1
|
BET-Independent Murine Leukemia Virus Integration Is Retargeted
In Vivo
and Selects Distinct Genomic Elements for Lymphomagenesis. Microbiol Spectr 2022; 10:e0147822. [PMID: 35852337 PMCID: PMC9431007 DOI: 10.1128/spectrum.01478-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Moloney murine leukemia virus (MLV) infects BALB/c mice and induces T-cell lymphoma in mice. Retroviral integration is mediated by the interaction of the MLV integrase (IN) with members of the bromodomain and extraterminal motif (BET) protein family (BRD2, BRD3, and BRD4). The introduction of the W390A mutation into MLV IN abolishes the BET interaction. Here, we compared the replication of W390A MLV to that of wild-type (WT) MLV in adult BALB/c mice to study the role of BET proteins in replication, integration, and tumorigenesis in vivo. Comparing WT and W390A MLV infections revealed similar viral loads in the blood, thymus, and spleen cells. Interestingly, W390A MLV integration was retargeted away from GC-enriched genomic regions. However, both WT MLV- and W390A MLV-infected mice developed T-cell lymphoma after similar latencies represented by an enlarged thymus and spleen and multiorgan tumor infiltration. Integration site sequencing from splenic tumor cells revealed clonal expansion in all WT MLV- and W390A MLV-infected mice. However, the integration profiles of W390A MLV and WT MLV differed significantly. Integrations were enriched in enhancers and promoters, but compared to the WT, W390A MLV integrated less frequently into enhancers and more frequently into oncogene bodies such as Notch1 and Ppp1r16b. We conclude that host factors direct MLV in vivo integration site selection. Although BET proteins target WT MLV integration preferentially toward enhancers and promoters, insertional lymphomagenesis can occur independently from BET, likely due to the intrinsically strong enhancer/promoter of the MLV long terminal repeat (LTR). IMPORTANCE In this study, we have shown that the in vivo replication of murine leukemia virus happens independently of BET proteins, which are key host determinants involved in retroviral integration site selection. This finding opens a new research line in the discovery of alternative viral or host factors that may complement the dominant host factor. In addition, our results show that BET-independent murine leukemia virus uncouples insertional mutagenesis from gene enhancers, although lymphomagenesis still occurs despite the lack of an interaction with BET proteins. Our findings also have implications for the engineering of BET-independent MLV-based vectors for gene therapy, which may not be a safe alternative.
Collapse
|
2
|
Bui PL, Nishimura K, Seminario Mondejar G, Kumar A, Aizawa S, Murano K, Nagata K, Hayashi Y, Fukuda A, Onuma Y, Ito Y, Nakanishi M, Hisatake K. Template Activating Factor-I α Regulates Retroviral Silencing during Reprogramming. Cell Rep 2019; 29:1909-1922.e5. [DOI: 10.1016/j.celrep.2019.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/02/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
|
3
|
Lee A, CingÖz O, Sabo Y, Goff SP. Characterization of interaction between Trim28 and YY1 in silencing proviral DNA of Moloney murine leukemia virus. Virology 2018; 516:165-175. [PMID: 29407374 DOI: 10.1016/j.virol.2018.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 10/18/2022]
Abstract
Moloney Murine Leukemia Virus (M-MLV) proviral DNA is transcriptionally silenced in embryonic cells by a large repressor complex tethered to the provirus by two sequence-specific DNA binding proteins, ZFP809 and YY1. A central component of the complex is Trim28, a scaffold protein that regulates many target genes involved in cell cycle progression, DNA damage responses, and viral gene expression. The silencing activity of Trim28, and its interactions with corepressors are often regulated by post-translational modifications such as sumoylation and phosphorylation. We defined the interaction domains of Trim28 and YY1, and investigated the role of sumoylation and phosphorylation of Trim28 in mediating M-MLV silencing. The RBCC domain of Trim28 was sufficient for interaction with YY1, and acidic region 1 and zinc fingers of YY1 were necessary and sufficient for its interaction with Trim28. Additionally, we found that residue K779 was critical for Trim28-mediated silencing of M-MLV in embryonic cells.
Collapse
Affiliation(s)
- Andreia Lee
- Department of Biological Sciences, United States
| | - Oya CingÖz
- Department of Biochemistry and Molecular Biophysics and Department of Microbiology and Immunology, Howard Hughes Medical Institute, Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| | - Yosef Sabo
- Department of Biochemistry and Molecular Biophysics and Department of Microbiology and Immunology, Howard Hughes Medical Institute, Columbia University Medical Center, Columbia University, New York, NY 10032, United States
| | - Stephen P Goff
- Department of Biochemistry and Molecular Biophysics and Department of Microbiology and Immunology, Howard Hughes Medical Institute, Columbia University Medical Center, Columbia University, New York, NY 10032, United States.
| |
Collapse
|
4
|
Tonelli FMP, Lacerda SMSN, Tonelli FCP, Costa GMJ, de França LR, Resende RR. Progress and biotechnological prospects in fish transgenesis. Biotechnol Adv 2017; 35:832-844. [PMID: 28602961 DOI: 10.1016/j.biotechadv.2017.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/04/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
The history of transgenesis is marked by milestones such as the development of cellular transdifferentiation, recombinant DNA, genetic modification of target cells, and finally, the generation of simpler genetically modified organisms (e.g. bacteria and mice). The first transgenic fish was developed in 1984, and since then, continuing technological advancements to improve gene transfer have led to more rapid, accurate, and efficient generation of transgenic animals. Among the established methods are microinjection, electroporation, lipofection, viral vectors, and gene targeting. Here, we review the history of animal transgenesis, with an emphasis on fish, in conjunction with major developments in genetic engineering over the past few decades. Importantly, spermatogonial stem cell modification and transplantation are two common techniques capable of revolutionizing the generation of transgenic fish. Furthermore, we discuss recent progress and future biotechnological prospects of fish transgenesis, which has strong applications for the aquaculture industry. Indeed, some transgenic fish are already available in the current market, validating continued efforts to improve economically important species with biotechnological advancements.
Collapse
Affiliation(s)
- Fernanda M P Tonelli
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nanocell, Divinópolis, MG, Brazil
| | - Samyra M S N Lacerda
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávia C P Tonelli
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Guilherme M J Costa
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Renato de França
- Laboratório de Biologia Celular, Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nacional de Pesquisas da Amazônia (INPA), Manaus, AM, Brazil.
| | - Rodrigo R Resende
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Instituto Nanocell, Divinópolis, MG, Brazil.
| |
Collapse
|
5
|
Transcriptional Silencing of Moloney Murine Leukemia Virus in Human Embryonic Carcinoma Cells. J Virol 2016; 91:JVI.02075-16. [PMID: 27795446 DOI: 10.1128/jvi.02075-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 11/20/2022] Open
Abstract
Embryonic carcinoma (EC) cells are malignant counterparts of embryonic stem (ES) cells and serve as useful models for investigating cellular differentiation and human embryogenesis. Though the susceptibility of murine EC cells to retroviral infection has been extensively analyzed, few studies of retrovirus infection of human EC cells have been performed. We tested the susceptibility of human EC cells to transduction by retroviral vectors derived from three different retroviral genera. We show that human EC cells efficiently express reporter genes delivered by vectors based on human immunodeficiency virus type 1 (HIV-1) and Mason-Pfizer monkey virus (M-PMV) but not Moloney murine leukemia virus (MLV). In human EC cells, MLV integration occurs normally, but no viral gene expression is observed. The block to MLV expression of MLV genomes is relieved upon cellular differentiation. The lack of gene expression is correlated with transcriptional silencing of the MLV promoter through the deposition of repressive histone marks as well as DNA methylation. Moreover, depletion of SETDB1, a histone methyltransferase, resulted in a loss of transcriptional silencing and upregulation of MLV gene expression. Finally, we provide evidence showing that the lack of MLV gene expression may be attributed in part to the lack of MLV enhancer function in human EC cells. IMPORTANCE Human embryonic carcinoma (EC) cells are shown to restrict the expression of murine leukemia virus genomes but not retroviral genomes of the lentiviral or betaretroviral families. The block occurs at the level of transcription and is accompanied by the deposition of repressive histone marks and methylation of the integrated proviral DNA. The host machinery required for silencing in human EC cells is distinct from that in murine EC cell lines: the histone methyltransferase SETDB1 is required, but the widely utilized corepressor TRIM28/Kap1 is not. A transcriptional enhancer element from the Mason-Pfizer monkey virus can override the silencing and promote transcription of chimeric proviral DNAs. The findings reveal novel features of human EC gene regulation not present in their murine counterparts.
Collapse
|
6
|
EBP1, a novel host factor involved in primer binding site-dependent restriction of moloney murine leukemia virus in embryonic cells. J Virol 2013; 88:1825-9. [PMID: 24227866 DOI: 10.1128/jvi.02578-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mouse embryonic cells are unable to support the replication of Moloney murine leukemia virus (MLV). The integrated viral DNA is transcriptionally silenced, largely due to binding of host transcriptional repressors to the primer binding site (PBS) of the provirus. We have previously shown that a PBS DNA-binding repressor complex contains ZFP809 and TRIM28. Here, we identified ErbB3-binding protein 1 (EBP1) to be a novel component of the ZFP809-TRIM28 silencing complex and show that EBP1 depletion reduces PBS-mediated retroviral silencing.
Collapse
|
7
|
Evaluation of residual promoter activity in γ-retroviral self-inactivating (SIN) vectors. Mol Ther 2011; 20:84-90. [PMID: 22008914 DOI: 10.1038/mt.2011.204] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Therapeutic gene delivery mediated by retroviral vectors has the advantage of stable integration into the host genome. A major safety concern for gene delivery achieved by murine leukemia virus (MLV)-based retroviral vectors is the activation of adjacent cellular genes including oncogenes following integration into the host genome. Self-inactivating (SIN) vectors lacking viral enhancers/promoters in their 3' long terminal repeat (LTR) have been proposed as a means of overcoming this safety concern. However the MLV-based SIN vectors currently used by laboratories to assess insertional mutagenesis, integration site selection, and the potency of transgene expression are not uniform in the composition of their 3' LTRs. We constructed a series of SIN vectors representative of the currently employed vectors, but lacking an internal promoter. Green fluorescent protein (GFP) was used as a reporter gene. Target cells exposed to these vectors were evaluated for number of integrants and GFP expression at the messenger RNA (mRNA) level and protein level. We found that viral promoter activity in the 3' LTR is not attenuated in many currently employed SIN vectors. These results suggest that the influence of strong residual promoter activity should be taken into consideration when interpreting experimental results obtained using SIN vectors in gene therapy research.
