1
|
Salati S, Prudente Z, Genovese E, Pennucci V, Rontauroli S, Bartalucci N, Mannarelli C, Ruberti S, Zini R, Rossi C, Bianchi E, Guglielmelli P, Tagliafico E, Vannucchi AM, Manfredini R. Calreticulin Affects Hematopoietic Stem/Progenitor Cell Fate by Impacting Erythroid and Megakaryocytic Differentiation. Stem Cells Dev 2018; 27:225-236. [PMID: 29258411 DOI: 10.1089/scd.2017.0137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Calreticulin (CALR) is a chaperone protein that localizes primarily to the endoplasmic reticulum (ER) lumen where it is responsible for the control of proper folding of neo-synthesized glycoproteins and the retention of calcium. Recently, mutations affecting exon 9 of the CALR gene have been described in approximately 40% of patients with myeloproliferative neoplasms (MPNs). Although the role of mutated CALR in the development of MPNs has begun to be clarified, there are still no data available on the function of wild-type (WT) CALR during physiological hematopoiesis. To shed light on the role of WT CALR during normal hematopoiesis, we performed gene silencing and overexpression experiments in hematopoietic stem progenitor cells (HSPCs). Our results showed that CALR overexpression is able to affect physiological hematopoiesis by enhancing both erythroid and megakaryocytic (MK) differentiation. In agreement with overexpression data, CALR silencing caused a significant decrease in both erythroid and MK differentiation of human HSPCs. Gene expression profiling (GEP) analysis showed that CALR is able to affect the expression of several genes involved in HSPC differentiation toward both the erythroid and MK lineages. Moreover, GEP data also highlighted the modulation of several genes involved in ER stress response, unfolded protein response (UPR), and DNA repair, and of several genes already described to play a role in MPN development, such as proinflammatory cytokines and hematological neoplasm-related markers. Altogether, our data unraveled a new and unexpected role for CALR in the regulation of normal hematopoietic differentiation. Moreover, by showing the impact of CALR on the expression of genes involved in several biological processes already described in cellular transformation, our data strongly suggest a more complex role for CALR in MPN development that goes beyond the activation of the THPO receptor and involves ER stress response, UPR, and DNA repair.
Collapse
Affiliation(s)
- Simona Salati
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Zelia Prudente
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Pennucci
- Institute for Cell and Gene Therapy & Center for Chronic Immunodeficiency, University of Freiburg, Freiburg, Germany
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Carmela Mannarelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Samantha Ruberti
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Rossi
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Guglielmelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
2
|
Vannucchi AM, Rotunno G, Bartalucci N, Raugei G, Carrai V, Balliu M, Mannarelli C, Pacilli A, Calabresi L, Fjerza R, Pieri L, Bosi A, Manfredini R, Guglielmelli P. Calreticulin mutation-specific immunostaining in myeloproliferative neoplasms: pathogenetic insight and diagnostic value. Leukemia 2014; 28:1811-8. [PMID: 24618731 PMCID: PMC4158831 DOI: 10.1038/leu.2014.100] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 02/20/2014] [Accepted: 02/28/2014] [Indexed: 01/01/2023]
Abstract
Mutations in the gene calreticulin (CALR) occur in the majority of JAK2- and MPL-unmutated patients with essential thrombocythemia (ET) and primary myelofibrosis (PMF); identifying CALR mutations contributes to the diagnostic pathway of ET and PMF. CALR mutations are heterogeneous spanning over the exon 9, but all result in a novel common protein C terminus. We developed a polyclonal antibody against a 17-amino-acid peptide derived from mutated calreticulin that was used for immunostaining of bone marrow biopsies. We show that this antibody specifically recognized patients harboring different types of CALR mutation with no staining in healthy controls and JAK2- or MPL-mutated ET and PMF. The labeling was mostly localized in megakaryocytes, whereas myeloid and erythroid cells showed faint staining, suggesting a preferential expression of calreticulin in megakaryocytes. Megakaryocytic-restricted expression of calreticulin was also demonstrated using an antibody against wild-type calreticulin and by measuring the levels of calreticulin RNA by gene expression analysis. Immunostaining using an antibody specific for mutated calreticulin may become a rapid, simple and cost-effective method for identifying CALR-mutated patients complementing molecular analysis; furthermore, the labeling pattern supports the preferential expansion of megakaryocytic cell lineage as a result of CALR mutation in an immature hematopoietic stem cell.
Collapse
Affiliation(s)
- A M Vannucchi
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - G Rotunno
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - N Bartalucci
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - G Raugei
- Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - V Carrai
- Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - M Balliu
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - C Mannarelli
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - A Pacilli
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - L Calabresi
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - R Fjerza
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - L Pieri
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - A Bosi
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - R Manfredini
- Centre for Regenerative Medicine 'Stefano Ferrari', Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - P Guglielmelli
- 1] Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy [2] Hematology Unit, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| |
Collapse
|
3
|
White R, Chenciner N, Bonello G, Salas M, Blancou P, Gauduin MC. Epithelial stem cells as mucosal antigen-delivering cells: A novel AIDS vaccine approach. Vaccine 2013; 33:6914-21. [PMID: 24286835 DOI: 10.1016/j.vaccine.2013.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/12/2013] [Accepted: 09/06/2013] [Indexed: 10/26/2022]
Abstract
A key obstacle limiting development of an effective AIDS vaccine is the inability to deliver antigen for a sufficient period of time resulting in weak and transient protection. HIV transmission occurs predominantly across mucosal surfaces; therefore, an ideal vaccine strategy would be to target HIV at mucosal entry sites to prevent infection. Such a novel strategy relies on the activation of mucosal immune response via presentation of viral antigens by the mucosal epithelial cells. The use of a terminally differentiated epithelial cell promoter to drive expression of antigens leading to viral protein production in the upper layers of the epithelium is central to the success of this approach. Our results show that when administered intradermally to mice, a GFP-reporter gene under the transcriptional control of the involucrin promoter is expressed in the upper layers of the epidermis and, although transduced cells were very low in number, high and sustained anti-GFP antibody production is observed in vivo. A subsequent experiment investigates the effectiveness of GFP-tagged replication-competent SIVdeltaNef and GFP-tagged replication-deficient SIVdeltaVifdeltaNef constructs under the transcriptional control of the involucrin promoter. Optimal conditions for production of pseudotyped VSV-G viral particles destined to transduce basal epithelial stem cells at the mucosal sites of entry of SIV in our animal model were determined. Altogether, the data demonstrate the feasibility of an epithelium-based vaccine containing involucrin-driven viral antigen encoding sequences that integrate into epithelial stem cells and show long-term expression in the upper layer of the epithelium even after multiple cycle of epithelia renewal. Such epithelium-based vaccine should elicit a long-term immunity against HIV/SIV infection at the site of entry of the virus.
Collapse
Affiliation(s)
- Robert White
- Texas Biomedical Research Institute, Department of Virology and Immunology, San Antonio, TX 78227, USA
| | - Nicole Chenciner
- Institut Pasteur, Unité de Rétrovirologie Moléculaire, CNRS URA 3015, 75724 Paris Cedex 15, France
| | - Gregory Bonello
- Texas Biomedical Research Institute, Department of Virology and Immunology, San Antonio, TX 78227, USA
| | - Mary Salas
- Texas Biomedical Research Institute, Department of Virology and Immunology, San Antonio, TX 78227, USA
| | - Philippe Blancou
- Institut National de la Santé et de la Recherche Médicale, University of Nice-Sophia Antipolis, Valbonne, France
| | - Marie-Claire Gauduin
- Texas Biomedical Research Institute, Department of Virology and Immunology, San Antonio, TX 78227, USA; Southwest National Primate Research Center, San Antonio, TX 78227, USA.
| |
Collapse
|
4
|
Miccio A, Poletti V, Tiboni F, Rossi C, Antonelli A, Mavilio F, Ferrari G. The GATA1-HS2 enhancer allows persistent and position-independent expression of a β-globin transgene. PLoS One 2011; 6:e27955. [PMID: 22164220 PMCID: PMC3229501 DOI: 10.1371/journal.pone.0027955] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 10/28/2011] [Indexed: 11/19/2022] Open
Abstract
Gene therapy of genetic diseases requires persistent and position-independent expression of a therapeutic transgene. Transcriptional enhancers binding chromatin-remodeling and modifying complexes may play a role in shielding transgenes from repressive chromatin effects. We tested the activity of the HS2 enhancer of the GATA1 gene in protecting the expression of a β-globin minigene delivered by a lentiviral vector in hematopoietic stem/progenitor cells. Gene expression from proviruses carrying GATA1-HS2 in both LTRs was persistent and resistant to silencing at most integration sites in the in vivo progeny of human hematopoietic progenitors and murine long-term repopulating stem cells. The GATA1-HS2-modified vector allowed correction of murine β-thalassemia at low copy number without inducing clonal selection of erythroblastic progenitors. Chromatin immunoprecipitation studies showed that GATA1 and the CBP acetyltransferase bind to GATA1-HS2, significantly increasing CBP-specific histone acetylations at the LTRs and β-globin promoter. Recruitment of CBP by the LTRs thus establishes an open chromatin domain encompassing the entire provirus, and increases the therapeutic efficacy of β-globin gene transfer by reducing expression variegation and epigenetic silencing.
