Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(9): 1702-1712, 2021 09.
Article in English | MEDLINE | ID: mdl-34158344

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive disease with poor outcomes, overwhelmingly due to relapse. Minimal residual disease (MRD), defined as the persistence of leukemic cells after chemotherapy treatment, is thought to be the major cause of relapse. The origins of relapse in AML have been traced to rare therapy-resistant leukemic stem cells (LSCs) that are already present at diagnosis. Effective treatment strategies for long-term remission are lacking, as it has been difficult to eliminate LSCs with conventional therapy. Here, we proposed a new approach based on the chimeric antigen receptor (CAR)-directed T lymphocytes, targeting T-cell immunoglobulin, and mucin domain 3 (TIM-3) to treat MRD in patients with AML. TIM-3 is selected as the target because it is highly expressed on AML blasts and LSCs in most subtypes regardless of the patient's genetic characteristics and treatment course. Moreover, it is absent in the normal hematopoietic stem cells, granulocytes, naïve lymphocytes, and most normal nonhematopoietic tissues. Using a naïve human Fab phage display library, we isolated an anti-human TIM-3 antibody and designed a second-generation anti-TIM-3. Our anti-TIM-3 CAR T cells exhibit potent antileukemic activity against AML cell lines and primary AML blasts, and in the mouse models. More importantly, we demonstrate efficient killing of the primary LSCs directly isolated from the patients. Hence, eradication of the LSCs present in the MRD by anti-TIM-3 CAR T-cell therapy following the first-line treatment may improve the clinical outcomes of patients with AML.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Cell Proliferation , Female , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
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.

3.
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
4.
Biol Blood Marrow Transplant ; 24(10): 1971-1980, 2018 10.
Article in English | MEDLINE | ID: mdl-29883796

ABSTRACT

The immunosuppressive properties of mesenchymal stromal cells (MSCs) have been clinically proven to be effective in treating graft-versus-host disease (GVHD). However, MSC therapy is limited by the need for laborious and expensive manufacturing processes that are fraught with batch-to-batch variability. Substitution of MSC therapy with key MSC-mediated immunomodulatory factors could be an option for GVHD treatment. Using a simulated in vitro model of the immunosuppressive effects of MSC on allogeneic graft reactions, a synergistic 2-factor combination (2FC) of CXCL5 and anti-CCL24 was identified from a panel of over 100 immunomodulatory factors as being superior to MSCs in the modulation of mixed lymphocyte reactions. This 2FC was superior to cyclosporine in ameliorating both moderate and severe GVHD while being equivalent to MSCs in moderate GVHD and superior to MSCs in severe GVHD. Its immunosuppressive efficacy could be further improved by extended treatment. Mechanistic studies revealed that in vitro the 2FC could only reduce the proliferation of Th 1 and Th 17, whereas in vivo CXCL5 acts in concert with anti-CCL24 antibody to reduce not only transplanted Th 1 and Th 17 but also cytotoxic T lymphocytes and natural killer cells to increase mouse immunosuppressive neutrophils without affecting human hematopoietic stem cell reconstitution. Concurrently, it reduced circulating human proinflammatory cytokines IFN-γ, IL-6, IL-17A, IL-8, macrophage inflammatory protein-1ß, and monocyte chemoattractant protein-1. Both in vitro and in vivo data suggest that CXCL5 and anti-CCL24 antibody act in concert to ameliorate GVHD via suppression of Th 1 and Th 17 responses. We propose that this novel 2FC could substitute for MSC therapy in GVHD treatment.