Collapse
|
8
|
Liu J, Balehosur D, Murray B, Kelly JM, Sumer H, Verma PJ. Generation and characterization of reprogrammed sheep induced pluripotent stem cells. Theriogenology 2011; 77:338-46.e1. [PMID: 21958637 DOI: 10.1016/j.theriogenology.2011.08.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 08/04/2011] [Accepted: 08/04/2011] [Indexed: 12/16/2022]
Abstract
Embryonic stem cells (ESCs) from domestic species have numerous potential applications in agricultural and biomedical sciences; however, despite intensive efforts, derivation of ESCs from sheep remains elusive. The objective was to derive sheep induced pluripotent stem cells (iPSCs), as an alternative pluripotent cell type to ESCs, from sheep fibroblasts by ectopic expression of heterologous transcription factors OCT4, SOX2, KLF4, and cMYC. Sheep fibroblasts were infected with pantropic retroviruses coding the four transcription factors and reprogrammed to pluripotency at a rate of 0.002%. The sheep iPSCs (siPSCs) reactivated endogenous OCT4 and SOX2 genes assessed by qRT-PCR and immuno-cytochemistry, retained normal karyotyping, and more importantly, concurrently silenced all exogenous transgenes. The siPSCs were enzymatically dissociated to single cells, making them amenable to efficient transfection and fluorescent-activated cell sorting techniques. Further, the siPSCs differentiated in vitro to form embryoid bodies, and in vivo to form robust teratomas, containing cells representative of the three germ layers. Moreover, when injected into diploid or tetraploid sheep embryos, siPSCs contributed to the inner cell mass of resulting blastocysts, suggesting true pluripotential. These reprogrammed siPSCs may constitute a robust pluripotent alternative to elusive sheep ESCs, with great potential for use in agriculture and pharmaceutical biotechnology.
Collapse
Affiliation(s)
- Jun Liu
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia
| | | | | | | | | | | |
Collapse
|
9
|
Rowe HM, Trono D. Dynamic control of endogenous retroviruses during development. Virology 2011; 411:273-87. [PMID: 21251689 DOI: 10.1016/j.virol.2010.12.007] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 02/07/2023]
Abstract
Close to half of the human genome encompasses mobile genetic elements, most of which are retrotransposons. These genetic invaders are formidable evolutionary forces that have shaped the architecture of the genomes of higher organisms, with some conserving the ability to induce new integrants within their hosts' genome. Expectedly, the control of endogenous retroviruses is tight and multi-pronged. It is most crucially established in the germ line and during the first steps of embryogenesis, primarily through transcriptional mechanisms that have likely evolved under their very pressure, but are now engaged in controlling gene expression at large, notably during early development.
Collapse
Affiliation(s)
- Helen M Rowe
- National Program, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | |
Collapse
|
10
|
Matsukage S, Kosugi I, Kawasaski H, Miura K, Kitani H, Tsutsui Y. Mouse embryonic stem cells are not susceptible to cytomegalovirus but acquire susceptibility during differentiation. ACTA ACUST UNITED AC 2009; 76:115-25. [PMID: 16470616 DOI: 10.1002/bdra.20233] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) is the most significant infectious cause of congenital anomalies of the central nervous system caused by intrauterine infection in humans. The timing of infection and the susceptibility of cells in early gestational stages are not well understood. In this study we investigated the susceptibility of embryonic stem (ES) cells to CMV infection during differentiation. METHODS ES cell lines were established from transgenic mice integrated with the murine CMV (MCMV) immediate-early (IE) promoter connected with a reporter lacZ gene. The susceptibility of the ES cells was analyzed in terms of viral gene expression and viral replication after induction of differentiation. RESULTS ES cells were nonpermissive to MCMV infection in the undifferentiated state. Upon differentiation, permissive cells appeared approximately 2 weeks after the leukemia inhibitory factor was removed. Upon neural differentiation by retinoic acid (RA), glial cells showed specific susceptibility in terms of expression of the viral antigen. The MCMV IE promoter was not activated in ES cells from the transgenic mice. Activation of the IE promoter was detected approximately 2 weeks after induction of differentiation and observed predominantly in glial cells. Upon MCMV infection of the ES cells, viral infection was correlated with the activation of the IE promoter. CONCLUSIONS ES cells are nonpermissive to MCMV infection and acquire permissiveness about 2 weeks after induction of differentiation, especially in glial cells. Acquisition of permissiveness in differentiated ES cells may be associated with activation of the IE promoter.
Collapse
Affiliation(s)
- Shoichi Matsukage
- Department of Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
Retroviruses are highly successful intracellular parasites, and as such they are found in nearly all branches of life. Some are relatively benign, but many are highly pathogenic and can cause either acute or chronic diseases. Therefore, there is tremendous selective pressure on the host to prevent retroviral replication, and for this reason cells have evolved a variety of restriction factors that act to inhibit or block the viruses. This review is a survey of the best-characterized restriction factors capable of inhibiting retroviral replication and aims to highlight the diversity of strategies used for this task.
Collapse
Affiliation(s)
- Daniel Wolf
- HHMI, Department of Biochemistry, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
12
|
TRIM28 mediates primer binding site-targeted silencing of Lys1,2 tRNA-utilizing retroviruses in embryonic cells. Proc Natl Acad Sci U S A 2008; 105:12521-6. [PMID: 18713861 DOI: 10.1073/pnas.0805540105] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Murine leukemia viruses (MLVs) and related retroelements are potently restricted in embryonic cells by postintegration transcriptional silencing, likely to protect the germ line from insertional mutagenesis. This silencing is in large part attributable to the presence of a nuclear repression complex, which targets a sequence element of the proviral DNA, the repressor-binding site. The repressor-binding site closely overlaps the tRNA primer binding site, a highly conserved sequence essential for virus replication and defining the site of initiation of DNA synthesis during reverse transcription. We have recently demonstrated that the cellular corepressor TRIM28 is recruited to the proline tRNA primer-binding site used by many MLVs and is required to mediate this silencing. Here, we show that TRIM28 is also required for the restriction of retroviruses using a completely distinct tRNA for the priming of their DNA synthesis, namely Lys-1,2 tRNA. These results generalize the role of TRIM28 in retroviral restriction and suggest that this system has evolved to restrict multiple retroviruses.
Collapse
|
13
|
TRIM28 mediates primer binding site-targeted silencing of murine leukemia virus in embryonic cells. Cell 2008; 131:46-57. [PMID: 17923087 DOI: 10.1016/j.cell.2007.07.026] [Citation(s) in RCA: 276] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 06/07/2007] [Accepted: 07/16/2007] [Indexed: 12/22/2022]
Abstract
Moloney murine leukemia virus (M-MLV) replication is restricted in embryonic carcinoma (EC) and embryonic stem (ES) cells, likely to protect the germ line from insertional mutagenesis. Proviral DNAs are potently silenced at the level of transcription in these cells. This silencing is largely due to an unidentified trans-acting factor that is thought to bind to the primer binding site (PBS) of M-MLV and repress transcription from the viral promoter. We have partially purified a large PBS-mediated silencing complex and identified TRIM28 (Kap-1), a known transcriptional silencer, as an integral component of the complex. We show that RNAi-mediated knockdown of TRIM28 in EC and ES cells relieves the restriction and that TRIM28 is bound to the PBS in vivo when restriction takes place. The identification of TRIM28 as a retroviral silencer adds to the growing body of evidence that many TRIM family proteins are involved in retroviral restriction.
Collapse
|
14
|
Primer binding site-dependent restriction of murine leukemia virus requires HP1 binding by TRIM28. J Virol 2008; 82:4675-9. [PMID: 18287239 DOI: 10.1128/jvi.02445-07] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
TRIM28 is a transcriptional corepressor which is required for primer binding site (PBS)-dependent restriction of murine leukemia virus (MLV) replication in embryonic stem and embryonic carcinoma (EC) cells. PBS-dependent restriction of MLV leads to transcriptional silencing of the integrated provirus and has been shown to correlate with TRIM28-mediated recruitment of HP1 to the silenced loci. Here we show, using a cell line with a point mutation in the HP1 binding domain of TRIM28, that interaction with HP1 is absolutely required for the PBS-dependent restriction of MLV in the F9 EC cell line.
Collapse
|
15
|
Jahid S, Bundy LM, Granger SW, Fan H. Chimeras between SRS and Moloney murine leukemia viruses reveal novel determinants in disease specificity and MCF recombinant formation. Virology 2006; 351:7-17. [PMID: 16616947 DOI: 10.1016/j.virol.2006.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Revised: 01/19/2006] [Accepted: 03/06/2006] [Indexed: 11/20/2022]
Abstract
SRS 19-6 MuLV is a murine retrovirus originally isolated in mainland China. A noteworthy feature of this virus (referred to as SRS MuLV here) induces tumors of multiple hematopoietic lineages, including myeloid, erythroid, T-lymphoid and B-lymphoid. To identify the determinants of disease specificity, chimeras between SRS and Moloney MuLV (M-MuLV) were generated by molecular cloning, and the pathogenic properties of the chimeras were investigated. The results indicated that, while the M-MuLV LTR can confer lymphoid specificity to SRS MuLV, the SRS LTR by itself was not sufficient to confer multiple lineage tumorigenesis to M-MuLV; additional sequences in gag or pol were also required. Thus, a secondary determinant for myeloid/erythroid leukemia in SRS MuLV is located in gag-pol. In these chimeras, an independent determinant for T-lymphoma was found in M-MuLV gag-pol. It was also interesting that insertion of M-MuLV env into SRS MuLV decreased the rate of leukemogenicity, while insertion of SRS env into M-MuLV (SEM) accelerated leukemogenesis. The enhanced pathogenicity of SEM was found to correlate with earlier formation of MCF recombinants. The basis for the accelerated MCF recombinant formation was investigated. The endogenous polytropic MuLV env sequences contributing to several SEM MCF recombinants were identified, and the cross-over points were identified. While no obvious differences in the relative homologies between SRS MuLV env and polytropic env vs. M-MuLV and polytropic envs suggested a reason for the more rapid MCF recombinant formation, an overlapping but different set of polytropic env proviruses were found to participate in MCF formation for M-MuLV vs. SEM. Thus, the mechanisms for MCF formation appear to differ for M-MuLV and SEM.