Collapse
Affiliation(s)
- Annarita Miccio
- H. San Raffaele-Telethon Institute for Gene Therapy (HSR-TIGET), Istituto Scientifico H. San Raffaele, Milan, Italy
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Poletti
- Laboratory of Gene Expression, Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Francesca Tiboni
- H. San Raffaele-Telethon Institute for Gene Therapy (HSR-TIGET), Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Claudia Rossi
- H. San Raffaele-Telethon Institute for Gene Therapy (HSR-TIGET), Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Antonella Antonelli
- H. San Raffaele-Telethon Institute for Gene Therapy (HSR-TIGET), Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Fulvio Mavilio
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Laboratory of Gene Expression, Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Giuliana Ferrari
- H. San Raffaele-Telethon Institute for Gene Therapy (HSR-TIGET), Istituto Scientifico H. San Raffaele, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
- * E-mail:
| |
Collapse
|
5
|
Tenedini E, Roncaglia E, Ferrari F, Orlandi C, Bianchi E, Bicciato S, Tagliafico E, Ferrari S. Integrated analysis of microRNA and mRNA expression profiles in physiological myelopoiesis: role of hsa-mir-299-5p in CD34+ progenitor cells commitment. Cell Death Dis 2011; 1:e28. [PMID: 21364636 PMCID: PMC3032330 DOI: 10.1038/cddis.2010.5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hematopoiesis entails a series of hierarchically organized events that proceed throughout cell specification and terminates with cell differentiation. Commitment needs the transcription factors' effort, which, in concert with microRNAs, drives cell fate and responds to promiscuous patterns of gene expression by turning on lineage-specific genes and repressing alternate lineage transcripts. We obtained microRNA profiles from human CD34+ hematopoietic progenitor cells and in vitro differentiated erythroblasts, megakaryoblasts, monoblasts and myeloblast precursors that we analyzed together with their gene expression profiles. The integrated analysis of microRNA-mRNA expression levels highlighted an inverse correlation between microRNAs specifically upregulated in one single-cell progeny and their putative target genes, which resulted in downregulation. Among the upregulated lineage-enriched microRNAs, hsa-miR-299-5p emerged as having a role in controlling CD34+ progenitor fate, grown in multilineage culture conditions. Gain- and loss-of-function experiments revealed that hsa-miR-299-5p participates in the regulation of hematopoietic progenitor fate, modulating megakaryocytic-granulocytic versus erythroid-monocytic differentiation.
Collapse
Affiliation(s)
- E Tenedini
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Abstract
After more than 1500 gene therapy clinical trials in the past two decades, the overall conclusion is that for gene therapy (GT) to be successful, the vector systems must still be improved in terms of delivery, expression and safety. The recent development of more efficient and stable vector systems has created great expectations for the future of GT. Impressive results were obtained in three primary immunodeficiencies and other inherited diseases such as congenital blindness, adrenoleukodystrophy or junctional epidermolysis bullosa. However, the development of leukemia in five children included in the GT clinical trials for X-linked severe combined immunodeficiency and the silencing of the therapeutic gene in the chronic granulomatous disease clearly showed the importance of improving safety and efficiency. In this review, we focus on the main strategies available to achieve physiological or tissue-specific expression of therapeutic transgenes and discuss the importance of controlling transgene expression to improve safety. We propose that tissue-specific and/or physiological viral vectors offer the best balance between efficiency and safety and will be the tools of choice for future clinical trials in GT of inherited diseases.
Collapse
|
7
|
A previously unreported function of β(1)B integrin isoform in caspase-8-dependent integrin-mediated keratinocyte death. J Invest Dermatol 2010; 130:2569-77. [PMID: 20613771 DOI: 10.1038/jid.2010.195] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Integrins regulate adhesive cell-matrix interactions and mediate survival signals. On the other hand, unligated or free cytoplasmic fragments of integrins induce apoptosis in many cell types (integrin-mediated death). We have previously shown that β(1) integrin expression protects keratinocyte stem cells from anoikis, whereas the role of the β(1)B integrin isoform has not been clarified. In this study we report that suspended keratinocytes undergo apoptosis through the activation of caspase-8, independently of the Fas/Fas ligand system. Indeed, anti-β(1) integrin-neutralizing antibodies induced apoptosis in short hairpin RNA Fas-associated death domain-treated cells. Moreover, before and during suspension, caspase-8 directly associated with β(1) integrin, which in turn internalized and progressively degraded, shedding the cytoplasmic domain. β(1)B was expressed only in the cytoplasm in a perinuclear manner and remained unaltered during suspension. At 24 hours, as β(1)A was located close to the nucleus, β(1)B colocalized with β(1)A and coimmunoprecipitated with caspase-8. Caspase-8 was activated earlier in β(1)B integrin-transfected keratinocytes, and these cells underwent a higher rate of apoptosis than mock cells. In contrast, caspase-8 was not activated in small interfering RNA (siRNA) β(1)B-transfected cells. These results indicate that when β(1)A is unligated, β(1)B is responsible for "integrin-mediated death" in human keratinocytes.
Collapse
|
8
|
Salsi V, Ferrari S, Ferraresi R, Cossarizza A, Grande A, Zappavigna V. HOXD13 binds DNA replication origins to promote origin licensing and is inhibited by geminin. Mol Cell Biol 2009; 29:5775-88. [PMID: 19703996 PMCID: PMC2772751 DOI: 10.1128/mcb.00509-09] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 05/26/2009] [Accepted: 08/12/2009] [Indexed: 12/24/2022] Open
Abstract
HOX DNA-binding proteins control patterning during development by regulating processes such as cell aggregation and proliferation. Recently, a possible involvement of HOX proteins in replication origin activity was suggested by results showing that a number of HOX proteins interact with the DNA replication licensing regulator geminin and bind a characterized human origin of replication. The functional significance of these observations, however, remained unclear. We show that HOXD13, HOXD11, and HOXA13 bind in vivo all characterized human replication origins tested. We furthermore show that HOXD13 interacts with the CDC6 loading factor, promotes pre-replication complex (pre-RC) proteins assembly at origins, and stimulates DNA synthesis in an in vivo replication assay. HOXD13 expression in cultured cells accelerates DNA synthesis initiation in correlation with the earlier pre-RC recruitment onto origins during G(1) phase. Geminin, which interacts with HOXD13 as well, blocks HOXD13-mediated assembly of pre-RC proteins and inhibits HOXD13-induced DNA replication. Our results uncover a function for Hox proteins in the regulation of replication origin activity and reveal an unforeseen role for the inhibition of HOX protein activity by geminin in the context of replication origin licensing.
Collapse
Affiliation(s)
- Valentina Salsi
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| | - Silvia Ferrari
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| | - Roberta Ferraresi
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| | - Andrea Cossarizza
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| | - Alexis Grande
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| | - Vincenzo Zappavigna
- Department of Animal Biology, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, Modena 41100, Italy
| |
Collapse
|
9
|
Lymboussaki A, Gemelli C, Testa A, Facchini G, Ferrari F, Mavilio F, Grande A. PPARdelta is a ligand-dependent negative regulator of vitamin D3-induced monocyte differentiation. Carcinogenesis 2008; 30:230-7. [PMID: 19056929 DOI: 10.1093/carcin/bgn272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
A number of reports indicate that peroxisome proliferator-activated receptor (PPAR) delta is involved in the molecular control of monocyte-macrophage differentiation. In this regard, the recent demonstration that PPARdelta is a primary response gene of 1alpha,25-dihydroxyvitamin D3 (VD), i.e. a powerful inducer of such process, allowed us to hypothesize the existence of a cross talk between PPARdelta and VD receptor pathways. To address this issue, we analyzed the effects promoted by stimulation with PPARdelta ligands and by overexpression of this nuclear receptor in monoblastic cell lines undergoing exposure to VD. The results obtained evidenced that, although promoting a weak differentiation effect by themselves, PPARdelta ligands efficiently co-operated with VD treatment. In spite of this, PPARdelta overexpression exerted a remarkable inhibitory effect on monocyte-macrophage differentiation induced by VD that was, at least partly, reverted by stimulation with a highly specific PPARdelta ligand. These data indicate that, although acting through a ligand-dependent modality, PPARdelta is a negative regulator of VD-mediated monocyte differentiation, allowing us to hypothesize a role of the investigated nuclear receptor in the differentiation block of M5 type (monoblastic) acute myeloid leukemias (AMLs). Bioinformatic analysis of a microarray database, containing the expression profiles of 285 AML cases, further supported this hypothesis demonstrating the existence of a subset of M5 type (monoblastic) AMLs that overexpress PPARdelta gene.
Collapse
Affiliation(s)
- Athina Lymboussaki
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | | | | | | | | | | | |
Collapse
|
10
|
Engineering human hematopoietic stem/progenitor cells to produce a broadly neutralizing anti-HIV antibody after in vitro maturation to human B lymphocytes. Blood 2008; 113:1422-31. [PMID: 19059876 DOI: 10.1182/blood-2008-09-177139] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Broadly neutralizing anti-HIV antibodies are rare and have proved hard to elicit with any immunogen. We have tested in vitro the notion that such antibodies or other antiviral proteins could be made by lentivirus-mediated gene transfer into human hematopoietic stem/progenitor cells (HSPCs), followed by differentiation of the transduced cells into B cells, the most potent antibody-producing cells. To do this, we have developed a highly efficient system for in vitro maturation of secreting B lymphocytes and plasma cells from CD34(+) HSPCs. It is a 3-stage, in vitro culture system that supports normal human B-lineage development from HSPCs to antibody-secreting plasmablasts (approximately 36%) and plasma cells (approximately 20%). By transducing human cord blood CD34(+) cells with lentiviral vectors encoding a secretory monoclonal anti-HIV antibody, b12 (IgG(1)), we were able to program human B cells to produce in vitro up to 1.5 microg/mL of this broadly neutralizing antibody. Our results suggest that an HIV vaccine might be delivered by autologous transplantation of in vitro-programmed HSPCs, which would develop into antibody-secreting B cells in vivo and provide a continuous supply of anti-HIV neutralizing antibodies.
