当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第3期 > 正文
编号:11342428
Transfer and Stable Transgene Expression of a Mammalian Artificial Chromosome into Bone Marrow-Derived Human Mesenchymal Stem Cells
http://www.100md.com 《干细胞学杂志》
     Chromos Molecular Systems Inc., Burnaby, British Columbia, Canada

    Key Words. Mammalian artificial chromosomes ? ACEs ? Gene therapy ? Cell therapy ? Mesenchymal stem cells

    Sandra Vanderbyl, MSc, Chromos Molecular Systems Inc., 8081 Lougheed Highway, Burnaby, British Columbia, Canada V5A 1W9. Telephone: 604-415-7100; Fax: 604-415-7151; e-mail: svanderbyl@chromos.com

    ABSTRACT

    Recent events in gene therapy applications have driven the development of vectors that maintain stable expression of therapeutic genes without the risk of cell transformation or the stimulation of the host immune system . However despite advances, current eukaryotic viral vector systems remain problematic. For example, integration into the host chromosome by retroviral-based vectors may lead to variegated gene expression, insertional mutagenesis, and oncogenesis . Similar concerns have also been raised regarding the integration of adeno-associated virus vectors and their association with chromosomal deletions and other rearrangements that are frequently located on human chromosome 19 . Furthermore, it has been demonstrated that retroviruses preferentially integrate near transcription start regions thus raising the likelihood of insertional mutagenesis with retrovirus-based vectors . Transient expression of therapeutic products is also a concern. Most notably, using nonintegrating vector systems such as adenoviral-based vectors may elicit a host immunological response to the vector, including death .

    Artificial chromosomes (ACEs) are being investigated as an alternative to current eukaryotic viral vector systems. Hadlaczky and colleagues developed a unique methodology to construct mammalian ACEs through the induction of large scale amplifications of "satellite" DNA sequences in pericentromeric heterochromatin . De novo centromeres and dicentric chromosomes were formed upon the integration of exogenous DNA sequences into the specific regions of acrocentric chromosomes—those containing both pericentromeric heterochromatin and the tandemly repeated ribosomal genes (rDNA). Ensuing breakage during mitosis generated new chromosomes ranging in size from 10 to 360 megabases . Considering that these ACEs, termed satellite DNA-based artificial chromosomes (SATACs), can be manipulated or engineered to contain a variety of exogenous sequences, they present a unique opportunity for a variety of applications, including gene therapy . SATACs have recently been engineered to contain multiple recombination acceptor sites in order to facilitate the insertion of desired transgenes onto the ACEs. These engineered SATACs are commonly referred to as Platform ACEs or ACEs.

    ACEs are safe, stable systems, and many numerous reviews have addressed their utility. In addition to ease of loading genes onto Platform ACEs, they can be isolated and stably transferred. This further expands their utility. As part of the overall ACE System, multiple transgenes (cDNAs or larger genomic encoding regions) can be efficiently targeted onto the Platform ACE through site-specific DNA recombination . Platform ACEs and transgene-loaded ACEs can be isolated by flow cytometry to high purities (>95%) and yields (>1,000,000 ACEs/hr) and can be transferred in vitro into a variety of mammalian cell lines and primary cells by cationic lipids and dendrimers . ACEs can also be used to generate transgenic mice through microinjection of purified ACEs into fertilized oocytes . Stability is exemplified by the fact the resulting transgenic mice were shown to stably maintain the ACE through four generations of the germline .