Collapse
Affiliation(s)
- Sohail Jahid
- Cancer Research Institute, Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | | | | | | |
Collapse
|
16
|
McGee-Estrada K, Palmarini M, Hallwirth C, Fan H. A Moloney murine leukemia virus driven by the Jaagsiekte sheep retrovirus enhancers shows enhanced specificity for infectivity in lung epithelial cells. Virus Genes 2005; 31:257-63. [PMID: 16175331 DOI: 10.1007/s11262-005-3239-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Accepted: 03/29/2005] [Indexed: 10/25/2022]
Abstract
Jaagsiekte sheep retrovirus (JSRV) is the etiologic agent of ovine pulmonary adenocarcinoma (OPA), a transmissible lung cancer in sheep. One of the unique features of this virus is that in infected animals, the only tissues that show expression of the virus are the tumor cells in the lung. We previously showed that the JSRV long terminal repeat (LTR) is preferentially active in murine lung epithelial cell lines (MLE-15 and mtCC1-2). To further explore the tissue specificity, we inserted the JSRV enhancer sequences from the U3 region of the LTR into a Moloney murine leukemia virus (M-MuLV) LTR lacking its own enhancer sequences, to give the chimeric LTR DeltaMo + JS. Transient transfection assays indicated that the DeltaMo + JS LTR is > 5-fold more active in lung epithelial cell lines than in non-lung lines, compared to the wild-type M-MuLV LTR. This was due to preferential activity of the JSRV enhancers in lung epithelial cells. Moreover, M-MuLV driven by the DeltaMo + JS LTR was > 3 logs more infectious in MLE-15 cells compared to non-lung cell lines. This chimeric virus may facilitate investigations of the tissue-specificity of JSRV.
Collapse
Affiliation(s)
- Kathleen McGee-Estrada
- Department of Molecular Biology and Biochemistry and Cancer Research Institute, University of California, Irvine, CA, 92697-3905, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Transgenic animal technology is one of the most fascinating technologies developed in the last two decades. It allows us to address questions in life sciences that no other methods have achieved. The impact on biomedical and biological research, as well as commercial interests are overwhelming. The questions accompanying this fast growing technology and its diversified applications attract the attention from a variety of entities. Still, one of the most fundamental problems remaining is the search for an efficient and reliable gene delivery system for creating transgenic animals. The traditional method of pronuclear microinjection has displayed great variability in success among species. While an acceptable efficiency in the production of transgenic mice has been attained, the relative low efficiency (<1%) in creating transgenic livestock has become one of the barriers for its application. In the past decades, improvements in producing transgenic livestock have made a slow progression, however, the recent advancement in cloning technology and the ability to create transgenic livestock in a highly efficient manner, have opened the gate to a new era in transgenic technology. Discoveries of new gene delivery systems have created an enthusiastic atmosphere that has made this technology so unique. This review focuses on gene delivery strategies as well as various approaches that may assist the advancement of transgenic efficiency in large animals.
Collapse
Affiliation(s)
- A W Chan
- Oregon Regional Primate Research Center, Oregon Health Sciences University, Beaverton, Oregon 97006, USA.
| |
Collapse
|
18
|
Amit M, Winkler ME, Menke S, Brüning E, Büscher K, Denner J, Haverich A, Itskovitz-Eldor J, Martin U. No evidence for infection of human embryonic stem cells by feeder cell-derived murine leukemia viruses. Stem Cells 2005; 23:761-71. [PMID: 15917472 DOI: 10.1634/stemcells.2004-0046] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Until recently, culture and expansion of nondifferentiated human embryonic stem cells (hESCs) depended on coculture with murine embryonic fibroblasts. Because mice are known to harbor a variety of pathogens, such culture conditions implicate the risk of xenozoonoses. Among these pathogens, endogenous retroviruses, including murine leukemia viruses (MuLVs), are of special importance. It is well known that some strains cause pathogenic (e.g., leukemic) effects and that xenotropic, polytropic, and amphotropic MuLVs are able to infect human cells. In view of potential clinical applications of hESC lines, it is therefore imperative to investigate potential infection of hESCs by mouse feeder cell-derived viruses. As a first step towards a comprehensive infection risk assessment, we have analyzed embryonic fibroblasts derived from different mouse strains for expression and release of xenotropic, polytropic, and amphotropic MuLVs. Moreover, several hESC lines have been investigated for expression of specific receptors for xenotropic/polytropic MuLVs, as well as for MuLV infection and expression. Evidence for expression of humantropic MuLVs was found in cultures of mouse embryonic fibroblasts (MEFs). Moreover, expression of specific receptors for xenotropic/ polytropic MuLV on human HEK293 and hESC lines and infection after coculture with an MuLV-producing mink cell line could be demonstrated. In contrast, no evidence of MuLV transmission from MEFs to human HEK293 cells or to the hESC lines I-3, I-6, I-8, and H-9 has been obtained. Our results suggest that recently established hESC lines are free of MuLV infections despite long-term close contact with MEFs.
Collapse
Affiliation(s)
- Michal Amit
- Department of Obstetrics and Gynecology, Rambam Medical Center, Faculty of Medicine, The Technion, Haifa, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yamada M, Onodera M, Mizuno Y, Mochizuki H. Neurogenesis in olfactory bulb identified by retroviral labeling in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated adult mice. Neuroscience 2004; 124:173-81. [PMID: 14960349 DOI: 10.1016/j.neuroscience.2003.10.040] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2003] [Indexed: 11/26/2022]
Abstract
Neurogenesis occurs during development and in the normal adult brain. Recent studies identified areas exhibiting postlesional selective neurogenesis and neuronal repair. In the olfactory bulb (OB), one of the most studied regions of the brain for neurogenesis, seizures and strong odor exposure are known to enhance neurogenesis. Here, we report enhanced neurogenesis in OB after dopaminergic neuronal loss induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a selective toxin for dopaminergic neurons. The neurogenesis has been previously confirmed mainly by the uptake of 5-bromodeoxyuridine (BrdU), a marker of proliferating cells, but methodological problems related to BrdU labeling might result in inaccurate findings with respect to specificity, toxicity and incorporation into normal/lesioned brain. For a better identification of neurogenesis, we used a retroviral vector. First, we investigated the population dynamics of newly formed neurons in different regions of OB including the glomerular layer, the most superficial layer of OB. Quantification of neurogenesis in OB revealed by our retroviral vector was substantially similar to that by BrdU-based method. One week after MPTP application and dopaminergic neuronal loss in OB, neurogenesis of dopaminergic neurons in OB increased by three-fold, but no such process was noted in non-dopaminergic neurons. Our results indicate selective dopaminergic neurogenesis in OB in response to neuronal damage/loss.
Collapse
Affiliation(s)
- M Yamada
- Research Institute for Diseases of Old Ages, Juntendo University, Tokyo, Japan
| | | | | | | |
Collapse
|
20
|
Haas DL, Lutzko C, Logan AC, Cho GJ, Skelton D, Jin Yu X, Pepper KA, Kohn DB. The Moloney murine leukemia virus repressor binding site represses expression in murine and human hematopoietic stem cells. J Virol 2003; 77:9439-50. [PMID: 12915559 PMCID: PMC187403 DOI: 10.1128/jvi.77.17.9439-9450.2003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Moloney murine leukemia virus (MLV) repressor binding site (RBS) is a major determinant of restricted expression of MLV in undifferentiated mouse embryonic stem (ES) cells and mouse embryonal carcinoma (EC) lines. We show here that the RBS repressed expression when placed outside of its normal MLV genome context in a self-inactivating (SIN) lentiviral vector. In the lentiviral vector genome context, the RBS repressed expression of a modified MLV long terminal repeat (MNDU3) promoter, a simian virus 40 promoter, and three cellular promoters: ubiquitin C, mPGK, and hEF-1a. In addition to repressing expression in undifferentiated ES and EC cell lines, we show that the RBS substantially repressed expression in primary mouse embryonic fibroblasts, primary mouse bone marrow stromal cells, whole mouse bone marrow and its differentiated progeny after bone marrow transplant, and several mouse hematopoietic cell lines. Using an electrophoretic mobility shift assay, we show that binding factor A, the trans-acting factor proposed to convey repression by its interaction with the RBS, is present in the nuclear extracts of all mouse cells we analyzed where expression was repressed by the RBS. In addition, we show that the RBS partially repressed expression in the human hematopoietic cell line DU.528 and primary human CD34(+) CD38(-) hematopoietic cells isolated from umbilical cord blood. These findings suggest that retroviral vectors carrying the RBS are subjected to high rates of repression in murine and human cells and that MLV vectors with primer binding site substitutions that remove the RBS may yield more-effective gene expression.
Collapse
Affiliation(s)
- Dennis L Haas
- Division of Research Immunology/BMT, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ploss M, Berdel B, Heber R, Reuss FU. Transcriptional inactivation of amphotropic murine leukemia virus replication in human cells. J Med Virol 2003; 69:267-72. [PMID: 12683417 DOI: 10.1002/jmv.10274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Amphotropic murine leukemia virus (MLV) replicates in cells from various mammalian species including humans and is a potential contaminant in MLV vector preparations for human gene transfer studies. Because MLV replication proceeds through an RNA genome that is generated under the control of viral enhancer and promoter elements, vectors were developed that delete such elements during transduction to reduce the generation of replication-competent virus. It was shown recently that replication of amphotropic MLV in certain human cells is possible without the 75 bp transcription enhancers. It is now demonstrated that enhancer-independent replication requires functional elements within U3 and is repressed by an extended deletion in the U3 region comprising enhancers, promoter and flanking sequences. It is concluded that the transcriptional inactivation of amphotropic MLV in human cells requires the combined deletion of enhancers and of additional elements in U3.