Collapse
|
11
|
Gemelli C, Orlandi C, Zanocco Marani T, Martello A, Vignudelli T, Ferrari F, Montanari M, Parenti S, Testa A, Grande A, Ferrari S. The vitamin D3/Hox-A10 pathway supports MafB function during the monocyte differentiation of human CD34+ hemopoietic progenitors. THE JOURNAL OF IMMUNOLOGY 2008; 181:5660-72. [PMID: 18832725 DOI: 10.4049/jimmunol.181.8.5660] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although a considerable number of reports indicate an involvement of the Hox-A10 gene in the molecular control of hemopoiesis, the conclusions of such studies are quite controversial given that they support, in some cases, a role in the stimulation of stem cell self-renewal and myeloid progenitor expansion, whereas in others they implicate this transcription factor in the induction of monocyte-macrophage differentiation. To clarify this issue, we analyzed the biological effects and the transcriptome changes determined in human primary CD34(+) hemopoietic progenitors by retroviral transduction of a full-length Hox-A10 cDNA. The results obtained clearly indicated that this homeogene is an inducer of monocyte differentiation, at least partly acting through the up-regulation of the MafB gene, recently identified as the master regulator of such a maturation pathway. By using a combined approach based on computational analysis, EMSA experiments, and luciferase assays, we were able to demonstrate the presence of a Hox-A10-binding site in the promoter region of the MafB gene, which suggested the likely molecular mechanism underlying the observed effect. Stimulation of the same cells with the vitamin D(3) monocyte differentiation inducer resulted in a clear increase of Hox-A10 and MafB transcripts, indicating the existence of a precise transactivation cascade involving vitamin D(3) receptor, Hox-A10, and MafB transcription factors. Altogether, these data allow one to conclude that the vitamin D(3)/Hox-A10 pathway supports MafB function during the induction of monocyte differentiation.
Collapse
Affiliation(s)
- Claudia Gemelli
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Santoni de Sio FR, Gritti A, Cascio P, Neri M, Sampaolesi M, Galli C, Luban J, Naldini L. Lentiviral vector gene transfer is limited by the proteasome at postentry steps in various types of stem cells. Stem Cells 2008; 26:2142-52. [PMID: 18483423 DOI: 10.1634/stemcells.2007-0705] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The isolation of human embryonic and somatic stem cells of different types has made it possible to design novel gene and cell replacement therapies. Vectors derived from retro/lentiviruses are used to stably introduce genes into stem cells and their progeny. However, the permissivity to retroviral infection varies among cell types. We previously showed that hematopoietic stem cells are poorly permissive to human immunodeficiency virus (HIV)-derived vectors and that pharmacological inhibition of the proteasome strongly enhances gene transfer. Here we report that the proteasome limits lentiviral gene transfer in all stem cell types tested, including embryonic, mesenchymal, and neural, of both human and mouse origin. Remarkably, this inhibitory activity was sharply reduced upon differentiation of the stem cells, suggesting that it represents a novel feature of the stem cell/immature progenitor phenotype. Proteasome-mediated inhibition was specific for lentiviral vectors and occurred at a postentry infection step. It was not mediated by activation of nuclear factor-kappaB, a major signaling pathway modulated by the proteasome, and did not correlate with high proteasome activity. Interaction of the virion core with cyclophilin A was required to maximize the effect of proteasome inhibitor on the infection pathway. These findings are relevant to uncover new mediators of HIV gene transfer and help in designing more effective protocols for the genetic modification of stem cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
|
13
|
Salati S, Zini R, Bianchi E, Testa A, Mavilio F, Manfredini R, Ferrari S. Role of CD34 antigen in myeloid differentiation of human hematopoietic progenitor cells. Stem Cells 2008; 26:950-9. [PMID: 18192237 DOI: 10.1634/stemcells.2007-0597] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CD34 is a transmembrane protein that is strongly expressed on hematopoietic stem/progenitor cells (HSCs); despite its importance as a marker of HSCs, its function is still poorly understood, although a role in cell adhesion has been demonstrated. To characterize the function of CD34 antigen on human HSCs, we examined, by both inhibition and overexpression, the role of CD34 in the regulation of HSC lineage differentiation. Our results demonstrate that CD34 silencing enhances HSC granulocyte and megakaryocyte differentiation and reduces erythroid maturation. In agreement with these results, the gene expression profile of these cells reveals the upregulation of genes involved in granulocyte and megakaryocyte differentiation and the downregulation of erythroid genes. Consistently, retroviral-mediated CD34 overexpression leads to a remarkable increase in erythroid progenitors and a dramatic decrease in granulocyte progenitors, as evaluated by clonogenic assay. Together, these data indicate that the CD34 molecule promotes the differentiation of CD34+ hematopoietic progenitors toward the erythroid lineage, which is achieved, at least in part, at the expense of granulocyte and megakaryocyte lineages.
Collapse
Affiliation(s)
- Simona Salati
- Department of Biomedical Sciences, Biological Chemistry Section, University of Modena and Reggio Emilia, Via Campi 287, 41100 Modena, Italy
| | | | | | | | | | | | | |
Collapse
|
14
|
Yilmaz A, Fernandez S, Lairmore MD, Boris-Lawrie K. Coordinate enhancement of transgene transcription and translation in a lentiviral vector. Retrovirology 2006; 3:13. [PMID: 16480517 PMCID: PMC1388234 DOI: 10.1186/1742-4690-3-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Accepted: 02/15/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coordinate enhancement of transgene transcription and translation would be a potent approach to significantly improve protein output in a broad array of viral vectors and nonviral expression systems. Many vector transgenes are complementary DNA (cDNA). The lack of splicing can significantly reduce the efficiency of their translation. Some retroviruses contain a 5' terminal post-transcriptional control element (PCE) that facilitates translation of unspliced mRNA. Here we evaluated the potential for spleen necrosis virus PCE to stimulate protein production from HIV-1 based lentiviral vector by: 1) improving translation of the internal transgene transcript; and 2) functionally synergizing with a transcriptional enhancer to achieve coordinate increases in RNA synthesis and translation. RESULTS Derivatives of HIV-1 SIN self-inactivating lentiviral vector were created that contain PCE and cytomegalovirus immediate early enhancer (CMV IE). Results from transfected cells and four different transduced cell types indicate that: 1) PCE enhanced transgene protein synthesis; 2) transcription from the internal promoter is enhanced by CMV IE; 3) PCE and CMV IE functioned synergistically to significantly increase transgene protein yield; 4) the magnitude of translation enhancement by PCE was similar in transfected and transduced cells; 5) differences were observed in steady state level of PCE vector RNA in transfected and transduced cells; 6) the lower steady state was not attributable to reduced RNA stability, but to lower cytoplasmic accumulation in transduced cells. CONCLUSION PCE is a useful tool to improve post-transcriptional expression of lentiviral vector transgene. Coordinate enhancement of transcription and translation is conferred by the combination of PCE with CMV IE transcriptional enhancer and increased protein yield up to 11 to 17-fold in transfected cells. The incorporation of the vector provirus into chromatin correlated with reduced cytoplasmic accumulation of PCE transgene RNA. We speculate that epigenetic modulation of promoter activity altered cotranscriptional recruitment of RNA processing factors and reduced the availability of fully processed transcript or the efficiency of export from the nucleus. Our results provide an example of the dynamic interplay between the transcription and post-transcription steps of gene expression and document that introduction of heterologous gene expression signals can yield disparate effects in transfected versus transduced cells.
Collapse
Affiliation(s)
- Alper Yilmaz
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Molecular, Cellular & Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Soledad Fernandez
- Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael D Lairmore
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Department of Molecular Virology, Immunology & Medical Genetics, The Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Molecular, Cellular & Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Kathleen Boris-Lawrie
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210, USA
- Department of Molecular Virology, Immunology & Medical Genetics, The Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Molecular, Cellular & Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
15
|
Gemelli C, Montanari M, Tenedini E, Zanocco Marani T, Vignudelli T, Siena M, Zini R, Salati S, Tagliafico E, Manfredini R, Grande A, Ferrari S. Virally mediated MafB transduction induces the monocyte commitment of human CD34+ hematopoietic stem/progenitor cells. Cell Death Differ 2006; 13:1686-96. [PMID: 16456583 DOI: 10.1038/sj.cdd.4401860] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Upregulation of specific transcription factors is a generally accepted mechanism to explain the commitment of hematopoietic stem cells along precise maturation lineages. Based on this premise, transduction of primary hematopoietic stem/progenitor cells with viral vectors containing the investigated transcription factors appears as a suitable experimental model to identify such regulators. Although MafB transcription factor is believed to play a role in the regulation of monocytic commitment, no demonstration is, to date, available supporting this function in normal human hematopoiesis. To address this issue, we retrovirally transduced cord blood CD34+ hematopoietic progenitors with a MafB cDNA. Immunophenotypic and morphological analysis of transduced cells demonstrated the induction of a remarkable monomacrophage differentiation. Microarray analysis confirmed these findings and disclosed the upregulation of macrophage-related transcription factors belonging to the AP-1, MAF, PPAR and MiT families. Altogether our data allow to conclude that MafB is a key regulator of human monocytopoiesis.
Collapse
Affiliation(s)
- C Gemelli
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Loewen N, Poeschla EM. Lentiviral vectors. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2005; 99:169-91. [PMID: 16568892 DOI: 10.1007/10_007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We review the use of lentiviral vectors in current human gene therapy applications that involve genetic modification of nondividing tissues with integrated transgenes. Safety issues, including insertional mutagenesis and replication-competent retroviruses, are discussed. Innate cellular defenses against retroviruses and their implications for human gene therapy with different lentiviral vectors are also addressed.
Collapse
Affiliation(s)
- Nils Loewen
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | |
Collapse
|
17
|
Urbinati F, Lotti F, Facchini G, Montanari M, Ferrari G, Mavilio F, Grande A. Competitive engraftment of hematopoietic stem cells genetically modified with a truncated erythropoietin receptor. Hum Gene Ther 2005; 16:594-608. [PMID: 15916484 DOI: 10.1089/hum.2005.16.594] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transplantation of genetically modified hematopoietic stem cells (HSCs) has therapeutic potential for a variety of blood genetic disorders. Engraftment of HSCs, however, requires toxic myeloablative treatments, which render this approach questionable for non-life-threatening disorders. A potential alternative is the use of transgenes, which allows positive selection of HSCs in vivo. We used retroviral vectors to express a truncated derivative of the erythropoietin receptor (tEpoR) in murine and human hematopoietic cells. Murine HSCs expressing tEpoR at different levels (1500 to 13,000 receptors/cell) acquire a competitive repopulation capacity in vivo upon transplantation into fully or partially myeloablated co-isogenic mouse recipients. Long-term analysis of transplanted mice showed that expression of tEpoR at paraphysiological levels (approximately 1500 receptors/cell) has no effect on steady-state hematopoiesis and induces no further expansion of transduced cells after the engraftment period. Human cord blood-derived CD34+ stem/progenitor cells transduced with a lentiviral vector expressing tEpoR expand their clonogenic capacity in vitro, and significantly increase their marrow repopulation capacity upon xenotransplantation into sublethally irradiated NOD-SCID mice, with no alteration in their phenotype, survival, and differentiation properties. These data indicate that expression of tEpoR is an effective strategy to promote selective engraftment of genetically modified HSCs upon transplantation in both myeloablative and nonmyeloablative conditions, without the use of toxic drugs for selection.
