to be used for this application and will speculate on their expected developments for the near future.
Collapse
Affiliation(s)
- Simon N Waddington
- Gene Therapy Research Group, Sir Alexander Fleming Building, Imperial College, South Kensington, London SW7 2AZ, UK
| | | | | | | | | | | | | |
Collapse
|
15
|
Sato M, Tanigawa M, Kikuchi N. Nonviral gene transfer to surface skin of mid-gestational murine embryos by intraamniotic injection and subsequent electroporation. Mol Reprod Dev 2005; 69:268-77. [PMID: 15349838 DOI: 10.1002/mrd.20124] [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] [Indexed: 11/11/2022]
Abstract
The surface epithelium of mid-gestational murine embryos is thought to be an attractive target for gene therapy in vivo, due to its visibility and accessibility from the external surface of the maternal uterus. Almost all studies of in utero gene transfer have adopted viral vectors for infection of fetal epithelium, and depended on intraamniotic introduction and simple incubation of vectors, leading to only infection of the surface layer (periderm) of fetal skin. Here we report a simple and convenient method of gene transfer of plasmid DNA into the deeper portion of surface skin of murine mid-gestational fetus. One to two microlitres of a solution containing a lacZ expression plasmid (0.5-1 microg) and trypan blue (0.05%) were placed onto the surface of a fetus (E 14.5) near the eye by a micropipette attached to a mouthpiece. This fetus was immediately electroporated by placing it between tweezer-type electrodes attached to a square-pulse generator. At 1 and 4 days after gene transfer, fetuses were subjected to histochemical staining for lacZ activity in the presence of X-Gal, a substrate for lacZ. Focal reactions were observed in the skin epidermal layers including periderm and basal layer 1 day after DNA introduction. However, lacZ-positive cells were limited to a skin surface layer, the stratum corneum, in the samples obtained 4 days after gene transfer. Similar observation was also made in the transgenic fetuses (carrying a lacZ gene placed immediately downstream of the loxP-flanked sequence) injected with Cre expression vector. These findings suggest rapid movement of fetal epidermal cells toward the surface during late developmental stages. This local gene transfer approach appears to be effective as a method for skin-targeted gene transfer, enabling study of the role of genes of interest and tracing of cell lineage during fetal skin development.
Collapse
Affiliation(s)
- Masahiro Sato
- The Institute of Medical Sciences, Tokai University, Bohseidai, Isehara, Kanagawa, Japan.
| | | | | |
Collapse
|
16
|
Meyerholz DK, Grubor B, Gallup JM, Lehmkuhl HD, Anderson RD, Lazic T, Ackermann MR. Adenovirus-mediated gene therapy enhances parainfluenza virus 3 infection in neonatal lambs. J Clin Microbiol 2004; 42:4780-7. [PMID: 15472341 PMCID: PMC522350 DOI: 10.1128/jcm.42.10.4780-4787.2004] [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: 11/20/2022] Open
Abstract
Parainfluenza viruses are a common cause of seasonal respiratory disease, but in high-risk individuals (e.g., young children) these viruses can cause severe clinical manifestations that require hospitalization. Beta-defensins are a subclass of antimicrobial peptides with antiviral activity. Use of adenovirus-mediated beta-defensin gene expression has been proposed as therapy for chronic bacterial infections commonly seen in cystic fibrosis patients; however, its use during parainfluenza virus 3 (PIV3) infection has not been evaluated. The hypothesis in this experiment was that adenovirus expression of human beta-defensin 6 (HBD6) would diminish concurrent PIV3 infection in neonatal lambs. The group infected with adenovirus HBD6 and PIV3 had increased levels of pulmonary neutrophil recruitment compared to those for the group infected with PIV3 or PIV3 and adenovirus, with an increased respiration rate and body temperature late in the course of the PIV3-adenovirus HBD6 infection. Interestingly, the adenovirus-treated groups had higher levels of immunohistochemical staining for PIV3 and syncytial cell formation than the group infected with PIV3, suggesting that treatment with the adenovirus vector, regardless of whether it was carrying a target gene, exacerbated the PIV3 infection. The levels of expression of mRNA for antimicrobial surfactant proteins A and D and sheep beta-defensin 1 were increased by PIV3 and adenovirus treatment, and the increased levels of expression roughly corresponded to the degree of inflammation. While pulmonary administration of a high-dose adenovirus vector has been associated with undesirable inflammation, this is the first study to show that it can exacerbate concurrent viral infection, a concern that needs to be addressed for future studies of adenovirus in the lung. Additionally, this study showed that adenovirus-mediated HBD6 expression increases neutrophil recruitment, a recently described attribute of beta-defensins, with mild accentuation of PIV3 activity and inflammation.
Collapse
Affiliation(s)
- David K Meyerholz
- Department of Veterinary Pathology, Iowa State University, Ames, Iowa 50011-1250, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Bouchard S, MacKenzie TC, Radu AP, Hayashi S, Peranteau WH, Chirmule N, Flake AW. Long-term transgene expression in cardiac and skeletal muscle following fetal administration of adenoviral or adeno-associated viral vectors in mice. J Gene Med 2004; 5:941-50. [PMID: 14601131 DOI: 10.1002/jgm.421] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In utero gene transfer may provide advantages for the correction of congenital genetic disorders. In the present study we compare the ability of adenovirus (AdCMVLacZ), and two serotypes of adeno-associated virus (AAVCMVLacZ serotypes 2 and 2/5), to target cardiac and skeletal muscle after prenatal systemic or intramuscular injection in mice and assess the immune response to the vectors. METHODS Day 14 gestation fetal mice underwent direct intraperitoneal or intramuscular injection of AdCMVLacZ, and AAVCMVLacZ serotypes 2 and 2/5 vectors. Tissues were processed for beta-galactosidase expression in frozen or high-resolution thin plastic sections at early and late time points. Neutralizing antibodies to Ad and AAV were analyzed in separate fetal experimental and neonatal or adult control groups after administration and re-administration of the vectors. RESULTS A single injection of each vector in utero resulted in sustained expression of beta-galactosidase transgene in skeletal and cardiac muscle. Transgene expression was detected for the length of the study, i.e. 86, 58, and 31 weeks after birth for AdCMVLacZ, and AAVCMVLacZ serotypes 2 and 2/5, respectively. High-level expression in the myocardium was observed independent of the vector or route of administration. Neutralizing antibody responses to AAV and Ad antigens were reduced and long-term expression in muscle was not ablated on postnatal re-administration of vector. CONCLUSIONS Sustained, high-level cardiac and skeletal muscle transgene expression can be obtained after prenatal gene transfer with each of these vectors. The potential for immune response to viral antigens is altered, but not entirely ablated after in utero exposure.