Subject(s)
Chemokine CCL24/pharmacology , Chemokine CXCL5/pharmacology , Cyclosporine/pharmacology , Graft vs Host Disease/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Animals , Disease Models, Animal , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Heterografts , Humans , Lymphocytes/immunology , Lymphocytes/pathology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Mice, SCID
5.
J Vis Exp ; (122)2017 04 08.
Article in English | MEDLINE | ID: mdl-28448034

ABSTRACT

The development of programmable genome-editing tools has facilitated the use of reverse genetics to understand the roles specific genomic sequences play in the functioning of cells and whole organisms. This cause has been tremendously aided by the recent introduction of the CRISPR/Cas9 system-a versatile tool that allows researchers to manipulate the genome and transcriptome in order to, among other things, knock out, knock down, or knock in genes in a targeted manner. For the purpose of knocking out a gene, CRISPR/Cas9-mediated double-strand breaks recruit the non-homologous end-joining DNA repair pathway to introduce the frameshift-causing insertion or deletion of nucleotides at the break site. However, an individual guide RNA may cause undesirable off-target effects, and to rule these out, the use of multiple guide RNAs is necessary. This multiplicity of targets also means that a high-volume screening of clones is required, which in turn begs the use of an efficient high-throughput technique to genotype the knockout clones. Current genotyping techniques either suffer from inherent limitations or incur high cost, hence rendering them unsuitable for high-throughput purposes. Here, we detail the protocol for using fluorescent PCR, which uses genomic DNA from crude cell lysate as a template, and then resolving the PCR fragments via capillary gel electrophoresis. This technique is accurate enough to differentiate one base-pair difference between fragments and hence is adequate in indicating the presence or absence of a frameshift in the coding sequence of the targeted gene. This precise knowledge effectively precludes the need for a confirmatory sequencing step and allows users to save time and cost in the process. Moreover, this technique has proven to be versatile in genotyping various mammalian cells of various tissue origins targeted by guide RNAs against numerous genes, as shown here and elsewhere.


Subject(s)
CRISPR-Cas Systems , Electrophoresis, Capillary/methods , Genotype , Polymerase Chain Reaction/methods , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , Genome , Genotyping Techniques , Hep G2 Cells , Humans , Mutation , RNA, Guide, Kinetoplastida/genetics
6.
Exp Hematol ; 48: 41-49, 2017 04.
Article in English | MEDLINE | ID: mdl-28087429

ABSTRACT

Xenograft models are transforming our understanding of the output capabilities of primitive human hematopoietic cells in vivo. However, many variables that affect posttransplantation reconstitution dynamics remain poorly understood. Here, we show that an equivalent level of human chimerism can be regenerated from human CD34+ cord blood cells transplanted intravenously either with or without additional radiation-inactivated cells into 2- to 6-month-old NOD-Rag1-/--IL2Rγc-/- (NRG) mice given a more radioprotective conditioning regimen than is possible in conventionally used, repair-deficient NOD-Prkdcscid/scid-IL2Rγc-/- (NSG) hosts. Comparison of sublethally irradiated and non-irradiated NRG mice and W41/W41 derivatives showed superior chimerism in the W41-deficient recipients, with some differential effects on different lineage outputs. Consistently superior outputs were observed in female recipients regardless of their genotype, age, or pretransplantation conditioning, with greater differences apparent later after transplantation. These results define key parameters for optimizing the sensitivity and minimizing the intraexperimental variability of human hematopoietic xenografts generated in increasingly supportive immunodeficient host mice.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins c-kit/genetics , Age Factors , Animals , Biomarkers , Cell Count , Female , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Sex Factors , Transplantation Chimera , Transplantation, Heterologous
7.
Sci Transl Med ; 8(359): 359ra129, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27708062

ABSTRACT

An in vitro drug-screening platform on patient samples was developed and validated to design personalized treatment for relapsed/refractory acute myeloid leukemia (AML). Unbiased clustering and correlation showed that homoharringtonine (HHT), also known as omacetaxine mepesuccinate, exhibited preferential antileukemia effect against AML carrying internal tandem duplication of fms-like tyrosine kinase 3 (FLT3-ITD). It worked synergistically with FLT3 inhibitors to suppress leukemia growth in vitro and in xenograft mouse models. Mechanistically, the effect was mediated by protein synthesis inhibition and reduction of short-lived proteins, including total and phosphorylated forms of FLT3 and its downstream signaling proteins. A phase 2 clinical trial of sorafenib and HHT combination treatment in FLT3-ITD AML patients resulted in complete remission (true or with insufficient hematological recovery) in 20 of 24 patients (83.3%), reduction of ITD allelic burden, and median leukemia-free and overall survivals of 12 and 33 weeks. The regimen has successfully bridged five patients to allogeneic hematopoietic stem cell transplantation and was well tolerated in patients unfit for conventional chemotherapy, including elderly and heavily pretreated patients. This study validated the principle and clinical relevance of in vitro drug testing and identified an improved treatment for FLT3-ITD AML. The results provided the foundation for phase 2/3 clinical trials to ascertain the clinical efficacy of FLT3 inhibitors and HHT in combination.


