Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 10(2): 254-275, 2018 02.
Article in English | MEDLINE | ID: mdl-29242210

ABSTRACT

Transferring large or multiple genes into primary human stem/progenitor cells is challenged by restrictions in vector capacity, and this hurdle limits the success of gene therapy. A paradigm is Duchenne muscular dystrophy (DMD), an incurable disorder caused by mutations in the largest human gene: dystrophin. The combination of large-capacity vectors, such as human artificial chromosomes (HACs), with stem/progenitor cells may overcome this limitation. We previously reported amelioration of the dystrophic phenotype in mice transplanted with murine muscle progenitors containing a HAC with the entire dystrophin locus (DYS-HAC). However, translation of this strategy to human muscle progenitors requires extension of their proliferative potential to withstand clonal cell expansion after HAC transfer. Here, we show that reversible cell immortalisation mediated by lentivirally delivered excisable hTERT and Bmi1 transgenes extended cell proliferation, enabling transfer of a novel DYS-HAC into DMD satellite cell-derived myoblasts and perivascular cell-derived mesoangioblasts. Genetically corrected cells maintained a stable karyotype, did not undergo tumorigenic transformation and retained their migration ability. Cells remained myogenic in vitro (spontaneously or upon MyoD induction) and engrafted murine skeletal muscle upon transplantation. Finally, we combined the aforementioned functions into a next-generation HAC capable of delivering reversible immortalisation, complete genetic correction, additional dystrophin expression, inducible differentiation and controllable cell death. This work establishes a novel platform for complex gene transfer into clinically relevant human muscle progenitors for DMD gene therapy.


Subject(s)
Chromosomes, Artificial, Human , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Cells, Cultured , Genetic Vectors , Humans , Mice , Models, Animal , Mutation
2.
Yonago Acta Med ; 58(1): 23-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26190894

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) hold promise for application in adult stem cell-mediated regenerative medicine in bone remodeling and fracture repair. MSCs in vitro can be directed to osteogenic lineage by dexamethasone (DEX); however, the use of DEX is not practical in clinical settings because of adverse side effects such as glucocorticoid-induced osteoporosis. For identifying substances that facilitate osteogenesis, a monitoring system, which detects the osteogenic differentiation stage of MSCs accurately and easily, is required. METHODS: By focusing on the human osteocalcin (OC) gene whose expression profile is described along with osteogenic differentiation, we constructed the luciferase (Luc) reporter gene driven by the enhancer/promoter sequence of the human OC gene (OC-Luc) utilizing a mammalian artificial chromosome. Mammalian artificial chromosome is a suitable platform for loading reporter constructs, because of its stable episomal maintenance in host cells, transferability into any cell and assurance of long-term physiological transgene expression. We loaded the OC-Luc on a mammalian artificial chromosome vector engineered from mouse chromosome (designated as mouse artificial chromosome, MAC) in Chinese hamster ovary cells (OC-Luc/MAC) and transferred this into human MSC cells via chromosome transfer. RESULTS: OC-Luc/MAC in human MSC cells are responsive to positive and negative stimulation by 1 alpha,25-dihydroxyvitamin D3 and DEX in differentiation stage of MSCs to osteoblasts, reflecting the manner of physiological expression. CONCLUSION: The OC-Luc/MAC reporter system may contribute not only to monitoring the osteogenic differentiation stage from MSC but also to identify novel osteogenic drugs.