Collapse
Affiliation(s)
- Sarah Bouchard
- Division of Pediatric General, Thoracic, and Fetal Surgery, The Children's Institute for Surgical Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Peebles D, Gregory LG, David A, Themis M, Waddington SN, Knapton HJ, Miah M, Cook T, Lawrence L, Nivsarkar M, Rodeck C, Coutelle C. Widespread and efficient marker gene expression in the airway epithelia of fetal sheep after minimally invasive tracheal application of recombinant adenovirus in utero. Gene Ther 2004; 11:70-8. [PMID: 14681699 DOI: 10.1038/sj.gt.3302130] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cystic fibrosis is a common lethal genetic disease caused by functional absence of the cystic fibrosis transmembrane conductance regulator (CFTR). Although a candidate disease for in utero gene therapy, demonstration of potentially therapeutic levels of transgene expression in the fetal airways after minimally invasive gene delivery is a mandatory prerequisite before application of this approach in humans can be considered. We report here on the delivery of a beta-galactosidase expressing adenovirus directly to the airways of fetal sheep in utero using ultrasound-guided percutaneous injection of the trachea in the fetal chest. Injection of adenoviral particles to the fetal airways was not associated with mortality and resulted in low-level expression in the peripheral airways. However, complexation of the virus with DEAE dextran, which confers a positive charge to the virus, and pretreatment of the airways with Na-caprate, which opens tight junctions, increased transgene expression, and a combination of these two enhancers resulted in widespread and efficient gene transfer of the fetal trachea and bronchial tree. Using a percutaneous ultrasound-guided injection technique, we have clearly demonstrated proof of principle for substantial transgene delivery to the fetal airways providing levels of gene expression that could be relevant for a therapeutic application of CFTR expressing vectors.
Collapse
Affiliation(s)
- D Peebles
- Department of Obstetrics and Gynaecology, Royal Free and University College Medical School, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
In the near future, prenatal therapy may include stem cell-based cellular therapy or gene therapy. There is considerable overlap in the rationale and potential applications for these 2 approaches. The purpose of this manuscript is to consider current progress in both areas relevant to prenatal treatment. Although clinical application is currently limited to a few highly selected disorders that are amenable to cellular therapy, there is reason to believe that a dramatic increase in application will occur in the near future.
Collapse
Affiliation(s)
- Alan W Flake
- The Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Boyle MP, Enke RA, Mogayzel PJ, Guggino WB, Martin DB, Agarwal S, Zeitlin PL. Effect of adeno-associated virus-specific immunoglobulin G in human amniotic fluid on gene transfer. Hum Gene Ther 2003; 14:365-73. [PMID: 12659677 DOI: 10.1089/104303403321208961] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intra-amniotic administration of adeno-associated virus (AAV) vector may be an effective way to deliver gene therapy for treatment of congenital pulmonary and intestinal disorders. In an effort to understand potential barriers to intra-amniotic gene therapy better, we determined whether human amniotic fluid (AF) could act as an inhibitor of AAV2-mediated gene transfer. AF samples were obtained from 21 different human pregnancies during routine amniocentesis at 16-20 weeks of gestation. An immortalized fetal human tracheal epithelial cell line (FHTE) was infected with AAV2 containing a luciferase reporter gene driven by the SV40 promoter in the presence and absence of each AF sample. Inhibition of transgene expression was observed in 8 (38%) of the AF samples (inhibitory AF) and resulted in luciferase levels of only 1.4% +/- 0.6% of those obtained with infection in normal media. Infections in 13 samples (62%) resulted in transgene expression comparable or in excess of infection in media alone (noninhibitory AF). Removal of immunoglobulin G (IgG) from inhibitory AF samples with Protein A returned luciferase expression to control levels (119% +/- 37% of control), suggesting the possible presence of inhibiting antibody. Eleven of the AF samples were evaluated by enzyme-linked immunosorbent assay (ELISA) for specific anti-AAV antibodies. All noninhibitory AF samples were negative (titers of < 1:20; n = 3), and 6 of the 8 inhibitory samples contained specific anti-AAV antibodies at titers ranging from 1:40 to 1:160. These studies demonstrate that AF from some individuals contains AAV-specific IgG that can inhibit gene transfer.
Collapse
Affiliation(s)
- Michael P Boyle
- Department of Medicine, The Johns Hopkins University School of Medicine, Jefferson B1-170, 600 North Wolfe Street, Baltimore, MD 21287-8922, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Kikuchi N, Nakamura S, Ohtsuka M, Kimura M, Sato M. Possible mechanism of gene transfer into early to mid-gestational mouse fetuses by tail vein injection. Gene Ther 2002; 9:1529-41. [PMID: 12407425 DOI: 10.1038/sj.gt.3301818] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2002] [Accepted: 05/10/2002] [Indexed: 11/08/2022]
Abstract
Our aim is to develop a simple gene transfer method into egg cylinder and mid-gestational murine embryos. We examined whether plasmid/lipid complexes injected into the tail veins of pregnant transgenic mice can be transferred to fetuses at E 4.5-13.5. When pregnant CETZ-17 mice carrying a transgene consisting of a ubiquitous promoter, floxed EGFP/CAT and the LacZ gene, were injected with a Cre expression vector DNA/lipid complex, Cre-mediated excision of the transgenes, as evaluated by X-gal staining, occurred in 10-50% of fetuses treated at E 11.5-13.5. Although younger embryos remained unstained, PCR analysis revealed low levels of the Cre vector DNA and recombined transgene. To examine the fate of a solution given intravenously, we injected trypan blue or fluorescence-labeled plasmid DNA/lipid complexes into females at E 5.5-11.5 and E 6.5, respectively. Both collected in the visceral endoderm (VE) lineage, but were undetectable in the embryo proper. These findings suggest that substances in maternal blood are delivered to post-implantation embryos via cells of the VE lineage and placenta, but that most are trapped in the VE. If significantly improved, gene transfer to fetuses by injection into the maternal circulation may become a promising tool in fetal gene therapy and embryological studies.