Collapse
Affiliation(s)
- Fabrizia Urbinati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | | | | | | | | | | | |
Collapse
|
18
|
Montanari M, Gemelli C, Tenedini E, Zanocco Marani T, Vignudelli T, Siena M, Zini R, Salati S, Chiossi G, Tagliafico E, Manfredini R, Grande A, Ferrari S. Correlation between differentiation plasticity and mRNA expression profiling of CD34+-derived CD14− and CD14+ human normal myeloid precursors. Cell Death Differ 2005; 12:1588-600. [PMID: 15947790 DOI: 10.1038/sj.cdd.4401679] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In spite of their apparently restricted differentiation potentiality, hematopoietic precursors are plastic cells able to trans-differentiate from a maturation lineage to another. To better characterize this differentiation plasticity, we purified CD14- and CD14+ myeloid precursors generated by 'in vitro' culture of human CD34+ hematopoietic progenitors. Morphological analysis of the investigated cell populations indicated that, as expected, they consisted of granulocyte and monocyte precursors, respectively. Treatment with differentiation inducers revealed that CD14- cells were bipotent granulo-monocyte precursors, while CD14+ cells appeared univocally committed to a terminal macrophage maturation. Flow cytometry analysis demonstrated that the conversion of granulocyte precursors to the mono-macrophage maturation lineage occurs through a differentiation transition in which the granulocyte-related myeloperoxidase enzyme and the monocyte-specific CD14 antigen are co-expressed. Expression profiling evidenced that the observed trans-differentiation process was accompanied by a remarkable upregulation of the monocyte-related MafB transcription factor.
Collapse
Affiliation(s)
- M Montanari
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The hemoglobin disorders of beta-thalassemia and sickle cell disease together constitute the most prevalent group of human monogenic diseases. Although curative allogeneic stem cell transplantation therapy and palliative therapies have been developed for these disorders, the majority of patients still suffer significant morbidity and early mortality. The development of therapeutic approaches based on genetic manipulation of autologous stem cells therefore remains an attractive alternative. In the past 4 years, significant advances have been made toward this goal using lentiviral vectors to obtain high-level expression of complex globin gene cassettes. Therapeutic correction in murine models of both beta-thalassemia and sickle cell anemia has been achieved using this approach. These advances, coupled with progress in the ability to achieve in vivo selection of genetically modified cells, can now be evaluated in the well-developed nonhuman primate autologous transplant model. The goal in these studies is to provide preclinical safety and efficacy data prior to human clinical trials in order to maximize the likelihood of success in the context of an acceptable risk to benefit ratio. Here we review progress in each of these areas.
Collapse
Affiliation(s)
- Derek A Persons
- Department of Hematology/Oncology, Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | | |
Collapse
|
20
|
Mountford JC, Vanin EF, Hayden RE, Bunce CM. All-trans retinoic acid increases transgene expression in MSCV-transduced cells, via a mechanism that is retinoid receptor dependent but independent of cellular differentiation. Hum Gene Ther 2005; 16:132-8. [PMID: 15703496 DOI: 10.1089/hum.2005.16.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Treatment of MSCV-GFP-transduced HL60 promyelocytic cells with all-trans retinoic acid (ATRA) resulted in a significant increase in GFP expression. The increased GFP expression was observed by 16 hr and was dependent on de novo protein production. This effect was specific to ATRA and unrelated to cell differentiation because it was not induced by dimethyl sulfoxide. Furthermore, a similar increase in GFP expression was observed in MSCV-GFP-transfected K562 cells, which do not differentiate when exposed to ATRA. Significantly increased GFP expression was seen at doses as low as 0.5 nM ATRA and was abrogated by AGN193109, an antagonist of retinoid signaling. We therefore conclude that this increase in gene expression is mediated by retinoic acid receptors. The long terminal repeat (LTR) region of MSCV contains candidate retinoic acid response elements and response elements for the ATRA-inducible transcription factor C/EBPalpha. We suggest that the increase in GFP expression is driven by the action of ATRA-activated host cell transcription factors. These findings offer a method to increase the expression of retroviral transgenes either in vitro or in vivo by treatment with low doses of retinoic acid that are clinically achievable and well tolerated. This use of inducible host cell transcription factors offers an alternative to engineering novel LTR regulatory sequences in order to increase transgene expression.
Collapse
Affiliation(s)
- Joanne C Mountford
- Section of Experimental Haematology and Haemopoietic Stem Cells, Division of Cancer Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom.
| | | | | | | |
Collapse
|
21
|
Malik P, Arumugam PI. Gene Therapy for beta-thalassemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2005:45-50. [PMID: 16304358 DOI: 10.1182/asheducation-2005.1.45] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Gene transfer for beta-thalassemia requires gene transfer into hematopoietic stem cells using integrating vectors that direct regulated expression of beta globin at therapeutic levels. Among integrating vectors, oncoretroviral vectors carrying the human beta-globin gene and portions of the locus control region (LCR) have suffered from problems of vector instability, low titers and variable expression. In recent studies, human immunodeficiency virus-based lentiviral (LV) vectors were shown to stably transmit the human beta-globin gene and a large LCR element, resulting in correction of beta-thalassemia intermedia in mice. Several groups have since demonstrated correction of the mouse thalassemia intermedia phenotype, with variable levels of beta-globin expression. These levels of expression were insufficient to fully correct the anemia in thalassemia major mouse model. Insertion of a chicken hypersensitive site-4 chicken insulator element (cHS4) in self-inactivating (SIN) LV vectors resulted in higher and less variable expression of human beta-globin, similar to the observations with cHS4-containing retroviral vectors carrying the human gamma-globin gene. The levels of beta-globin expression achieved from insulated SIN-LV vectors were sufficient to phenotypically correct the thalassemia phenotype from 4 patients with human thalassemia major in vitro, and this correction persisted long term for up to 4 months, in xeno-transplanted mice in vivo. In summary, LV vectors have paved the way for clinical gene therapy trials for Cooley's anemia and other beta-globin disorders. SIN-LV vectors address several safety concerns of randomly integrating viral vectors by removing viral transcriptional elements and providing lineage-restricted expression. Flanking the proviral cassette with chromatin insulator elements, which additionally have enhancer-blocking properties, may further improve SIN-LV vector safety.
Collapse
Affiliation(s)
- Punam Malik
- Children's Hospital Los Angeles, 4650 Sunset Blvd., M.S. #45, Los Angeles, CA 90027, USA.
| | | |
Collapse
|
22
|
Werner M, Kraunus J, Baum C, Brocker T. B-cell-specific transgene expression using a self-inactivating retroviral vector with human CD19 promoter and viral post-transcriptional regulatory element. Gene Ther 2004; 11:992-1000. [PMID: 15029232 DOI: 10.1038/sj.gt.3302255] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Retroviral gene transfer resulting in transgene expression selectively restricted to specific cell lineages would be desirable for many gene therapeutic applications. Such transcriptional targeting of retroviruses can be accomplished by employing eukaryotic control elements in self-inactivating (SIN) retroviral vectors, but use of these vectors is complicated by an accompanying reduction in viral titers. To overcome this restriction and address the influence of the post-transcriptional regulatory element of the Woodchuck hepatitis virus (WPRE) on viral titers and transgene expression, we developed SIN-vectors with and without WPRE. Using the enhancer-promoter of the Spleen Focus Forming virus (SFFV) to direct eGFP expression to multiple hematopoietic lineages, we show that WPRE significantly (>10 x) increased viral titers (>10(6) per ml of unconcentrated supernatant) and transgene expression in NIH3T3 cells in vitro. Gene expression in vivo was significantly lowered in lymphoid cells, but not in myeloid cells when WPRE was present. Furthermore, the use of WPRE in combination with the B-cell lineage-specific CD19 promoter significantly increased viral titers and allowed targeting of transgene expression by SIN-vectors specifically to B cells throughout their development in primary and secondary lymphoid organs.
Collapse
Affiliation(s)
- M Werner
- Institute for Immunology, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | |
Collapse
|
23
|
Richardson TB, Kaspers J, Porter CD. Retroviral hybrid LTR vector strategy: functional analysis of LTR elements and generation of endothelial cell specificity. Gene Ther 2004; 11:775-83. [PMID: 14999228 DOI: 10.1038/sj.gt.3302220] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transcriptional targeting is an important aspect of developing gene therapy vectors in order to restrict transgene expression to selected target cells. One approach, when using retroviral vectors, is to replace viral transcriptional control elements within the long terminal repeat (LTR) with sequences imparting the desired specificity. We have developed such hybrid LTR retroviruses, incorporating sequences from each of the human promoters for flt-1, ICAM-2 and KDR, as part of our antivascular cancer gene therapy strategy targeting tumour endothelial cells. The chosen fragments were used to replace the enhancer or combined enhancer and proximal promoter regions of the viral LTR. All showed activity in primary human breast microvascular endothelial cells, with viruses incorporating ICAM-2 sequences exhibiting the greatest specificity versus nonendothelial cells in vitro and a marked alteration of specificity towards endothelial cells in a subcutaneous xenograft model in vivo. Moreover, our study documents the effect of enhancer and/or proximal promoter deletion on LTR activity and reports that differential dependence in different cell lines can give the false impression of specificity if experiments are not adequately controlled. This finding also has implications for other retroviral vector designs seeking to provide transcriptional specificity and for their safety with respect to prevention of gene activation at sites of proviral integration.
