Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Genome Res ; 21(3): 477-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173034

ABSTRACT

We have streamlined the process of transferring plasmids into any yeast strain library by developing a novel mating-based, high-throughput method called selective ploidy ablation (SPA). SPA uses a universal plasmid donor strain that contains conditional centromeres on every chromosome. The plasmid-bearing donor is mated to a recipient, followed by removal of all donor-strain chromosomes, producing a haploid strain containing the transferred plasmid. As proof of principle, we used SPA to transfer plasmids containing wild-type and mutant alleles of DNA topoisomerase I (TOP1) into the haploid yeast gene-disruption library. Overexpression of Top1 identified only one sensitive mutation, rpa34, while overexpression of top1-T(722)A allele, a camptothecin mimetic, identified 190 sensitive gene-disruption strains along with rpa34. In addition to known camptothecin-sensitive strains, this set contained mutations in genes involved in the Rpd3 histone deacetylase complex, the kinetochore, and vesicle trafficking. We further show that mutations in several ESCRT vesicle trafficking components increase Top1 levels, which is dependent on SUMO modification. These findings demonstrate the utility of the SPA technique to introduce plasmids into the haploid gene-disruption library to discover new interacting pathways.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Gene Regulatory Networks , High-Throughput Screening Assays/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Alleles , Camptothecin/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , DNA Topoisomerases, Type I/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression , Genomic Library , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mutation , Plasmids/genetics , Ploidies , Saccharomyces cerevisiae/drug effects , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Transformation, Genetic
2.
Blood ; 116(16): 2915-20, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20606166

ABSTRACT

Fanconi anemia (FA) is an inherited chromosomal instability syndrome characterized by bone marrow failure, myelodysplasia (MDS), and acute myeloid leukemia (AML). Eight FA proteins associate in a nuclear core complex to monoubiquitinate FANCD2/FANCI in response to DNA damage. Additional functions have been described for some of the core complex proteins; however, in vivo genetic proof has been lacking. Here we show that double-mutant Fancc(-/-);Fancg(-/-) mice develop spontaneous hematologic sequelae including bone marrow failure, AML, MDS and complex random chromosomal abnormalities that the single-mutant mice do not. This genetic model provides evidence for unique core complex protein function independent of their ability to monoubiquitinate FANCD2/FANCI. Importantly, this model closely recapitulates the phenotypes found in FA patients and may be useful as a preclinical platform to evaluate the molecular pathogenesis of spontaneous bone marrow failure, MDS and AML in FA.


Subject(s)
Bone Marrow/physiopathology , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Animals , Chromosome Aberrations , Mice , Mice, Inbred C57BL
3.
Genetics ; 180(4): 1799-808, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832360

ABSTRACT

We have created a resource to rapidly map genetic traits to specific chromosomes in yeast. This mapping is done using a set of 16 yeast strains each containing a different chromosome with a conditionally functional centromere. Conditional centromere function is achieved by integration of a GAL1 promoter in cis to centromere sequences. We show that the 16 yeast chromosomes can be individually lost in diploid strains, which become hemizygous for the destabilized chromosome. Interestingly, most 2n - 1 strains endoduplicate and become 2n. We also demonstrate how chromosome loss in this set of strains can be used to map both recessive and dominant markers to specific chromosomes. In addition, we show that this method can be used to rapidly validate gene assignments from screens of strain libraries such as the yeast gene disruption collection.


Subject(s)
Chromosomes, Fungal/genetics , Saccharomyces cerevisiae/genetics , Chromosome Mapping , Diploidy , Loss of Heterozygosity , Meiosis , Models, Genetic , Phenotype , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
Blood ; 112(12): 4458-65, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18684868

ABSTRACT

Fanconi anemia (FA) is a complex genetic disorder characterized by congenital abnormalities, bone marrow failure, and myeloid malignancies. Identification of 13 FA genes has been instrumental to explore gene transfer technologies aimed at correction of autologous FA-deficient stem cells. To date, 3 human FA stem cell gene therapy trials with standard 4-day transduction protocols using gammaretroviral vectors failed to provide clinical benefit. In addition, 2- to 4 day ex vivo manipulation of bone marrow from mice containing a disruption of the homologue of human FANCC (Fancc) results in a time-dependent increase in apoptosis and a risk for malignant transformation of hematopoietic cells. Here, we show that a 14-hour transduction period allows a foamyviral vector construct expressing the human FANCC cDNA to efficiently transduce murine FA stem cells with 1 to 2 proviral integrations per genome. Functionally, the repopulating activity of Fancc(-/-) stem cells from reconstituted mice expressing the recombinant FANCC transgene was comparable with wild-type controls. Collectively, these data provide evidence that short-term transduction of c-kit(+) cells with a foamyviral vector is sufficient for functional correction of a stem cell phenotype in a murine FA model. These data could have implications for future gene therapy trials for FA patients.