Collapse
Affiliation(s)
- N Kikuchi
- Division of Molecular Life Science, School of Medicine, Tokai University, Isehara, Japan
| | | | | | | | | |
Collapse
|
22
|
Waszak P, Franco-Montoya ML, Jacob MP, Deprez I, Levame M, Lafuma C, Harf A, Delacourt C. Effect of intratracheal adenoviral vector administration on lung development in newborn rats. Hum Gene Ther 2002; 13:1873-85. [PMID: 12396619 DOI: 10.1089/104303402760372963] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Local overexpression of genes that promote lung defense or repair may be helpful in protecting the immature neonatal lung from injuries, but whether the vectors used to administer these genes affect physiological postnatal lung growth has not been investigated. We explored the effect on alveolarization of E1-deleted Adnull vector (Ad5-LMP-null) given intratracheally to 3-day-old rats. Three Adnull doses were evaluated 10(8), 5 x 10(8), and 10(9) TCID(50). Lung morphometry on day 21 showed significant growth disorders with the two higher doses. With 5 x 10(8) TCID(50), absolute lung volume increased significantly (+16%), as did absolute (+20%) and specific (+32%) alveolar airspace volumes, whereas alveolar surface density decreased by 13% (p < 0.009 for all parameters). Lung inflammation was mild, nonsignificant, and occurred mainly with the highest Adnull dose, indicating that it was unlikely to contribute to our results. Adnull instillation induced a significant#10; decrease in terminal bronchiolar cell proliferation as evaluated by proliferating cell nuclear antigen immunostaining (p = 0.02), as well as a 23% decrease in absolute parenchyma elastic fiber length (p = 0.02). Furthermore, lung tropoelastin mRNA content decreased by 25% (p < 0.02). In conclusion, E1-deleted adenoviral vectors can induce lung growth disorders when instilled into the airways of neonatal rats. Interactions with lung matrix turnover may be the main explanation to these deleterious effects.
Collapse
Affiliation(s)
- Paul Waszak
- INSERM U492, Faculté de Médecine, 94010 Créteil, France
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- Alan W Flake
- Center for Fetal Diagnosis and Therapy, The Children's Institute for Surgical Science, Children's Hospital of Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
24
|
Galan HL, Bennett ML, Tyson RW, Owens G, Regnault TRH, Accurso F, Hobbins JC, Schaack J. Inefficient transduction of sheep in utero after intra-amniotic injection of retroviral producer cells. Am J Obstet Gynecol 2002; 187:469-74. [PMID: 12193945 DOI: 10.1067/mob.2002.123601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Our purpose was to test the hypothesis that the intra-amniotic injection of a retroviral vector producer cell line into pregnant sheep will result in retrovirus-mediated transduction and stable gene transfer to the ovine fetus. STUDY DESIGN Thirteen pregnant ewes at various gestational ages underwent amniocentesis and injection of cells producing the retrovirus vector LIRESgeo, which is derived from Maloney murine leukemia virus and encodes Escherichia coli LacZ (beta-galactosidase) as a marker gene. Pregnant ewes and fetuses were killed, and amniotic fluid, placenta, and fetal tissues were collected and assayed for transgene expression 7 to 77 days after intraamniotic injection. In addition, serum was collected and analyzed for evidence of specific immune responses against the producer cells, and amniotic fluid was collected and analyzed for deleterious effects on producer cell viability, vector production, and vector transduction. RESULTS Only 1 of 10 fetuses exposed to the retroviral producer cells demonstrated beta-galactosidase activity that correlated with positive immunohistochemistry for LacZ in lung, trachea, pancreas, and small intestine. However, the presence of the LacZ gene could not be confirmed by polymerase chain reaction. Thus, we could not confirm transduction after any of the injections. The retroviral producer cells survived well in amniotic fluid and continued to produce high levels of retroviral vector after intra-amniotic injection, although amniotic fluid inhibited the transducing activity of the vector in a manner dependent on gestational age. CONCLUSIONS Intra-amniotic retroviral gene transfer with the use of these amphotropic producer cells does not result in reproducible gene transfer in the ovine fetus although amniotic fluid sustains producer cell viability and vector production. Possible reasons for the inefficient transduction are discussed.
Collapse
Affiliation(s)
- Henry L Galan
- Department of Obstetrics and Gynecology, University of Colorado Health Sciences Center, Denver, 80262, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Shen JS, Meng XL, Ohashi T, Eto Y. Adenovirus-mediated prenatal gene transfer to murine central nervous system. Gene Ther 2002; 9:819-23. [PMID: 12040464 DOI: 10.1038/sj.gt.3301700] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2001] [Accepted: 02/18/2002] [Indexed: 11/08/2022]
Abstract
In some lysosomal storage disorders pathological alterations in the central nervous system (CNS) occur as early as the prenatal period and the neuropathology progresses rapidly soon after birth. In these diseases, postnatal therapies alone are often insufficient. Therefore prenatal gene therapy to the CNS may be necessary. In order to investigate the feasibility of gene transfer to the CNS prenatally, we administered recombinant adenovirus carrying LacZ gene to rat embryos from embryonic day 9 to 12 (E9-E12). Results showed that efficient transduction of the reporter gene to the CNS was achieved when adenoviruses were injected at E12. The regions where the reporter gene was transduced mainly localized at the telencephalon and hypophysis of the embryo, and the gene expression persisted at least 1 week after birth. In addition, when adenoviruses were injected at E9, E10 and E11, no transgene expression was detected in the CNS, but was mainly observed in the liver, the heart and the skin, respectively.
Collapse
Affiliation(s)
- J-S Shen
- Department of Gene Therapy, Institute of DNA Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
26
|
Endoh M, Koibuchi N, Sato M, Morishita R, Kanzaki T, Murata Y, Kaneda Y. Fetal gene transfer by intrauterine injection with microbubble-enhanced ultrasound. Mol Ther 2002; 5:501-8. [PMID: 11991740 DOI: 10.1006/mthe.2002.0577] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intrauterine injection of naked DNA expressing luciferase, green fluorescent protein (GFP), or beta-galactosidase (beta-gal) and fluorescein isothiocyanate-labeled oligodeoxynucleotide (FITC-ODN), in combination with microbubble-enhanced ultrasound (US), referred to as the "shotgun method" (SGM), produced high-level protein expression in fetal mice. With the SGM, luciferase expression increased approximately 10(3)-fold in comparison with expression after injection of naked DNA alone. Electron microscopic analysis demonstrated transient formation of pores on the skin surface after intraamniotic (i.a.) injection with the SGM. Widespread expression of GFP and beta-gal and delivery of FITC-ODN were observed in multiple fetal tissues adjacent to the injection points. PCR analysis indicated that germline transfection was only transient following intraperitoneal (i.p) injection, and there was no evidence of transfer of the reporter gene to the offspring. Thus, SGM might provide a useful means to clarify the molecular mechanisms of genetic diseases in utero, as well as a tool to develop gene therapies in utero.
