Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Stem Cells Transl Med ; 6(3): 864-876, 2017 03.
Article in English | MEDLINE | ID: mdl-28297579

ABSTRACT

Successful expansion of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) would benefit many HSPC transplantation and gene therapy/editing applications. However, current expansion technologies have been limited by a loss of multipotency and self-renewal properties ex vivo. We hypothesized that an ex vivo vascular niche would provide prohematopoietic signals to expand HSPCs while maintaining multipotency and self-renewal. To test this hypothesis, BM autologous CD34+ cells were expanded in endothelial cell (EC) coculture and transplanted in nonhuman primates. CD34+ C38- HSPCs cocultured with ECs expanded up to 17-fold, with a significant increase in hematopoietic colony-forming activity compared with cells cultured with cytokines alone (colony-forming unit-granulocyte-erythroid-macrophage-monocyte; p < .005). BM CD34+ cells that were transduced with green fluorescent protein lentivirus vector and expanded on ECs engrafted long term with multilineage polyclonal reconstitution. Gene marking was observed in granulocytes, lymphocytes, platelets, and erythrocytes. Whole transcriptome analysis indicated that EC coculture altered the expression profile of 75 genes in the BM CD34+ cells without impeding the long-term engraftment potential. These findings show that an ex vivo vascular niche is an effective platform for expansion of adult BM HSPCs. Stem Cells Translational Medicine 2017;6:864-876.


Subject(s)
Bone Marrow Cells/cytology , Endothelial Cells/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Cell Lineage , Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Profiling , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Primates , Time Factors
2.
Hum Gene Ther ; 26(7): 443-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26068008

ABSTRACT

Genome editing using the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated 9 (Cas9) technology is revolutionizing the study of gene function and likely will give rise to an entire new class of therapeutics for a wide range of diseases. Achieving this goal requires not only characterization of the technology for efficacy and specificity but also optimization of its delivery to the target cells for each disease indication. In this review we survey the various methods by which the CRISPR-Cas9 components have been delivered to cells and highlight some of the more clinically relevant approaches. Additionally, we discuss the methods available for assessing the specificity of Cas9 editing; an important safety consideration for development of the technology.


Subject(s)
CRISPR-Cas Systems , Genetic Therapy/trends , Bacterial Proteins , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases , Humans
3.
J Clin Invest ; 125(3): 1243-54, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25664855

ABSTRACT

Pluripotent stem cells (PSCs) represent an alternative hematopoietic stem cell (HSC) source for treating hematopoietic disease. The limited engraftment of human PSC-derived (hPSC-derived) multipotent progenitor cells (MPP) has hampered the clinical application of these cells and suggests that MPP require additional cues for definitive hematopoiesis. We hypothesized that the presence of a vascular niche that produces Notch ligands jagged-1 (JAG1) and delta-like ligand-4 (DLL4) drives definitive hematopoiesis. We differentiated hes2 human embryonic stem cells (hESC) and Macaca nemestrina-induced PSC (iPSC) line-7 with cytokines in the presence or absence of endothelial cells (ECs) that express JAG1 and DLL4. Cells cocultured with ECs generated substantially more CD34+CD45+ hematopoietic progenitors compared with cells cocultured without ECs or with ECs lacking JAG1 or DLL4. EC-induced cells exhibited Notch activation and expressed HSC-specific Notch targets RUNX1 and GATA2. EC-induced PSC-MPP engrafted at a markedly higher level in NOD/SCID/IL-2 receptor γ chain-null (NSG) mice compared with cytokine-induced cells, and low-dose chemotherapy-based selection further increased engraftment. Long-term engraftment and the myeloid-to-lymphoid ratio achieved with vascular niche induction were similar to levels achieved for cord blood-derived MPP and up to 20-fold higher than those achieved with hPSC-derived MPP engraftment. Our findings indicate that endothelial Notch ligands promote PSC-definitive hematopoiesis and production of long-term engrafting CD34+ cells, suggesting these ligands are critical for HSC emergence.