Collapse
|
24
|
Testa A, Lotti F, Cairns L, Grande A, Ottolenghi S, Ferrari G, Ronchi A. Deletion of a Negatively Acting Sequence in a Chimeric GATA-1 Enhancer-Long Terminal Repeat Greatly Increases Retrovirally Mediated Erythroid Expression. J Biol Chem 2004; 279:10523-31. [PMID: 14701820 DOI: 10.1074/jbc.m313638200] [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: 11/06/2022] Open
Abstract
The locus control region of the beta-globin gene cluster has been used previously to direct erythroid expression of globin genes from retroviral vectors for the purpose of gene therapy. Short erythroid regulatory elements represent a potentially valuable alternative to the locus control region. Among them, the GATA-1 enhancer HS2 was used to replace the retroviral enhancer within the 3'-long terminal repeat (LTR) of the retroviral vector SFCM, converting it into an erythroid-specific regulatory element. In this work, we have functionally studied an additional GATA-1 enhancer, HS1. HS1 participates in the transcriptional autoregulation of GATA-1 through an essential GATA-binding site that is footprinted in vivo. In this work we identified within HS1 a new in vivo footprinted region, and we showed that this sequence indeed binds a nuclear protein in vitro. Addition of HS1 to HS2 within the LTR of SFCM significantly improves the expression of a reporter gene. The deletion of the newly identified footprinted sequence in the retroviral construct further increases expression up to a level almost equal to that of the wild type retroviral LTR, without loss of erythroid specificity, suggesting that this sequence may act as a negative regulatory element. An improved vector backbone, MDeltaN, allows even better expression from the new GATA cassette. These results suggest that substantial improvement of overall expression can be achieved by the combination of multiple changes in both regulatory elements and vectors.
Collapse
Affiliation(s)
- Anna Testa
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, 20126 Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Hlavaty J, Stracke A, Klein D, Salmons B, Günzburg WH, Renner M. Multiple modifications allow high-titer production of retroviral vectors carrying heterologous regulatory elements. J Virol 2004; 78:1384-92. [PMID: 14722293 PMCID: PMC321378 DOI: 10.1128/jvi.78.3.1384-1392.2004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor-specific expression of therapeutic genes is a prerequisite in many approaches to retrovirus-mediated cancer gene therapy. However, tissue specificity is often associated with a reduction in viral titer. To overcome this problem, we constructed a series of murine leukemia virus (MLV)-based retroviral promoter conversion (ProCon) vectors carrying either the simian virus 40 poly(A) signal trimer (3pA) inserted in the 3' long terminal repeat (LTR) of these vectors or the human cytomegalovirus enhancer region (CMVe) inserted 5' and 3' of the retroviral LTRs. Furthermore, an extended AT stretch/attachment site (AT/att) of wild-type MLV was introduced into the vector. In the vector-producing cells, insertion of the CMVe and/or the 3pA resulted in a three- to fourfold-enhanced marker gene expression compared to the parental vector, whereas insertion of the AT/att gave a slight decrease in expression. The combination of all three modifications had no additional effects. In contrast, however, neomycin selection of infected cells revealed only a slight increase in virus titer with vectors carrying the 3pA modification; the titer was increased by 1 with vectors containing the extended AT/att, although the viral DNA copy numbers in infected cells were similar with both types of vectors. Thus, insufficient integration rather than insufficient reverse transcription and/or production of virus RNA is the major cause for the low titer obtained with the ProCon vectors. The combination of all three modifications resulted in a 2- to 3-log increase in the virus titer. These modifications result in expression targeted ProCon vectors with titers similar to those of nonmodified MLV-based vectors.
Collapse
Affiliation(s)
- Juraj Hlavaty
- Institute of Virology, University of Veterinary Medicine, A-1210 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
26
|
Sadat MA, Pech N, Saulnier S, Leroy BA, Hossle JP, Grez M, Dinauer MC. Long-term high-level reconstitution of NADPH oxidase activity in murine X-linked chronic granulomatous disease using a bicistronic vector expressing gp91phox and a Delta LNGFR cell surface marker. Hum Gene Ther 2003; 14:651-66. [PMID: 12804147 DOI: 10.1089/104303403321618164] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A murine model of X-linked chronic granulomatous disease (X-CGD), an inherited immune deficiency with absent phagocyte NADPH oxidase activity caused by defects in the gp91(phox) gene, was used to evaluate a bicistronic retroviral vector in which expression of human gp91(phox) and a linked gene for Delta LNGFR, a truncated form of human low-affinity nerve growth factor receptor, are under the control of a spleen focus-forming virus long-terminal repeat (LTR). Four independent cohorts of 11-Gy irradiated X-CGD mice (total, 22 mice) were transplanted with or without preselection of transduced X-CGD bone marrow (BM). Transplanted mice had high-level correction of neutrophil gp91(phox) expression and reconstitution of NADPH oxidase activity. Expression lasted for at least 14 months in primary transplants, and persisted in secondary and tertiary transplants. Both gp91(phox) and Delta LNGFR were detected on circulating granulocytes, lymphocytes, lymphoid, and (for Delta LNGFR) red blood cells. Mice receiving transduced bone marrow [BM] preselected ex vivo for Delta LNGFR expression had high-level (= 80%) reconstitution with transduced cells, with an improved fraction of oxidase-corrected neutrophils posttransplant. Analysis of secondary and tertiary CFU-S showed that silencing of individual provirus integrants can occur even after preselection for Delta LNGFR prior to transplantation, and that persistent provirus expression was associated with multiple integration sites in most cases. No obvious adverse consequences of transgenic protein expression were observed.
Collapse
Affiliation(s)
- Mohammed A Sadat
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics (Hematology/Oncology), James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Cairns L, Cirò M, Minuzzo M, Morlé F, Starck J, Ottolenghi S, Ronchi A. Induction of globin mRNA expression by interleukin-3 in a stem cell factor-dependent SV-40 T-antigen-immortalized multipotent hematopoietic cell line. J Cell Physiol 2003; 195:38-49. [PMID: 12599207 DOI: 10.1002/jcp.10241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Erythropoiesis requires the stepwise action on immature progenitors of several growth factors, including stem cell factor (SCF), interleukin 3 (IL-3), and erythropoietin (Epo). Epo is required to sustain proliferation and survival of committed progenitors and might further modulate the level of expression of several erythroid genes, including globin genes. Here we report a new SCF-dependent immortalized mouse progenitor cell line (GATA-1 ts SCF) that can also grow in either Epo or IL-3 as the sole growth factor. When grown in SCF, these cells show an "open" chromatin structure of the beta-globin LCR, but do not significantly express globin. However, Epo or IL-3 induce globin expression and are required for its maintainance. This effect of IL-3 is unexpected as IL-3 was previously reported either to be unable to induce hemoglobinization, or even to antagonize it. This suggests that GATA-1 ts SCF cells may have progressed to a stage in which globin genes are already poised for expression and only require signal(s) that can be elicited by either Epo or IL-3. Through the use of inhibitors, we suggest that p38 may be one of the molecules modulating induction and maintenance of globin expression.
Collapse
Affiliation(s)
- Linda Cairns
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Tagliafico E, Siena M, Zanocco-Marani T, Manfredini R, Tenedini E, Montanari M, Grande A, Ferrari S. Requirement of the coiled-coil domains of p92(c-Fes) for nuclear localization in myeloid cells upon induction of differentiation. Oncogene 2003; 22:1712-23. [PMID: 12642874 DOI: 10.1038/sj.onc.1206279] [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: 11/08/2022]
Abstract
The nonreceptor tyrosine kinase Fes is implicated in myeloid cells differentiation. It has been observed that its localization can be cytoplasmic, perinuclear, or nuclear. To further characterize this point, we studied Fes subcellular localization in myeloid cell lines (HL60 and K562) and in COS1 cells. Fes was observed in both the nucleus and the cytoplasm of HL60, K562 cells overexpressing Fes and only in the cytoplasm of COS1 cells, suggesting that nuclear localization is cell context dependent. Moreover, in myeloid cells, the treatment with differentiation-inducing agents such as retinoic acid, phorbol esters and vitamin D, is followed by an increase of the oligomeric form of Fes in the nucleus. In fact, oligomerization seems to be necessary for translocation to occur, since Fes mutants missing the coiled-coil domains are not able to form oligomers and fail to localize in the nucleus. The active form of Fes is tyrosine phosphorylated; however, phosphorylation is not required for Fes to localize in the nucleus, since tyrosine kinase inhibitors do not block the translocation process.
Collapse
Affiliation(s)
- Enrico Tagliafico
- Dipartimento di Scienze Biomediche, Universitá di Modena e Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ballas CB, Zielske SP, Gerson SL. Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem 2002; 38:20-8. [PMID: 12046846 DOI: 10.1002/jcb.10127] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
There is excitement generated almost daily about the possible uses of stem cells to treat human disease. Much of the interest of late is generated by embryonic stem cells (ESCs). As exciting as ESCs may be, they are quite controversial for moral reasons, given their source. They are also scientifically controversial since they are much less well understood than the original, long-standing, and clinically successful hematopoietic stem cell (HSC). HSCs have the distinct advantage of being reasonably well characterized and have been proven in the clinic. They can be isolated by simple procedures directly from the bone marrow or from peripheral blood after being stimulated (mobilized). They can then be manipulated and delivered to a patient, often producing a cure. Their biology provides the paradigm by which all other stem cells are judged, and they have little in the way of moral controversy surrounding them given they are isolated from adults who have consented to the procedure. Another putative stem cell has gained momentum in the last few years; the mesenchymal stem cell (MSC). MSCs appear to have much in common with HSCs. They were originally characterized from bone marrow, are capable of differentiating along multiple lineages and, at least in vitro, have significant expansion capability. Unlike HSCs, they have not yet been definitively shown to function as stem cells, despite their ability to differentiate into various mesenchymal cell types under the right culture conditions. Still, there is mounting evidence these cells may be useful, if not as true stem cells then at least as vehicles for emerging cell and gene therapies, especially in the field of tissue engineering. While this is an important endpoint, it is more important to thoroughly understand stem cell biology. That understanding can then be applied toward the ultimate goal of using these cells not just for various forms of therapy, but rather as a tool to discover the mechanisms and means to bring about directed repair and regeneration of damaged or diseased tissues and organs. The excitement of HSCs and MSCs has been muted somewhat by the excitement surrounding ESCs, primarily due to the fact HSCs and MSCs are viewed as limited to specific cell types while ESCs could potentially be applied to any cell type. Recent information indicates HSCs, MSCs, and other cells in general may have more universal differentiation abilities than previously thought.