Collapse
Affiliation(s)
- Martin Ploss
- Deutsches Krebsforschungszentrum (DKFZ), Angewandte Tumorvirologie F0400, Heidelberg, Germany
| | | | | | | |
Collapse
|
22
|
Reuss FU, Berdel B, Heber R, Bantel-Schaal U. Replication of enhancer-deficient amphotropic murine leukemia virus in human fibrosarcoma but not in primary human fibroblasts. J Med Virol 2002; 68:278-84. [PMID: 12210420 DOI: 10.1002/jmv.10202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Amphotropic murine leukemia virus (MLV) replicates in cells from various mammalian species including humans and is a potential contaminant in MLV vector preparations for human gene transfer studies. In general, MLV replication depends on the expression of viral genes under the control of 75 bp enhancer elements in the long terminal repeat. However, in specific human fibrosarcoma and lymphoma lines replication of amphotropic MLV is possible without these enhancers. Fibrosarcomas are malignant tumors of fibroblast origin. To test the replication potential of intact and enhancerless amphotropic MLV in untransformed cells, infection studies with these viruses were carried out in three types of primary human fibroblasts. Replication of amphotropic MLV is observed in two of three tested fibroblast strains. None of these primary human fibroblasts is permissive for enhancer-deficient MLV, suggesting that replication of this virus may be limited to transformed cells.
Collapse
Affiliation(s)
- Frank U Reuss
- Deutsches Krebsforschungszentrum, Angewandte Tumorvirologie F0400, Heidelberg, Germany.
| | | | | | | |
Collapse
|
23
|
Prasad Alur RK, Foley B, Parente MK, Tobin DK, Heuer GG, Avadhani AN, Pongubala J, Wolfe JH. Modification of multiple transcriptional regulatory elements in a Moloney murine leukemia virus gene transfer vector circumvents silencing in fibroblast grafts and increases levels of expression of the transferred enzyme. Gene Ther 2002; 9:1146-54. [PMID: 12170378 DOI: 10.1038/sj.gt.3301777] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2002] [Accepted: 03/07/2002] [Indexed: 11/09/2022]
Abstract
Down-regulation of retroviral vector expression occurs in a number of cell types after transplantation. Although a number of vector elements have been shown to affect expression in specific experimental situations, the results can vary depending on the specific cDNA being expressed, the individual retroviral elements included in vectors, the promoter, or the inclusion of selectable markers. In previous experiments with the lysosomal enzyme beta-glucuronidase, silencing has occurred in more than 95% of transduced cells regardless of the position of the expression unit within the vector, whether a eukaryotic or viral promoter was used, whether a bacterial selectable marker gene was present or not, the target cell type, or the species of the host. It has been a consistent finding that a small number of continuously expressing cells persist for long periods after transplantation. In this study we found that deletion of all the transcriptional regulatory elements from the vector LTR, inclusion of a permissive primer binding site sequence, and use of a eukaryotic housekeeping promoter could greatly increase the number of expressing cells in fibroblast grafts in subcutaneous neo-organs and in the brain. Furthermore, the level of enzyme expression was increased five-fold on a per positive cell basis, indicating that the deleted regulatory elements were exerting a negative effect on expression in the few cells that were positive before modification of the vector. This resulted in more than a 50-fold increase in total activity compared with the previous highest expressing vector.
Collapse
Affiliation(s)
- R K Prasad Alur
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Reuss FU, Berdel B, Heber R, Ploss M. Enhancer-deficient amphotropic murine leukemia virus and recombinants with heterologous transcription elements can be efficiently amplified and detected in Mus dunni fibroblasts. Gene Ther 2002; 9:1183-8. [PMID: 12170382 DOI: 10.1038/sj.gt.3301785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2001] [Accepted: 03/23/2002] [Indexed: 11/09/2022]
Abstract
Amphotropic murine leukemia virus (MLV) replicates in cells from various mammalian species, including humans, and is a potential contaminant in MLV vector preparations for human gene transfer studies. Mus dunni fibroblasts are routinely used for amplification and detection of contaminating virus. We have recently characterized an amphotropic MLV mutant lacking the 75-bp viral enhancer elements and spontaneous MLV-(RCMV) recombinants that have acquired cytomegalovirus (CMV) transcription elements. Both of these viruses replicate in specific human cell types. To test whether the formation of such viruses can be detected and controlled with current routine procedures, we have analyzed the replication of these amphotropic MLV mutants in Mus dunni fibroblasts. We find that M. dunni cells are permissive for enhancer-deficient and CMV promoter-recombinant MLV from several human cell lines. Thus, M. dunni fibroblasts are suitable for the amplification and subsequent detection of enhancer-deficient and enhancer-recombinant MLV in vector preparations.
Collapse
Affiliation(s)
- F U Reuss
- Deutsches Krebsforschungszentrum (DKFZ), Angewandte Tumorvirologie F0400, Heidelberg, Germany
| | | | | | | |
Collapse
|
25
|
Suzuki A, Obi K, Urabe T, Hayakawa H, Yamada M, Kaneko S, Onodera M, Mizuno Y, Mochizuki H. Feasibility of ex vivo gene therapy for neurological disorders using the new retroviral vector GCDNsap packaged in the vesicular stomatitis virus G protein. J Neurochem 2002; 82:953-60. [PMID: 12358801 DOI: 10.1046/j.1471-4159.2002.01048.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal progenitor cells (NPC) are particularly suited as the target population for genetic and cellular therapy of neurological disorders such as Parkinson's disease or stroke. However, genetic modification of these cells using retroviral vectors remains a great challenge because of the low transduction rate and the need for fetal calf serum (FCS) during the transduction process that induces the cell differentiation to mature neurons. To overcome these problems, we developed a new retrovirus production system in which the simplified retroviral vector GCDNsap engineered to be resistant to denovo methylation was packaged in the vesicular stomatitis virus G protein (VSV-G), concentrated by centrifugation, and resuspended in serum-free medium (StemPro-34 SFM). In transduction experiments using enhanced green fluorescent protein (EGFP) as a marker, the concentrated FCS-free virus supernatant infected NPC at a high rate, while maintaining the ability of these cells to self-renew and differentiate in vitro. When such cells were grafted into mouse brains, EGFP-expressing NPC were detected in the region around the injection site at 8 weeks post transplantation. These findings suggest that the gene transfer system described here may provide a useful tool to genetically modify NPC for treatments of neurological disorders.
Collapse
Affiliation(s)
- A Suzuki
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Swindle CS, Klug CA. Mechanisms that regulate silencing of gene expression from retroviral vectors. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:449-56. [PMID: 12183830 DOI: 10.1089/15258160260090915] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The propensity of retroviruses toward transcriptional silencing limits their value as gene therapy vectors. Silencing has been shown to be particularly robust when stem cells are used for transduction, posing a significant problem for gene therapy of hematologic diseases. Stability of proviral expression with newer generation vectors is significantly improved over that obtainable with original vectors based on Moloney murine leukemia virus (MoMLV). However, strategies to increase resistance further to retroviral silencing are needed, because newer generation vectors have been shown to remain prone to a significant degree of silencing that could limit their efficacy as gene therapy vectors. Proviral silencing has been attributed to known mechanisms of cellular gene repression, such as DNA methylation and histone modification, as well as uncharacterized mechanisms that act independently of DNA methylation. A further understanding of transcriptional silencing that occurs in stem cells and during hematopoietic development is needed for design of effective vectors for gene therapy of hematologic diseases.
Collapse
Affiliation(s)
- C Scott Swindle
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294-3300, USA
| | | |
Collapse
|
27
|
Reuss FU, Heber R, Ploss A, Berdel B. Amphotropic murine leukemia virus replication in human mammary epithelial cells and the formation of cytomegalovirus-promoter recombinants. Virology 2001; 291:91-100. [PMID: 11878879 DOI: 10.1006/viro.2001.1199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amphotropic murine leukemia virus (MLV) can replicate in human cells and is a potential contaminant in vector preparations for human gene transfer studies. We have recently shown that replication of amphotropic MLV in specific human sarcoma and lymphoma lines is possible in the absence of the viral 75-bp transcription enhancer elements. Here, we have tested the replication of an amphotropic MLV, MLV-(MOA), and an enhancer-deficient mutant of this virus in human breast carcinoma-derived cell lines. The proviral expression plasmids use a cytomegalovirus (CMV) promoter for the initial transcription of virus RNA. We found that all cells analyzed are permissive for replication of MLV-(MOA). Enhancer-deficient virus is unable to replicate. However, in two lines the replication defect can be rescued by the spontaneous insertion of a CMV promoter and enhancer into the U3 region. This recombinant virus MLV-(RCMV) replicates with kinetics similar to that of MLV-(MOA) but is restricted to specific cell lines. The potential formation of RCMV recombinants during MLV vector preparation must be considered.
Collapse
Affiliation(s)
- F U Reuss
- Angewandte Tumorvirologie F0400, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 242, Heidelberg, 69120, Germany.
| | | | | | | |
Collapse
|
28
|
Reuss FU, Berdel B, Ploss M, Heber R. Replication of enhancer-deficient amphotropic murine leukemia virus in human cells. Proc Natl Acad Sci U S A 2001; 98:10898-903. [PMID: 11535815 PMCID: PMC58571 DOI: 10.1073/pnas.191182098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Amphotropic murine leukemia virus (MLV) replicates in cells from various mammalian species, including humans, and is a potential contaminant in MLV vector preparations for human gene transfer studies. The generation of replication-competent virus is considered less likely with vectors that delete the viral transcription elements. This conclusion is based on data obtained in rodents, where MLV replication depends on the expression of viral genes under the control of 75-bp enhancer elements in the long terminal repeat. We demonstrate here that in some human cells replication of amphotropic MLV is possible in the absence of these enhancer elements. Replication of the enhancer-deficient virus MLV-(MOA)Delta E is observed in selected human sarcoma and B lymphoma lines and proceeds at a lower rate than that of the intact virus. No insertion of a foreign promoter or enhancer into the long terminal repeat was detected. Our data suggest the presence of a secondary enhancer element within the MLV provirus that can in selected human cells mediate virus transcription and replication in the absence of the 75-bp U3 enhancers.