Subject(s)
Endothelium, Vascular/cytology , Hematopoietic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Multipotent Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Endothelial Cells/physiology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans , Macaca nemestrina , Male , Mice, Inbred NOD , Mice, SCID , Stem Cell Niche
4.
J Clin Invest ; 124(9): 4082-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25105369

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is one of the most potent chemotherapy agents for the treatment of glioblastoma. Unfortunately, almost half of glioblastoma tumors are TMZ resistant due to overexpression of methylguanine methyltransferase (MGMT(hi)). Coadministration of O6-benzylguanine (O6BG) can restore TMZ sensitivity, but causes off-target myelosuppression. Here, we conducted a prospective clinical trial to test whether gene therapy to confer O6BG resistance in hematopoietic stem cells (HSCs) improves chemotherapy tolerance and outcome. METHODS: We enrolled 7 newly diagnosed glioblastoma patients with MGMT(hi) tumors. Patients received autologous gene-modified HSCs following single-agent carmustine administration. After hematopoietic recovery, patients underwent O6BG/TMZ chemotherapy in 28-day cycles. Serial blood samples and tumor images were collected throughout the study. Chemotherapy tolerance was determined by the observed myelosuppression and recovery following each cycle. Patient-specific biomathematical modeling of tumor growth was performed. Progression-free survival (PFS) and overall survival (OS) were also evaluated. RESULTS: Gene therapy permitted a significant increase in the mean number of tolerated O6BG/TMZ cycles (4.4 cycles per patient, P < 0.05) compared with historical controls without gene therapy (n = 7 patients, 1.7 cycles per patient). One patient tolerated an unprecedented 9 cycles and demonstrated long-term PFS without additional therapy. Overall, we observed a median PFS of 9 (range 3.5-57+) months and OS of 20 (range 13-57+) months. Furthermore, biomathematical modeling revealed markedly delayed tumor growth at lower cumulative TMZ doses in study patients compared with patients that received standard TMZ regimens without O6BG. CONCLUSION: These data support further development of chemoprotective gene therapy in combination with O6BG and TMZ for the treatment of glioblastoma and potentially other tumors with overexpression of MGMT. TRIAL REGISTRATION: Clinicaltrials.gov NCT00669669. FUNDING: R01CA114218, R01AI080326, R01HL098489, P30DK056465, K01DK076973, R01HL074162, R01CA164371, R01NS060752, U54CA143970.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Adult , Bone Marrow/drug effects , Brain Neoplasms/mortality , Carmustine/adverse effects , Combined Modality Therapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Female , Glioblastoma/mortality , Guanine/analogs & derivatives , Guanine/pharmacology , Hematopoietic Stem Cell Transplantation , Humans , Male , Middle Aged , Models, Biological , Prospective Studies , Temozolomide , Tumor Suppressor Proteins/genetics
5.
Gastroenterology ; 145(5): 966-969.e7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23891978

ABSTRACT

The narrow species tropism of hepatitis C virus (HCV) limits animal studies. We found that pigtail macaque (Macaca nemestrina) hepatic cells derived from induced pluripotent stem cells support the entire HCV life cycle, although infection efficiency was limited by defects in the HCV cell entry process. This block was overcome by either increasing occludin expression, complementing the cells with human CD81, or infecting them with a strain of HCV with less restricted requirements for CD81. Using this system, we can modify viral and host cell genetics to make pigtail macaques a suitable, clinically relevant model for the study of HCV infection.