    Human mesenchymal stem cells (hMSCs) have emerged as a promising strategy for ex vivo gene therapy . Improvements in isolation, in vitro cultivation of hMSCs , and in vivo studies on differentiation into functional cells of multiple tissue types make them attractive delivery vehicles for autologous cell-mediated gene therapy. Characteristics that make them appealing cell hosts include ease of purification from clinical bone marrow (BM) aspirates (frequency of 1/105) and the ability to culture and expand hMSCs in vitro without apparent loss of differentiation potential into multiple cell types . Additionally MSCs have been shown to confer immune privilege by not presenting alloantigens and by suppressing T-cell lymphocyte proliferation . These features may enable a cellular therapy scenario whereby hMSCs can be derived from a "universal" donor (irrespective of their major histocompatability complex haplotype), expanded, and prepared as an "off-the-shelf" reagent.

    hMSCs are being evaluated as cellular delivery vehicles for therapeutic genes for a variety of clinical indications. This list includes the replacement of genes to correct acquired or inherited disorders of bone, muscle, or cartilage by transfer of genes encoding bone morphogenic proteins -2, -4, -9 , the transfer of genes encoding the blood coagulation factors VIII and IX for the treatment of hemophilia , human growth hormone , insulin-like growth factor 1 , and interleukin-3 . This list also includes: erythropoietin , -l-iduronidase for treatment of mucopolysaccharidosis type I , pro2 collagen (I) for treatment of osteogenesis imperfecta , sox9 to enhance chondrogenesis, and interferon to treat tumors . In several of these studies retroviral-based vectors were utilized, thereby increasing the risk of inducing insertional mutagenesis. The risk of insertional mutagenesis can be reduced prior to patient treatment by physical screening of selected recombinants for vector integration into genetically safe regions; however, this approach is problematic when transducing committed stem cells (SCs) with limited cell doublings. Furthermore, the integrated vectors often result in pronounced transgene silencing leading to short-term therapeutic gain. Theoretically, the ACE System would overcome these issues as ACEs are nonintegrating and can be engineered to optimize long-term transgene expression.

    Cell therapy regimes are restricted by the ability to target host cells, the expression and stability of the inserted constructs, the delivery efficiency of vectors, and safety. When considering the application of ACE-based cell therapies in the clinic, a limitation that requires further investigation is the delivery efficiency of protein-DNA complexes. When compared to viral vectors, which can reach transduction efficiencies of 100%, the transfer efficiency of the ACEs is at least an order of magnitude lower . Therefore, strategies that facilitate the enrichment of cells harboring a transferred ACE must be simultaneously developed. To this end, the use of human cell surface or drug selectable marker genes is being investigated as a means to enrich for the ACE-containing cells within a population.

    In addition to the enrichment and ex vivo delivery strategies for ACE-containing cells, we are currently initiating animal studies to investigate the potential for application of ACE-based cell therapies in disease models along with the relative safety and tolerability of these modified cells. With respect to the latter, current data suggest that ACEs are mitotically and meiotically stable, nontumorigenic, and nonmutagenic, as transgenic mice containing an ACE were phenotypically normal, as were their offspring through four generations . Although these observations will need to be substantiated in the context of cellular therapies, our current plans include these and additional cellular assays to determine cell fate, stability of the ACE, duration of transgene expression, and immune response. Current animal studies focus on the utility of ACE-modified MSCs in a small animal model of disease that, if successful, will represent the first demonstration of a therapeutic benefit using a chromosome-based ex vivo cell therapy.

    In this work we evaluated the ability of BM-derived hMSCs to be genetically modified with ACEs expressing the red fluorescent protein (RFP). An intact ACE chromosome was introduced into the hMSCs, and reporter gene expression was detected in the transfected hMSCs. hMSCs containing the ACEs maintained their multipotential capacity as evidenced by their ability to be induced to differentiate along osteogenic and adipogenic lineages. Furthermore, RFP expression was detected in the differentiated cells suggesting that the ACE chromosomal environment is well suited for transgene expression in differentiating SC populations. To our knowledge this is the first report of the stable expression of a transgene introduced into human adult SCs by mammalian ACE. This finding demonstrates the potential utility of the ACE technology for human adult SC ex vivo gene therapy and provides a novel approach for a broad range of tissue engineering applications.