Collapse
Affiliation(s)
- F U Reuss
- Deutsches Krebsforschungszentrum, Angewandte Tumorvirologie F0400, 69120 Heidelberg, Germany.
| | | | | | | |
Collapse
|
29
|
Modin C, Pedersen FS, Duch M. Lack of shielding of primer binding site silencer-mediated repression of an internal promoter in a retrovirus vector by the putative insulators scs, BEAD-1, and HS4. J Virol 2000; 74:11697-707. [PMID: 11090169 PMCID: PMC112452 DOI: 10.1128/jvi.74.24.11697-11707.2000] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A major determinant for transcriptional incompetence of murine leukemia virus (MLV) and MLV-derived vectors in embryonal cells is located at the proline primer binding site (PBS). The mechanism of silencing is unknown, yet the effect is capable of spreading to adjacent promoters. Based on a retroviral vector containing an internal promoter and the escape mutant B2 PBS with expressional capacity in embryonal cells, we have developed an assay to test the ability of putative insulators to shield the silencer at the PBS. Since the B2 PBS reverts to the wild-type PBS at high frequency, a shielding ability of a putative insulator can be assessed from the ratio of expressing B2 PBS to proline PBS proviruses in the target embryonal carcinoma cell population as measured by primer extension. Our results show that none of the possible insulators, scs, BEAD-1, or HS4, is able to shield an internal promoter from the repressive effect of the silencer at the PBS region when inserted between the silencer and the promoter.
Collapse
Affiliation(s)
- C Modin
- Department of Molecular and Structural Biology, University of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | |
Collapse
|
30
|
Zentilin L, Qin G, Tafuro S, Dinauer MC, Baum C, Giacca M. Variegation of retroviral vector gene expression in myeloid cells. Gene Ther 2000; 7:153-66. [PMID: 10673720 DOI: 10.1038/sj.gt.3301057] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We have comparatively evaluated the efficiency of a series of retroviral vectors transducing the gp91-phox gene, whose defects are responsible for impaired production of superoxide anion (O2-) by phagocytic cells and lead to the X-linked form of chronic granulomatous disease (X-CGD). These vectors included four constructs based on the MoMuLV backbone and expressing gp91-phox from the viral long terminal repeat (LTR) or from internal promoters, and one construct based on the myelotropic FMEV vector. Expression of the therapeutic gene from the MoMuLV LTR was unsatisfactory after transduction of the PLB985 X-CGD knockout cell line and of primary CD34+ hematopoietic progenitors from X-CGD patients. The presence of either constitutive or inducible internal promoters did not result in important improvements in the efficiency of O2- production and lowered the titers of the viral preparations. In contrast, sustained levels of superoxide generation were obtained upon transduction with the FMEV vector. To analyze the efficiency of transgene expression at the single cell level, over 150 cellular clones were generated from bulk cultures of PLB985 X-CGD cells transduced with this vector, each one representative of an individual transduction event. These clones revealed a markedly heterogeneous pattern of gp91-phox expression, ranging from complete silencing to full restoration of superoxide production. Within each clone, expression of the therapeutic gene correlated with the number of expressing cells rather than with the average levels of expression from each cell, indicating that at the single cell level, the proviral promoter is regulated by a binary, on/off mechanism. Moreover, both transduced bulk and clonal cell populations displayed a tendency to a progressive extinction of expression over time, with a mechanism involving LTR methylation. The design of novel retroviral vectors escaping silencing is highly desirable for efficient gene therapy.
Collapse
Affiliation(s)
- L Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
This article reviews 1) the use of gene transfer methods to genetically manipulate hematopoietic stem cell targets, 2) recent advances in technology that are addressing problems that have prevented widespread successful translation of gene transfer approaches for the cure of disease, and 3) recent regulatory issues related to human gene therapy trials.In Section I, Dr. Nienhuis describes the use of alternative viral envelopes and vector systems to improve efficiency of transduction of hematopoietic stem cells. Major limitations of stem cell transduction are related to low levels of viral receptors on the stem cells of large animal species and the low frequency of cycling stem cells in the bone marrow. Attempts to circumvent these limitations by exploiting non-oncoretroviral vectors and pseudotyping of Moloney vectors with alternative envelopes are discussed.In Section II, Dr. Hawley addresses new strategies to improve the expression of transgenes in cells derived from long-term reconstituting hematopoietic stem cells. Transgene silencing in transduced hematopoietic stem cells remains an obstacle to gene therapy for some gene sequences. New generations of retroviral backbones designed to both improve expression and reduce silencing in primary cells are explored.In Section III, Drs. Smith and Cornetta update regulatory issues related to human gene therapy trials. Increased scrutiny of human trials has led to changes in requirements and shifts in emphasis of existing regulations, which apply to human gene therapy trials. The current Food and Drug Administration's structure and regulations and the roles of the Recombinant DNA Advisory Committee of the NIH and other sponsors and partners in gene therapy trials are reviewed.
Collapse
|
32
|
Abstract
Abstract
This article reviews 1) the use of gene transfer methods to genetically manipulate hematopoietic stem cell targets, 2) recent advances in technology that are addressing problems that have prevented widespread successful translation of gene transfer approaches for the cure of disease, and 3) recent regulatory issues related to human gene therapy trials.
In Section I, Dr. Nienhuis describes the use of alternative viral envelopes and vector systems to improve efficiency of transduction of hematopoietic stem cells. Major limitations of stem cell transduction are related to low levels of viral receptors on the stem cells of large animal species and the low frequency of cycling stem cells in the bone marrow. Attempts to circumvent these limitations by exploiting non-oncoretroviral vectors and pseudotyping of Moloney vectors with alternative envelopes are discussed.
In Section II, Dr. Hawley addresses new strategies to improve the expression of transgenes in cells derived from long-term reconstituting hematopoietic stem cells. Transgene silencing in transduced hematopoietic stem cells remains an obstacle to gene therapy for some gene sequences. New generations of retroviral backbones designed to both improve expression and reduce silencing in primary cells are explored.
In Section III, Drs. Smith and Cornetta update regulatory issues related to human gene therapy trials. Increased scrutiny of human trials has led to changes in requirements and shifts in emphasis of existing regulations, which apply to human gene therapy trials. The current Food and Drug Administration's structure and regulations and the roles of the Recombinant DNA Advisory Committee of the NIH and other sponsors and partners in gene therapy trials are reviewed.
Collapse
|
33
|
Linney E, Hardison NL, Lonze BE, Lyons S, DiNapoli L. Transgene expression in zebrafish: A comparison of retroviral-vector and DNA-injection approaches. Dev Biol 1999; 213:207-16. [PMID: 10452858 DOI: 10.1006/dbio.1999.9376] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To assess alternative methods for introducing expressing transgenes into the germ line of zebrafish, transgenic fish that express a nuclear-targeted, enhanced, green fluorescent protein (eGFP) gene were produced using both pseudotyped retroviral vector infection and DNA microinjection of embryos. Germ-line transgenic founders were identified and the embryonic progeny of these founders were evaluated for the extent and pattern of eGFP expression. To compare the two modes of transgenesis, both vectors used the Xenopus translational elongation factor 1-alpha enhancer/promoter regulatory cassette. Several transgenic founder fish which transferred eGFP expression to their progeny were identified. The gene expression patterns are described and compared for the two modes of gene transfer. Transient expression of eGFP was detected 1 day after introducing the transgenes via either DNA microinjection or retroviral vector infection. In both cases of gene transfer, transgenic females produced eGFP-positive progeny even before the zygotic genome was turned on. Therefore, GFP was being provided by the oocyte before fertilization. A transgenic female revealed eGFP expression in her ovarian follicles. The qualitative patterns of gene expression in the transgenic progeny embryos after zygotic induction of gene expression were similar and independent of the mode of transgenesis. The appearance of newly synthesized GFP is detectable within 5-7 h after fertilization. The variability of the extent of eGFP expression from transgenic founder to transgenic founder was wider for the DNA-injection transgenics than for the retroviral vector-produced transgenics. The ability to provide expressing germ-line transgenic progeny via retroviral vector infection provides both an alternative mode of transgenesis for zebrafish work and a possible means of easily assessing the insertional mutagenesis frequency of retroviral vector infection of zebrafish embryos. However, because of the transfer of GFP from oocyte to embryo, the stability of GFP may create problems of analysis in embryos which develop as quickly as those of zebrafish.
Collapse
Affiliation(s)
- E Linney
- Department of Microbiology, Duke University Medical Center, Duke University, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
34
|
Granger SW, Bundy LM, Fan H. Tandemization of a subregion of the enhancer sequences from SRS 19-6 murine leukemia virus associated with T-lymphoid but not other leukemias. J Virol 1999; 73:7175-84. [PMID: 10438804 PMCID: PMC104241 DOI: 10.1128/jvi.73.9.7175-7184.1999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most simple retroviruses induce tumors of a single cell type when infected into susceptible hosts. The SRS 19-6 murine leukemia virus (MuLV), which originated in mainland China, induces leukemias of multiple cellular origins. Indeed, infected mice often harbor more than one tumor type. Since the enhancers of many MuLVs are major determinants of tumor specificity, we tested the role of the SRS 19-6 MuLV enhancers in its broad disease specificity. The enhancer elements of the Moloney MuLV (M-MuLV) were replaced by the 170-bp enhancers of SRS 19-6 MuLV, yielding the recombinants DeltaMo+SRS(+) and DeltaMo+SRS(-) M-MuLV. M-MuLV normally induces T-lymphoid tumors in all infected mice. Surprisingly, when neonatal mice were inoculated with DeltaMo+SRS(+) or DeltaMo+SRS(-) M-MuLV, all tumors were of T-lymphoid origin, typical of M-MuLV rather than SRS 19-6 MuLV. Thus, the SRS 19-6 MuLV enhancers did not confer the broad disease specificity of SRS 19-6 MuLV to M-MuLV. However, all tumors contained DeltaMo+SRS M-MuLV proviruses with common enhancer alterations. These alterations consisted of tandem multimerization of a subregion of the SRS 19-6 enhancers, encompassing the conserved LVb and core sites and adjacent sequences. Moreover, when tumors induced by the parental SRS 19-6 MuLV were analyzed, most of the T-lymphoid tumors had similar enhancer alterations in the same region whereas tumors of other lineages retained the parental SRS 19-6 MuLV enhancers. These results emphasize the importance of a subregion of the SRS 19-6 MuLV enhancer in induction of T-cell lymphoma. The relevant sequences were consistent with crucial sequences for T-cell lymphomagenesis identified for other MuLVs such as M-MuLV and SL3-3 MuLV. These results also suggest that other regions of the SRS 19-6 MuLV genome contribute to its broad leukemogenic spectrum.