Subject(s)
Disease Models, Animal , Hepacivirus/pathogenicity , Hepatitis C/virology , Hepatocytes/virology , Induced Pluripotent Stem Cells/virology , Macaca nemestrina , Animals , Cell Line , Cells, Cultured , Hepatitis C/pathology , Hepatitis C/physiopathology , Hepatocytes/pathology , Host-Pathogen Interactions/genetics , Humans , Induced Pluripotent Stem Cells/pathology , Occludin/physiology , Tetraspanin 28/deficiency , Tetraspanin 28/physiology , Virus Internalization , Virus Replication/physiology
6.
J Gene Med ; 15(6-7): 233-41, 2013.
Article in English | MEDLINE | ID: mdl-23666780

ABSTRACT

BACKGROUND: Nonmyeloablative allogeneic hematopoietic stem cell (HSC) transplantation can cure malignant and nonmalignant diseases affecting the hematopoietic system, such as severe combined immunodeficiencies, aplastic anemia and hemoglobinopathies. Although nonmyeloablative is favored over myeloablative transplantation for many patients, graft rejection remains problematic. One strategy for decreasing rejection is to protect donor activated T cells in the graft from methotrexate (MTX) by genetically modifying the cells to express MTX-resistant dihydrofolate reductase (Tyr22-DHFR), leaving the immunosuppressive effects of MTX to act solely on activated host T lymphocytes, shifting the balance to favor allogeneic engraftment. METHODS: To evaluate MTX resistance of Tyr22-DHFR(+) T lymphocytes in vivo, we transplanted dogs with autologous CD34(+) cells modified with yellow fluorescent protein (YFP) and DHFR-green fluorescent protein (GFP) lentivirus vectors. Dogs were then treated with a standard MTX regimen days 1, 3, 6 and 11) following immune activation with a foreign antigen as a surrogate assay to mimic early transplantation. RESULTS: DHFR-GFP(+) gene marking was maintained in CD3(+) CD25(+) and CD4(+) T lymphocytes after MTX treatment, whereas the level of T lymphocytes that expressed only a fluorescent reporter (YFP(+) ) decreased. These data show that Tyr22-DHFR expression protects T lymphocytes from MTX toxicity in dogs, highlighting a clinically relevant application for preserving donor T lymphocytes during post-transplantation immunosuppression. CONCLUSIONS: The findings of the present study have implications for the clinical translation of MTX-resistant T cells to facilitate engraftment of allogeneic cells following nonmyeloablative conditioning and to minimize the risk of rejection. In summary, Tyr22-DHFR expression in T lymphocytes provides chemoprotection from MTX-mediated elimination in the context of immune activation in vivo.


Subject(s)
Drug Resistance/genetics , Methotrexate/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Dogs , Enzyme Inhibitors/pharmacology , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Polymerase Chain Reaction , Sheep , T-Lymphocytes/cytology
7.
Blood ; 120(13): e35-44, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22898598

ABSTRACT

Induced pluripotent stem cell (iPSC) therapeutics are a promising treatment for genetic and infectious diseases. To assess engraftment, risk of neoplastic formation, and therapeutic benefit in an autologous setting, testing iPSC therapeutics in an appropriate model, such as the pigtail macaque (Macaca nemestrina; Mn), is crucial. Here, we developed a chemically defined, scalable, and reproducible specification protocol with bone morphogenetic protein 4, prostaglandin-E2 (PGE2), and StemRegenin 1 (SR1) for hematopoietic differentiation of Mn iPSCs. Sequential coculture with bone morphogenetic protein 4, PGE2, and SR1 led to robust Mn iPSC hematopoietic progenitor cell formation. The combination of PGE2 and SR1 increased CD34(+)CD38(-)Thy1(+)CD45RA(-)CD49f(+) cell yield by 6-fold. CD34(+)CD38(-)Thy1(+)CD45RA(-)CD49f(+) cells isolated on the basis of CD34 expression and cultured in SR1 expanded 3-fold and maintained this long-term repopulating HSC phenotype. Purified CD34(high) cells exhibited 4-fold greater hematopoietic colony-forming potential compared with unsorted hematopoietic progenitors and had bilineage differentiation potential. On the basis of these studies, we calculated the cell yields that must be achieved at each stage to meet a threshold CD34(+) cell dose that is required for engraftment in the pigtail macaque. Our protocol will support scale-up and testing of iPSC-derived CD34(high) cell therapies in a clinically relevant nonhuman primate model.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Blotting, Western , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Lineage , Cells, Cultured , Colony-Forming Units Assay , Dinoprostone/genetics , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocytes/cytology , Lymphocytes/metabolism , Macaca , Myeloid Cells/cytology , Myeloid Cells/metabolism , Purines/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Internalization , Wnt Proteins/genetics , Wnt Proteins/metabolism
8.
Mol Ther ; 19(9): 1667-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21587213