3.
BMC Biotechnol ; 15: 58, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26088202

ABSTRACT

BACKGROUND: Human artificial chromosome (HAC) vectors have some unique characteristics as compared with conventional vectors, carrying large transgenes without size limitation, showing persistent expression of transgenes, and existing independently from host genome in cells. With these features, HACs are expected to be promising vectors for modifications of a variety of cell types. However, the method of introduction of HACs into target cells is confined to microcell-mediated chromosome transfer (MMCT), which is less efficient than other methods of vector introduction. Application of Measles Virus (MV) fusogenic proteins to MMCT instead of polyethylene glycol (PEG) has partly solved this drawback, whereas the tropism of MV fusogenic proteins is restricted to human CD46- or SLAM-positive cells. RESULTS: Here, we show that retargeting of microcell fusion by adding anti-Transferrin receptor (TfR) single chain antibodies (scFvs) to the extracellular C-terminus of the MV-H protein improves the efficiency of MV-MMCT to human fibroblasts which originally barely express both native MV receptors, and are therefore resistant to MV-MMCT. Efficacy of chimeric fusogenic proteins was evaluated by the evidence that the HAC, tagged with a drug-resistant gene and an EGFP gene, was transferred from CHO donor cells into human fibroblasts. Furthermore, it was demonstrated that no perturbation of either the HAC status or the functions of transgenes was observed on account of retargeted MV-MMCT when another HAC carrying four reprogramming factors (iHAC) was transferred into human fibroblasts. CONCLUSIONS: Retargeted MV-MMCT using chimeric H protein with scFvs succeeded in extending the cell spectrum for gene transfer via HAC vectors. Therefore, this technology could facilitate the systematic cell engineering by HACs.


Subject(s)
Chromosomes, Artificial, Human/genetics , Measles virus/genetics , Viral Fusion Proteins/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Fibroblasts , Gene Transfer Techniques , Humans , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
Chromosome Res ; 23(1): 111-33, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25657031

ABSTRACT

Microcell-mediated chromosome transfer (MMCT) is a technique to transfer a chromosome from defined donor cells into recipient cells and to manipulate chromosomes as gene delivery vectors and open a new avenue in somatic cell genetics. However, it is difficult to uncover the function of a single specific gene via the transfer of an entire chromosome or fragment, because each chromosome or fragment contains a set of numerous genes. Thus, alternative tools are human artificial chromosome (HAC) and mouse artificial chromosome (MAC) vectors, which can carry a gene or genes of interest. HACs/MACs have been generated mainly by either a "top-down approach" (engineered creation) or a "bottom-up approach" (de novo creation). HACs/MACs with one or more acceptor sites exhibit several characteristics required by an ideal gene delivery vector, including stable episomal maintenance and the capacity to carry large genomic loci plus their regulatory elements, thus allowing the physiological regulation of the introduced gene in a manner similar to that of native chromosomes. The MMCT technique is also applied for manipulating HACs and MACs in donor cells and delivering them to recipient cells. This review describes the lessons learned and prospects identified from studies on the construction of HACs and MACs, and their ability to drive exogenous gene expression in cultured cells and transgenic animals via MMCT. New avenues for a variety of applications to bio-medical challenges are also proposed.


Subject(s)
Biomedical Technology/methods , Chromosomes, Artificial/genetics , Epigenesis, Genetic , Gene Transfer Techniques , Genetic Engineering/methods , Models, Genetic , Animals , Genetic Vectors/genetics , Humans , Mice
5.
FEBS J ; 281(11): 2623-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24712640

ABSTRACT

Mitotic catastrophe, a form of cell death that occurs during mitosis and after mitotic slippage to a tetraploid state, plays important roles in the efficacy of cancer cell killing by microtubule inhibitors (MTIs). Prolonged mitotic arrest by the spindle assembly checkpoint is a well-known requirement for mitotic catastrophe, and thus for conferring sensitivity to MTIs. We previously reported that turning off spindle assembly checkpoint activation after a defined period of time is another requirement for efficient postslippage death from a tetraploid state, and we identified SIRT2, a member of the sirtuin protein family, as a regulator of this process. Here, we investigated whether SIRT2 regulates basal autophagy and whether, in that case, autophagy regulation by SIRT2 is required for postslippage death, by analogy with previous insights into SIRT1 functions in autophagy. We show, by combined knockdown of autophagy genes and SIRT2, that SIRT2 serves this function at least partially by suppressing basal autophagy levels. Notably, increased autophagy induced by rapamycin and mild starvation caused mitotic arrest for an abnormally long period of time in the presence of MTIs, and this was followed by delayed postslippage death, which was also observed in cells with SIRT2 knockdown. These results underscore a causal association among increased autophagy levels, mitotic arrest for an abnormally long period of time after exposure to MTIs, and resistance to MTIs. Although autophagy acts as a tumor suppressor mechanism, this study highlights its negative aspects, as increased autophagy may cause mitotic catastrophe malfunction. Thus, SIRT2 offers a novel target for tumor therapy.