    MATERIALS AND METHODS

    ACE-Transgene Expression in hMSCs

    As a first step pursuant to the use of ACEs in a gene therapy context, we tested the ability to introduce flow-sorted and concentrated RFP-ACEs into a purified hMSC population. The RFP-ACEs were transfected into limited passaged hMSCs using commercially available transfection reagents. After transfer of the RFP-ACEs, the cells were passaged in 10-cm dishes, and the level of transgene expression (RFP fluorescence) was monitored by flow cytometry. From 4 to 5 days post-transfection, RFP expression was detected in 11 ± 4% (n = 3; scoring a minimum of 10,000 cells) of the total hMSC population. Intact RFP-ACEs, as determined by FISH analysis, were detected in 5 ± 3% (n = 2; scoring a minimum of 50 metaphase chromosome spreads) of total hMSC population. A representative example of FISH analysis of the RFP-ACEs-modified hMSCs can be seen in Figure 1A. Since the frequency of intact delivered RFP-ACEs/transfected hMSC, as determined by FISH analyses, was not statistically different from the observed frequency of RFP-expressing hMSCs in the total cell population that was transfected (p-value 0.05), this suggested that in general the transfection of the RFP-ACEs into the hMSCs resulted in the delivery of an intact chromosome.

    Figure 1. FISH analyses. A) Untransfected control hMSCs (i) and ACE-modified hMSCs (ii) were incubated with colchicine, fixed to microscope slides, and hybridized to digoxigenin-labeled mouse major satellite DNA (red signal). Note that the ACE is intact and is maintained as an autonomous chromosome. B) ACE-modified hMSCs were induced to differentiate into either osteocytes (i) or adipocytes (ii) analysis done with interphase cells. Cells were fixed and hybridized to digoxigenin-labeled mouse major satellite DNA (red signal).

    ACE-Transgene Expression During hMSC Differentiation

    We evaluated whether the hMSCs carrying the RFP-ACEs still maintained their potential to differentiate into unique cell lineages. To do so, we elected to monitor reporter gene expression in the hMSC population as the cells differentiated along adipogenic and osteogenic lineages. These two pathways were chosen because the differentiated cells were visually distinctive, based on microscopic examination, and simple biochemical identification assays were available—staining lipids with Oil Red "O" or measuring insoluble calcium deposition to identify adipocytes or osteocytes, respectively. Initially differentiation was monitored in heterogeneous cell populations prior to enrichment of RFP-ACEs containing hMSCs (population with 11% RFP-expressing hMSCs). In this case, the RFP-ACE-transfected hMSCs appear to differentiate at a rate comparable to the untransfected controls. Greater than 95% of the cells had differentiated 2–3 weeks post-induction into either adipogenic or osteogenic lineages (visual inspection, data not shown). Furthermore, during differentiation RFP expression was maintained (visual inspection). FISH analyses were performed on terminally differentiated cultures 2–3 weeks post-induction in order to evaluate the integrity of the transferred RFP-ACEs during differentiation (Fig. 1B). The frequencies of RFP-ACEs were scored as 25% ± 7% (n = 2) in adipocytes and 6% ± 4% (n = 2) in osteocytes. These preliminary experiments indicate that the hMSCs stably maintained RFP-ACEs and concomitant RFP expression after osteogenic and adipogenic differentiation (Fig. 2A, 2B).

    Figure 2. A) RFP-expressing adipocytes at 2 weeks post induction in the adipocyte differentiation pathway. Panel (i): Phase microscopic view from a representative field. Panel (ii): RFP-expressing cells from the identical microscopic field as depicted in panel (i). B) RFP-expressing osteocytes at 2 weeks post induction in the osteocyte differentiation pathway. Panel (i): Phase microscopic view from a representative field. Panel (ii): RFP-expressing cells from the identical microscopic field as depicted in panel (i).

    RT-PCR analyses of selected gene markers (PPAR2, LDL, osteopontin) from induced and non-induced cultures were performed to further confirm the differentiated phenotype of the mesenchymal cultures containing the RFP-ACEs. Previous reports have utilized the expression of PPAR2 and LDL as hallmarks of adipogenic differentiation and osteopontin as a biomarker for osteogenic differentiation .