ABSTRACT

The development of technology to generate induced pluripotent stem (iPS) cells constitutes one of the most exciting scientific breakthroughs because of the enormous potential for regenerative medicine. However, the safety of iPS cell-related products is a major concern for clinical translation. Insertional mutagenesis, possible oncogenic transformation of iPS cells or their derivatives, or the contamination of differentiated iPS cells with undifferentiated cells, resulting in the formation of teratomas, have remained considerable obstacles. Here, we demonstrate the utility of suicide genes to safeguard iPS cells and their derivatives. We found suicide genes can control the cell fate of iPS cells in vitro and in vivo without interfering with their pluripotency and self-renewal capacity. This study will be useful to evaluate the safety of iPS cell technology in a clinically highly relevant, large animal model and further benefit the clinical use of human iPS cells.


Subject(s)
Genes, Transgenic, Suicide , Genetic Vectors , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Teratoma/metabolism , Animals , Blotting, Southern , Cell Differentiation , Cell Line , Cell Proliferation , Cloning, Molecular , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Macaca/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Models, Animal , Mutagenesis, Insertional , Regenerative Medicine , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
9.
Bioeng Bugs ; 1(6): 434-6, 2010.
Article in English | MEDLINE | ID: mdl-21468213

ABSTRACT

Human embryonic stem cells (hES Cs) are an attractive alternative cell source for hematopoietic gene therapy applications as the cells are easily modified with lentiviral or other vectors and can be subsequently induced to differentiate into hematopoietic progenitor cells. However, demonstration of the full hematopoietic potential of hESC-derived progeny is challenging due to low marrow engraftment and the difficulty of detecting cells in the peripheral blood of human/mouse xenografts. Methotrexate (MTX) chemotherapy coupled with expression of a drug resistant dihydrofolate reductase such as Tyr22 (Tyr22DHFR) has the potential to selectively increase engraftment of gene-modified human hematopoietic cells in mice, which would allow for better phenotypic characterization of hESC-derived cells in vivo. We showed that hES Cs transduced with Tyr22DHFR-GFP encoding lentivirus vectors differentiate into MTX resistant (MTXr) hemato-endothelial cells. MTX treatment of immunodeficient mice infused with Tyr22DHFR hESC-derived hemato-endothelial cells increased the long-term engraftment of human cells in the bone marrow of MTX-treated mice. In contrast to previous studies, these results indicate that MTX administration has the potential to support in vivo selection that is maintained after cessation of treatment. The MTX/Tyr22DHFR system may therefore be useful for enrichment of gene-modified cell populations in human stem cell and gene therapy applications.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Methotrexate/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Cell Differentiation/genetics , Drug Resistance , Embryonic Stem Cells/enzymology , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Humans , Mice , Stem Cell Transplantation/methods , Tetrahydrofolate Dehydrogenase/genetics
10.
J Pharmacol Exp Ther ; 322(3): 989-97, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17586722