Subject(s)
Gene Duplication , Harringtonines/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Adult , Aged , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Culture Techniques , Cell Line, Tumor , Cluster Analysis , Drug Evaluation, Preclinical , Drug Synergism , Female , Harringtonines/pharmacology , Homoharringtonine , Humans , Male , Mice , Middle Aged , Models, Biological , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Biosynthesis/drug effects , Remission Induction , Sorafenib , Treatment Outcome , Young Adult , fms-Like Tyrosine Kinase 3/chemistry , fms-Like Tyrosine Kinase 3/metabolism
8.
Exp Hematol ; 44(7): 635-40, 2016 07.
Article in English | MEDLINE | ID: mdl-27090409

ABSTRACT

The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.


Subject(s)
Blood Platelets/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myelopoiesis , Neutrophils/metabolism , Thrombopoiesis , Animals , Cell Differentiation/drug effects , Cells, Cultured , Graft Survival , Hematopoietic Cell Growth Factors/metabolism , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Myelopoiesis/drug effects , Thrombopoiesis/drug effects
9.
Sci Rep ; 5: 15587, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26498861

ABSTRACT

Recent advances in the engineering of sequence-specific synthetic nucleases provide enormous opportunities for genetic manipulation of gene expression in order to study their cellular function in vivo. However, current genotyping methods to detect these programmable nuclease-induced insertion/deletion (indel) mutations in targeted human cells are not compatible for high-throughput screening of knockout clones due to inherent limitations and high cost. Here, we describe an efficient method of genotyping clonal CRISPR/Cas9-mediated mutants in a high-throughput manner involving the use of a direct lysis buffer to extract crude genomic DNA straight from cells in culture, and fluorescent PCR coupled with capillary gel electrophoresis. This technique also allows for genotyping of multiplexed gene targeting in a single clone. Overall, this time- and cost-saving technique is able to circumvent the limitations of current genotyping methods and support high-throughput screening of nuclease-induced mutants.


Subject(s)
CRISPR-Cas Systems , Electrophoresis, Capillary/methods , Gene Targeting , Genotyping Techniques/methods , Polymerase Chain Reaction/methods , Cell Line , Clone Cells , Humans , INDEL Mutation
10.
Cell Stem Cell ; 14(2): 253-63, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24440600

ABSTRACT

Cellular barcoding offers a powerful approach to characterize the growth and differentiation activity of large numbers of cotransplanted stem cells. Here, we describe a lentiviral genomic-barcoding and analysis strategy and its use to compare the clonal outputs of transplants of purified mouse and human basal mammary epithelial cells. We found that both sources of transplanted cells produced many bilineage mammary epithelial clones in primary recipients, although primary clones containing only one detectable mammary lineage were also common. Interestingly, regardless of the species of origin, many clones evident in secondary recipients were not detected in the primary hosts, and others that were changed from appearing luminal-restricted to appearing bilineage. This barcoding methodology has thus revealed conservation between mice and humans of a previously unknown diversity in the growth and differentiation activities of their basal mammary epithelial cells stimulated to grow in transplanted hosts.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Stem Cell Transplantation , Stem Cells/cytology , Animals , Cell Lineage , Cell Proliferation , Cell Size , Clone Cells , Epithelial Cells/cytology , Epithelial Cells/transplantation , Female , High-Throughput Nucleotide Sequencing , Humans , Mice , Regeneration
11.
Blood ; 122(18): 3129-37, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24030380