    In order to detect transgene expression and monitor cytogenetic events during differentiation, the RFP-expressing hMSCs were enriched by flow cytometry 3–5 days post-transfection, and plated into three sets of wells. One sample (non-induced control) was maintained under hMSC culturing conditions, whereas parallel cultures were induced to differentiate into osteogenic and adipogenic lineages. Samples of induced cells were harvested at various times during the 3-week induction period. The cultures were analyzed by RT-PCR to measure the induced expression of lineage specific marker genes and by FISH. RT-PCRs for ?-actin transcripts were conducted as measures of RNA integrity for all culturing conditions since ?-actin expression is ubiquitous for both undifferentiated and differentiated hMSCs (top panels Fig. 3, lanes C-F). Only hMSCs induced to the adipogenic lineage expressed LDL or PPAR2 mRNA (Summary RT-PCR results , representative LDL profile Fig. 3 bottom panels I-M). RT-PCR detected expression of the LDL and PPAR2 genes in the genetically modified adipocyte cultures as early as 1 week post induction. Parallel osteogenic-induced cultures were tested for the presence of adipogenic lineage marker genes LDL or PPAR2. For all of the osteogenic-induced cultures, expression of LDL or PPAR2 genes was not detected (data not shown). Similar results showing lineage specificity were found when assessing osteogenic-induced cultures for expression of the osteogenic lineage marker, osteopontin, 13 days post-induction (Table 1). Overall, the RFP-ACE-modified hMSCs continued to express the RFP transgene and to maintain the integrity of the RFP-ACE after in vitro differentiation.

    Figure 3: RT-PCR analyses using the ubiquitous cell lineage marker ?-actin (row B-F) and the specific adipose marker LPL (row I-M) from adipocyte mRNA. A ?-actin PCR product was found in all samples except for the negative PCR control. Top row: 515-bp ?-actin products. A) 100-bp ladder; B) negative control (no RNA); C) transfected hMSC (undifferentiated); D) transfected, 7-days post adipocyte induction; E) transfected, 13-days post adipocyte induction; F) nontransfected, 13-days post adipocyte induction; G) 100-bp ladder. Bottom row: 276-bp LPL products; H) 100-bp ladder; I) negative control (no RNA); J) transfected hMSC (undifferentiated); K) transfected, 7-days post adipocyte induction; L) transfected, 13-days post adipocyte induction; M) nontransfected, 13-days post adipocyte induction; N) 100-bp ladder. For summary of RT-PCR analyses refer to Table 1.

    Table 1. Summary of RT-PCR of hMSCs cells

    DISCUSSION

    Chromos Molecular Systems, Inc. was responsible for all material and financial support for this research. We would like to thank Diane Monteith and Tom Stodola, for purifying ACEs and technical support; Harry Ledebur and Joseph Zendegui for critical discussions; and Shamila Gorjian for manuscript preparation.

    REFERENCES

    Lipps HJ, Jenke AC, Nehlsen K et al. Chromosome-based vectors for gene therapy. Gene 2003;304:23–33.

    Hacein-Bey-Abina S, von Kalle C, Schmidt M et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 2003;348:255–256.

    Kaiser J. Gene therapy. Seeking the cause of the induced leukemias in X-SCID trial. Science 2003;299:495.

    Marshall E. Gene therapy. Second child in French trial is found to have leukemia. Science 2003;299:320.

    Miller DG, Rutledge EA, Russell DW. Chromosomal effects of adeno-associated virus vector integration. Nat Genet 2002;30:147–148.

    Wu X, Li Y, Crise B et al. Transcription start regions in the human genome are favored targets for MLV integration. Science 2003;300:1749–1751.

    Raper SE, Chirmule N, Lee FS et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 2003;80:148–158.

    Hadlaczky G, Praznovszky T, Cserpan I et al. Centromere formation in mouse cells cotransformed with human DNA and a dominant marker gene. Proc Natl Acad Sci USA 1991;88:8106–8110.

    Praznovszky T, Kereso J, Tubak V et al. De novo chromosome formation in rodent cells. Proc Natl Acad Sci USA 1991;88:11042–11046.

    Kereso J, Praznovszky T, Cserpan I et al. De novo chromosome formations by large-scale amplification of the centromeric region of mouse chromosomes. Chromosome Res 1996;4:226–239.