Collapse
Affiliation(s)
- Masayuki Endoh
- Department of Gene Therapy Science, Osaka University Medical School, Yamada-oka 2-2, Suita, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Stammberger U, Uduehi AN, Kubisa B, Roth T, Schmid RA. Non-viral gene delivery to atelectatic and ventilated lungs. Ann Thorac Surg 2002; 73:432-6; discussion 436-7. [PMID: 11845855 DOI: 10.1016/s0003-4975(01)03388-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Three different nonviral vectors and naked DNA were evaluated for in vivo transfer of plasmid DNA to rat lungs through airways in either atelectatic or ventilated lungs. METHODS The F344 rats underwent instillation of 300 microg DNA (pCIluc, luciferase) to the left lung. Naked DNA, linear polyethylenimine, branched polyethylenimine, and lipid GL-67 (in either atelectatic or ventilated lungs) were assessed (n = 5 per group). After 24 hours, left lung PaO2 (mm Hg) and luciferase activity (RLU/mg) were measured. The median (range) was given, and the analysis of variance was applied, followed by the planned comparison on log-transformed data. RESULTS In atelectatic lungs, lipid GL-67 was best (927 [330 to 4112] RLU/mg; p < 0.001 versus other groups of atelectatic lung; p < 0.001 versus all other groups), but highest luciferase activity in all groups was measured in ventilated lungs using linear polyethylenimine (1,240 [922 to 2519] RLU/mg; p < 0.001 versus other groups of ventilated lung; p < 0.001 versus all other groups). In comparison with naked DNA, all nonviral vector systems significantly impaired PaO2 24 hours after airway transfection (p < 0.001; naked DNA versus all other groups). Regardless of transfection technique, PaO2 was worst in lungs transfected by linear polyethylenimine. CONCLUSIONS Highest transfection was achieved with GL-67 in atelectatic lungs and with linear polyethylenimine in ventilated lungs. All gene delivery systems impaired gas exchange of the transduced lung in comparison with naked DNA.
Collapse
Affiliation(s)
- Uz Stammberger
- Division of General Thoracic Surgery, University Hospital, Berne, Switzerland
| | | | | | | | | |
Collapse
|
28
|
Schneider H, Mühle C, Douar AM, Waddington S, Jiang QJ, von der Mark K, Coutelle C, Rascher W. Sustained delivery of therapeutic concentrations of human clotting factor IX--a comparison of adenoviral and AAV vectors administered in utero. J Gene Med 2002; 4:46-53. [PMID: 11828387 DOI: 10.1002/jgm.233] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Prenatal somatic gene therapy has been considered for genetic disorders presenting with morbidity at birth. Haemophilia is associated with an increased risk of catastrophic perinatal bleeding complications such as intracranial haemorrhage, which could be prevented by gene transfer in utero. Prenatal gene therapy may be more promising than postnatal treatment, as the fetus may be more amenable to uptake and integration of therapeutic DNA and the immaturity of its immune system may permit life-long immune tolerance of the transgenic protein, thus avoiding the dominant problem in haemophilia treatment, the formation of inhibitory antibodies. METHODS Adenovirus serotype 5-derived or AAV serotype 2-derived vectors carrying human clotting factor IX (hfIX) cDNA or a reporter gene were administered intramuscularly, intraperitoneally or intravascularly to late-gestation mouse fetuses. Both vector types were evaluated with respect to the kinetics of hfIX delivery to the systemic circulation and possible immune responses against the vector or the transgene product. RESULTS Mice treated in utero by intramuscular injection of an adenoviral vector carrying hfIX cDNA exhibited high-level gene expression at birth and therapeutic--albeit continuously decreasing--plasma concentrations of hfIX over the entire 6 months of the study. Adenoviral vector spread to multiple organs was detected by polymerase chain reaction (PCR). Intramuscular, intraperitoneal or intravascular application of AAV vectors carrying hfIX cDNA led to much lower plasma concentrations of hfIX shortly after birth, which appeared to decline during the first month of life but stabilized in some of the mice at detectable levels. No signs of immune responses were found, either against the different viral vectors or against hfIX. CONCLUSION This study demonstrates for the first time that sustained systemic delivery of a therapeutic protein can be achieved by prenatal gene transfer. It thus shows the feasibility of gene therapy in utero and provides a basis for considering this concept as a preventive therapeutic strategy for haemophilia and perhaps also for other plasma protein deficiencies.
Collapse
Affiliation(s)
- Holm Schneider
- Children's Hospital, University of Erlangen-Nuernberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Boyle MP, Enke RA, Adams RJ, Guggino WB, Zeitlin PL. In utero AAV-mediated gene transfer to rabbit pulmonary epithelium. Mol Ther 2001; 4:115-21. [PMID: 11482982 DOI: 10.1006/mthe.2001.0428] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In utero intra-amniotic administration of adeno-associated virus (AAV) for treatment of cystic fibrosis (CF) has the potential to be an efficient way to target the rapidly dividing undifferentiated cells of the fetal pulmonary epithelium, while simultaneously treating other tissues involved in CF (such as the intestines), but has never before been studied. Intra-amniotic administration of 1x10(12) particles of AAV-luciferase vector to 110 fetal rabbits at 24-25 days gestation resulted in transgene expression in amniotic membranes, trachea, and pulmonary epithelium. The highest level of transgene expression was found in amniotic membranes. Transgene expression peaked in the lungs 10 days after vector delivery, decreased at day 17, and was no longer detectable after 24 days. The number of pulmonary cells transduced was approximately 1 in 500 and immunohistochemical analysis showed expression in varying cell types, including alveolar cells. Transgene expression was not detected in fetal rabbit intestines, skin or liver, nor in maternal ovaries or liver. Intra-amniotic administration of AAV does not result in the tissue inflammation and fetal loss previously documented with in utero adenoviral administration, and results in high levels of transgene expression in amniotic membranes with lower levels in fetal pulmonary epithelium.
Collapse
Affiliation(s)
- M P Boyle
- Department of Medicine, The Johns Hopkins University School Of Medicine, Baltimore, Maryland 21205, USA.
| | | | | | | | | |
Collapse
|
30
|
Tran ND, Porada CD, Almeida-Porada G, Glimp HA, Anderson WF, Zanjani ED. Induction of stable prenatal tolerance to beta-galactosidase by in utero gene transfer into preimmune sheep fetuses. Blood 2001; 97:3417-23. [PMID: 11369632 DOI: 10.1182/blood.v97.11.3417] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The successful transduction of hematopoietic stem cells and long-term (28 months) transgene expression within the hematopoietic system following the direct injection of high-titer retroviral vectors into preimmune fetal sheep was previously demonstrated. The present studies extended these analyses for 40 months postinjection and evaluated whether the longevity of transgene expression in this model system was the result of induction of prenatal tolerance to the transgene product. The intraperitoneal injection of retroviral vectors into preimmune sheep fetuses transduces thymic epithelial cells thought to present antigen and thus define self during immune system development. To directly demonstrate induction of tolerance, postnatal sheep were boosted with purified beta-galactosidase and showed that the peripheral blood lymphocytes from in utero-transduced sheep exhibited significantly lower stimulation indices to transduced autologous cells than did control animals and that the in utero-transduced sheep had a reduced ability to mount an antibody response to the vector-encoded beta-galactosidase protein compared with control sheep. Collectively, our results provide evidence that the direct injection of retroviral vectors into preimmune sheep fetuses induces cellular and humoral tolerance to the vector/transgene products and provide an explanation for the duration and stability of transgene expression seen in this model. These results also suggest that even relatively low levels of gene transfer in utero may render the recipient tolerant to the exogenous gene and thus potentially permit the successful postnatal treatment of the recipient. (Blood. 2001;97:3417-3423)
Collapse
Affiliation(s)
- N D Tran
- Department of Veterans Affairs Medical Center, 1000 Locust St., Reno, NV 89502-2597, USA
| | | | | | | | | | | |
Collapse
|
31
|
Ye X, Mitchell M, Newman K, Batshaw ML. Prospects for prenatal gene therapy in disorders causing mental retardation. MENTAL RETARDATION AND DEVELOPMENTAL DISABILITIES RESEARCH REVIEWS 2001; 7:65-72. [PMID: 11241884 DOI: 10.1002/1098-2779(200102)7:1<65::aid-mrdd1009>3.0.co;2-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Advances in understanding the genetics and pathogenesis of disease and in prenatal diagnosis have lead to an exploration of ways to intervene earlier and earlier in the disease process. The possibility of prenatal gene therapy for severe genetic and developmental disorders has sparked new research and debate as to its feasibility, reliability, and ethics as a therapeutic option. Recent animal studies have demonstrated the feasibility of introducing a vector into the developing fetus. The optimal timing and best mode of delivery, however, have yet to be defined. Whether or not this research should be pursued also has been the subject of recent bioethical debates. There is additional concern with the possibility of in utero gene transfer inducing mutagenesis and subsequent tumor formation. This review will provide a summary of the current state of knowledge in the field of prenatal gene therapy and possible directions for the future research.