Subject(s)
Cell Proliferation , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia/therapy , Hematopoietic Stem Cells/physiology , Spumavirus/genetics , Transduction, Genetic/methods , Animals , Cells, Cultured , Circadian Rhythm , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia/veterinary , Fanconi Anemia Complementation Group C Protein/metabolism , Genetic Therapy , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/genetics , Time Factors
5.
Exp Hematol ; 36(9): 1084-90, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495331

ABSTRACT

Fanconi anemia (FA) is a heterogeneous inherited disorder characterized by a progressive bone marrow (BM) failure and susceptibility to myeloid leukemia. Genetic correction using gene-transfer technology is one potential therapy. A major hurdle in applying this technology in FA patients is the inability of granulocyte colony-stimulating factor (G-CSF) to mobilize sufficient numbers of hematopoietic stem (HSC)/progenitor cells (HPC) from the BM to the peripheral blood. Whether the low number of CD34(+) cells is a result of BM hypoplasia or an inability of G-CSF to adequately mobilize FA HSC/HPC remains incompletely understood. Here we use competitive repopulation of lethally irradiated primary and secondary recipients to show that in two murine models of FA, AMD3100 synergizes with G-CSF resulting in a mobilization of HSC, whereas G-CSF alone fails to mobilize stem cells even in the absence of hypoplasia.


Subject(s)
Bone Marrow Transplantation , Fanconi Anemia/surgery , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Peripheral Blood Stem Cell Transplantation , Animals , Benzylamines , Cells, Cultured/drug effects , Colony-Forming Units Assay , Cyclams , Drug Synergism , Drug Therapy, Combination , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Graft Survival , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera , Transplantation, Homologous
6.
Blood ; 108(13): 4283-7, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16946306

ABSTRACT

Fanconi anemia (FA) is a heterogeneous genetic disorder characterized by bone marrow (BM) failure and cancer susceptibility. Identification of the cDNAs of FA complementation types allows the potential of using gene transfer technology to introduce functional cDNAs as transgenes into autologous stem cells and provide a cure for the BM failure in FA patients. However, strategies to enhance the mobilization, transduction, and engraftment of exogenous stem cells are required to optimize efficacy prior to widespread clinical use. Hypersensitivity of Fancc-/- cells to interferon-gamma (IFN-gamma), a nongenotoxic immune-regulatory cytokine, enhances engraftment of syngeneic wild-type (WT) cells in Fancc-/- mice. However, whether this phenotype is of broad relevance in other FA complementation groups is unresolved. Here we show that primitive and mature myeloid progenitors in Fanca-/- and Fancg-/- mice are hypersensitive to IFN-gamma and that in vivo infusion of IFN-gamma at clinically relevant concentrations was sufficient to allow consistent long-term engraftment of isogenic WT repopulating stem cells. Given that FANCA, FANCC, and FANCG complementation groups account for more than 90% of all FA patients, these data provide evidence that IFN-gamma conditioning may be a useful nongenotoxic strategy for myelopreparation in FA patients.


Subject(s)
Antiviral Agents/pharmacology , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/therapy , Genetic Therapy , Interferon-gamma/pharmacology , Myeloid Progenitor Cells/transplantation , Animals , Fanconi Anemia/genetics , Genetic Therapy/methods , Graft Enhancement, Immunologic/methods , Graft Survival/drug effects , Graft Survival/genetics , Hematopoietic Stem Cell Mobilization/methods , Humans , Mice , Mice, Knockout , Transduction, Genetic/methods , Transplantation, Autologous , Transplantation, Isogeneic
7.
Carcinogenesis ; 27(3): 446-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16258176