Collapse
Affiliation(s)
- Christopher B Ballas
- Division of Hematology/Oncology, Comprehensive Cancer Center at University Hospitals of Cleveland and Case Western Reserve University, Ohio, USA
| | | | | |
Collapse
|
30
|
Ghazizadeh S, Doumeng C, Taichman LB. Durable and stratum-specific gene expression in epidermis. Gene Ther 2002; 9:1278-85. [PMID: 12224010 DOI: 10.1038/sj.gt.3301800] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2002] [Accepted: 05/07/2002] [Indexed: 11/08/2022]
Abstract
A number of genetic disorders are manifested in cutaneous epithelium and gene therapy approaches for treatment of such diseases are being considered. A successful gene therapy protocol requires durable and correctly targeted gene expression within the tissue. The continuous renewal and high levels of compartmentalization in epidermis are two challenges for a successful gene therapy of skin disorders. For those disorders which affect the upper layers of epidermis, vectors must be available that target stem cells, but remain silent until the progeny of these cells undergo differentiation. To explore the potential of long-term and targeted vector expression in epidermis, a hybrid retroviral vector encoding the reporter enhanced green fluorescent protein (EGFP) was constructed. The viral enhancer in the long terminal repeat of the vector was replaced with a 510-bp enhancer element of the human involucrin promoter. Keratinocyte-specific expression directed by the hybrid vector was demonstrated in culture and suprabasal-specific expression was observed in organotypic human epidermal cultures. In vivo transduction of mouse skin with this hybrid vector indicated long-term and stratum-specific expression of the transgene in mouse epidermis. The design of similar vectors for various gene therapy applications constitutes an important step toward clinically effective gene therapy.
Collapse
Affiliation(s)
- S Ghazizadeh
- Department of Oral Biology and Pathology, Westchester Hall (Room 100), SUNY at Stony Brook, Stony Brook, NY 11794-8702, USA
| | | | | |
Collapse
|
31
|
Li Z, Fehse B, Schiedlmeier B, Düllmann J, Frank O, Zander AR, Ostertag W, Baum C. Persisting multilineage transgene expression in the clonal progeny of a hematopoietic stem cell. Leukemia 2002; 16:1655-63. [PMID: 12200677 DOI: 10.1038/sj.leu.2402619] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2002] [Accepted: 04/23/2002] [Indexed: 11/08/2022]
Abstract
Many applications of hematopoietic gene therapy require selection for clones with active transgene expression. However, it was unclear whether the clonal progeny of a retrovirally transduced hematopoietic stem cell would be capable of maintaining transgene expression through serial repopulation and multilineage differentiation. Such investigations require simultaneous analyses of clonality, multilineage activity and transgene copy numbers. Using a mouse model, the present study demonstrates that a single hematopoietic stem cell expressing a marker gene from one or two insertions of a simple retroviral vector actively maintains multilineage transgene expression in the vast majority (80-99%) of bone marrow and peripheral blood cells. Gene expression persisted through serial transplantations for at least 97 weeks post gene transfer and was observed in the lymphoid (B, T and NK cells), myeloid (CD11b(+), Gr-1(+)), erythroid (Ter119(+), mature red blood cells) and megakaryocytic (as indicated by platelets) progeny. Therefore, a single immunoselection for hematopoietic stem cells expressing the transgene in vivo was sufficient to establish a completely chimeric hematopoiesis. These observations imply that the retroviral vectors used in this study contain cis-elements that mediate expression through massive clonal expansion and multilineage differentiation, provided the insertion occurred in genetic loci permissive for expression in hematopoietic stem cells.
Collapse
Affiliation(s)
- Z Li
- Experimental Cell Therapy, Department of Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Ijichi S, Ijichi N, Osame M, Hall WW. In vivo induction of human immunodeficiency virus type 1 entry into nucleus-free cells by CD4 gene transfer to hematopoietic stem cells: a hypothetical possible strategy for therapeutic intervention. Med Hypotheses 2002; 59:24-34. [PMID: 12160677 DOI: 10.1016/s0306-9877(02)00194-9] [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: 11/22/2022]
Abstract
As a useful alternative to employing soluble CD4 to inhibit binding of human immunodeficiency virus type 1 (HIV-1) to target cells, the introduction of CD4-bearing erythrocyte has been proposed by two study groups (see Refs. (5,6)). Prominently, Nicolau and colleagues demonstrated that the electroinserted CD4 molecules in the membranes of erythrocytes are capable of mediating HIV-1 entry. The implications of the studies are that inactivation of the integration-dependent retrovirus by the facilitation of entry into the nucleus-free cells, referred to as 'fake host trap' or 'host cell decoy', may be a possible therapeutic approach. Here we expand this concept to include genetic modification of autologous hematopoietic stem cells and review the relevant theoretical basis. Effective application of molecular technologies to induce partial replacement of hematopoiesis may be critical for this strategy.
Collapse
Affiliation(s)
- S Ijichi
- Nagahama Shinryojyo, Shimokoshiki-mura, Satsuma-gun, Kagoshima, Japan.
| | | | | | | |
Collapse
|
33
|
Matthes TW, Kindler V, Leuba F, French LE, Chapuis B, Beris P, Piguet V. Optimized lentiviral transduction of erythroid precursors from healthy adults and patients with myelodysplastic syndromes. Leukemia 2002; 16:1319-23. [PMID: 12094256 DOI: 10.1038/sj.leu.2402527] [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] [Received: 08/09/2001] [Accepted: 01/22/2002] [Indexed: 11/09/2022]
Abstract
Lentivectors, derived from human immunodeficiency virus-1 (HIV-1), represent a novel investigational and therapeutic tool for targeting hematopoietic progenitor cells. We describe a new protocol whereby we achieved a highly efficient lentiviral transduction of erythroid precursor cells originating from the bone marrow of healthy adults and patients with myelodysplastic syndromes (MDS). CD34(+) stem cells from healthy subjects were cultured with erythropoietin, IL-3 and stem cell factor, and thereby expanded approximately 300-fold. When these cultures were transduced with a lentiviral vector expressing GFP as a reporter gene, 70% glycophorin(+) cells were GFP(+). Although proliferation and levels of transduction were reduced in cultures of CD34(+) stem cells from patients with myelodysplastic syndromes, 50% of glycophorin(+) cells became GFP(+), amongst which 30% were sideroblastic erythroid precursors. This study demonstrates that lentiviral vectors are capable of efficiently transducing MDS precursors and offers new perspectives to investigate the influence of specific genes on normal erythroid differentiation. This may eventually help to correct defects in patients suffering from myelodysplastic syndromes.
Collapse
Affiliation(s)
- T W Matthes
- Division of Hematology, Department of Internal Medicine, University Hospital Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
34
|
Urban C, Preisegger KH, Krugluger W, Hopmeier P, Schwinger W, Lackner H, Kerbl R, Dornbusch HJ, Benesch M. Allogeneic bone marrow transplantation in a child with hemoglobinopathy olmsted. J Pediatr Hematol Oncol 2002; 24:417-9. [PMID: 12142796 DOI: 10.1097/00043426-200206000-00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors describe the first successful bone marrow transplant for the treatment of hemoglobinopathy Olmsted in a boy who presented with severe transfusion-dependent hemolytic anemia and jaundice at age 4 months. He received bone marrow from an HLA-identical sibling with normal hemoglobin electrophoresis after conditioning with busulfan, cyclophosphamide, and antithymocyte globulin when he was 18 months old. The posttransplant course was uneventful. Two years after transplantation the patient has a normal hemoglobin level without evidence of hemolysis. DNA analysis shows 100% chimerism of donor cell origin, confirming full engraftment with normal hematopoietic cells.