ABSTRACT

Methotrexate (MTX) dose-escalation studies were conducted in C57BL/6 mice to determine the chemoprotective effect of transplantation using bone marrow transduced with lentivirus vectors expressing a drug-resistant variant of murine dihydrofolate reductase (DHFR). Methotrexate-resistant dihydrofolate reductase [tyrosine-22 (Tyr22)DHFR] and enhanced green fluorescent protein (GFP) coding sequences were inserted into self-inactivating lentiviral vectors as part of a genetic fusion or within the context of a bicistronic expression cassette. MTX-treated animals that received Tyr22DHFR-transduced marrow recovered to normal hematocrit levels by 3 weeks post-transplant and exhibited significant GFP marking in myeloid and lymphoid lineage-derived peripheral blood mononuclear cells (PBMCs). In contrast, MTX-treated animals transplanted with control GFP-transduced marrow exhibited extremely reduced hematocrits with severe marrow hypoplasia and did not survive MTX dose escalation. To minimize cell manipulation, we treated unfractionated marrow in an overnight exposure. Transduction at a multiplicity of infection of 10 resulted in up to 11% vector-modified PBMCs in primary recipients and successful repopulation of secondary recipients with vector-marked cells. Experimental cohorts exhibited sustained proviral expression with stable GFP fluorescence intensity. These results demonstrate the effectiveness of lentivirus vectors for chemoprotection in a well developed animal model, with the potential for further preclinical development toward human application.


Subject(s)
Antidotes/administration & dosage , Drug Resistance/genetics , Methotrexate/toxicity , Tetrahydrofolate Dehydrogenase/administration & dosage , Tetrahydrofolate Dehydrogenase/pharmacology , Animals , Bone Marrow Diseases/chemically induced , Bone Marrow Transplantation , Genetic Vectors , Hematocrit , Lentivirus , Mice , Mice, Inbred C57BL , Mutation , Tetrahydrofolate Dehydrogenase/genetics , Transduction, Genetic
11.
Biosci Rep ; 24(6): 577-94, 2004 Dec.
Article in English | MEDLINE | ID: mdl-16158196

ABSTRACT

Sleeping Beauty (SB) is a gene-insertion system reconstructed from transposon sequences found in teleost fish and is capable of mediating the transposition of DNA sequences from transfected plasmids into the chromosomes of vertebrate cell populations. The SB system consists of a transposon, made up of a gene of interest flanked by transposon inverted repeats, and a source of transposase. Here we carried out a series of studies to further characterize SB-mediated transposition as a tool for gene transfer to chromosomes and ultimately for human gene therapy. Transfection of mouse 3T3 cells, HeLa cells, and human A549 lung carcinoma cells with a transposon containing the neomycin phosphotransferase (NEO) gene resulted in a several-fold increase in drug-resistant colony formation when co-transfected with a plasmid expressing the SB transposase. A transposon containing a methotrexate-resistant dihydrofolate reductase gene was also found to confer an increased frequency of methotrexate-resistant colony formation when co-transfected with SB transposase-encoding plasmid. A plasmid containing a herpes simplex virus thymidine kinase gene as well as a transposon containing a NEO gene was used for counterselection against random recombinants (NEO+TK+) in medium containing G418 plus ganciclovir. Effective counterselection required a recovery period of 5 days after transfection before shifting into medium containing ganciclovir to allow time for transiently expressed thymidine kinase activity to subside in cells not stably transfected. Southern analysis of clonal isolates indicated a shift from random recombination events toward transposition events when clones were isolated in medium containing ganciclovir as well as G418. We found that including both transposon and transposase functions on the same plasmid substantially increased the stable gene transfer frequency in Huh7 human hepatoma cells. The results from these experiments contribute technical and conceptual insight into the process of transposition in mammalian cells, and into the optimal provision of transposon and transposase functions that may be applicable to gene therapy studies.


Subject(s)
DNA Transposable Elements/genetics , Transposases/genetics , Transposases/metabolism , 3T3 Cells , Animals , Cell Line , Drug Resistance/genetics , Gene Transfer Techniques , HeLa Cells , Humans , Kanamycin Kinase/genetics , Mice , Plasmids/genetics , Recombination, Genetic , Selection, Genetic , Thymidine Kinase/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...