ABSTRACT

Mitomycin C (MMC) induces various types of DNA damages that cause significant cytotoxicity to cells. Accordingly, repair of MMC-induced damages involves multiple repair pathways such as nucleotide excision repair, homologous recombination repair and translesion bypass repair pathways. Nonetheless, repair of the MMC-induced DNA damages in mammals have not been fully delineated. In this study, we investigated potential roles for Xeroderma pigmentosum (XP) proteins in the repair of MMC-induced DNA damages using an assay that detects the ssDNA patches generated following treatment with MMC or 8'-methoxy-psoralen (8-MOP) + UVA (ultraviolet light A). Human wild-type cells formed distinctive ssDNA foci following treatment with MMC or 8-MOP + UVA, but not with those inducing alkylation damage, oxidative damage or strand-break damage, suggesting that the foci represent ssDNA patches formed during the crosslink repair. In contrast to wild-type cells, mutant defective in XPE orXPG did not form the ssDNA foci following MMC treatment, while XPF mutant cells showed a significantly delayed response in forming the foci. A positive role for XPG in the repair of MMC-induced DNA damages was further supported by observations that cells treated with MMC induced a tight association of XPG with chromatin, and a targeted inhibition of XPG abolished MMC-induced ssDNA foci formation, rendering cells hypersensitive to MMC. Together, our results suggest that XPG along with XPE and XPF play unique role(s) in the repair of MMC-induced DNA damages.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA Damage , DNA Repair , Mitomycin/pharmacology , Cell Culture Techniques , Chromatin/metabolism , DNA-Binding Proteins/physiology , Endonucleases/physiology , Fibroblasts , Humans , Nuclear Proteins/physiology , Oxidative Stress , Transcription Factors/physiology
8.
Blood ; 105(9): 3465-71, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15644418

ABSTRACT

Current strategies for genetic therapy using Moloney retroviruses require ex vivo manipulation of hematopoietic cells to facilitate stable integration of the transgene. While many studies have evaluated the impact of ex vivo culture on normal murine and human stem/progenitor cells, the cellular consequences of ex vivo manipulation of stem cells with intrinsic defects in genome stability are incompletely understood. Here we show that ex vivo culture of Fancc(-/-) bone marrow cells results in a time-dependent increase in apoptosis of primitive Fancc(-/-) progenitor cells in conditions that promote the proliferation of wild-type stem/progenitor cells. Further, recipients reconstituted with the surviving Fancc(-/-) cells have a high incidence of cytogenetic abnormalities and myeloid malignancies that are associated with an acquired resistance to tumor necrosis factor alpha (TNF-alpha). Collectively, these data indicate that the intrinsic defects in the genomic stability of Fancc(-/-) stem/progenitor cells provide a selective pressure for cells that are resistant to apoptosis and have a propensity for the evolution to clonal hematopoiesis and malignancy. These studies could have implications for the design of genetic therapies for treatment of Fanconi anemia and potentially other genetic diseases with intrinsic defects in genome stability.


Subject(s)
Apoptosis , Chromosome Aberrations , DNA-Binding Proteins/deficiency , Hematologic Neoplasms/etiology , Hematopoietic Stem Cells/pathology , Nuclear Proteins/deficiency , Animals , Bone Marrow Cells/pathology , Cell Cycle Proteins/genetics , Cells, Cultured , Clone Cells , DNA-Binding Proteins/genetics , Fanconi Anemia/pathology , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Mice, Knockout , Nuclear Proteins/genetics , Risk , Tumor Necrosis Factor-alpha/pharmacology
9.
J Biol Chem ; 279(49): 50986-93, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15377654

ABSTRACT

Fanconi anemia (FA) is a complex, heterogeneous genetic disorder composed of at least 11 complementation groups. The FA proteins have recently been found to functionally interact with the cell cycle regulatory proteins ATM and BRCA1; however, the function of the FA proteins in cell cycle control remains incompletely understood. Here we show that the Fanconi anemia complementation group C protein (Fancc) is necessary for proper function of the DNA damage-induced G2/M checkpoint in vitro and in vivo. Despite apparently normal induction of the G2/M checkpoint after ionizing radiation, murine and human cells lacking functional FANCC did not maintain the G2 checkpoint as compared with wild-type cells. The increased rate of mitotic entry seen in Fancc-/-mouse embryo fibroblasts correlated with decreased inhibitory phosphorylation of cdc2 kinase on tyrosine 15. An increased inability to maintain the DNA damage-induced G2 checkpoint was observed in Fancc -/-; Trp53 -/-cells compared with Fancc -/-cells, indicating that Fancc and p53 cooperated to maintain the G2 checkpoint. In contrast, genetic disruption of both Fancc and Atm did not cooperate in the G2 checkpoint. These data indicate that Fancc and p53 in separate pathways converge to regulate the G2 checkpoint. Finally, fibroblasts lacking FANCD2 were found to have a G2 checkpoint phenotype similar to FANCC-deficient cells, indicating that FANCD2, which is activated by the FA complex, was also required to maintain the G2 checkpoint. Because a proper checkpoint function is critical for the maintenance of genomic stability and is intricately related to the function and integrity of the DNA repair process, these data have implications in understanding both the function of FA proteins and the mechanism of genomic instability in FA.