Subject(s)
Sirtuin 2/genetics , Autophagy/drug effects , Autophagy/genetics , HCT116 Cells , Humans , Mitosis/drug effects , Mitosis/genetics , Sirtuin 1/deficiency , Sirtuin 1/genetics , Sirtuin 2/deficiency , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tubulin Modulators/pharmacology
6.
Cytotechnology ; 65(5): 803-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23494082

ABSTRACT

Microcell-mediated chromosome transfer (MMCT) technology enables a single and intact mammalian chromosome or megabase-sized chromosome fragments to be transferred from donor to recipient cells. The conventional MMCT method is performed immediately after the purification of microcells. The timing of the isolation of microcells and the preparation of recipient cells is very important. Thus, ready-made microcells can improve and simplify the process of MMCT. Here, we established a cryopreservation method to store microcells at -80 °C, and compared these cells with conventionally- (immediately-) prepared cells with respect to the efficiency of MMCT and the stability of a human artificial chromosome (HAC) transferred to human HT1080 cells. The HAC transfer in microcell hybrids was confirmed by FISH analysis. There was no significant difference between the two methods regarding chromosome transfer efficiency and the retention rate of HAC. Thus, cryopreservation of ready-to-use microcells provides an improved and simplified protocol for MMCT.

7.
PLoS One ; 6(10): e25961, 2011.
Article in English | MEDLINE | ID: mdl-21998730

ABSTRACT

Human artificial chromosomes (HACs) have unique characteristics as gene-delivery vectors, including episomal transmission and transfer of multiple, large transgenes. Here, we demonstrate the advantages of HAC vectors for reprogramming mouse embryonic fibroblasts (MEFs) into induced pluripotent stem (iPS) cells. Two HAC vectors (iHAC1 and iHAC2) were constructed. Both carried four reprogramming factors, and iHAC2 also encoded a p53-knockdown cassette. iHAC1 partially reprogrammed MEFs, and iHAC2 efficiently reprogrammed MEFs. Global gene expression patterns showed that the iHACs, unlike other vectors, generated relatively uniform iPS cells. Under non-selecting conditions, we established iHAC-free iPS cells by isolating cells that spontaneously lost iHAC2. Analyses of pluripotent markers, teratomas and chimeras confirmed that these iHAC-free iPS cells were pluripotent. Moreover, iHAC-free iPS cells with a re-introduced HAC encoding Herpes Simplex virus thymidine kinase were eliminated by ganciclovir treatment, indicating that the HAC safeguard system functioned in iPS cells. Thus, the HAC vector could generate uniform, integration-free iPS cells with a built-in safeguard system.


Subject(s)
Cell Engineering/methods , Chromosomes, Artificial, Human/genetics , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Animals , CHO Cells , Cricetinae , Cricetulus , Fibroblasts/cytology , Genes, Transgenic, Suicide/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Simplexvirus/genetics , Thymidine Kinase/genetics
8.
Genes Cells ; 16(1): 34-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21059157

ABSTRACT

We previously reported that sirtuin 2 (SIRT2), a mammalian member of the NAD+-dependent protein deacetylases, participates in mitotic regulation, specifically, in efficient mitotic cell death caused by the spindle checkpoint. Here, we describe a novel function of SIRT2 that is different from mitotic regulation. SIRT2 down-regulation using siRNA caused apoptosis in cancer cell lines such as HeLa cells, but not in normal cells. The apoptosis was caused by p53 accumulation, which is mediated by p38 MAPK activation-dependent degradation of p300 and the subsequent MDM2 degradation. Sirtuin inhibitors are emerging as antitumor drugs, and this function has been ascribed to the inhibition of SIRT1, the most well-characterized sirtuin that deacetylases p53 to promote cell survival and also binds to other proteins in response to genotoxic stress. This study suggests that SIRT2 can be a novel molecular target for cancer therapy and provides a molecular basis for the efficacy of SIRT2 for future cancer therapy.