Collapse
Affiliation(s)
- X Ye
- George Washington University School of Medicine, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | | | | | | |
Collapse
|
32
|
Coutelle C, Themis M, Schneider H, Kiserud T, Cook T, Douar AM, Hanson M, Pavirani A, Rodeck C. Fetal somatic gene therapy--a preventive approach to the treatment of genetic disease: the case for. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2001:99-114. [PMID: 11105256 DOI: 10.1007/978-3-662-04469-8_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- C Coutelle
- Section of Molecular Genetics, Imperial College School of Medicine, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lipshutz GS, Gruber CA, Hardy J, Contag CH, Gaensler KM. In utero delivery of adeno-associated viral vectors: intraperitoneal gene transfer produces long-term expression. Mol Ther 2001; 3:284-92. [PMID: 11273769 DOI: 10.1006/mthe.2001.0267] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recombinant adeno-associated viruses (rAAV) are promising gene transfer vectors that produce long-term expression without toxicity. To investigate future approaches for in utero gene delivery, the efficacy and safety of prenatal administration of rAAV were determined. Using luciferase as a reporter, expression was assessed by whole-body imaging and by analysis of luciferase activity in tissue extracts, at the time of birth and monthly thereafter. Transgene expression was detected in all injected animals. Highest levels of luciferase activity were detected at birth in the peritoneum and liver, while the heart, brain, and lung demonstrated low-level expression. In vivo luciferase imaging revealed persistent peritoneal expression for 18 months after in utero injection and provided a sensitive whole-body assay, useful in identifying tissues for subsequent analyses. There was no detectable hepatocellular injury. Antibodies that reacted with either luciferase or rAAV were not found. AAV sequences were not detected in germ-line tissues of injected animals or in tissues of their progeny. In utero AAV-mediated gene transfer in this animal model demonstrates that novel therapeutic vectors and strategies can be rapidly tested in vivo and that rAAV may be developed to ameliorate genetic diseases with perinatal morbidity and mortality.
Collapse
Affiliation(s)
- G S Lipshutz
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143-0793, USA
| | | | | | | | | |
Collapse
|
34
|
Hegyi L, Hardwick SJ, Siow RC, Skepper JN. Macrophage death and the role of apoptosis in human atherosclerosis. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:27-42. [PMID: 11276357 DOI: 10.1089/152581601750098192] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The arterial disease atherosclerosis is responsible for severe morbidity and is the most common cause of death in the Western population. The complete pathogenesis of the disease is unknown, but multiple risk factors have been identified that correlate with the development of its complications such as heart attack and stroke. Evidence suggests that atherosclerosis is an inflammatory disease and the major cell types involved are smooth muscle cells, macrophages, and T lymphocytes. In this paper, we review the function of macrophages in the context of atherosclerosis and we also discuss the role and significance of macrophage death, including apoptosis. There is much evidence, certainly in vitro, suggesting that low-density lipoprotein becomes atherogenic when it undergoes cell-mediated oxidation within the artery wall. Besides inducing apoptosis in vitro, oxidized low-density lipoprotein may also cause extensive DNA damage in intimal cells, which might presage apoptosis. We review the results of experimental and clinical studies, which may indicate how the complications of atherosclerosis could be prevented by using different therapeutical strategies including bone marrow transplantation and gene therapy.
Collapse
Affiliation(s)
- L Hegyi
- Division of Cardiovascular Medicine, Department of Medicine, ACCI, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK.
| | | | | | | |
Collapse
|
35
|
Mitchell M, Jerebtsova M, Batshaw ML, Newman K, Ye X. Long-term gene transfer to mouse fetuses with recombinant adenovirus and adeno-associated virus (AAV) vectors. Gene Ther 2000; 7:1986-92. [PMID: 11175309 DOI: 10.1038/sj.gt.3301332] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We have developed a micro-injection technique to deliver recombinant adenovirus and AAV to mouse fetuses at day 15 after conception. Several routes of delivery, including injections to the amniotic fluid, the front limb, the placenta, the liver, and the retro-orbital venus plexus, were tested using an E1-deleted recombinant adenovirus (Ad.CBlacZ) or a recombinant adeno-associated virus (AAV.CMVlacZ) carrying a beta-galactosidase (lacZ) gene. Injection of Ad.CBlacZ into the amniotic cavity led to transgene expression in the skin and in the digestive tract of the fetuses. Injection of Ad.CBlacZ in the front limb resulted in LacZ expression in all major muscle groups around the injection site and at low levels in the liver. The other three routes of delivery, ie intra-placental, intra-hepatic and retro-orbital injections of Ad.CBlacZ, all led to lacZ expression predominantly in the liver. Further studies revealed a maximal tolerant dose (defined as the highest viral dose with < or =20% mortality in the injected fetuses) of 1 x 10(9) particles per fetus for intra- hepatic injections, 3 x 10(9) particles per fetus for intra-placental injection, 1 x 1010 particles per fetus for retro-orbital and intra-amniotic injections, and 2 x 10(10) particle per fetus for intra-muscular injection. The adenovirus-mediated lacZ expression in liver and muscle persisted for at least 6 weeks. Intra-muscular injection of AAV.CMVlacZ also resulted in lacZ expression in the muscle up to 3 months after birth with no indication of cellular immune response at the injection site. Taken together, our results demonstrated that prolonged transgene expression can be achieved by in utero gene transfer using either adenoviral or AAV vectors. The distribution of virus-mediated gene transfer appeared to determined mostly by the route of viral administration.