ABSTRACT

Human cord blood (CB) offers an attractive source of cells for clinical transplants because of its rich content of cells with sustained repopulating ability in spite of an apparent deficiency of cells with rapid reconstituting ability. Nevertheless, the clonal dynamics of nonlimiting CB transplants remain poorly understood. To begin to address this question, we exposed CD34+ CB cells to a library of barcoded lentiviruses and used massively parallel sequencing to quantify the clonal distributions of lymphoid and myeloid cells subsequently detected in sequential marrow aspirates obtained from 2 primary NOD/SCID-IL2Rγ(-/-) mice, each transplanted with ∼10(5) of these cells, and for another 6 months in 2 secondary recipients. Of the 196 clones identified, 68 were detected at 4 weeks posttransplant and were often lympho-myeloid. The rest were detected later, after variable periods up to 13 months posttransplant, but with generally increasing stability throughout time, and they included clones in which different lineages were detected. However, definitive evidence of individual cells capable of generating T-, B-, and myeloid cells, for over a year, and self-renewal of this potential was also obtained. These findings highlight the caveats and utility of this model to analyze human hematopoietic stem cell control in vivo.


Subject(s)
Cell Differentiation , Cell Proliferation , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Animals , Antigens, CD34/metabolism , Base Sequence , Cell Lineage , Clone Cells/classification , Clone Cells/cytology , Clone Cells/metabolism , DNA Barcoding, Taxonomic , Fetal Blood/metabolism , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunophenotyping , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Kinetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Molecular Sequence Data , Oligonucleotides/genetics , Time Factors , Transplantation, Heterologous
12.
Blood ; 121(5): e1-4, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23233660

ABSTRACT

UNLABELLED: Better methods to characterize normal human hematopoietic cells with short-term repopulating activity cells (STRCs) are needed to facilitate improving recovery rates in transplanted patients.We now show that 5-fold more human myeloid cells are produced in sublethally irradiated NOD/SCID-IL-2Receptor-γchain-null (NSG) mice engineered to constitutively produce human interleukin-3, granulocyte-macrophage colony-stimulating factor and Steel factor (NSG-3GS mice) than in regular NSG mice 3 weeks after an intravenous injection of CD34 human cord blood cells. Importantly, the NSG-3GS mice also show a concomitant and matched increase in circulating mature human neutrophils. Imaging NSG-3GS recipients of lenti-luciferase-transduced cells showed that human cells being produced 3 weeks posttransplant were heterogeneously distributed, validating the blood as a more representative measure of transplanted STRC activity. Limiting dilution transplants further demonstrated that the early increase in human granulopoiesis in NSG-3GS mice reflects an expanded output of differentiated cells per STRC rather than an increase in STRC detection. KEY POINTS: NSG-3GS mice support enhanced clonal outputs from human short-term repopulating cells (STRCs) without affecting their engrafting efficiency. Increased human STRC clone sizes enable their more precise and efficient measurement by peripheral blood monitoring.


Subject(s)
Cord Blood Stem Cell Transplantation , Graft Survival , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-3/biosynthesis , Myelopoiesis , Neutrophils/metabolism , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-3/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neutrophils/cytology , Time Factors , Transplantation, Heterologous
13.
Blood ; 119(15): 3431-9, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22374695

ABSTRACT

Delayed recovery of mature blood cells poses a serious, expensive, and often life-threatening problem for many stem cell transplantation recipients, particularly if heavily pretreated and serving as their own donor, or having a CB transplantation as the only therapeutic option. Importantly, the different cells required to ensure a rapid, as well as a permanent, hematopoietic recovery in these patients remain poorly defined. We now show that human CB and mobilized peripheral blood (mPB) collections contain cells that produce platelets and neutrophils within 3 weeks after being transplanted into sublethally irradiated NOD/scid-IL-2Rγc-null mice. The cells responsible for these 2 outputs are similarly distributed between the aldehyde dehydrogenase-positive and -negative subsets of lineage marker-negative CB and mPB cells, but their overall frequencies vary independently in individual samples. In addition, their total numbers can be seen to be much (> 30-fold) lower in a single "average" CB transplantation compared with a single "average" mPB transplantation (normalized for a similar weight of the recipient), consistent with the published differential performance in adult patients of these 2 transplantation products. Experimental testing confirmed the clinical relevance of the surrogate xenotransplantation assay for quantifying cells with rapid platelet regenerative activity, underscoring its potential for future applications.