Subject(s)
Apoptosis/genetics , Down-Regulation , Sirtuin 2/metabolism , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , E1A-Associated p300 Protein/metabolism , HeLa Cells , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sirtuins/genetics , Sirtuins/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
9.
BMC Biotechnol ; 10: 37, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20444293

ABSTRACT

BACKGROUND: Microcell-mediated chromosome transfer (MMCT) is a technique by which a chromosome(s) is moved from donor to recipient cells by microcell fusion. Polyethylene glycol (PEG) has conventionally been used as a fusogen, and has been very successful in various genetic studies. However, PEG is not applicable for all types of recipient cells, because of its cell type-dependent toxicity. The cytotoxicity of PEG limits the yield of microcell hybrids to low level (10-6 to 10-5 per recipient cells). To harness the full potential of MMCT, a less toxic and more efficient fusion protocol that can be easily manipulated needs to be developed. RESULTS: Microcell donor CHO cells carrying a human artificial chromosome (HAC) were transfected with genes encoding hemagglutinin (H) and fusion (F) proteins of an attenuated Measles Virus (MV) Edmonston strain. Mixed culture of the CHO transfectants and MV infection-competent human fibrosarcoma cells (HT1080) formed multinucleated syncytia, suggesting the functional expression of the MV-H/F in the CHO cells. Microcells were prepared and applied to HT1080 cells, human immortalized mesenchymal stem cells (hiMSC), and primary fibroblasts. Drug-resistant cells appeared after selection in culture with Blasticidin targeted against the tagged selection marker gene on the HAC. The fusion efficiency was determined by counting the total number of stable clones obtained in each experiment. Retention of the HAC in the microcell hybrids was confirmed by FISH analyses. The three recipient cell lines displayed distinct fusion efficiencies that depended on the cell-surface expression level of CD46, which acts as a receptor for MV. In HT1080 and hiMSC, the maximum efficiency observed was 50 and 100 times greater than that using conventional PEG fusion, respectively. However, the low efficiency of PEG-induced fusion with HFL1 was not improved by the MV fusogen. CONCLUSIONS: Ectopic expression of MV envelope proteins provides an efficient recipient cell-oriented MMCT protocol, facilitating extensive applications for studies of gene function and genetic corrections.


Subject(s)
Cell Fusion/methods , Membrane Cofactor Protein/chemistry , Viral Envelope Proteins/chemistry , Animals , CHO Cells , Cell Line, Tumor , Chromosomes, Artificial, Human , Cricetinae , Cricetulus , Genetic Vectors , Humans , Measles virus
10.
Cell Cycle ; 8(8): 1279-91, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19282667

ABSTRACT

We previously identified SIRT2, a deacetylase for tubulin and histone H4, as a protein downregulated in gliomas, and reported that exogenously-expressed SIRT2 arrests the cell cycle prior to entry into mitosis to prevent chromosomal instability in response to microtubule inhibitors (MTIs) such as nocodazole, characteristics previously reported for the CHFR protein. We herein investigated the effects of SIRT2 downregulation on sensitivity to MTIs using HCT116 cells, a mitotic checkpoint-proficient near-diploid cancer cell line used for studying checkpoints. We found that SIRT2 downregulation confers resistance to MTIs as well as that of BubR1, a well-characterized mitotic checkpoint protein, though by a different mechanism. While BubR1 suppression abolished spindle checkpoint functions, which is a requirement for cell death after release from the spindle checkpoint, SIRT2 downregulation prolonged chronic mitotic arrest from sustained activation of the mitotic checkpoint and consequently prevented a shift to secondary outcomes, including cell death, after release from chronic mitotic arrest. Consistent with this notion, BubR1 downregulation was dominant over SIRT2 knockdown in regard to mitotic regulation in the presence of nocodazole. These results suggest that SIRT2 functions to release chronic mitotic arrest in cells treated with MTIs, leading to other outcomes. We also found that SIRT2 downregulation caused centrosome fragmentation in response to nocodazole prior to the alteration in spindle checkpoint function, implying not only a novel function of SIRT2 for centrosome maintenance upon exposure to mitotic stress caused by MTIs, but also the existence of a centrosome-mediated signaling pathway to sustain the spindle checkpoint. Therefore, this study highlights a novel pathway leading to resistance to MTIs, in which SIRT2 downregulation participates.


