Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cancer Res ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38832931

ABSTRACT

DNA methyltransferase inhibitors (DNMTi), most commonly cytidine analogs, are compounds that decrease 5'-cytosine methylation. DNMTi are used clinically based on the hypothesis that cytosine demethylation will lead to re-expression of tumor suppressor genes. 5-Aza-4'-thio-2'-deoxycytidine (Aza TdCyd or ATC) is a recently described thiol substituted DNMTi that has been shown to have anti-tumor activity in solid tumor models. Here, we investigated the therapeutic potential of ATC in a murine transplantation model of myelodysplastic syndrome. ATC treatment led to transformation of transplanted wild-type bone marrow nucleated cells into lymphoid leukemia, and healthy mice treated with ATC also developed lymphoid leukemia. Whole exome sequencing revealed thousands of acquired mutations, almost all of which were C>G transversions in a specific 5'-NCG-3' context. These mutations involved dozens of genes involved in human lymphoid leukemia, such as Notch1, Pten, Pax5, Trp53, and Nf1. Human cells treated in vitro with ATC showed thousands of acquired C>G transversions in a similar context. Deletion of Dck, the rate-limiting enzyme for the cytidine salvage pathway, eliminated C>G transversions. Taken together, these findings demonstrate a highly penetrant mutagenic and leukemogenic phenotype associated with ATC.

2.
Res Sq ; 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38168433

ABSTRACT

DNA methyltransferase inhibitors (DNMTi), most commonly cytidine analogs, are compounds that are used clinically to decrease 5'-cytosine methylation, with the aim of re-expression of tumor suppressor genes. We used a murine pre-clinical model of myelodysplastic syndrome based on transplantation of cells expressing a NUP98::HOXD13 transgene to investigate 5-Aza-4'-thio-2'-deoxycytidine (Aza TdCyd or ATC), a thiol substituted DNMTi, as a potential therapy. We found that ATC treatment led to lymphoid leukemia in wild-type recipient cells; further study revealed that healthy mice treated with ATC also developed lymphoid leukemia. Whole exome sequencing revealed thousands of acquired mutations, almost all of which were C > G transversions in a previously unrecognized, specific 5'-NCG-3' context. These mutations involved dozens of genes well-known to be involved in human lymphoid leukemia, such as Notch1, Pten, Pax5, Trp53 , and Nf1 . Treatment of human cells in vitro showed thousands of acquired C > G transversions in a similar context. Deletion of Dck , the rate-limiting enzyme for the cytidine salvage pathway, eliminated C > G transversions. Taken together, these findings demonstrate that DNMTi can be potent mutagens in human and mouse cells, both in vitro and in vivo .

3.
FASEB J ; 36(9): e22430, 2022 09.
Article in English | MEDLINE | ID: mdl-35920299

ABSTRACT

Minichromosome maintenance proteins (Mcm2-7) form a hexameric complex that unwinds DNA ahead of a replicative fork. The deficiency of Mcm proteins leads to replicative stress and consequent genomic instability. Mice with a germline insertion of a Cre cassette into the 3'UTR of the Mcm2 gene (designated Mcm2Cre ) have decreased Mcm2 expression and invariably develop precursor T-cell lymphoblastic leukemia/lymphoma (pre-T LBL), due to 100-1000 kb deletions involving important tumor suppressor genes. To determine whether mice that were protected from pre-T LBL would develop non-T-cell malignancies, we used two approaches. Mice engrafted with Mcm2Cre/Cre Lin- Sca-1+ Kit+ hematopoietic stem/progenitor cells did not develop hematologic malignancy; however, these mice died of hematopoietic stem cell failure by 6 months of age. Placing the Mcm2Cre allele onto an athymic nu/nu background completely prevented pre-T LBL and extended survival of these mice three-fold (median 296.5 vs. 80.5 days). Ultimately, most Mcm2Cre/Cre ;nu/nu mice developed B-cell precursor acute lymphoblastic leukemia (BCP-ALL). We identified recurrent deletions of 100-1000 kb that involved genes known or suspected to be involved in BCP-ALL, including Pax5, Nf1, Ikzf3, and Bcor. Moreover, whole-exome sequencing identified recurrent mutations of genes known to be involved in BCP-ALL progression, such as Jak1/Jak3, Ptpn11, and Kras. These findings demonstrate that an Mcm2Cre/Cre hypomorph can induce hematopoietic dysfunction via hematopoietic stem cell failure as well as a "deletor" phenotype affecting known or suspected tumor suppressor genes.