Collapse
Affiliation(s)
- S W Granger
- Department of Molecular Biology and Biochemistry and Cancer Research Institute, University of California, Irvine, California 92697-3900, USA
| | | | | |
Collapse
|
35
|
Lander JK, Chesebro B, Fan H. Appearance of mink cell focus-inducing recombinants during in vivo infection by moloney murine leukemia virus (M-MuLV) or the Mo+PyF101 M-MuLV enhancer variant: implications for sites of generation and roles in leukemogenesis. J Virol 1999; 73:5671-80. [PMID: 10364317 PMCID: PMC112626 DOI: 10.1128/jvi.73.7.5671-5680.1999] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
One hallmark of murine leukemia virus (MuLV) leukemogenesis in mice is the appearance of env gene recombinants known as mink cell focus-inducing (MCF) viruses. The site(s) of MCF recombinant generation in the animal during Moloney MuLV (M-MuLV) infection is unknown, and the exact roles of MCF viruses in disease induction remain unclear. Previous comparative studies between M-MuLV and an enhancer variant, Mo+PyF101 MuLV, suggested that MCF generation or early propagation might take place in the bone marrow under conditions of efficient leukemogenesis. Moreover, M-MuLV induces disease efficiently following both intraperitoneal (i.p.) and subcutaneous (s.c.) inoculation but leukemogenicity by Mo+PyF101 M-MuLV is efficient following i.p. inoculation but attenuated upon s. c. inoculation. Time course studies of MCF recombinant appearance in the bone marrow, spleen, and thymus of wild-type and Mo+PyF101 M-MuLV i.p.- and s.c.-inoculated mice were carried out by performing focal immunofluorescence assays. Both the route of inoculation and the presence of the PyF101 enhancer sequences affected the patterns of MCF generation or early propagation. The bone marrow was a likely site of MCF recombinant generation and/or early propagation following i.p. inoculation of M-MuLV. On the other hand, when the same virus was inoculated s.c., the primary site of MCF generation appeared to be the thymus. Also, when Mo+PyF101 M-MuLV was inoculated i.p., MCF generation appeared to occur primarily in the thymus. The time course studies indicated that MCF recombinants are not involved in preleukemic changes such as splenic hyperplasia. On the other hand, MCFs were detected in tumors from Mo+PyF101 M-MuLV s. c.-inoculated mice even though they were largely undetectable at preleukemic times. These results support a role for MCF recombinants late in disease induction.
Collapse
Affiliation(s)
- J K Lander
- Department of Molecular Biology and Biochemistry and Cancer Research Institute, University of California, Irvine, California 92697, USA
| | | | | |
Collapse
|
36
|
Okimoto MA, Fan H. Moloney murine leukemia virus infects cells of the developing hair follicle after neonatal subcutaneous inoculation in mice. J Virol 1999; 73:2509-16. [PMID: 9971836 PMCID: PMC104498 DOI: 10.1128/jvi.73.3.2509-2516.1999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/1998] [Accepted: 11/03/1998] [Indexed: 11/20/2022] Open
Abstract
The nature of Moloney murine leukemia virus (M-MuLV) infection after a subcutaneous (s.c.) inoculation was studied. We have previously shown that an enhancer variant of M-MuLV, Mo+PyF101 M-MuLV, is poorly leukemogenic when used to inoculate mice s.c., but not when inoculated intraperitoneally. This attenuation of leukemogenesis correlated with an inability of Mo+PyF101 M-MuLV to establish infection in the bone marrow of mice at early times postinfection. These results suggested that a cell type(s) is infected in the skin by wild-type but not Mo+PyF101 M-MuLV after s.c. inoculation and that this infection is important for the delivery of infection to the bone marrow, as well as for efficient leukemogenesis. To determine the nature of the cell types infected by M-MuLV and Mo+PyF101 M-MuLV in the skin after a s.c. inoculation, immunohistochemistry with an anti-M-MuLV CA antibody was performed. Cells of developing hair follicles, specifically cells of the outer root sheath (ORS), were extensively infected by M-MuLV after s.c. inoculation. The Mo+PyF101 M-MuLV variant also infected cells of the ORS but the level of infection was lower. By Western blot analysis, the level of infection in skin by Mo+PyF101 M-MuLV was approximately 4- to 10-fold less than that of wild-type M-MuLV. Similar results were seen when a mouse keratinocyte line was infected in vitro with both viruses. Cells of the ORS are a primary target of infection in vivo, since a replication defective M-MuLV-based vector expressing beta-galactosidase also infected these cells after a s.c. inoculation.
Collapse
Affiliation(s)
- M A Okimoto
- Department of Molecular Biology and Biochemistry and Cancer Research Institute, University of California, Irvine, California 92697-3900, USA
| | | |
Collapse
|
37
|
Okimoto MA, Fan H. Identification of directly infected cells in the bone marrow of neonatal moloney murine leukemia virus-infected mice by use of a moloney murine leukemia virus-based vector. J Virol 1999; 73:1617-23. [PMID: 9882368 PMCID: PMC103987 DOI: 10.1128/jvi.73.2.1617-1623.1999] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/1998] [Accepted: 10/17/1998] [Indexed: 11/20/2022] Open
Abstract
Early bone marrow infection of Moloney murine leukemia virus (M-MuLV)-infected mice was studied. Previous experiments indicated that early bone marrow infection is essential for the efficient development of T lymphoma. In order to identify the cellular pathway of infection in the bone marrow, infection of mice with a helper-free replication-defective M-MuLV-based retroviral vector was carried out. Such a vector will undergo only one round of infection, without spreading to other cells; thus, cells infected by the initially injected virus (directly infected cells) can be identified. For these experiments, the BAG vector that expresses bacterial beta-galactosidase was employed. Neonatal NIH/Swiss mice were inoculated intraperitoneally with ca. 10(6) infectious units of a BAG vector pseudotyped with M-MuLV proteins, and bone marrow cells were recovered 2 to 12 days postinfection. Single-cell suspensions were tested for infection by staining with X-Gal (5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside) or by immunofluorescence with an anti-beta-galactosidase antibody. Two sizes of infected cells were evident: large multinucleated cells and small nondescript (presumptively hematopoietic) cells. Secondary stains for lineage-specific markers indicated that the large cells were osteoclasts. Some of the small cells expressed nonspecific esterase, which placed them in the myeloid lineage, but they lacked markers for hematopoietic progenitors (mac-1, gr-1, sca-1, and CD34). These results provide evidence for primary M-MuLV infection of osteoclasts or osteoclast progenitors in the bone marrow, and they suggest that known hematopoietic progenitors are not primary targets for infection. However, the subsequent spread of infection to hematopoietic progenitors was indicated, since bone marrow from mice infected in parallel with replication-competent wild-type M-MuLV showed detectable infection in small cells positive for mac-1 or CD34, as well as in osteoclasts.
Collapse
Affiliation(s)
- M A Okimoto
- Department of Molecular Biology and Biochemistry and Cancer Research Institute, University of California, Irvine, California 92697-3900, USA
| | | |
Collapse
|
38
|
Robbins PB, Skelton DC, Yu XJ, Halene S, Leonard EH, Kohn DB. Consistent, persistent expression from modified retroviral vectors in murine hematopoietic stem cells. Proc Natl Acad Sci U S A 1998; 95:10182-7. [PMID: 9707621 PMCID: PMC21482 DOI: 10.1073/pnas.95.17.10182] [Citation(s) in RCA: 120] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/1998] [Indexed: 01/15/2023] Open
Abstract
Retroviral vectors based on the Moloney murine leukemia virus (MoMuLV) have shown inconsistent levels and duration of expression as well as a propensity for the acquisition of de novo methylation in vivo. MoMuLV-based vectors are known to contain sequences that are capable of suppressing or preventing expression from the long terminal repeat. Previously, we constructed a series of modified retroviral vectors and showed that they function significantly better than MoMuLV-based vectors in vitro. To test the efficacy of the modified vectors in hematopoietic stem cells in vivo, we examined gene expression and proviral methylation in differentiated hematopoietic colonies formed in the spleens of mice after serial transplantation with transduced bone marrow (2 degreesCFU-S). We found a significant increase in the frequency of expression with our modified vectors (>90% expression in vector DNA containing 2 degreesCFU-S) over the frequency observed with the standard MoMuLV-based vector (28% expression in vector containing 2 degreesCFU-S). Expression from the modified vectors was highly consistent, with expression in >50% of the vector-containing 2 degreesCFU-S from all 20 transplant recipients analyzed, whereas expression from the standard MoMuLV-based vector was inconsistent, with expression in 0-10% of the vector containing 2 degreesCFU-S from 8 recipients and expression in >50% of the vector-containing 2 degreesCFU-S from 4 other recipients. In addition, we established that the modified vectors had a lower level of DNA methylation than the control vector. These findings represent significant advances in the development and evaluation of effective retroviral vectors for application in vivo.