Subject(s)
Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Microtubules/drug effects , Mitosis/drug effects , Sirtuins/metabolism , Tubulin Modulators/pharmacology , Centrosome/drug effects , Centrosome/metabolism , HCT116 Cells , Humans , Microtubules/metabolism , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Sirtuin 2
11.
Biochem Biophys Res Commun ; 369(3): 807-11, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18328257

ABSTRACT

Telomerase-mediated life-span extension enables the expansion of normal cells without malignant transformation, and thus has been thought to be useful in cell therapies. Currently, integrating vectors including the retrovirus are used for human telomerase reverse transcriptase (hTERT)-mediated expansion of normal cells; however, the use of these vectors potentially causes unexpected insertional mutagenesis and/or activation of oncogenes. Here, we established normal human fibroblast (hPF) clones retaining non-integrating human artificial chromosome (HAC) vectors harboring the hTERT expression cassette. In hTERT-HAC/hPF clones, we observed the telomerase activity and the suppression of senescent-associated SA-beta-galactosidase activity. Furthermore, the hTERT-HAC/hPF clones continued growing beyond 120days after cloning, whereas the hPF clones retaining the silent hTERT-HAC senesced within 70days. Thus, hTERT-HAC-mediated episomal expression of hTERT allows the extension of the life-span of human primary cells, implying that gene delivery by non-integrating HAC vectors can be used to control cellular proliferative capacity of primary cultured cells.


Subject(s)
Cell Proliferation , Chromosomes, Artificial, Human/genetics , Genetic Vectors/genetics , Telomerase/genetics , Animals , CHO Cells , Cell Division , Cellular Senescence , Cricetinae , Cricetulus , Fibroblasts/enzymology , Fibroblasts/physiology , Gene Transfer Techniques , Humans , Telomerase/analysis
12.
Reprod Biomed Online ; 16(1): 57-69, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18252049

ABSTRACT

Human chromosome fragments and human artificial chromosomes (HAC) represent feasible gene delivery vectors via microcell-mediated chromosome transfer. Strategies to construct HAC involve either 'build up' or 'top-down' approaches. For each approach, techniques for manipulating HAC in donor cells in order to deliver HAC to recipient cells are required. The combination of chromosome fragments or HAC with microcell-mediated chromosome transfer has facilitated human gene mapping and various genetic studies. The recent emergence of stem cell-based tissue engineering has opened up new avenues for gene and cell therapies. The task now is to develop safe and effective vectors that can deliver therapeutic genes into specific stem cells and maintain long-term regulated expression of these genes. Although the transfer-efficiency needs to be improved, HAC possess several characteristics that are required for gene therapy vectors, including stable episomal maintenance and the capacity for large gene insets. HAC can also carry genomic loci with regulatory elements, which allow for the expression of transgenes in a genetic environment similar to the natural chromosome. This review describes the lessons and prospects learned, mainly from recent studies in developing HAC and HAC-mediated gene expression in embryonic and adult stem cells, and in transgenic animals.


Subject(s)
Chromosomes, Artificial, Human/genetics , Embryonic Stem Cells , Gene Transfer Techniques , Genetic Vectors/genetics , Adult Stem Cells , Animals , Animals, Genetically Modified , Humans , Mice
13.
Biochim Biophys Acta ; 1770(2): 206-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17184921

