Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Leukemia ; 33(6): 1542, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30787431

ABSTRACT

In the original version of this article there was a mistake in the spelling of the author Sujoy Ghosh, originally spelt Sujoy Gosh. This has now been corrected in both the PDF and HTML versions of the article.

2.
Leukemia ; 33(6): 1487-1500, 2019 06.
Article in English | MEDLINE | ID: mdl-30575819

ABSTRACT

The study of myelodysplastic syndromes (MDS) in murine models has now indicated the possible involvement of the bone marrow microenvironment in the generation of dysplastic hematopoietic cells. However, there is scant work on patient samples and the role of hypomethylating agents on the bone marrow stromal cells of MDS patients is unclear. We show that human MDS-MSCs exhibit phenotypic, transcriptomic and epigenetic abnormalities. Stimuli provided by MDS-MSCs impaired the growth and function of healthy HSPCs, which is further sustained autonomously in HSPCs for significant periods of time resulting in a failure for active hematopoietic engraftment across primary and secondary transplant recipients (chimerism: 0.34-91% vs 2.78%, engraftment frequencies: at 0.06 ± 0.02 vs full engraftment for MDS-MSC vs healthy groups, respectively). Hypomethylation of MDS-MSCs improved overall engraftment in most of the MDS-MSC groups tested (2/7 with p < 0.01, 3/7 with p < 0.05 and 2/7 with no significant difference). MDS-MSCs that fail to respond to hypomethylating therapy are associated with patients with rapid adverse disease transformation and this further suggests that MDS-MSCs may be an integral part of disease progression and have prognostic value as well as potential as a therapeutic target.


Subject(s)
Azacitidine/pharmacology , Biomarkers, Tumor/metabolism , DNA Methylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hematopoiesis/drug effects , Mesenchymal Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Proliferation , Epigenesis, Genetic , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Exp Hematol ; 62: 45-55, 2018 06.
Article in English | MEDLINE | ID: mdl-29605545

ABSTRACT

Major hemoglobinopathies place tremendous strain on global resources. Intrauterine hemopoietic cell transplantation (IUHCT) and gene transfer (IUGT) can potentially reduce perinatal morbidities with greater efficacy than postnatal therapy alone. We performed both procedures in the thalassemic HbbTh3/+ mouse. Intraperitoneal delivery of co-isogenic cells at embryonic days13-14 produced dose-dependent chimerism. High-dose adult bone marrow (BM) cells maintained 0.2-3.1% chimerism over ~24 weeks and treated heterozygotes (HET) demonstrated higher chimerism than wild-type (WT) pups (1.6% vs. 0.7%). Fetalliver (FL) cells produced higher chimerism than BM when transplanted at thesame doses, maintaining 1.8-2.4% chimerism over ~32 weeks. We boosted transplanted mice postnatally with BM cells after busulfan conditioning. Engraftment was maintained at >1% only in chimeras. IUHCT-treated nonchimeras and non-IUHCT mice showed microchimerism or no chimerism. Improved engraftment was observed with a higher initial chimerism, in HET mice and with the addition of fludarabine. Chimeric HET mice expressed 2.2-15.1% engraftment with eventual decline at 24 weeks (vs. <1% in nonchimeras) and demonstrated improved hematological indices and smaller spleens compared with untreated HETmice. Intravenous delivery of GLOBE lentiviral-vector expressing human ß-globin (HBB) resulted in a vector concentration of 0.001-0.6 copies/cell. Most hematological indices were higher in treated than untreated HET mice, including hemoglobin and mean corpuscular volume, but were still lower than in WT. Therefore, direct IUGT and IUHCT strategies can be used to achieve hematological improvement but require further dose optimization. IUHCT will be useful combined with postnatal transplantation to further enhance engraftment.


Subject(s)
Fetal Therapies , Genetic Therapy , Genetic Vectors/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Lentivirus/genetics , beta-Thalassemia/therapy , Animals , Bone Marrow Transplantation/methods , Busulfan , Cell Survival , Cellular Microenvironment , Disease Models, Animal , Female , Fetal Therapies/methods , Genetic Therapy/methods , Hepatocytes/transplantation , Liver/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Transplantation Chimera , Transplantation Conditioning , Vidarabine/analogs & derivatives , beta-Thalassemia/embryology , beta-Thalassemia/genetics
4.
Stem Cells Transl Med ; 7(5): 376-393, 2018 05.
Article in English | MEDLINE | ID: mdl-29392885