Subject(s)
Blood Cells/cytology , Blood Cells/physiology , Blood Platelets/physiology , Hematopoietic Stem Cell Transplantation , Neutrophils/physiology , Adult , Animals , Blood Cells/classification , Humans , Infant, Newborn , Leukocyte Count , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Phenotype , Platelet Count , Transplantation, Heterologous
14.
Blood ; 119(22): 5133-43, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22368270

ABSTRACT

Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 (FLT3) gene occurs in 30% of acute myeloid leukemias (AMLs) and confers a poor prognosis. Thirteen relapsed or chemo-refractory FLT3-ITD(+) AML patients were treated with sorafenib (200-400 mg twice daily). Twelve patients showed clearance or near clearance of bone marrow myeloblasts after 27 (range 21-84) days with evidence of differentiation of leukemia cells. The sorafenib response was lost in most patients after 72 (range 54-287) days but the FLT3 and downstream effectors remained suppressed. Gene expression profiling showed that leukemia cells that have become sorafenib resistant expressed several genes including ALDH1A1, JAK3, and MMP15, whose functions were unknown in AML. Nonobese diabetic/severe combined immunodeficiency mice transplanted with leukemia cells from patients before and during sorafenib resistance recapitulated the clinical results. Both ITD and tyrosine kinase domain mutations at D835 were identified in leukemia initiating cells (LICs) from samples before sorafenib treatment. LICs bearing the D835 mutant have expanded during sorafenib treatment and dominated during the subsequent clinical resistance. These results suggest that sorafenib have selected more aggressive sorafenib-resistant subclones carrying both FLT3-ITD and D835 mutations, and might provide important leads to further improvement of treatment outcome with FLT3 inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzenesulfonates/administration & dosage , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Pyridines/administration & dosage , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Adult , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/adverse effects , Benzenesulfonates/adverse effects , Bone Marrow/enzymology , Bone Marrow/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Humans , Janus Kinase 3/biosynthesis , Janus Kinase 3/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Male , Matrix Metalloproteinase 15/biosynthesis , Matrix Metalloproteinase 15/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Structure, Tertiary , Pyridines/adverse effects , Retinal Dehydrogenase , Sorafenib , Time Factors , Transplantation, Heterologous , fms-Like Tyrosine Kinase 3/metabolism
15.
Leuk Res ; 34(10): 1390-4, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20170959

ABSTRACT

We evaluated the NOD/SCID engraftment of CD34(+) cells from polycythemia vera (PV) and secondary polycythemia patients (SP) and the JAK2V617F clone before and after transplantation. Peripheral blood CD34(+) cells were transplanted intra-femorally. In the injected BM, successful engraftment (>0.1%) occurred in 8/26 mice transplanted with CD34+ cells from 5/13 PV patients (median: 4.26%, range: 0.3-5.56%), in contrast to 0/14 mice from 9 SP patients (P=0.017). The engrafting PV cells were of multi-lineage. JAK2V617F/total JAK2 ratios decreased after transplantation (initial: 65.9% versus 6-week: 13.0%, P=0.001). Essential thrombocythemia (ET) BM cells also exhibited a similar decrease in JAK2V617F clone. The results suggested that events in addition to JAK2V617F are involved in the pathogenesis of PV and ET.