Subject(s)
Cell Cycle Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Alleles , Animals , Bromodeoxyuridine/pharmacology , CDC2 Protein Kinase/metabolism , Cell Division , Cell Line , Cells, Cultured , Coloring Agents/pharmacology , DNA/metabolism , DNA Repair , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group D2 Protein , Fanconi Anemia Complementation Group Proteins , Female , Fibroblasts/metabolism , Flow Cytometry , G2 Phase , Histones/chemistry , Humans , Immunoblotting , Keratinocytes/metabolism , Male , Mice , Mice, Transgenic , Mitosis , Mutation , Phosphorylation , Protein Binding , Radiation, Ionizing , Time Factors , Transgenes , Tyrosine/chemistry
10.
J Biol Chem ; 279(29): 30053-9, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15138265

ABSTRACT

Fanconi anemia (FANC) is a heterogeneous genetic disorder characterized by a hypersensitivity to DNA-damaging agents, chromosomal instability, and defective DNA repair. Eight FANC genes have been identified so far, and five of them (FANCA, -C, -E, -F, and -G) assemble in a multinuclear complex and function at least in part in a complex to activate FANCD2 by monoubiquitination. Here we show that FANCA and FANCG are redox-sensitive proteins that are multimerized and/or form a nuclear complex in response to oxidative stress/damage. Both FANCA and FANCG proteins exist as monomers under non-oxidizing conditions, whereas they become multimers following H2O2 treatment. Treatment of cells with oxidizing agent not only triggers the multimeric complex of FANCA and FANCG in vivo but also induces the interaction between FANCA and FANCG. N-Ethylmaleimide treatment abolishes multimerization and interaction of FANCA and FANCG in vitro. Taken together, our results lead us to conclude that FANCA and FANCG uniquely respond to oxidative damage by forming complex(es) via intermolecular disulfide linkage(s), which may be crucial in forming such complexes and in determining their function.


Subject(s)
DNA-Binding Proteins/physiology , Fanconi Anemia/metabolism , Oxidative Stress , Proteins/physiology , Animals , Blotting, Western , COS Cells , Cloning, Molecular , DNA Damage , DNA Repair , DNA, Complementary/metabolism , Dimerization , Disulfides , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Ethylmaleimide/pharmacology , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group G Protein , Glutathione Transferase/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology , Mitomycin/pharmacology , Models, Biological , Oxidants/pharmacology , Oxidation-Reduction , Oxygen/metabolism , Precipitin Tests , Protein Structure, Tertiary
11.
J Biol Chem ; 279(7): 6046-55, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14617623

ABSTRACT

Ku70-Ku80 complex is the regulatory subunit of DNA-dependent protein kinase (DNA-PK) and plays an essential role in double-strand break repair following ionizing radiation (IR). It preferentially interacts with chromosomal breaks and protects DNA ends from nuclease attack. Here we show evidence that cells defective in Ku80 exhibit a significantly slow S phase progression following DNA damage. IR-induced retardation in S phase progression in Ku80-/- cells was not due to the lack of DNA-PK kinase activity because both wild-type cells and DNA-PKcs-deficient cells showed no such symptom. Instead, proliferating cell nuclear antigen (PCNA) dissociated from chromosomes following IR in Ku80-deficient cells but not in wild-type or DNA-PKcs-deficient cells. Treatment of HeLa cells with IR induced colocalization of the Ku complex with PCNA on chromosomes. Together, these results suggest that binding of the Ku complex at chromosomal breaks may be necessary to maintain the sliding clamps (PCNA) on chromatin, which would allow cells to resume DNA replication without a major delay following IR.