Subject(s)
Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Minichromosome Maintenance Complex Component 2 , Animals , DNA Replication , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Minichromosome Maintenance Complex Component 2/genetics , Mutation , Repressor Proteins/genetics , Transcription Factors/metabolism
4.
Cancer Res ; 81(19): 5033-5046, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34321240

ABSTRACT

Mutations in the isocitrate dehydrogenase 1 (IDH1) and IDH2 genes are frequently observed in a wide variety of hematologic malignancies, including myeloid and T-cell leukemias. In this study, we generated Idh2R140Q transgenic mice to examine the role of the Idh2R140Q mutation in leukemia. No leukemia developed in Idh2R140Q transgenic mice, suggesting a need for additional genetic events for leukemia development. Because myeloid cells from NUP98-HOXD13 fusion (NHD13) transgenic mice frequently acquire somatic Idh mutations when they transform to acute myeloid leukemia, we generated Idh2R140Q/NHD13 double transgenic mice. Idh2R140Q/NHD13 transgenic mice developed an immature T-cell leukemia with an immunophenotype similar to double-negative 1 (DN1) or DN2 thymocytes. Idh2R140Q/NHD13 leukemic cells were enriched for an early thymic precursor transcriptional signature, and the gene expression profile for Idh2R140Q/NHD13 DN1/DN2 T-ALL closely matched that of human early/immature T-cell precursor (EITP) acute lymphoblastic leukemia (ALL). Moreover, recurrent mutations found in patients with EITP ALL, including KRAS, PTPN11, JAK3, SH2B3, and EZH2 were also found in Idh2R140Q/NHD13 DN1/DN2 T-ALL. In vitro treatment of Idh2R140Q/NHD13 thymocytes with enasidenib, a selective inhibitor of mutant IDH2, led to a marked decrease in leukemic cell proliferation. These findings demonstrate that Idh2R140Q/NHD13 mice can serve as a useful in vivo model for the study of early/immature thymocyte precursor acute lymphoblastic leukemia development and therapy. SIGNIFICANCE: T-cell leukemia induced in Idh2R140Q/NUP98-HOXD13 mice is immunophenotypically, transcriptionally, and genetically similar to human EITP ALL, providing a model for studying disease development and treatment.


Subject(s)
Homeodomain Proteins/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Thymocytes/metabolism , Animals , Biomarkers, Tumor , Cell Differentiation/genetics , Cell Line, Tumor , Computational Biology/methods , DNA Methylation , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Heterografts , Humans , Immunophenotyping , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymocytes/pathology , Transcriptome
5.
J Cell Biol ; 220(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33620383

ABSTRACT

Chromosomal instability (CIN) is a hallmark of many cancers. Restricting the localization of centromeric histone H3 variant CENP-A to centromeres prevents CIN. CENP-A overexpression (OE) and mislocalization have been observed in cancers and correlate with poor prognosis; however, the molecular consequences of CENP-A OE on CIN and aneuploidy have not been defined. Here, we show that CENP-A OE leads to its mislocalization and CIN with lagging chromosomes and micronuclei in pseudodiploid DLD1 cells and xenograft mouse model. CIN is due to reduced localization of proteins to the kinetochore, resulting in defects in kinetochore integrity and unstable kinetochore-microtubule attachments. CENP-A OE contributes to reduced expression of cell adhesion genes and higher invasion of DLD1 cells. We show that CENP-A OE contributes to aneuploidy with karyotypic heterogeneity in human cells and xenograft mouse model. In summary, our results provide a molecular link between CENP-A OE and aneuploidy, and suggest that karyotypic heterogeneity may contribute to the aggressive phenotype of CENP-A-overexpressing cancers.


Subject(s)
Aneuploidy , Centromere Protein A/biosynthesis , Chromosomal Instability , Kinetochores/metabolism , Micronuclei, Chromosome-Defective , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Cell Line, Tumor , Centromere Protein A/genetics , Heterografts , Humans , Kinetochores/pathology , Mice , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology
6.
Sci Rep ; 9(1): 17213, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748606

ABSTRACT

Transgenic mice that express either a NUP98-PHF23 (NP23) or NUP98-HOXD13 (NHD13) fusion in the hematopoietic compartment develop a wide spectrum of leukemias, including myeloid, erythroid, megakaryocytic and lymphoid, at age 9-14 months. NP23-NHD13 double transgenic mice were generated by interbreeding NP23 and NHD13 mice. Remarkably, 100% of the NP23-NHD13 double transgenic mice developed acute myeloid leukemia (AML) within three months, characterized by replacement of the thymus with leukemic myeloblasts. The marked infiltration of thymus led to the intriguing hypothesis that AML generated in NP23-NHD13 mice arose in the thymus, as opposed to the bone marrow (BM). Transplantation of CD4-CD8- double negative (DN) thymocytes (which were also negative for Mac1 and Gr1) from leukemic NHD13/NP23 mice demonstrated that DN thymocytes could transmit AML, and limiting dilution studies showed that leukemia initiating cells were increased 14-fold in the thymus compared to BM. Further thymocyte fractionation demonstrated that DN1 and DN2, but not DN3 or DN4 fractions transmitted AML, and a marked expansion (100-fold) of Lineage-Sca1 + Kit + (LSK) cells in the thymus of the NP23-NHD13 mice. Taken together, these results show that the thymus of NP23-NHD13 mice acts as a reservoir for AML initiating cells and that thymic progenitors can transmit AML.