Subject(s)
Antigens, CD34/analysis , Hematopoietic Stem Cell Transplantation , Janus Kinase 2/genetics , Mutation , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/genetics , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Polycythemia Vera/etiology , Primary Myelofibrosis/etiology , Thrombocythemia, Essential/etiology
16.
Exp Hematol ; 38(1): 3-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19837128

ABSTRACT

OBJECTIVE: Xenogeneic transplantation has been the gold standard for enumeration of leukemia initiating cells in acute myeloid and lymphoblastic leukemia (ALL). Most transplantation models have required conditioning in which the recipients were either irradiated or treated with chemotherapy prior to injection of human leukemia cells. In this study, we reported an undescribed model in which adult ALL cells were injected into unconditioned newborn nonobese diabetic severe combined immunodeficient mice via an intrahepatic route. MATERIALS AND METHODS: Bone marrow (BM) and peripheral blood were collected from patients with ALL at diagnosis or relapse. CD34(+) selected lymphoblasts or mononuclear cells were transplanted as mentioned previously. Cells were also transplanted into sublethally irradiated adult mice via intravenous route for comparison. Leukemia engraftment was enumerated from mouse BM 6 to 18 weeks after transplantation. Clonality of the engrafting cells was examined based on IGH rearrangement and fluorescent in situ hybridization. RESULTS: Five of 13 ALL samples engrafted into the recipient BM 6 to 18 weeks after transplantation. Engrafted cells recapitulated the immunophenotype and cytogenetic characteristics of the original samples. Engraftment in BM and peripheral blood was significantly correlated. Importantly, there was significant correlation of engraftment between this and the conventional adult nonobese diabetic severe combined immunodeficient mouse model involving irradiation. CONCLUSION: Our results demonstrated that this unconditioned newborn mouse model could be used for enumeration of leukemia initiating cells in ALL and should be further evaluated.


Subject(s)
Neoplasm Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adult , Animals , Animals, Newborn , Base Sequence , Cell Lineage , DNA Primers , Female , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Spectral Karyotyping , Transplantation, Heterologous
17.
Leuk Res ; 34(1): 119-22, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19683812

ABSTRACT

In this study, we tested if FLT3/internal tandem duplication (ITD) in acute myeloid leukemia (AML) might occur at different hierarchical stages during leukemogenesis. In 56 AML cases, 10 showed FLT3/ITD (single ITD=5; multiple ITD=5). Myeloblasts from seven cases (CD34-selected=4; unselected=3) were transplanted into NOD/SCID mice. Five cases engrafted successfully into 14 mice. Two patients carried single FLT3/ITD subclones, which were maintained during primary and secondary transplantations. In three patients with multiple FLT3/ITD subclones, some subclones persisted or expanded while others diminished upon transplantation. Their different engraftment capabilities in NOD/SCID mice supported the proposition that FLT3/ITD might occur at different stages during leukemogenesis.


Subject(s)
Gene Duplication , Leukemia, Myeloid, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3/chemistry
18.
Br J Haematol ; 144(4): 484-91, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19055666

ABSTRACT

To examine the differences between primitive bone marrow (BM) and peripheral blood (PB) myeloblasts in acute myeloid leukaemia (AML), we compared CD34(+) myeloblasts of paired BM and PB samples from 14 AML patients in terms of surface phenotype, homing and engraftment in a xenogeneic transplantation model, and gene expression, based on microarray studies and quantitative polymerase chain reaction. While there was no significant difference in surface phenotypes between these two populations, in vivo assay showed significantly better homing potential of PB CD34(+) cells than BM CD34(+) cells. Significant correlation between homing and engraftment in AML samples was also noted. In addition, gene expression profiling of CD34(+) cells from five paired BM and PB leukaemic samples showed that genes involved in G-protein and prostaglandin signalling, chemotaxis and stress response, cell proliferation and apoptosis were down-regulated in PB CD34(+) myeloblasts. These data suggested that circulating primitive myeloblasts in AML are functionally different from those residing in the marrow compartment, and such differences may be partly regulated by the BM microenvironment.