Collapse
Affiliation(s)
- P B Robbins
- Department of Molecular Microbiology and Immunology, Division of Research Immunology/Bone Marrow Transplantation, Childrens Hospital Los Angeles, University of Southern California School of Medicine, Los Angeles, CA 90027, USA
| | | | | | | | | | | |
Collapse
|
39
|
Robbins PB, Yu XJ, Skelton DM, Pepper KA, Wasserman RM, Zhu L, Kohn DB. Increased probability of expression from modified retroviral vectors in embryonal stem cells and embryonal carcinoma cells. J Virol 1997; 71:9466-74. [PMID: 9371608 PMCID: PMC230252 DOI: 10.1128/jvi.71.12.9466-9474.1997] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gene expression from the Moloney murine leukemia retrovirus (Mo-MuLV) is highly restricted in embryonic carcinoma (EC) and embryonic stem (ES) cells. We compared levels of expression in PA317 fibroblasts, F9 (EC) cells, and CCE (ES) cells by Mo-MuLV-based vectors and vectors based on our previously reported MND backbone, which has alterations to address three viral elements implicated as repressors of expression by Mo-MuLV: the enhancer, the primer binding site, and the negative-control region. Expression was evaluated with three reporter genes, the chloramphenicol acetyltransferase (CAT) gene, whose expression was measured by enzymatic assay and by Northern blotting; a truncated nerve growth factor receptor (tNGFR), whose expression was measured by fluorescence-activated cell sorting (FACS) as a cell surface protein; and the enhanced green fluorescent protein (EGFP), whose expression was measured intracellularly by flow cytometry. We found significantly higher levels of CAT activity (5- to 300-fold) and greater quantities of vector-specific transcripts in ES and EC cells transduced with the modified MND-CAT-SN vector than in those transduced with L-CAT-SN. Northern blot analysis indicated that long terminal repeat transcripts from MND-CAT-SN are >80 times more abundant than the L-CAT-SN transcripts. FACS analysis of tNGFR expression from a pair of vectors, L-tNGFR-SN and MND-tNGFR-SN, indicated that only 1.04% of the CCE cells containing the L-tNGFR-SN vector expressed the cell surface reporter, while the MND-tNGFR-SN vector drove expression in 99.54% of the CCE cells. Of the F9 cells containing the L-tNGFR-SN vector, 13.32% expressed tNGFR, while 99.89% of the F9 cells transduced with MND-tNGFR-SN showed expression. Essentially identical results were produced with an analogous pair of vectors encoding EGFP. In unselected pools of F9 cells 48 h posttransduction, the L-EGFP-SN vector drove expression in only 5% of the population while the MND-EGFP-SN vector drove expression in 88% of the cells. After more than 3 weeks in culture without selection, the proportion of cells showing expression from L-EGFP-SN decreased slightly to 3% while expression from the MND-EGFP-SN vector persisted in 80% of the cells. Interestingly, in the few ES and EC cells which did show expression from the L-tNGFR-SN or L-EGFP-SN vectors, the magnitude of reporter expression was similar to that from the MND-tNGFR-SN or MND-EGFP-SN vector in nearly all cells, suggesting that the MND vectors are far less susceptible to position-dependent variegation of expression than are the Mo-MuLV-based vectors. Therefore, the modified retroviral vector, MND, achieves higher net levels of expression due to a greater frequency of expression, which may be useful for the expression of exogenous genes in EC and ES cells.
Collapse
Affiliation(s)
- P B Robbins
- Department of Molecular Microbiology and Immunology, University of Southern California School of Medicine, Los Angeles, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Kaptein LC, Van Beusechem VW, Rivière I, Mulligan RC, Valerio D. Long-term in vivo expression of the MFG-ADA retroviral vector in rhesus monkeys transplanted with transduced bone marrow cells. Hum Gene Ther 1997; 8:1605-10. [PMID: 9322093 DOI: 10.1089/hum.1997.8.13-1605] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have tested the recombinant human adenosine deaminase (hADA) retroviral vector MFG-ADA for its efficacy in transducing hemopoietic stem cells of nonhuman primates and its expression level in the hematopoietic system. The percentage of provirus-positive granulocytes 1 year after transplantation of bone marrow transduced with MFG-ADA was 0.1%, which was equivalent to previously obtained results with the hADA virus-producing cell line POC-1. However, in MFG-ADA monkeys, significantly more peripheral blood mononuclear cells carried the hADA gene (1% versus 0.1%). Human ADA expression levels in peripheral blood mononuclear cells were different between POC-1 and MFG-ADA monkeys using samples with equal numbers of provirus copies per cell. In contrast, in total red blood cell lysates of MFG-ADA monkeys, the hADA expression was higher (approximately 10-fold) and could be detected longer (20 weeks and up to more than 1 year after bone marrow transplantation in 2 monkeys) than in POC-1 monkeys that were only positive for up to 12 weeks at the most. At 3 years after bone marrow transplantation, the MFG-ADA provirus could still be detected in 0.1% of bone marrow cells and peripheral blood cells and in 1% of cultured T cells. These results show that MFG-ADA virus can give rise to long-term in vivo expression of hADA in the primate hematopoietic system. However, transduction efficiencies remain low.
Collapse
Affiliation(s)
- L C Kaptein
- Department of Molecular Cell Biology, University of Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
41
|
Kotomura N, Ninomiya Y, Umesono K, Niwa O. Transcriptional regulation by competition between ELP isoforms and nuclear receptors. Biochem Biophys Res Commun 1997; 230:407-12. [PMID: 9016793 DOI: 10.1006/bbrc.1996.5972] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
ELP is a transcription factor belonging to the nuclear receptor superfamily. The consensus binding sequence for ELP contains a half site of the nuclear receptor recognition element. We demonstrated previously that ELP1, the repressor type isoform of ELP, competes for binding with the retinoic acid receptor and represses retinoic acid-induced transactivation. In this study, competitive repression by ELP1 was investigated for several other nuclear receptors. As in the case of the retinoic acid receptor, binding of vitamin D receptor, thyroid hormone receptor, and estrogen receptor could be competed by ELP1, resulting in repression of their ligand-dependent transactivation. Interestingly, the activator-type ELP isoforms were capable of repressing retinoic acid-induced transactivation through binding to the retinoic acid receptor binding element. These data suggest that competition for target DNA binding is a general mechanism of transcriptional repression by ELP isoforms.
Collapse
Affiliation(s)
- N Kotomura
- Department of Molecular Pathology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Mimami-ku, Japan
| | | | | | | |
Collapse
|
42
|
Lund A, Duch M, Pedersen F. Transcriptional Silencing of Retroviral Vectors. J Biomed Sci 1996; 3:365-378. [PMID: 11725119 DOI: 10.1007/bf02258042] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Although retroviral vector systems have been found to efficiently transduce a variety of cell types in vitro, the use of vectors based on murine leukemia virus in preclinical models of somatic gene therapy has led to the identification of transcriptional silencing in vivo as an important problem. Extinction of long-term vector expression has been observed after implantation of transduced hematopoietic cells as well as fibroblasts, myoblasts and hepatocytes. Here we review the influence of vector structure, integration site and cell type on transcriptional silencing. While down-regulation of proviral transcription is known from a number of cellular and animal models, major insight has been gained from studies in the germ line and embryonal cells of the mouse. Key elements for the transfer and expression of retroviral vectors, such as the viral transcriptional enhancer and the binding site for the tRNA primer for reverse transcription may have a major influence on transcriptional silencing. Alterations of these elements of the vector backbone as well as the use of internal promoter elements from housekeeping genes may contribute to reduce transcriptional silencing. The use of cell culture and animal models in the testing and improvement of vector design is discussed. Copyright 1996 S. Karger AG, Basel
Collapse
Affiliation(s)
- A.H. Lund
- Department of Molecular and Structural Biology, University of Aarhus, Denmark
| | | | | |
Collapse
|
43
|
Vernet M, Cebrian J. cis-acting elements that mediate the negative regulation of Moloney murine leukemia virus in mouse early embryos. J Virol 1996; 70:5630-3. [PMID: 8764077 PMCID: PMC190523 DOI: 10.1128/jvi.70.8.5630-5633.1996] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have addressed the question of the nature of Moloney murine leukemia virus (MoMuLV) repression in mouse embryos by assaying for the transient expression of MoMuLV-derived constructs microinjected into early cleavage embryos. We show that the same cis-acting DNA sequences responsible for the block in MoMuLV expression in embryonal carcinoma cell lines operate in early embryos: (i) the MoMuLV long terminal repeat is nonfunctional, and (ii) the +147 to +163 repressor binding site, or negative regulatory element, negatively regulates the expression from an active promoter.
Collapse
Affiliation(s)
- M Vernet
- Institut Cochin de Génétique Molécularire, Laboratoire de Génétique et Pathologie Expérimentales, Unité 380 de l'Institut National de la Santé et de la Recherche Médicale, Paris, France
| | | |
Collapse
|
44
|
Ferrari G, Salvatori G, Rossi C, Cossu G, Mavilio F. A retroviral vector containing a muscle-specific enhancer drives gene expression only in differentiated muscle fibers. Hum Gene Ther 1995; 6:733-42. [PMID: 7548273 DOI: 10.1089/hum.1995.6.6-733] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Genetically modified myogenic cells have a number of potentially relevant applications for gene therapy of genetic defects. Retroviral vectors proved to be a safe and efficient tool to transfer and express genes into satellite cells and their differentiated progeny, although muscle-specific regulation of the transferred gene is very difficult to achieve in a conventional vector framework. We modified a Moloney murine leukemia virus (MoMLV)-derived retroviral vector containing a bacterial beta-galactosidase (beta-Gal) reporter gene by inserting a muscle creatinine kinase (MCK) enhancer element into the U3 region of the viral long terminal repeat (LTR). The resulting vector (mLBSN) was transferred into cells of different histological origin, including undifferentiated murine and human myogenic cells, which were unable to express the transgene at detectable levels. Instead, gene expression from the modified LTR was obtained in a mouse myogenic cell line and in human primary satellite cells upon induction of differentiation into myotubes in culture, and correlated with the activation of the muscle differentiation program. beta-Gal-negative, mLBSN-transduced human satellite cells were also transplanted into the quadricep muscle of immunodeficient mice, where activation of the transgene expression was observed in vivo after differentiation and fusion into muscle fibers. These results show that retroviral vectors carrying LTRs modified in the enhancer sequences may be used to target tissue- and differentiation-specific gene expression into the muscle. For practical purposes, satellite cells engineered by muscle-specific retroviral vectors might represent an effective tool to deliver expression of a given gene product specifically into the muscle tissue, avoiding undesired protein accumulation in mononucleated cells. More generally, this type of vector might be useful whenever regulated expression of a transferred gene is necessary in a target cell or tissue.