Collapse
Affiliation(s)
- Christian Urban
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Graz School of Medicine, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- L E Ailles
- Laboratory for Gene Transfer and Therapy, Institute for Cancer Research and Treatment, University of Torino Medical School, Candiolo, Torino, Italy
| | | |
Collapse
|
36
|
Grande A, Montanari M, Tagliafico E, Manfredini R, Marani TZ, Siena M, Tenedini E, Gallinelli A, Ferrari S. Physiological levels of 1α, 25 dihydroxyvitamin D3 induce the monocytic commitment of CD34+ hematopoietic progenitors. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.4.641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Alexis Grande
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Monica Montanari
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Enrico Tagliafico
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Rossella Manfredini
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Tommaso Zanocco Marani
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Michela Siena
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Elena Tenedini
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| | - Andrea Gallinelli
- Dipartimento di Scienze Ginecologiche, Ostetriche e Pediatriche, Università di Modena e Reggio Emilia, Modena, Italy
| | - Sergio Ferrari
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy and
| |
Collapse
|
37
|
Lotti F, Menguzzato E, Rossi C, Naldini L, Ailles L, Mavilio F, Ferrari G. Transcriptional targeting of lentiviral vectors by long terminal repeat enhancer replacement. J Virol 2002; 76:3996-4007. [PMID: 11907239 PMCID: PMC136069 DOI: 10.1128/jvi.76.8.3996-4007.2002] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene therapy of many genetic diseases requires permanent gene transfer into self-renewing stem cells and restriction of transgene expression to specific progenies. Human immunodeficiency virus (HIV)-derived lentiviral vectors are very effective in transducing rare, nondividing stem cell populations (e.g., hematopoietic stem cells) without altering their long-term repopulation and differentiation capacities. We developed a strategy for transcriptional targeting of lentiviral vectors based on replacing the viral long terminal repeat (LTR) enhancer with cell lineage-specific, genomic control elements. An upstream enhancer (HS2) of the erythroid-specific GATA-1 gene was used to replace most of the U3 region of the LTR, immediately upstream of the HIV type 1 (HIV-1) promoter. The modified LTR was used to drive the expression of a reporter gene (the green fluorescent protein [GFP] gene), while a second gene (a truncated form of the p75 nerve growth factor receptor [DeltaLNGFR]) was placed under the control of an internal constitutive promoter to monitor cell transduction, or to immunoselect transduced cells, independently from the expression of the targeted promoter. The transcriptionally targeted vectors were used to transduce cell lines, human CD34+ hematopoietic stem-progenitor cells, and murine bone marrow (BM)-repopulating stem cells. Gene expression was analyzed in the stem cell progeny in vitro and in vivo after xenotransplantation into nonobese diabetic-SCID mice or BM transplantation in coisogenic mice. The modified LTR directed high levels of transgene expression specifically in mature erythroblasts, in a TAT-independent fashion and with no alteration in titer, infectivity, and genomic stability of the lentiviral vector. Expression from the modified LTR was higher, better restricted, and showed less position-effect variegation than that obtained by the same combination of enhancer-promoter elements placed in a conventional, internal position. Cloning of the woodchuck hepatitis virus posttranscriptional regulatory element at a defined position in the targeted vector allowed selective accumulation of the genomic transcripts with respect to the internal RNA transcript, with no loss of cell-type restriction. A critical advantage of this targeting strategy is the use of a spliced, major viral transcript to express a therapeutic gene and that of an internal, independently regulated promoter to express an additional gene for either cell marking or in vivo selection purposes.
Collapse
Affiliation(s)
- Francesco Lotti
- TIGET, Istituto Scientifico H. San Raffaele, 20132 Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Cui Y, Golob J, Kelleher E, Ye Z, Pardoll D, Cheng L. Targeting transgene expression to antigen-presenting cells derived from lentivirus-transduced engrafting human hematopoietic stem/progenitor cells. Blood 2002; 99:399-408. [PMID: 11781219 DOI: 10.1182/blood.v99.2.399] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) represent an important target for the treatment of various blood disorders. As the source of critical cells within the immune system, genetic modification of HSCs can also be used to modulate immune responses. The effectiveness of HSC-mediated gene therapy largely depends on efficient gene delivery into long-term repopulating progenitors and targeted transgene expression in an appropriate progeny of the transduced pluripotent HSCs. Self-inactivating (SIN) lentiviral vectors have been demonstrated to be capable of transducing mitotically inactive cells, including HSCs, and accommodating a nonviral promoter to control the transgene expression in transduced cells. In this study, we constructed 2 SIN lentiviral vectors, EF.GFP and DR.GFP, to express the green fluorescent protein (GFP) gene controlled solely by the promoter of either a housekeeping gene EF-1alpha or the human HLA-DRalpha gene, which is selectively expressed in antigen-presenting cells (APCs). We demonstrated that both vectors efficiently transduced human pluripotent CD34+ cells capable of engrafting nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. When the EF.GFP vector was used, constitutive high-level GFP expression was obtained in all the human HSC progeny detectable in NOD/SCID mice and in subsequent in vitro differentiation assays, indicating that engrafting human HSCs have been transduced. In contrast, the DR.GFP vector mediated transgene expression specifically in human HLA-DR+ cells and highly in differentiated dendritic cells (DCs), which are critical in regulating immunity. Furthermore, human DCs derived from transduced and engrafted human cells potently stimulated allogeneic T-cell proliferation. This study demonstrated successful targeting of transgene expression to APCs/DCs after stable gene transduction of pluripotent HSCs.
Collapse
Affiliation(s)
- Yan Cui
- Division of Immunology and Hematopoiesis, Johns Hopkins Oncology Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
39
|
Wahlers A, Zipfel PF, Schwieger M, Ostertag W, Baum C. In vivo analysis of retroviral enhancer mutations in hematopoietic cells: SP1/EGR1 and ETS/GATA motifs contribute to long terminal repeat specificity. J Virol 2002; 76:303-12. [PMID: 11739695 PMCID: PMC135690 DOI: 10.1128/jvi.76.1.303-312.2002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The objective of this work was to identify, in the context of chromosomally integrated DNA, the contribution of defined transcription factor binding motifs to the function of a complex retrovirus enhancer in hematopoietic cells in vivo. Repopulating murine hematopoietic cells were transduced with equal gene dosages of replication-incompetent retrovirus vectors encoding enhanced green fluorescent protein. Enhancer sequences were derived from mouse spleen focus-forming virus. Destruction of GC-rich sites representing overlapping targets for SP1 or EGR1 uniformly attenuated gene expression (approximately 25 to 70% of wild-type levels) in all hematopoietic lineages, as shown by multicolor flow cytometry of peripheral blood and bone marrow cells at various time points posttransplantation. In contrast, a point mutation within a dual ETS/GATA motif that abolished transactivation by ETS factors but not by GATA-1 slightly increased activity in erythroid cells and significantly attenuated enhancer function in T lymphocytes. This study shows that controlled gene transfer in transplantable hematopoietic cells allows a functional analysis of distinct cis elements within a complex retrovirus enhancer, as required for the characterization and engineering of various cellular and viral regulatory sequences in basic research and gene therapy.
Collapse
Affiliation(s)
- Anke Wahlers
- Department of Cell and Virus Genetics, Heinrich Pette Institute, D-20251 Hamburg, Germany
| | | | | | | | | |
Collapse
|
40
|
Abstract
Current treatment of solid tumors is limited by severe adverse effects, resulting in a narrow therapeutic index. Therefore, cancer gene therapy has emerged as a targeted approach that would significantly reduce undesired side effects in normal tissues. This approach requires a clear understanding of the molecular biology of both the malignant clone and the biological vectors that serve as vehicles to target cancer cells. In this review we discuss novel approaches for conditional gene expression in cancer cells. Targeting transgene expression to malignant tissues requires the use of specific regulatory elements including promoters based on tumor biology, tissue-specific promoters and inducible regulatory elements. We also discuss the regulation of both replication and transgene expression by conditionally-replicative viruses. These approaches have the potential to restrict the expression of transgenes exclusively to tissues of interest and thereby to increase the therapeutic index of future vectors for cancer gene therapy.
Collapse
Affiliation(s)
- Y S Haviv
- Division of Human Gene Therapy, Departments of Medicine, Surgery and Pathology, University of Alabama at Birmingham, 1824 6th Avenue South, Birmingham, AL 35294, USA
| | | |
Collapse
|
41
|
Fairbairn LJ, Ewing JC. Prospects for gene therapy using haemopoietic stem cells. Best Pract Res Clin Haematol 2001; 14:823-34. [PMID: 11924924 DOI: 10.1053/beha.2001.0175] [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: 01/25/2023]
Abstract
Gene therapy has thus far promised much and delivered little. Much of this has been due to deficiencies in the reagents and methodologies employed in early clinical trials. Recent technological advances in vectors and haemopoietic stem cell manipulation, coupled with improved pre-clinical assays of gene transfer and expression in re-populating stem cells give cause for greater optimism. Here we review these advances and indicate areas requiring further development before clinical gene therapy in the haemopoietic system becomes a widely applicable treatment modality.
Collapse
Affiliation(s)
- L J Fairbairn
- Cancer Research Campaign Gene Therapy Group, Paterson Institute for Cancer Research, Manchester, UK
| | | |
Collapse
|
42
|
Moreau-Gaudry F, Xia P, Jiang G, Perelman NP, Bauer G, Ellis J, Surinya KH, Mavilio F, Shen CK, Malik P. High-level erythroid-specific gene expression in primary human and murine hematopoietic cells with self-inactivating lentiviral vectors. Blood 2001; 98:2664-72. [PMID: 11675336 DOI: 10.1182/blood.v98.9.2664] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Use of oncoretroviral vectors in gene therapy for hemoglobinopathies has been impeded by low titer vectors, genetic instability, and poor expression. Fifteen self- inactivating (SIN) lentiviral vectors using 4 erythroid promoters in combination with 4 erythroid enhancers with or without the woodchuck hepatitis virus postregulatory element (WPRE) were generated using the enhanced green fluorescent protein as a reporter gene. Vectors with high erythroid-specific expression in cell lines were tested in primary human CD34(+) cells and in vivo in the murine bone marrow (BM) transplantation model. Vectors containing the ankyrin-1 promoter showed high-level expression and stable proviral transmission. Two vectors containing the ankyrin-1 promoter and 2 erythroid enhancers (HS-40 plus GATA-1 or HS-40 plus 5-aminolevulinate synthase intron 8 [I8] enhancers) and WPRE expressed at levels higher than the HS2/beta-promoter vector in bulk unilineage erythroid cultures and individual erythroid blast-forming units derived from human BM CD34(+) cells. Sca1(+)/lineage(-) Ly5.1 mouse hematopoietic cells, transduced with these 2 ankyrin-1 promoter vectors, were injected into lethally irradiated Ly5.2 recipients. Eleven weeks after transplantation, high-level expression was seen from both vectors in blood (63%-89% of red blood cells) and erythroid cells in BM (70%-86% engraftment), compared with negligible expression in myeloid and lymphoid lineages in blood, BM, spleen, and thymus (0%-4%). The I8/HS-40-containing vector encoding a hybrid human beta/gamma-globin gene led to 43% to 113% human gamma-globin expression/copy of the mouse alpha-globin gene. Thus, modular use of erythroid-specific enhancers/promoters and WPRE in SIN-lentiviral vectors led to identification of high-titer, stably transmitted vectors with high-level erythroid-specific expression for gene therapy of red cell diseases.