ABSTRACT

Umbilical cord blood (UCB) transplants in adults have slower hematopoietic recovery compared to bone marrow (BM) or peripheral blood (PB) stem cells mainly due to low number of total nucleated cells and hematopoietic stem and progenitor cells (HSPC). As such in this study, we aimed to perform ex vivo expansion of UCB HSPC from non-enriched mononucleated cells (MNC) using novel azole-based small molecules. Freshly-thawed UCB-MNC were cultured in expansion medium supplemented with small molecules and basal cytokine cocktail. The effects of the expansion protocol were measured based on in vitro and in vivo assays. The proprietary library of >50 small molecules were developed using structure-activity-relationship studies of SB203580, a known p38-MAPK inhibitor. A particular analog, C7, resulted in 1,554.1 ± 27.8-fold increase of absolute viable CD45+ CD34+ CD38- CD45RA- progenitors which was at least 3.7-fold higher than control cultures (p < .001). In depth phenotypic analysis revealed >600-fold expansion of CD34+ /CD90+ /CD49f+ rare HSPCs coupled with significant (p < .01) increase of functional colonies from C7 treated cells. Transplantation of C7 expanded UCB grafts to immunodeficient mice resulted in significantly (p < .001) higher engraftment of human CD45+ and CD45+ CD34+ cells in the PB and BM by day 21 compared to non-expanded and cytokine expanded grafts. The C7 expanded grafts maintained long-term human multilineage chimerism in the BM of primary recipients with sustained human CD45 cell engraftment in secondary recipients. In conclusion, a small molecule, C7, could allow for clinical development of expanded UCB grafts without pre-culture stem cell enrichment that maintains in vitro and in vivo functionality. Stem Cells Translational Medicine 2018;7:376-393.


Subject(s)
Antigens, CD34/metabolism , Azoles/pharmacology , Fetal Blood/drug effects , Hematopoietic Stem Cells/drug effects , Integrin alpha6/metabolism , Stem Cells/drug effects , Thy-1 Antigens/metabolism , Animals , Cells, Cultured , Fetal Blood/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Imidazoles/pharmacology , Mice , Mice, SCID , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Stem Cells/metabolism , Structure-Activity Relationship
5.
PLoS One ; 9(8): e104805, 2014.
Article in English | MEDLINE | ID: mdl-25118036

ABSTRACT

Hematopoietic Stem Cell (HSC) targeted gene transfer is an attractive treatment option for a number of hematopoietic disorders caused by single gene defects. However, extensive methylation of promoter sequences results in silencing of therapeutic gene expression. The choice of an appropriate promoter is therefore crucial for reproducible, stable and long-term transgene expression in clinical gene therapy. Recent studies suggest efficient and stable expression of transgenes from the ubiquitous chromatin opening element (UCOE) derived from the human HNRPA2B1-CBX3 locus can be achieved in murine HSC. Here, we compared the use of HNRPA2B1-CBX3 UCOE (A2UCOE)-mediated transgene regulation to two other frequently used promoters namely EF1α and PGK in human fetal liver-derived HSC (hflHSC). Efficient transduction of hflHSC with a lentiviral vector containing an HNRPA2B1-CBX3 UCOE-eGFP (A2UCOE-eGFP) cassette was achieved at higher levels than that obtained with umbilical cord blood derived HSC (3.1x; p<0.001). While hflHSC were readily transduced with all three test vectors (A2UCOE-eGFP, PGK-eGFP and EF1α-eGFP), only the A2-UCOE construct demonstrated sustained transgene expression in vitro over 24 days (p<0.001). In contrast, within 10 days in culture a rapid decline in transgene expression in both PGK-eGFP and EF1α-eGFP transduced hflHSC was seen. Subsequently, injection of transduced cells into immunodeficient mice (NOD/SCID/Il2rg-/-) demonstrated sustained eGFP expression for the A2UCOE-eGFP group up to 10 months post transplantation whereas PGK-eGFP and EF1α-eGFP transduced hflHSC showed a 5.1 and 22.2 fold reduction respectively over the same time period. We conclude that the A2UCOE allows a more efficient and stable expression in hflHSC to be achieved than either the PGK or EF1α promoters and at lower vector copy number per cell.