    Hollo G, Kereso J, Praznovszky T et al. Evidence for a megareplicon covering megabases of centromeric chromosome segments. Chromosome Res 1996;4:240–247.

    Csonka E, Cserpan I, Fodor K et al. Novel generation of human satellite DNA-based artificial chromosomes in mammalian cells. J Cell Sci 2000;113:3207–3216.

    Perez C, de Jong G, Drayer J. Satellite DNA-based artificial chromosomes—chromosomal vectors. Trends Biotechnol 2000;18:402–403.

    Hadlaczky G. Satellite DNA-based artificial chromosomes for use in gene therapy. Curr Opin Mol Ther 2001;3:125–132.

    Stewart S, MacDonald N, Perkins E et al. Retrofitting of a satellite repeat DNA-based murine artificial chromosome (ACes) to contain loxP recombination sites. Gene Ther 2002;9:719–723.

    de Jong G, Telenius AH, Telenius H et al. Mammalian artificial chromosome pilot production facility: large-scale isolation of functional satellite DNA-based artificial chromosomes. Cytometry 1999;35:129–133.

    de Jong G, Telenius A, Vanderbyl S et al. Efficient in-vitro transfer of a 60-Mb mammalian artificial chromosome into murine and hamster cells using cationic lipids and dendrimers. Chromosome Res 2001;9:475–485.

    Vanderbyl S, MacDonald N, de Jong G. A flow cytometry technique for measuring chromosome-mediated gene transfer. Cytometry 2001;44:100–105.

    Co DO, Borowski AH, Leung JD et al. Generation of transgenic mice and germline transmission of a mammalian artificial chromosome introduced into embryos by pronuclear microinjection. Chromosome Res 2000;8:183–191.

    Monteith D, Leung J, Borowski A et al. Pronuclear microinjection of purified artificial chromosomes for generation of transgenic mice. In: Sgaramella V, Eridani S, eds. Methods in Molecular Biology: Chapter 12. Natural and Artificial Chromosomes. Totowa, New Jersey: Humana Press, 2003:229–246.

    Turgeman G, Pittman DD, Muller R et al. Engineered human mesenchymal stem cells: a novel platform for skeletal cell mediated gene therapy. J Gene Med 2001;3:240–251.

    Ballas CB, Zielske SP, Gerson SL. Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem 2002;38(suppl):20–28.

    Pittenger M, Vanguri P, Simonetti D et al. Adult mesenchymal stem cells: potential for muscle and tendon regeneration and use in gene therapy. J Musculoskel Neuron Interact 2002;2:309–320.

    Van Damme A, Driessche T, Collen D et al. Bone marrow stromal cells as targets for gene therapy. Curr Gene Ther 2002;2:195–209.

    Caplan AI. Mesenchymal stem cells and gene therapy. Clin Orthop 2000;379(suppl):S67–S70.

    Caplan AI, Bruder SP. Mesenchymal stem cells: building blocks for molecular medicine in the 21st century. Trends Mol Med 2001;7:259–264.

    Haynesworth SE, Goshima J, Goldberg VM et al. Characterization of cells with osteogenic potential from human marrow. Bone 1992;13:81–88.

    Jiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41–49.

    Bartholomew A, Sturgeon C, Siatskas M et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002;30:42–48.

    Krampera M, Glennie S, Dyson J et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood 2003;101:3722–3729.

    Di Nicola M, Carlo-Stella C, Magni M et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002;99:3838–3843.

    Lieberman JR, Le LQ, Wu L et al. Regional gene therapy with a BMP-2-producing murine stromal cell line induces heterotopic and orthotopic bone formation in rodents. J Orthop Res 1998;16:330–339.

    Musgrave DS, Bosch P, Lee JY et al. Ex vivo gene therapy to produce bone using different cell types. Clin Orthop 2000;Sep:290–305.

    Moutsatsos IK, Turgeman G, Zhou S et al. Exogenously regulated stem cell-mediated gene therapy for bone regeneration. Mol Ther 2001;3:449–461.