Collapse
Affiliation(s)
- F Moreau-Gaudry
- Children's Hospital Los Angeles, University of Southern California School of Medicine, Los Angeles 90027, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Inherited disorders of hemoglobin remain desirable targets for genetically based therapies. That stem cell replacement reverses the phenotype of both thalassemia and sickle cell anemia has been well established through allogeneic bone marrow transplantation studies, yet significant toxicities and finite donor availability limit this approach to a minority of affected individuals. Genetically based strategies that have as their goal addition of a normal copy of the human beta-globin gene along with key regulatory sequences to autologous hematopoietic stem cells represent a viable alternative to allogeneic transplantation, but this approach has been impeded by formidable obstacles over the last decade. Large animal models have become the standard for the development of clinically relevant gene addition strategies, and significant progress in the techniques used to deliver potentially therapeutic genes has been achieved. The clinical application of such strategies may be close at hand, at least for disorders in which modest level, constitutive expression is sufficient to correct the phenotype. For the thalassemias and hemoglobinopathies, complex, regulated, lineage specific expression of the beta-globin gene at relatively high levels will be required. The discovery of the beta-globin locus control region renewed interest in the thalassemias and sickle cell anemia as targets for gene transfer, but difficulties in attaining high-titer vectors along with a tendency toward rearrangement when segments of the locus control region (LCR) were incorporated into retroviral vectors stalled further progress. Recent advances in vector construction have circumvented this problem and others limiting both gene transfer efficiency and regulation of transgene expression, offering new hope for clinical application.
Collapse
Affiliation(s)
- J Tisdale
- Molecular and Clinical Hematology Branch, National Institute of Diabetes and Digestive and Kidney Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
44
|
Wahlers A, Schwieger M, Li Z, Meier-Tackmann D, Lindemann C, Eckert HG, von Laer D, Baum C. Influence of multiplicity of infection and protein stability on retroviral vector-mediated gene expression in hematopoietic cells. Gene Ther 2001; 8:477-86. [PMID: 11313827 DOI: 10.1038/sj.gt.3301426] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2000] [Accepted: 01/15/2001] [Indexed: 11/08/2022]
Abstract
Using retroviral vectors encoding enhanced green fluorescent protein (EGFP), we addressed to what extent expression of retroviral transgenes in hematopoietic cells depends on the multiplicity of infection (MOI) and on the half-life of the encoded protein. We show that an elevation of the MOI not only elevates the frequency of transduced cells, but also increases transgene expression levels and reduces interanimal variability in vivo (hematopoietic cells of C57BL/6J mice analyzed 13 weeks after transplantation). This suggests that the MOI has to be carefully controlled and should be adapted as desired for clinical studies when evaluating vector performance in preclinical models. The impact of protein stability is demonstrated by comparing vectors expressing EGFP or a destabilized variant with a C-terminal PEST-sequence, d2EGFP. The loss of expression with d2EGFP was more pronounced in terminally differentiated cells of the peripheral blood (>30 fold) than in progenitor cells (five- to 10-fold), indicating a stronger transcription of the retroviral promoter in progenitor cells and a predominant role of protein inheritance over de novo synthesis of transgenic protein in mature blood cells. This analysis reveals an important and differentiation-dependent contribution of protein half-life to the expression of retroviral vectors in hematopoietic cells, establishes d2EGFP as a more accurate reporter for determination of vector transcription, and also suggests that preclinical data obtained under conditions of high transduction rates or with vectors expressing stable reporter proteins require careful interpretation.
Collapse
Affiliation(s)
- A Wahlers
- Department Cell and Virus Genetics, Heinrich-Pette-Institute, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Grande A, Montanari M, Manfredini R, Tagliafico E, Zanocco-Marani T, Trevisan F, Ligabue G, Siena M, Ferrari S, Ferrari S. A functionally active RARalpha nuclear receptor is expressed in retinoic acid non responsive early myeloblastic cell lines. Cell Death Differ 2001; 8:70-82. [PMID: 11313705 DOI: 10.1038/sj.cdd.4400771] [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] [Received: 02/03/2000] [Revised: 07/24/2000] [Accepted: 08/25/2000] [Indexed: 11/09/2022] Open
Abstract
Although all-trans retinoic acid (ATRA) can restore the differentiation capacity of leukemic promyelocytes, early leukemic myeloblasts are conversely not responsive to ATRA induced granulocytic differentiation. To assess whether this resistance to ATRA is related to an impaired function of the Retinoic Acid Receptor alpha (RARalpha), we performed an analysis of RARalpha expression and transactivation activity, in several myeloid leukemic cell lines, representative of different types of spontaneous acute myeloid leukemias. Our results indicate that a functionally active RARalpha nuclear receptor is expressed in all the analyzed cell lines, regardless of their differentiation capacity following exposure to ATRA. The observation that ATRA treatment is able to induce the expression of retinoic acid target genes, in late- but not in early-myeloblastic leukemic cells, raises the possibility that the differentiation block of these cells is achieved through a chromatin mediated mechanism. Acetylation is apparently not involved in this process, since the histone deacetylase inhibitor trichostatin A, is not able to restore the differentiation capacity of early leukemic myeloblasts. Further investigation is needed to clarify whether myeloid transcription factors, distinct to RARalpha, play a role in the resistance of these cells to ATRA treatment.
Collapse
MESH Headings
- Alkaline Phosphatase/pharmacology
- Blotting, Northern
- Blotting, Western
- Cell Differentiation/drug effects
- Cell Nucleus/chemistry
- Cell Nucleus/metabolism
- DNA/metabolism
- Dimerization
- Enhancer Elements, Genetic
- Flow Cytometry
- Gene Expression/drug effects
- Genetic Vectors/genetics
- Genetic Vectors/metabolism
- Genetic Vectors/pharmacology
- Humans
- Hydroxamic Acids/pharmacology
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Macrophage-1 Antigen/biosynthesis
- Phosphorylation/drug effects
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Retinoid X Receptors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcriptional Activation/physiology
- Tretinoin/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A Grande
- Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università di Modena e Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pan D, Aronovich E, McIvor RS, Whitley CB. Retroviral vector design studies toward hematopoietic stem cell gene therapy for mucopolysaccharidosis type I. Gene Ther 2000; 7:1875-83. [PMID: 11110421 DOI: 10.1038/sj.gt.3301298] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To optimize a gene transfer system for hematopoietic stem cell gene therapy of patients with mucopolysaccharidosis (MPS) type I, 10 retroviral vectors were constructed to express the human alpha-L-iduronidase (IDUA) cDNA. These vectors were designed to evaluate the potential effects of specific promoters, the addition of selectable markers, and the use of multiple promoters versus an internal ribosome entry site for expression of IDUA and selectable maker genes. The effect of vector design was investigated in primary patient fibroblasts (F(MPS)) or murine fibroblast cell lines; while overall comparison of transgene expression was determined in patients' peripheral blood lymphocytes (PBL(MPS)) and CD34+ progenitors (PBPC(MPS)). We observed that the human PGK promoter introduced the highest IDUA activity per 1% relative transgene frequency in F(MPS). Use of the same promoter to separately regulate both the therapeutic gene and a drug-resistance gene resulted in decreased expression of the unselected gene. Co-selection using bicistronic vectors not only increased the number of transductants, but also elevated transgene expression under selective pressure in transgene-positive progenitors. Bicistronic vector LP1CD overcame down-regulation and practically introduced the highest IDUA level in unselected PBL(MPS) and an intermediate level in PBPC(MPS). These studies provide a better understanding of factors contributing to efficient gene expression in hematopoietic cells.
Collapse
Affiliation(s)
- D Pan
- Department of Pediatrics, and Institute of Human Genetics, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
47
|
Halene S, Kohn DB. Gene therapy using hematopoietic stem cells: Sisyphus approaches the crest. Hum Gene Ther 2000; 11:1259-67. [PMID: 10890736 DOI: 10.1089/10430340050032366] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gene transfer targeting cells of the blood and immune system was one of the first areas of investigation in the field of gene therapy. Despite the encouraging results achieved in early studies using murine bone marrow, the task of gene transfer into human hematopoietic stem cells proved to be far more difficult. As a result, progress has been disappointingly slow and initial clinical trials generally failed to achieve significant levels of gene marking. The continued application of new advances in vectorology and hematopoietic stem cell biology has now led to improvements in preclinical models that are being translated into clinical trials. The progress and remaining problems are discussed in this review article.
Collapse
Affiliation(s)
- S Halene
- Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California School of Medicine, CA 90027, USA
| | | |
Collapse
|
48
|
Mavria G, Jäger U, Porter CD. Generation of a high titre retroviral vector for endothelial cell-specific gene expression in vivo. Gene Ther 2000; 7:368-76. [PMID: 10694818 DOI: 10.1038/sj.gt.3301093] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tumour growth is dependent upon a blood supply and is associated with the switch to the angiogenic phenotype. We are developing strategies for targeting gene expression to endothelial cells in the tumour vasculature. Recombinant retroviruses have been generated that incorporate regulatory sequences of the prepro-endothelin-1 (ppET1) promoter. Following reverse transcription and integration these modifications are duplicated in the proviral 5' LTR for transcription of the internal beta-galactosidase reporter gene. The titres and endothelial specificity of retroviral vectors harbouring different modifications have been analysed. In the optimal strategy, replacing the MLV enhancer with ppET1 promoter sequences containing the GATA and AP1 elements whilst maintaining sequences from the viral promoter resulted in endothelial cell-specific expression of the reporter gene, and viral titres comparable to those of the unmodified vector. A panel of endothelial and non-endothelial cells infected with the modified virus from a high titre producer clone showed a pattern of expression consistent with the activity of the endogenous ppET1 promoter. The modified LTR retained specificity in vivo, in subcutaneous tumours arising from the co-injection of tumour cells and irradiated virus producer cells. This simple model achieves high efficiency of transduction and can be used routinely for the screening of targeted retroviral vectors. Gene Therapy (2000) 7, 368-376.
Collapse
Affiliation(s)
- G Mavria
- Chester Beatty Laboratories, Institute of Cancer Research, 237, Fulham Road, London SW3 6JB, UK
| | | | | |
Collapse
|