ABSTRACT

Human artificial chromosome (HAC) vectors possess several characteristics sufficient for the requirements of gene therapy vectors, including stable episomal maintenance and mediation of long-term transgene expression. In this study, we adopted an antigen-mediated genetically modified cell amplification (AMEGA) system employing an antibody/cytokine receptor chimera that triggers a growth signal in response to a cognate non-toxic antigen, and applied it to growth control of HAC-transferred cells by adding an antigen that differed from cytokines that may manifest pleiotropic effects. We previously constructed a novel HAC vector, 21 Delta qHAC, derived from human chromosome 21, housed in CHO cells. Here, we constructed an HAC vector harboring an ScFv-gp130 chimera responsive to fluorescein-conjugated BSA (BSA-FL) as well as a model transgene, enhanced green fluorescent protein (EGFP), in CHO cells. The modified HAC was transferred into interleukin (IL)-6-dependent hybridoma 7TD1 cells by microcell-mediated chromosome transfer, and the cells were subsequently found to show BSA-FL-dependent cell growth and sustained expression of EGFP in the absence of IL-6. The AMEGA system in combination with HAC technology will be useful for increasing the efficacy of gene therapy by conferring a growth advantage on the genetically modified cells.


Subject(s)
Cell Division/physiology , Chromosomes, Artificial, Human/physiology , Hybridomas/cytology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Gene Transfer Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Interleukin-6/pharmacology , Mice , Recombinant Fusion Proteins , Transfection
14.
Stem Cell Rev ; 2(1): 43-50, 2006.
Article in English | MEDLINE | ID: mdl-17142886

ABSTRACT

The recent emergence of stem cell-based tissue engineering has now opened up new venues for gene therapy. The task now is to develop safe and effective vectors that can deliver therapeutic genes into specific stem cell lines and maintain long-term regulated expression of these genes. Human artificial chromosomes (HACs) possess several characteristics that require gene therapy vectors, including a stable episomal maintenance, and the capacity for large gene inserts. HACs can also carry genomic loci with regulatory elements, thus allowing for the expression of transgenes in a genetic environment similar to the chromosome. Currently, HACs are constructed by a two prone approaches. Using a top-down strategy, HACs can be generated from fragmenting endogenous chromosomes. By a bottom-up strategy, HACs can be created de novo from cloned chromosomal components using chromosome engineering. This review describes the current advances in developing HACs, with the main focus on their applications and potential value in gene delivery, such as HAC-mediated gene expression in embryonic, adult stem cells, and transgenic animals.


Subject(s)
Chromosomes, Artificial, Human , Gene Transfer Techniques , Stem Cells/physiology , Animals , Genetic Engineering/methods , Humans , Mice
15.
Biochem Biophys Res Commun ; 340(4): 1053-61, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16403445

ABSTRACT

Human artificial chromosomes (HACs) behave as independent minichromosomes and are potentially useful as a way to achieve safe, long-term expression of a transgene. In this study, we sought to elucidate the potential of HAC vectors carrying the human proinsulin transgene for gene therapy of insulin-dependent diabetes mellitus (IDDM) using non-beta-cells as a host for the vector. To facilitate the production of mature insulin in non-beta-cells and to safely regulate the level of transgene expression, we introduced furin-cleavable sites into the proinsulin coding region and utilized the heat shock protein 70 (Hsp70) promoter. We used Cre-loxP-mediated recombination to introduce the gene cassettes onto 21DeltapqHAC, a HAC vector whose structure is completely defined, present in human fibrosarcoma HT1080 cells. We observed long-term expression and stable retention of the transgene without aberrant translocation of the HAC constructs. As expected, the Hsp70 promoter allowed us to regulate gene expression with temperature, and the production and secretion of intermediates of mature insulin were made possible by the furin-cleavable sites we had introduced into proinsulin. This study can be an initial step on the application of HAC vectors on the gene delivery to non-beta-cells, which might provide a direction for future treatment for diabetes.