    Peng H, Chen ST, Wergedal JE et al. Development of an MFG-based retroviral vector system for secretion of high levels of functionally active human BMP4. Mol Ther 2001;4:95–104.

    Olmsted-Davis EA, Gugala Z, Gannon FH et al. Use of a chimeric adenovirus vector enhances BMP2 production and bone formation. Hum Gene Ther 2002;13:1337–1347.

    Dumont RJ, Dayoub H, Li JZ et al. Ex vivo bone morphogenetic protein-9 gene therapy using human mesenchymal stem cells induces spinal fusion in rodents. Neurosurgery 2002;51:1239–1244.

    Gelse K, von der Mark K, Aigner T et al. Articular cartilage repair by gene therapy using growth factor-producing mesenchymal cells. Arthritis Rheum 2003;48:430–441.

    Tsuda H, Wada T, Ito Y et al. Efficient BMP2 gene transfer and bone formation of mesenchymal stem cells by a fiber-mutant adenoviral vector. Mol Ther 2003;7:354–365.

    Tsuchida H, Hashimoto J, Crawford E et al. Engineered allogeneic mesenchymal stem cells repair femoral segmental defect in rats. J Orthop Res 2003;21:44–53.

    Cherington V, Chiang GG, McGrath CA et al. Retroviral vector-modified bone marrow stromal cells secrete biologically active factor IX in vitro and transiently deliver therapeutic levels of human factor IX to the plasma of dogs after reinfusion. Hum Gene Ther 1998;9:1397–1407.

    Chuah MK, Brems H, Vanslembrouck V et al. Bone marrow stromal cells as targets for gene therapy of hemophilia A. Hum Gene Ther 1998;9:353–365.

    Chuah MK, Van Damme A, Zwinnen H et al. Long term persistence of human bone marrow stromal cells transduced with factor VIII-retroviral vectors and transient production of therapeutic levels of human factor VIII in nonmyeloablated immunodeficient mice. Hum Gene Ther 2000;11:729–738.

    Chiang GG, Rubin HL, Cherington V et al. Bone marrow stromal cell-mediated gene therapy for hemophilia A: in vitro expression of human factor VIII with high biological activity requires the inclusion of the proteolytic site at amino acid 1648. Hum Gene Ther 1999;10:61–76.

    Gordon EM, Skotzko M, Kundu RK et al. Capture and expansion of bone marrow-derived mesenchymal progenitor cells with a transforming growth factor-beta1-von Willebrand’s factor fusion protein for retrovirus-mediated delivery of coagulation factor IX. Hum Gene Ther 1997;8:1385–1394.

    Hurwitz DR, Kirchgesser M, Merrill W et al. Systemic delivery of human growth hormone or human factor IX in dogs by reintroduced genetically modified autologous bone marrow stroma cells. Hum Gene Ther 1997;8:137–156.

    Krebsbach PH, Zhang K, Malik AK et al. Bone marrow stromal cells as a genetic platform for systemic delivery of therapeutic proteins in vivo: human factor IX model. J Gene Med 2003;5:11–17.

    Suzuki K, Oyama M, Faulcon L et al. In vivo expression of human growth hormone by genetically modified murine bone marrow stromal cells and its effect on the cells in vitro. Cell Transplant 2000;9:319–327.

    Shen FH, Visger JM, Balian G et al. Systemically administered mesenchymal stromal cells transduced with insulin-like growth factor-I localize to a fracture site and potentiate healing. J Orthop Trauma 2002;16:651–659.

    Lee K, Majumdar MK, Buyaner D et al. Human mesenchymal stem cells maintain transgene expression during expansion and differentiation. Mol Ther 2001;3:857–866.

    Bartholomew A, Patil S, Mackay A et al. Baboon mesenchymal stem cells can be genetically modified to secrete human erythropoietin in vivo. Hum Gene Ther 2001;12:1527–1541.