Collapse
Affiliation(s)
- M Mitchell
- Centers for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center, Washington DC, USA
| | | | | | | | | |
Collapse
|
36
|
Man Y, Hart VJ, Ring CJ, Sanjar S, West MR. Loss of epithelial integrity resulting from E-cadherin dysfunction predisposes airway epithelial cells to adenoviral infection. Am J Respir Cell Mol Biol 2000; 23:610-7. [PMID: 11062139 DOI: 10.1165/ajrcmb.23.5.4046] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Epithelial intercellular adhesion is fundamental to the formation of the airway epithelial protective barrier. In this respect, cadherins are important because these adhesion molecules regulate formation and maintenance of epithelial intercellular junctions. To study the importance of airway epithelial integrity in determining susceptibility to virus infection, we used a replication-incompetent adenovirus, RAd35, and an E-cadherin specific function-blocking antibody, SHE78-7, to disrupt intercellular contacts in human bronchial epithelial cell line 16HBE14o- and primary bronchial epithelial cells. After exposure of 16HBE14o- cell cultures to SHE78-7, disruption of the transepithelial permeability barrier was indicated by a loss of transepithelial electrical resistance and an associated increase of mannitol, inulin, and dextran paracellular flux. Subsequent exposure of SHE78-7-treated cell cultures to RAd35 showed a remarkable increase in adenoviral infection as assessed by beta-galactosidase reporter gene expression. In cultures exposed to SHE78-7, disruption of E-cadherin function resulted in infection equivalent to that in control cultures using 16-fold lower viral titers. These studies show that manipulation of E-cadherin function provides a specific means of altering epithelial integrity that in turn determines resistance of airway epithelia to adenoviral infection.
Collapse
Affiliation(s)
- Y Man
- Cell Biology Department and Clinical Virology Department, Medicines Research Centre, GlaxoWellcome, Stevenage, United Kingdom.
| | | | | | | | | |
Collapse
|
37
|
Lipshutz GS, Flebbe-Rehwaldt L, Gaensler KM. Reexpression following readministration of an adenoviral vector in adult mice after initial in utero adenoviral administration. Mol Ther 2000; 2:374-80. [PMID: 11020353 DOI: 10.1006/mthe.2000.0136] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenovirus-mediated gene delivery is limited by the induction of immune responses that produce toxicity and prevent reexpression. To determine whether adenoviral delivery in the preimmune fetus would produce tolerance, we assessed luciferase (luc) expression following sequential pre- and postnatal adenoviral-mediated gene delivery. Day 15 fetuses were injected intrahepatically with 1 x 10(7) pfu of an adenoviral-luc vector (Ad-luc). Following in utero injection, hepatic luc expression persisted 1 month postnatally. No humoral response to adenovirus or luc was detected. Adult mice, previously injected in utero, were reinjected intravenously with 5 x 10(8) pfu of Ad-luc at 3 months of age and again at 6 months with either 5 x 10(8) pfu of Ad-luc or cationic liposome-DNA complexes (CLDC). Following the first postnatal injection, animals injected in utero had levels of luc comparable to those of age-matched naive controls. However, both control and experimental animals subsequently developed antibodies to adenovirus and luc. No further expression was achieved with a second postnatal injection of Ad-luc or with delivery of CLDC-luc. These studies demonstrate that the delivery of adenoviral vectors in utero at E15 does not elicit an immune response. However, delivery of recombinant adenovirus postnatally results in brisk and limiting immune responses regardless of the in utero exposure.
Collapse
Affiliation(s)
- G S Lipshutz
- Department of Surgery and, University of California San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
38
|
Abstract
The potential of gene therapy for treatment of lung disease remains unrealised. Early model systems often resulted in promising efficiency of gene transfer, only to prove irreproducible in the clinic. While problems such as induction of host immune responses and duration of expression also need to be addressed, it is now widely believed that alternative, relevant models which more accurately reflect gene transfer efficiencies in human lungs are urgently required. We report here on a human lung slice culture system to assess gene transfer to adult lung epithelium. A lacZ-expressing adenovirus (AdCA35lacZ) was used as a reporter vector. A solution of AdCA35lacZ was instilled via bronchioles into resected lung tissue, a route analogous to clinical administration. Following a 1 h incubation, the tissue was inflated with a 0.4% agarose solution, instilled via the same bronchioles. Once solidified, 500 microm slices of the tissue were prepared and cultured for 4 days. beta-Galactosidase staining revealed lacZ transgene expression in bronchiolar and alveolar cells of the lung slices throughout the 4 days in culture. This system, which can also be used to study other viral and liposome vectors, could prove to be a useful alternative model for assessing gene delivery to adult human lung epithelium.
Collapse
Affiliation(s)
- S McBride
- CRC Laboratories, Pathology Department, University of Edinburgh Medical School, Teviot Place, Edinburgh EH8 9AG, Scotland, UK
| | | | | |
Collapse
|
39
|
Senoo M, Matsubara Y, Fujii K, Nagasaki Y, Hiratsuka M, Kure S, Uehara S, Okamura K, Yajima A, Narisawa K. Adenovirus-mediated in utero gene transfer in mice and guinea pigs: tissue distribution of recombinant adenovirus determined by quantitative TaqMan-polymerase chain reaction assay. Mol Genet Metab 2000; 69:269-76. [PMID: 10870844 DOI: 10.1006/mgme.2000.2984] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal somatic cell gene therapy could become an attractive solution for some congenital genetic diseases or the disorders which manifest themselves during the fetal period. We performed adenovirus-mediated gene transfer to mice and guinea pig fetuses in utero and evaluated the efficiency of gene transfer by histochemical analysis and a quantitative TaqMan-polymerase chain reaction (TaqMan-PCR) assay. We first injected a replication-deficient recombinant adenovirus containing the Escherichia coli LacZ gene driven by a CAG promoter (AxCALacZ) into pregnant mice through the amniotic space, placenta, or intraperitoneal space of the fetus. Histochemical analysis showed limited transgene expression in fetal tissues. We then administered AxCALacZ to guinea pig fetuses in the late stage of pregnancy through the umbilical vein. The highest beta-galactosidase expression was observed in liver followed by moderate expression in heart, spleen, and adrenal gland. The transgene expression was also present in kidney, intestine, and placenta to a lesser degree. No positively stained cells were observed in lung, muscle, or pancreas except in the vascular endothelium of these organs. Quantitative measurement of recombinant adenoviral DNA by the TaqMan-PCR assay showed that the vast majority of the injected viruses was present in liver. The current study indicated that adenovirus-mediated gene transfer into guinea pig fetus through the umbilical vein is feasible and results in efficient transgene expression in fetal tissues. The experimental procedures using pregnant guinea pigs might serve as a good experimental model for in utero gene transfer. Since our TaqMan-PCR assay detects the LacZ gene, one of the most widely used reporter genes, it may be generally applicable to adenovirus quantification in various gene transfer experiments.