Subject(s)
Chromosomes, Artificial, Human/genetics , Fibrosarcoma/metabolism , HSP70 Heat-Shock Proteins/genetics , Insulin/biosynthesis , Insulin/metabolism , Transfection/methods , Cell Line, Tumor , Fibrosarcoma/genetics , Genetic Vectors , Hot Temperature , Humans , Insulin/genetics , Insulin Secretion , Promoter Regions, Genetic/genetics
16.
J Hum Genet ; 51(2): 147-150, 2006.
Article in English | MEDLINE | ID: mdl-16333524

ABSTRACT

A number of gene delivery systems are currently being developed for potential use in gene therapy. Here, we demonstrate the feasibility of 21deltaqHAC, a newly developed human artificial chromosome (HAC), as a gene delivery system. We first introduced a 21deltaqHAC carrying an EGFP reporter gene and a geneticin-resistant gene (EGFP-21deltaqHAC) into hematopoietic cells by microcell-mediated chromosome transfer. These HAC-containing hematopoietic cells showed resistance to geneticin, expressed EGFP and retained the ability to differentiate into various lineages, and the EGFP-21deltaqHAC was successfully transduced into primary hematopoietic cells. Hematopoietic cells harboring the EGFP-21deltaqHAC could still be detected at two weeks post-transplantation in immunodeficient mice. We also showed effective expansion of hematopoietic cells by introducing the 21deltaqHAC containing ScFvg, a gp130-based chimeric receptor that transmits growth signals in response to specific-antigen of this receptor. All of these results demonstrate the usefulness of HAC in gene therapy.


Subject(s)
Chromosomes, Artificial, Human/genetics , Chromosomes, Human, Pair 21/genetics , Cord Blood Stem Cell Transplantation/methods , Gene Expression , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Animals , DNA Primers , Flow Cytometry , Genetic Vectors/genetics , Gentamicins , Green Fluorescent Proteins/metabolism , Humans , Mice
17.
J Hum Genet ; 50(11): 592-599, 2005.
Article in English | MEDLINE | ID: mdl-16231070

ABSTRACT

Following the recent completion of the human genome sequence, genomics research has shifted its focus to understanding gene complexity, expression, and regulation. However, in order to investigate such issues, there is a need to develop a practical system for genomic DNA expression. Transformation-associated recombination (TAR) cloning has proven to be a convenient tool for selective isolation of a genetic locus from a complex genome as a circular YAC using recombination in yeast. The human artificial chromosome (HAC) vector containing an acceptor loxP site has served as a platform for the reproducible expression of transgenes. In this study, we describe a system that efficiently expresses a genetic locus in mammalian cells by retrofitting a TAR-YAC with the donor loxP site and loading it onto the HAC vector by the Cre/loxP system. In order to demonstrate functional expression of genomic loci, the entire human hypoxanthine phosphoribosyl transferase (HPRT) locus contained in a 100 kb YAC was loaded onto the HAC vector and was shown to complement the genetic defect in Hprt-deficient CHO cells. Thus, the combination of TAR cloning and the HAC vector may serve as a powerful tool for functional genomic studies.


Subject(s)
Chromosomes, Artificial, Human/genetics , Cloning, Molecular/methods , Gene Expression Profiling/methods , Genome, Human/genetics , Recombination, Genetic/genetics , Transformation, Genetic/genetics , Animals , Blotting, Southern , CHO Cells , Cricetinae , Cricetulus , DNA Primers , Electrophoresis, Gel, Pulsed-Field , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , In Situ Hybridization, Fluorescence , Polymerase Chain Reaction , Restriction Mapping
18.
Stem Cells ; 23(10): 1608-16, 2005.
Article in English | MEDLINE | ID: mdl-16141362

ABSTRACT

Mesenchymal stem cells (MSCs) hold promise for use in adult stem cell-mediated gene therapy. One of the major aims of stem cell-mediated gene therapy is to develop vectors that will allow appropriate levels of expression of therapeutic genes along differentiation under physiological regulation of the specialized cells. Human artificial chromosomes (HACs) are stably maintained as independent chromosomes in host cells and should be free from potential insertional mutagenesis problems of conventional transgenes. Therefore, HACs have been proposed as alternative implements to cell-mediated gene therapy. Previously, we constructed a novel HAC, termed 21 Deltapq HAC, with a loxP site in which circular DNA can be reproducibly inserted by the Cre/loxP system. We here assessed the feasibility of lineage-specific transgene expression by the 21Deltapq HAC vector using an in vitro differentiation system with an MSC cell line, hiMSCs, which has potential for osteogenic, chondrogenic, and adipogenic differentiation. An enhanced green fluorescent protein (EGFP) gene driven by a promoter for osteogenic lineage-specific osteopontin (OPN) gene was inserted onto the 21 Deltapq HAC and then transferred into hiMSC. The expression cassette was flanked by the chicken HS4 insulators to block promoter interference from adjacent drug-resistant genes. The EGFP gene was specifically expressed in the hiMSC that differentiated into osteocytes in coordination with the transcription of endogenous OPN gene but was not expressed after adipogenic differentiation induction or in noninduction culture. These results suggest that use of the HAC vector is suitable for regulated expression of transgenes in stem cell-mediated gene therapy.