    Baxter MA, Wynn RF, Deakin JA et al. Retrovirally mediated correction of bone marrow-derived mesenchymal stem cells from patients with mucopolysaccharidosis type I. Blood 2002;99:1857–1859.

    Niyibizi C, Smith P, Mi Z et al. Transfer of proalpha2 (I) cDNA into cells of a murine model of human Osteogenesis Imperfecta restores synthesis of type I collagen comprised of alpha1 (I) and alpha2 (I) heterotrimers in vitro and in vivo. J Cell Biochem 2001;83:84–91.

    Tsuchiya H, Kitoh H, Sugiura F et al. Chondrogenesis enhanced by over expression of sox9 gene in mouse bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun 2003;301:338–343.

    Studeny M, Marini FC, Champlin RE et al. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-? delivery into tumors. Cancer Res 2002;62:3603–3608.

    Pinkel D, Straume T, Gray JW. Cytogenetic analysis using quantitative, high-sensitivity, fluorescence hybridization. Proc Natl Acad Sci USA 1986;83:2934–2938.

    Hung SC, Chen NJ, Hsieh SL et al. Isolation and characterization of size-sieved stem cells from human bone marrow. STEM CELLS 2002;20:249–258.

    Caterson EJ, Nesti LJ, Danielson KG et al. Human marrow-derived mesenchymal progenitor cells: isolation, culture expansion, and analysis of differentiation. Mol Biotechnol 2002;20:245–256.

    Ringe J, Kaps C, Burmester GR et al. Stem cells for regenerative medicine: advances in the engineering of tissues and organs. Naturwissenschaften 2002;89:338–351.

    Tocci A, Forte L. Mesenchymal stem cell: use and perspectives. Hematol J 2003;4:92–96.

    Jiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41–49.

    Groth AC, Olivares EC, Thyagarajan B et al. A phage integrase directs efficient site-specific integration in human cells. Proc Natl Acad Sci USA 2000;97:5995–6000.

    Kaminski JM, Huber MR, Summers JB et al. Design of a nonviral vector for site-selective, efficient integration into the human genome. FASEB J 2002;16:1242–1247.

    Kolot M, Silberstein N, Yagil E. Site-specific recombination in mammalian cells expressing the Int recombinase of bacteriophage HK022. Mol Biol Rep 1999;26:207–213.

    Lorbach E, Christ N, Schwikardi M et al. Site-specific recombination in human cells catalyzed by phage lambda integrase mutants. J Mol Biol 2000;296:1175–1181.

    Olivares EC, Hollis RP, Calos MP. Phage R4 integrase mediates site-specific integration in human cells. Gene 2001;278:167–176.

    Stoll SM, Ginsburg DS, Calos MP. Phage TP901-1 site-specific integrase functions in human cells. J Bacteriol 2002;184:3657–3663.

    Thyagarajan B, Olivares EC, Hollis RP et al. Site-specific genomic integration in mammalian cells mediated by phage phiC31 integrase. Mol Cell Biol 2001;21:3926–3934.

    Gorman C, Bullock C. Site-specific gene targeting for gene expression in eukaryotes. Curr Opin Biotechnol 2000;11:455–460.

    Kolb AF. Genome engineering using site-specific recombinases. Cloning Stem Cells 2002;4:65–80.

    Shen C, Buck AK, Liu X et al. Gene silencing by adenovirus-delivered siRNA. FEBS Lett 2003;539:111–114.

    Shuey DJ, McCallus DE, Giordano T. RNAi: gene-silencing in therapeutic intervention. Drug Discov Today 2002;7:1040–1046.

    Yu JY, DeRuiter SL, Turner DL. RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells. Proc Natl Acad Sci USA 2002;99:6047–6052.

    Robinson KA, Beverley SM. Improvements in transfection efficiency and tests of RNA interference (RNAi) approaches in the protozoan parasite Leishmania. Mol Biochem Parasitol 2003;128:217–228.

    Xia H, Mao Q, Paulson HL et al. siRNA-mediated gene silencing in vitro and in vivo. Nat Biotechnol 2002;20:1006–1010.(S. Vanderbyl, G. N. MacDo)