Subject(s)
Antigens, CD34/analysis , Bone Marrow Cells/physiology , Granulocyte Precursor Cells/physiology , Leukemia, Myeloid, Acute/pathology , Adult , Aged , Animals , Antigens, Neoplasm/analysis , Female , Gene Expression Profiling/methods , Graft Survival , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/blood , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Transplantation , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods , Transplantation, Heterologous
19.
J Biol Chem ; 283(24): 16545-53, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18408007

ABSTRACT

The Forkhead box transcription factor FoxM1 is expressed in proliferating cells. When it was depleted in mice and cell lines, cell cycle defects and chromosomal instability resulted. Premature senescence was observed in embryonic fibroblasts derived from FoxM1 knock-out mice, but the underlying cause has remained unclear. To investigate whether FoxM1 can protect cells against stress-induced premature senescence, we established NIH3T3 lines with doxycycline-inducible overexpression of FoxM1c. Treatment of these lines with sublethal doses (20 and 100 microm) of H(2)O(2) induced senescence with senescence-associated beta-galactosidase expression and elevated levels of p53 and p21. Induction of FoxM1c expression markedly suppressed senescence and expression of p53 and p21. Consistent with down-regulation of the p19(Arf)-p53 pathway, p19(Arf) levels decreased while expression of the Polycomb group protein Bmi-1 was induced. That Bmi-1 is a downstream target of FoxM1c was further supported by the dose-dependent induction of Bmi-1 by FoxM1c at both the protein and mRNA levels, and FoxM1 and Bmi-1 reached maximal levels in cells at the G(2)/M phase. Depletion of FoxM1 by RNA interference decreased Bmi-1 expression. Using Bmi-1 promoter reporters with wild-type and mutated c-Myc binding sites and short hairpin RNAs targeting c-Myc, we further demonstrated that FoxM1c activated Bmi-1 expression via c-Myc, which was recently reported to be regulated by FoxM1c. Our results reveal a functional link between FoxM1c, c-Myc, and Bmi-1, which are major regulators of tumorigenesis. This link has important implications for the regulation of cell proliferation and senescence by FoxM1 and Bmi-1.


Subject(s)
Forkhead Transcription Factors/physiology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/physiology , Oxidative Stress , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Animals , Anti-Bacterial Agents/pharmacology , Binding Sites , Cellular Senescence , Doxycycline/pharmacology , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
20.
Exp Hematol ; 35(1): 56-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198874

ABSTRACT

OBJECTIVE: We have previously shown that all-trans retinoic acid (ATRA) enhanced the maintenance of early human hematopoietic progenitor cells (HPCs) in the presence of an irradiated stromal cell line AFT024. In this study, we examined the effects of ATRA on the stromal cell component with particular reference to cellular proliferation and gene expression. METHODS: Irradiated AFT024 cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with fetal bovine serum and were incubated with ATRA at 1 mumol/L up to 21 days. The cells were examined in terms of immunostaining for proliferative cell nuclear antigen (PCNA) and BrdU incorporation, apoptosis assay, cell cycle analysis, and gene expression using semiquantitative reverse-transcriptase polymerase chain reaction. RESULTS: In the control experiments, AFT024 cells lost their confluence in culture after 15-Gy irradiation and were arrested in G2/M phase on days 7 and 21. ATRA restored the cellular confluence with an increase in proliferation on day 21 (BrdU incorporation: 20.6-fold; PCNA staining: 51.7-fold) with reversal of cell cycle arrest (S phase: 2.7-fold increase; G2/M phase: 2.0-fold decrease). There was no effect on apoptosis as shown by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining. ATRA significantly upregulated the expression of cell cycle genes for checkpoint transition, including cyclin A2, B2, and aurora kinase B, as well as genes associated with a putative role in HPC maintenance, including osteopontin, HoxA5, enhancer of zeste homolog 2, and peroxisome proliferator-activated receptor gamma. CONCLUSION: We concluded that ATRA induced cellular proliferation of irradiated AFT024 cells and expression of a number of genes whose relevance to HPC homeostasis would have to be further examined.


Subject(s)
Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Stromal Cells/cytology , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , G2 Phase/drug effects , G2 Phase/radiation effects , Gene Expression Regulation/genetics , Genes, cdc/drug effects , Mice , Stem Cells/cytology , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...