Collapse
Affiliation(s)
- G Ferrari
- DIBIT-Istituto Scientifico H.S. Raffaele, Milano, Italy
| | | | | | | | | |
Collapse
|
45
|
Hoatlin ME, Kozak SL, Spiro C, Kabat D. Amplified and tissue-directed expression of retroviral vectors using ping-pong techniques. J Mol Med (Berl) 1995; 73:113-20. [PMID: 7633947 DOI: 10.1007/bf00198238] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Ping-pong amplification is an efficient process by which helper-free retrovirions replicate in cocultures of cell lines that package retroviruses into distinct host-range envelopes [11]. Transfection of a retroviral vector DNA into these cocultures results in massive virus production, with potentially endless cross-infection between different types of packaging cells. Because the helper-free virus spreads efficiently throughout the coculture, it is unnecessary to use dominant selectable marker genes, and the retroviral vectors can be simplified and optimized for expressing a single gene of interest. The most efficient ping-pong vector, pSFF, derived from the Friend erythroleukemia virus, has been used for high-level expression of several genes that could not be expressed with commonly employed two-gene retroviral vectors. Contrary to previous claims, problems of vector recombination are not inherent to ping-pong methods. Indeed, the pSFF vector has not formed replication-competent recombinants as shown by stringent assays. Here we review these methods, characterize the ping-pong process using the human erythropoietin gene as a model, and describe a new vector (pSFY) designed for enhanced expression in T lymphocytes. Factors that limit tissue-specific expression are reviewed.
Collapse
Affiliation(s)
- M E Hoatlin
- Department of Biochemistry and Molecular Biology, Oregon Health Sciences University, Portland 97201-3098, USA
| | | | | | | |
Collapse
|
46
|
Challita PM, Skelton D, el-Khoueiry A, Yu XJ, Weinberg K, Kohn DB. Multiple modifications in cis elements of the long terminal repeat of retroviral vectors lead to increased expression and decreased DNA methylation in embryonic carcinoma cells. J Virol 1995; 69:748-55. [PMID: 7815539 PMCID: PMC188638 DOI: 10.1128/jvi.69.2.748-755.1995] [Citation(s) in RCA: 171] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Infection by murine retroviruses in embryonic carcinoma (EC) and embryonic stem cells is highly restricted. The transcriptional unit of the Moloney murine leukemic virus (MoMuLV) long terminal repeat (LTR) is inactive in EC and embryonic stem cells in association with increased proviral methylation. In this study, expression in F9 EC cells was achieved from novel retroviral vectors containing three modifications in the MoMuLV-based retroviral vector: presence of the myeloproliferative sarcoma virus LTR, substitution of the primer binding site, and either deletion of a negative control region at the 5' end of the LTR or insertion of a demethylating sequence. We conclude that inhibition of expression from the MoMuLV LTR in EC cells is mediated through the additive effects of multiple cis-acting elements affecting the state of methylation of the provirus.
Collapse
Affiliation(s)
- P M Challita
- Department of Microbiology, University of Southern California School of Medicine, Los Angeles
| | | | | | | | | | | |
Collapse
|
47
|
John HA. Variable efficiency of retroviral-mediated gene transfer into early-passage cultures of fetal lamb epithelial, mesenchymal, and neuroectodermal tissues. Hum Gene Ther 1994; 5:283-93. [PMID: 8018744 DOI: 10.1089/hum.1994.5.3-283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The relative efficiency of retroviral-mediated gene transfer into early-passage cultures of different tissues of fetal lamb was investigated. Monolayer cultures prepared by plating 1 x 10(6) cells were infected with the Moloney murine leukemia (MoMLV)-based vector pZIP Neo at a multiplicity of infection (moi) of approximately 1 pfu per 2 x 10(2) recipient cells prior to selection for neomycin resistance. At the low moi used, cells from different tissues showed marked differences in efficiency of colony formation in the descending order: brain > kidney > muscle, lung > skin. Brain cells were transduced at least an order of magnitude more efficiently than other cell types, despite the doubling time of brain cell cultures being five times as long. Cultures were analyzed by morphological and immunocytological criteria to determine whether any particular cell types were transduced. A wide variety of morphologically distinct neuron-like and glial-like brain cells were neomycin resistant. The majority of muscle cell colonies were myogenic. Approximately half of the large kidney colonies were epithelial-like. The majority of lung colonies consisted of fibroblasts. The results suggest that cells originating from the surface embryonic germ layer (ectoderm) and/or occupying positions near the fetal external surface have a markedly lower susceptibility to retroviral-mediated gene transduction.
Collapse
Affiliation(s)
- H A John
- Moredun Research Institute, Edingburgh, UK
| |
Collapse
|
48
|
Castanotto D, Rossi JJ, Sarver N. Antisense catalytic RNAs as therapeutic agents. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1994; 25:289-317. [PMID: 8204504 DOI: 10.1016/s1054-3589(08)60435-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- D Castanotto
- Division of Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | | | | |
Collapse
|
49
|
Brightman BK, Farmer C, Fan H. Escape from in vivo restriction of Moloney mink cell focus-inducing viruses driven by the Mo+PyF101 long terminal repeat (LTR) by LTR alterations. J Virol 1993; 67:7140-8. [PMID: 8230436 PMCID: PMC238176 DOI: 10.1128/jvi.67.12.7140-7148.1993] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Mo+PyF101 M-MuLV is a variant Moloney murine leukemia virus containing polyomavirus F101 enhancers inserted just downstream from the M-MuLV enhancers in the long terminal repeat (LTR). The protein coding sequences for this virus are identical to those of M-MuLV. Mo+PyF101 M-MuLV induces T-cell disease with a much lower incidence and longer latency than wild-type M-MuLV. We have previously shown that Mo+PyF101 M-MuLV is defective in preleukemic events induced by wild-type M-MuLV, including splenic hematopoietic hyperplasia, bone marrow depletion, and generation of recombinant mink cell focus-inducing viruses (MCFs). We also showed that an M-MCF virus driven by the Mo+PyF101 LTR is infectious in vitro but does not propagate in mice. However, in these experiments, when a pseudotypic mixture of Mo+PyF101 M-MuLV and Mo+PyF101 MCF was inoculated into newborn NIH Swiss mice, they died of T-cell leukemia at times almost equivalent to those induced by wild-type M-MuLV. Tumor DNAs from Mo+PyF101 M-MuLV-Mo+PyF101 MCF-inoculated mice were examined by Southern blot analysis. The predominant forms of Mo+PyF101 MCF proviruses in these tumors contained added sequences in the U3 region of the LTR. The U3 regions of representative tumor-derived variant Mo+PyF101 MCFs were cloned by polymerase chain reaction amplification, and sequencing indicated that they had acquired an additional copy of the M-MuLV 75-bp tandem repeat in the enhancer region. NIH 3T3 cell lines infected with altered viruses were obtained from representative Mo+PyF101 M-MuLV-Mo+PyF101 MCF-induced tumors, and mice were inoculated with the recovered viruses. Leukemogenicity was approximately equivalent to that in the original Mo+PyF101 M-MuLV-Mo+PyF101 MCF viral stock. Southern blot analysis on the resulting tumors now predominantly revealed loss of the polyomavirus sequences. These results suggest that the suppressive effects of the PyF101 sequences on M-MuLV-induced disease and potentially on MCF propagation were overcome in two ways: by triplication of the M-MuLV direct repeats and by loss of the polyomavirus sequences.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Base Sequence
- Cloning, Molecular
- DNA, Neoplasm/genetics
- Genetic Variation
- Leukemia, Experimental/etiology
- Leukemia, Experimental/genetics
- Leukemia, Experimental/microbiology
- Leukemia, T-Cell/etiology
- Leukemia, T-Cell/genetics
- Leukemia, T-Cell/microbiology
- Mice
- Mice, Inbred Strains
- Mink Cell Focus-Inducing Viruses/genetics
- Molecular Sequence Data
- Moloney murine leukemia virus/genetics
- Moloney murine leukemia virus/pathogenicity
- Proviruses/genetics
- Repetitive Sequences, Nucleic Acid/genetics
- Retroviridae Infections/genetics
- Sequence Analysis, DNA
- Sequence Homology, Nucleic Acid
- Tumor Virus Infections/genetics
- Virulence
Collapse
Affiliation(s)
- B K Brightman
- Department of Molecular Biology and Biochemistry, University of California, Irvine 92717
| | | | | |
Collapse
|
50
|
Music L, Sauer G. Inhibition of Moloney Murine Leukaemia Virus Transcription by a Phospholipase-C Inhibitor Affecting Trans-Acting Factors. Antivir Chem Chemother 1992. [DOI: 10.1177/095632029200300506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The propagation and the transcription of Moloney murine leukaemia virus (Mo-MuLV) can be inhibited by the antiviral compound tricyclodecan-9-yl-xanthate (code name: D609), which inhibits phospholipase-C (PLC) and, as a consequence, the activation of protein kinase-C (PKC) isoenzyme(s). The frans-acting factors LVa, LVb, and LVc were shown to be affected; it was not possible to retrieve them after treatment with D609 from Mo-MuLV producer cells, by virtue of the binding affinity to their consensus sequences. In contrast, the binding efficiency of the other three known transacting factors (core, NF1 and GRE), which in addition to the viral transcription, play a role in the regulation of cellular mRNA synthesis remained unimpaired. Neither LVa, LVb, nor LVc was found to be phosphorylated, which suggests that these are not targets of PKC. Only one phosphorylated DNA-binding protein was identified with an apparent molecular weight of 34kDa. This protein co-purified irrespective of the recognition sequences that we used (LVa, LVb, LVc, core, and NF1). Direct evidence is provided for the inhibition of the TPA-induced phosphorylation of the 34 kDa protein by D609. We suggest that the binding of LVa, LVb, and LVc to the DNA is mediated by the 34 kDa protein in its phosphorylated state.
Collapse
Affiliation(s)
- L. Music
- Department of Molecular Biology of DNA Tumour Viruses, German Cancer Research Centre, 6900 Heidelberg, Germany
| | - G. Sauer
- Department of Molecular Biology of DNA Tumour Viruses, German Cancer Research Centre, 6900 Heidelberg, Germany
| |
Collapse
|