Subject(s)
Cell Lineage , Chromosomes, Artificial, Human/genetics , Chromosomes, Artificial, Human/metabolism , Gene Expression , Genetic Vectors , Mesenchymal Stem Cells/cytology , Transgenes , Antineoplastic Combined Chemotherapy Protocols , Cell Differentiation , Cyclophosphamide , Doxorubicin , Gene Deletion , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Osteopontin , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Vincristine
19.
Biochem Biophys Res Commun ; 329(3): 1018-25, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15752757

ABSTRACT

Efficient regulation of transgene would greatly facilitate the analysis of gene function in biological systems for basic research and clinical applications. The tetracycline-regulatable system (TRS) has proven to be a promising tool for such purposes. Despite their widespread application, a number of challenges are still associated with the use of TRS, including clonal variability in the regulation and copy number. We have recently constructed a novel human artificial chromosome (HAC) called 21DeltaqHAC. By housing a TRS-based DNA-PKcs expression cassette in this HAC, we were able to circumvent the problems associated with conventional TRS-based vectors. We achieved tight control of DNA-PKcs expression and rescued the radiosensitive phenotype of DNA-PKcs-deficient CHO cells. The combined use of HAC and the TRS serves as a model for controllable and fixed copy number expression vectors. Our study also demonstrates the suitability of the HAC to accommodate multi-subunit constructs such as that of the TRS.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Cloning, Molecular/methods , DNA-Binding Proteins/genetics , Genetic Engineering/methods , Protein Serine-Threonine Kinases/genetics , Transfection/methods , Animals , CHO Cells , Cricetinae , Cricetulus , DNA-Activated Protein Kinase , DNA-Binding Proteins/biosynthesis , Gene Transfer Techniques , Genetic Vectors , Humans , Nuclear Proteins , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/biosynthesis , Recombinant Proteins/biosynthesis
20.
Oncol Rep ; 12(6): 1273-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15547750

ABSTRACT

Paternally expressed imprinted gene 1/mesoderm-specific transcript (PEG1/MEST) is an imprinted gene expressed from the paternal allele. Recently, frequent loss of imprinting (LOI) of PEG1/MEST has been reported in lung adenocarcinomas. It is suggested that the LOI may be involved in pathogenesis of lung adenocarcinoma. In the present study, incidence of LOI of PEG1/MEST was examined in lung cancer cell lines, including small cell lung cancer (SCLC). Among 50 cell lines tested, 20 cell lines were heterozygous for the AflIII site of the PEG1/MEST gene. In these heterozygotes, biallelic expression was observed in 9 cell lines (45%), monoallelic in 11 (55%). In cell lines of non-small cell lung cancer (NSCLC), 62% (8 of 13) exhibited biallelic expression. In SCLC, only 1 of 7 cell lines (14%) showed biallelic expression. LOI of PEG1/MEST in the NSCLC cell line is significantly frequent compared with that in SCLC cell lines (p=0.043). This result supports the possibility that LOI may be related to tumorigenesis and malignant transformation, especially in NSCLC.


Subject(s)
Genomic Imprinting , Lung Neoplasms/genetics , Proteins/genetics , Adenocarcinoma/genetics , Carcinoma, Giant Cell/genetics , Carcinoma, Large Cell/genetics , Carcinoma, Small Cell/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Humans , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...