Collapse
Affiliation(s)
- M Senoo
- Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ye X, Jerebtsova M, Ray PE. Liver bypass significantly increases the transduction efficiency of recombinant adenoviral vectors in the lung, intestine, and kidney. Hum Gene Ther 2000; 11:621-7. [PMID: 10724040 DOI: 10.1089/10430340050015806] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant adenoviruses have great potential as gene delivery systems because of their ability to infect a wide range of target cells. However, systemic delivery of viral vectors to tissues other than liver and spleen has been inefficient because of the rapid clearance of the circulating virus by the liver. In the present study we tested the hypothesis that a systemic administration of E1-deleted recombinant adenovirus vectors that bypasses the hepatic circulation will lead to enhanced expression of these vectors in extrahepatic tissues. The portal vein and hepatic artery in B6/129 F1 mice were clamped and an E1-deleted recombinant adenovirus carrying the beta-galactosidase gene (Ad.CBlacZ) was then administered through the retroorbital venous plexus. The clamp was released 30 min after viral injection with no major chronic ischemic consequences noted. High levels of LacZ expression were detected predominantly in the vessels and capillaries of the lung, intestinal wall, and renal glomeruli 7 days after viral infusion. The transgene expression persisted for at least 21 days. Intense LacZ staining was also observed in the liver, suggesting that liver infection occurred after the portal clamp was released. A retroorbital infusion of anti-adenovirus neutralizing antibodies 5 min before the release of the portal clamp significantly reduced postclamp viral infection to the liver, while LacZ expression in lung and intestine persisted after the antibody treatment. Taken together, these results suggest that liver bypass can significantly improve the transduction efficiency in the other target organs. This method could be used to develop animal models of human diseases that predominantly affect the vessels of the lung, intestine, and kidney.
Collapse
Affiliation(s)
- X Ye
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.
| | | | | |
Collapse
|
41
|
Tran ND, Porada CD, Zhao Y, Almeida-Porada G, Anderson WF, Zanjani ED. In utero transfer and expression of exogenous genes in sheep. Exp Hematol 2000; 28:17-30. [PMID: 10658673 DOI: 10.1016/s0301-472x(99)00133-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE We have previously reported that directly injecting low-titer retroviral vector supernatant into pre-immune sheep fetuses resulted in the transfer and long-term expression of the bacterial NeoR gene within the hematopoietic system of these animals for over 5 years. In the present studies, we investigated whether using a higher titer vector would enable more efficient transduction and expression of the transgenes within the hematopoetic cells in sheep injected in utero. MATERIALS AND METHODS Sixteen pre-immune sheep fetuses were injected intraperitoneally with the G1nBgSvNa8.1 helper-free retroviral vector supernatant encoding the bacterial NeoR and LacZ genes (titer: 1x10(7) cfu/mL). RESULTS Over the 2-year time course of these studies, the presence and expression of the NeoR and LacZ genes were demonstrated in 12 of the 14 animals evaluated by several immunological and biochemical methods. Seven of the 12 sheep examined by flow cytometric analysis contained > or =6% transduced peripheral blood lymphocytes. Vector distribution was widespread without any detectable pathology. Importantly, PCR analyses and breeding experiments demonstrated that the germ line was not altered. CONCLUSIONS These studies confirmed that direct injection of an engineered retrovirus is a feasible means of safely delivering foreign genes into a developing fetus and thus achieving long-term expression of the transgenes within the recipient's hematopoietic cells. Furthermore, expression of the NeoR gene from these studies was higher than that reported in our previous study in which a lower titer vector was used.
Collapse
Affiliation(s)
- N D Tran
- Department of Veterans Affairs Medical Center, University of Nevada, Reno 89520, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Advances in prenatal diagnosis and gene transfer technology have allowed consideration of prenatal gene therapy. A compelling argument can be made for this strategy in treating genetic diseases that are fatal in the prenatal or perinatal period. In other diseases, the fetal environment may offer unique biological advantages that favor a prenatal gene therapy strategy over treatment after birth. Although issues of safety and efficacy must be resolved before clinical application, the development of fetal gene therapy may become a new molecular therapeutic arm in the field of prenatal intervention.
Collapse
Affiliation(s)
- E Y Yang
- Department of Surgery, the Center for Fetal Diagnosis and Treatment, Children's Hospital of Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
43
|
Gaensler KM, Tu G, Bruch S, Liggitt D, Lipshutz GS, Metkus A, Harrison M, Heath TD, Debs RJ. Fetal gene transfer by transuterine injection of cationic liposome-DNA complexes. Nat Biotechnol 1999; 17:1188-92. [PMID: 10585716 DOI: 10.1038/70729] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In utero injection of cationic liposome-DNA complexes (CLDCs) containing chloramphenicol acetyltransferase, beta-galactosidase (beta-gal), or human granulocyte colony-stimulating factor (hG-CSF) expression plasmids produced high-level gene expression in fetal rats. Tissues adjacent to the injection site exhibited the highest levels of gene expression. Chloramphenicol acetyltransferase expression persisted for at least 14 days and was reexpressed following postnatal reinjection of CLDCs. Intraperitoneal administration of the hG-CSF gene produced high serum hG-CSF levels. X-gal staining demonstrated widespread beta-gal expression in multiple fetal tissues and cell types. No toxic or inflammatory responses were observed, nor was there evidence of fetal-maternal or maternal-fetal gene transfer, suggesting that CLDCs may provide a useful alternative to viral vectors for in utero gene transfer.
Collapse
Affiliation(s)
- K M Gaensler
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang G, Slepushkin V, Zabner J, Keshavjee S, Johnston JC, Sauter SL, Jolly DJ, Dubensky TW, Davidson BL, McCray PB. Feline immunodeficiency virus vectors persistently transduce nondividing airway epithelia and correct the cystic fibrosis defect. J Clin Invest 1999; 104:R55-62. [PMID: 10587528 PMCID: PMC483477 DOI: 10.1172/jci8390] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/1999] [Accepted: 10/22/1999] [Indexed: 11/17/2022] Open
Abstract
Several problems limit the application of gene transfer to correct the cystic fibrosis (CF) Cl(-) transport defect in airway epithelia. These include inefficient transduction with vectors applied to the apical surface, a low rate of division by airway epithelial cells, failure of transgene expression to persist, and immune responses to vectors or vector-encoded proteins. To address these issues, we used a feline immunodeficiency virus-based (FIV-based) vector. FIV vector formulated with a calcium chelator transduced fully differentiated, nondividing human airway epithelia when applied to the apical surface. FIV-based vector encoding the cystic fibrosis transmembrane conductance regulator cDNA corrected the Cl(-) transport defect in differentiated CF airway epithelia for the life of the culture (>3 months). When this approach was applied in vivo, FIV vector expressing beta-galactosidase transduced 1-14% of adult rabbit airway epithelia. Transduced cells were present in the conducting airways, bronchioles, and alveoli. Importantly, gene expression persisted, and cells with progenitor capacity were targeted. FIV-based lentiviral vectors may be useful for the treatment of genetic lung diseases such as CF. This article may have been published online in advance of the print edition.