Subject(s)
Gene Transfer Techniques , Hematopoietic Stem Cells/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Molecular Targeted Therapy/methods , Animals , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Liver/cytology , Liver/embryology , Liver/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphoglycerate Kinase/genetics , Promoter Regions, Genetic/genetics , Transgenes/genetics
6.
Mol Ther ; 21(2): 324-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23299800

ABSTRACT

Genotoxicity models are extremely important to assess retroviral vector biosafety before gene therapy. We have developed an in utero model that demonstrates that hepatocellular carcinoma (HCC) development is restricted to mice receiving nonprimate (np) lentiviral vectors (LV) and does not occur when a primate (p) LV is used regardless of woodchuck post-translation regulatory element (WPRE) mutations to prevent truncated X gene expression. Analysis of 839 npLV and 244 pLV integrations in the liver genomes of vector-treated mice revealed clear differences between vector insertions in gene dense regions and highly expressed genes, suggestive of vector preference for insertion or clonal outgrowth. In npLV-associated clonal tumors, 56% of insertions occurred in oncogenes or genes associated with oncogenesis or tumor suppression and surprisingly, most genes examined (11/12) had reduced expression as compared with control livers and tumors. Two examples of vector-inserted genes were the Park 7 oncogene and Uvrag tumor suppressor gene. Both these genes and their known interactive partners had differential expression profiles. Interactive partners were assigned to networks specific to liver disease and HCC via ingenuity pathway analysis. The fetal mouse model not only exposes the genotoxic potential of vectors intended for gene therapy but can also reveal genes associated with liver oncogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Damage , Fetus/pathology , Genetic Therapy/adverse effects , Infectious Anemia Virus, Equine/genetics , Liver/pathology , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Genome , HIV/genetics , Immunohistochemistry , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Mutagenesis , Mutagenesis, Insertional , Mutation , Real-Time Polymerase Chain Reaction
7.
Mol Ther ; 19(11): 1950-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21629224

ABSTRACT

Intrauterine gene transfer (IUGT) offers ontological advantages including immune naiveté mediating tolerance to the vector and transgenic products, and effecting a cure before development of irreversible pathology. Despite proof-of-principle in rodent models, expression efficacy with a therapeutic transgene has yet to be demonstrated in a preclinical nonhuman primate (NHP) model. We aimed to determine the efficacy of human Factor IX (hFIX) expression after adeno-associated-viral (AAV)-mediated IUGT in NHP. We injected 1.0-1.95 × 10(13) vector genomes (vg)/kg of self-complementary (sc) AAV5 and 8 with a LP1-driven hFIX transgene intravenously in 0.9G late gestation NHP fetuses, leading to widespread transduction with liver tropism. Liver-specific hFIX expression was stably maintained between 8 and 112% of normal activity in injected offspring followed up for 2-22 months. AAV8 induced higher hFIX expression (P = 0.005) and milder immune response than AAV5. Random hepatocellular integration was found with no hotspots. Transplacental spread led to low-level maternal tissue transduction, without evidence of immunotoxicity or germline transduction in maternal oocytes. A single intravenous injection of scAAV-LP1-hFIXco to NHP fetuses in late-gestation produced sustained clinically-relevant levels of hFIX with liver-specific expression and a non-neutralizing immune response. These data are encouraging for conditions where gene transfer has the potential to avert perinatal death and long-term irreversible sequelae.


Subject(s)
Dependovirus/genetics , Factor IX/genetics , Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Hemophilia B/therapy , Animals , Cell Line , Dependovirus/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacokinetics , HEK293 Cells , Hemophilia B/genetics , Humans , Injections , Macaca fascicularis , Placenta/metabolism , Pregnancy , Transduction, Genetic , Virus Integration
9.
Mol Ther ; 12(4): 763-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16084128

ABSTRACT

Gene therapy by use of integrating vectors carrying therapeutic transgene sequences offers the potential for a permanent cure of genetic diseases by stable vector insertion into the patients' chromosomes. However, three cases of T cell lymphoproliferative disease have been identified almost 3 years after retrovirus gene therapy for X-linked severe combined immune deficiency. In two of these cases vector insertion into the LMO2 locus was implicated in leukemogenesis, demonstrating that a more profound understanding is required of the genetic and molecular effects imposed on the host by vector integration or transgene expression. In vivo models to test for retro- and lentiviral vector safety prior to clinical application are therefore needed. Here we present a high incidence of lentiviral vector-associated tumorigenesis following in utero and neonatal gene transfer in mice. This system may provide a highly sensitive model to investigate integrating vector safety prior to clinical application.


Subject(s)
Genetic Therapy/adverse effects , Lentivirus/genetics , Liver Neoplasms/etiology , Animals , Animals, Newborn , Fetus , Gene Transfer Techniques , Genetic Vectors/genetics , HIV-1/genetics , Liver/pathology , Liver Neoplasms/pathology , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...