Subject(s)
Antigens, Nuclear/physiology , DNA Helicases , DNA Replication , DNA-Binding Proteins/physiology , Animals , Antigens, Nuclear/biosynthesis , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/metabolism , Cytosol/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/biosynthesis , Dimerization , Dose-Response Relationship, Radiation , HeLa Cells , Humans , Ku Autoantigen , Mice , Microscopy, Confocal , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Radiation, Ionizing , Time Factors
12.
Blood ; 102(12): 4146-52, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12855557

ABSTRACT

Fanconi anemia (FA) is a recessive genomic instability syndrome characterized by developmental defects, progressive bone marrow failure, and cancer. FA is genetically heterogeneous, however; the proteins encoded by different FA loci interact functionally with each other and with the BRCA1, BRCA2, and ATM gene products. Although patients with FA are highly predisposed to the development of myeloid leukemia and solid tumors, the alterations in biochemical pathways responsible for the progression of tumorigenesis in these patients remain unknown. FA cells are hypersensitive to a range of genotoxic and cellular stresses that activate signaling pathways mediating apoptosis. Here we show that ionizing radiation (IR) induces modestly elevated levels of p53 in cells from FA type C (Fancc) mutant mice and that inactivation of Trp53 rescues tumor necrosis factor alpha-induced apoptosis in myeloid cells from Fancc-/- mice. Further, whereas Fancc-/- mice failed to form hematopoietic or solid malignancies, mice mutant at both Fancc and Trp53 developed tumors more rapidly than mice mutant at Trp53 alone. This shortened latency was associated with the appearance of tumor types that are found in patients with FA but not in mice mutant at Trp53 only. Collectively, these data demonstrate that p53 and Fancc interact functionally to regulate apoptosis and tumorigenesis in Fancc-deficient cells.


Subject(s)
Apoptosis , Cell Cycle Proteins , DNA-Binding Proteins , Fanconi Anemia/pathology , Neoplasms/etiology , Nuclear Proteins , Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Embryo, Mammalian/cytology , Fanconi Anemia/complications , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Fibroblasts/pathology , Genetic Predisposition to Disease , Genotype , Inheritance Patterns , Mice , Mice, Knockout , Mice, Mutant Strains , Neoplasms/genetics , Neoplasms/pathology , Proteins/genetics , Radiation, Ionizing , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/radiation effects
13.
Blood ; 101(4): 1299-307, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12393504

ABSTRACT

Fanconi anemia (FA) is a chromosomal instability disorder characterized by a progressive bone marrow (BM) failure and an increased incidence of myeloid leukemias. Children with FA are currently being enrolled in clinical trials to evaluate the safety of retroviral-mediated gene transfer. Previously, we used Fancc(-/-) mice to show that Fancc(-/-) hematopoietic stem cells (HSCs) have a profound defect in repopulating ability. Here, we examined whether retroviral-mediated gene transfer of recombinant Fancc (rFancc) would restore the repopulating ability of Fancc(-/-) HSC to wild-type levels. Fancc(-/-) HSCs transduced with a retrovirus encoding rFancc exhibited a repopulating ability that approached wild-type levels. Interestingly, approximately 30% of primary recipients (7 of 22) transplanted with uncorrected Fancc(-/-) cells developed a range of hematopoietic abnormalities including pancytopenia and BM hypoplasia similar to individuals with FA. Hematopoietic abnormalities were detected in only 1 of 22 mice transplanted with Fancc(-/-) cells transduced with a retrovirus encoding rFancc. Moreover, several mice with hematopoietic defects had progenitors that displayed a marked resistance to IFN-gamma, TNF-alpha, and MIP-1alpha compared to both Fancc(-/-) progenitors, which are uniquely hypersensitive to these cytokines, and wild-type progenitors. These data are analogous to studies using progenitors from patients with myelodysplasia and provide functional support for clonal evolution in these mice. Collectively, these data show that gene transfer can enhance HSC repopulating ability and suppresses the tendency for clonal evolution. These studies also reveal potential detrimental effects of ex vivo manipulation for untransduced Fancc(-/-) HSCs.


Subject(s)
Cell Cycle Proteins , Clone Cells , DNA-Binding Proteins , Gene Expression , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Nuclear Proteins , Proteins/genetics , Retroviridae/genetics , Transfection , Animals , Blotting, Western , Bone Marrow/pathology , Cell Division , Disease Models, Animal , Fanconi Anemia , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Hematopoiesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Primary Myelofibrosis/pathology , Recombinant Proteins , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...