Collapse
Affiliation(s)
- G Wang
- Program in Gene Therapy, Department of Pediatrics, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang G, Slepushkin VA, Bodner M, Zabner J, van Es HHG, Thomas P, Jolly DJ, Davidson BL, McCray PB. Keratinocyte growth factor induced epithelial proliferation facilitates retroviral–mediated gene transfer to distal lung epithelia in vivo. J Gene Med 1999. [DOI: 10.1002/(sici)1521-2254(199901/02)1:1<22::aid-jgm1>3.0.co;2-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
46
|
Lipshutz GS, Sarkar R, Flebbe-Rehwaldt L, Kazazian H, Gaensler KM. Short-term correction of factor VIII deficiency in a murine model of hemophilia A after delivery of adenovirus murine factor VIII in utero. Proc Natl Acad Sci U S A 1999; 96:13324-9. [PMID: 10557319 PMCID: PMC23946 DOI: 10.1073/pnas.96.23.13324] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Development of in utero gene transfer approaches may provide therapies for genetic disorders with perinatal morbidity. In hemophilia A, prenatal and postnatal bleeding may be catastrophic, and modest increments in factor VIII (FVIII) activity are therapeutic. We performed transuterine i.p. gene transfer at day 15 of gestation in a murine model of hemophilia A. Normal, carrier (X(H)X), and FVIII-deficient (X(H)Y and X(H)X(H)) fetuses injected with adenoviral vectors carrying luciferase or beta-galactosidase reporter genes showed high-level gene expression with 91% fetal survival. The live-born rates of normal and FVIII-deficient animals injected in utero with adenovirus murine FVIII (3.3 x 10(5) plaque-forming units) was 87%. FVIII activity in plasma was 50.7 +/- 10.5% of normal levels at day 2 of life, 7.2 +/- 2.2% by day 15 of life, and no longer detectable at day 21 of life in hemophilic animals. Injection of higher doses of murine FVIII adenovirus at embryonic day 15 produced supranormal levels of FVIII activity in the neonatal period. PCR analysis identified viral genomes primarily in the liver, intestine, and spleen, although adenoviral DNA was detected in distal tissues when higher doses of adenovirus were administered. These studies show that transuterine i.p. injection of adenoviral vectors produces therapeutic levels of circulating FVIII throughout the neonatal period. The future development of efficient and persisting vectors that produce long-term gene expression may allow for in utero correction of genetic diseases originating in the fetal liver, hematopoietic stem cells, as well as other tissues.
Collapse
Affiliation(s)
- G S Lipshutz
- Department of Surgery, Third and Parnassus Avenues, University of California, San Francisco, CA 94143-0793, USA
| | | | | | | | | |
Collapse
|
47
|
Baumgartner TL, Baumgartner BJ, Hudon L, Moise KJ. Ultrasonographically guided direct gene transfer in utero: successful induction of beta-galactosidase in a rabbit model. Am J Obstet Gynecol 1999; 181:848-52. [PMID: 10521740 DOI: 10.1016/s0002-9378(99)70312-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We sought to determine whether the transfer of enzyme-encoding genes in utero can be detected after birth. STUDY DESIGN An adenoviral vector carrying the gene for beta-galactosidase was injected under ultrasonographic guidance into the livers of 4 rabbit fetuses per litter (3 litters total) at 27 days' gestation. On delivery of the pups 2 to 3 days later, the livers were analyzed for beta-galactosidase activity by using 5-bromo-4-chloro-3-indolyl-beta-D -galactopyranoside (X-gal) staining. Polymerase chain reaction was also performed on liver extracts as an additional independent measure of successful vector delivery. RESULTS Successful targeting of the livers of fetal rabbits was demonstrated by beta-galactosidase activity in the nuclei of liver serosal cells, parenchymal hepatocytes, or columnar cells of the gallbladder in 7 (58%) of 12 injected pups and by polymerase chain reaction in liver extracts from 10 (83%) of 12 injected pups. CONCLUSIONS These results suggest that vectors that carry genes for specific enzymes can be delivered to fetal organs in utero and that expression of the enzyme can be detected after delivery.
Collapse
Affiliation(s)
- T L Baumgartner
- Department of Obstetrics, University of Texas Health Science Center at Houston, Division of Neurosurgery, Houston, Texas, USA
| | | | | | | |
Collapse
|
48
|
Türkay A, Saunders T, Kurachi K. Intrauterine gene transfer: gestational stage-specific gene delivery in mice. Gene Ther 1999; 6:1685-94. [PMID: 10516717 DOI: 10.1038/sj.gt.3301007] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intrauterine gene transfer in mice by intraplacental microinjection of recombinant adenoviral or retroviral vectors carrying beta-galactosidase (beta-gal) reporter gene was analyzed in relation to gestational stage, viral titer and promoters. After injections of viral vectors on days 9.5, 11.5 or 14.5 post coitum (p.c.), embryos, fetuses and adult animals were analyzed for beta-gal expression on days 13.5 p.c., 18.5 p.c. and at 2 months of age, respectively. Injection of adenoviral vectors on day 9.5 or day 11.5 p.c. resulted in high beta-gal expression in the heart or liver, respectively. Injection on either day also gave expression in other tissues including vasculature and hindbrain. This temporal pattern of adenoviral transduction correlated with the expression level of integrin beta3 subunit, which is known to be a component involved in adenoviral transduction. Adenovirus-mediated intrauterine gene transfer resulted in persistent beta-gal expression in multiple cell foci in the liver and hearts of 2-month-old adult animals treated in utero, indicating stable integration of the transgene into the host cell genome at a low frequency. Although at low efficiency, injection of retroviral vector on day 9.5 and 11.5 p.c. resulted in beta-gal expression in embryonic liver, while day 9.5 p.c. injection resulted in persistent beta-gal expression in 2-month-old adult heart. This is the first study to show gestational stage-specific gene transfer via intraplacental microinjection of adenoviral vectors.
Collapse
Affiliation(s)
- A Türkay
- Department of Human Genetics, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
49
|
Allen CM, Sharman WM, Madeleine CL, Weber JM, Langlois R, Ouellet R, Lier JE. Photodynamic Therapy: Tumor Targeting with Adenoviral Proteins. Photochem Photobiol 1999. [DOI: 10.1111/j.1751-1097.1999.tb08246.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
50
|
|