Subject(s)
Cell Transformation, Neoplastic/pathology , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Nuclear Pore Complex Proteins/metabolism , Stem Cell Transplantation/methods , Thymocytes/cytology , Transcription Factors/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Transgenic , Nuclear Pore Complex Proteins/genetics , Transcription Factors/genetics
7.
Blood ; 133(24): 2610-2614, 2019 06 13.
Article in English | MEDLINE | ID: mdl-30992267

ABSTRACT

Approximately 10% of NUP98-PHF23 (NP23) mice develop an aggressive acute lymphoblastic leukemia of B-1 lymphocyte progenitor origin (pro-B1 ALL), accompanied by somatic frameshift mutations of the BCL6 interacting corepressor (Bcor) gene, most commonly within a 9-bp "hotspot" in Bcor exon 8. To determine whether experimentally engineered Bcor mutations would lead to pro-B1 ALL, we used clustered, regularly interspaced, short palindromic repeats-associated protein 9 to introduce a Bcor frameshift mutation into NP23 hematopoietic stem and progenitor cells through the use of Bcor small guide RNAs (Bcor sgRNAs). Recipient mice transplanted with NP23 bone marrow or fetal liver cells that had been transduced with a Bcor sgRNA developed pro-B1 ALL, characterized by a B-1 progenitor immunophenotype, clonal Igh gene rearrangement, and Bcor indel mutation, whereas control recipients did not. Similar to a subset of human B-cell precursor ALL, the murine pro-B1 ALL had acquired somatic mutations in Jak kinase genes. JAK inhibitors (ruxolitinib and tofacitinib) inhibited the growth of pro-B1 ALL cell lines established from Bcor sgRNA/NP23 recipients at clinically achievable concentrations (100 nM). Our results demonstrate that Bcor mutations collaborate with NP23 to induce pro-B1 ALL, and that JAK inhibitors are potential therapies for pro-B1 ALL.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Repressor Proteins/genetics , Animals , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Frameshift Mutation , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Mice , Mice, Transgenic , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology
8.
J Vis Exp ; (140)2018 10 03.
Article in English | MEDLINE | ID: mdl-30346380

ABSTRACT

Myelodysplastic syndromes (MDS) are a diverse group of hematopoietic stem cell disorders that are defined by ineffective hematopoiesis, peripheral blood cytopenias, dysplasia, and a propensity for transformation to acute leukemia. NUP98-HOXD13 (NHD13) transgenic mice recapitulate human MDS in terms of peripheral blood cytopenias, dysplasia, and transformation to acute leukemia. We previously demonstrated that MDS could be transferred from a genetically engineered mouse with MDS to wild-type recipients by transplanting MDS bone marrow nucleated cells (BMNC). To more clearly understand the MDS cell of origin, we have developed approaches to transplant specific, immunophenotypically defined hematopoietic subsets. In this article, we describe the process of isolating and transplanting specific populations of hematopoietic stem and progenitor cells. Following transplantation, we describe approaches to assess the efficiency of transplantation and persistence of the donor MDS cells.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/therapy , Animals , Cell Separation , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL
9.
Cell Stem Cell ; 23(3): 355-369.e9, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146412

ABSTRACT

Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is derived from aberrant clonal hematopoietic stem/progenitor cells (HSPCs) that persist after conventional therapies. Defining the mechanisms underlying MDS HSPC maintenance is critical for developing MDS therapy. The deacetylase SIRT1 regulates stem cell proliferation, survival, and self-renewal by deacetylating downstream proteins. Here we show that SIRT1 protein levels were downregulated in MDS HSPCs. Genetic or pharmacological activation of SIRT1 inhibited MDS HSPC functions, whereas SIRT1 deficiency enhanced MDS HSPC self-renewal. Mechanistically, the inhibitory effects of SIRT1 were dependent on TET2, a safeguard against HSPC transformation. SIRT1 deacetylated TET2 at conserved lysine residues in its catalytic domain, enhancing TET2 activity. Our genome-wide analysis identified cancer-related genes regulated by the SIRT1/TET2 axis. SIRT1 activation also inhibited functions of MDS HSPCs from patients with TET2 heterozygous mutations. Altogether, our results indicate that restoring TET2 function through SIRT1 activation represents a promising means to target MDS HSPCs.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Myelodysplastic Syndromes/metabolism , Proto-Oncogene Proteins/metabolism , Sirtuin 1/metabolism , Animals , DNA-Binding Proteins/genetics , Dioxygenases , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured
10.
PLoS One ; 12(9): e0185219, 2017.
Article in English | MEDLINE | ID: mdl-28953912

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (A-HSCT) remains the only curative option for patients with myelodysplastic syndrome (MDS). We used the NUP98-HOXD13 (NHD13) murine model for MDS to study HSCT in a pre-clinical setting. NHD13 recipients transplanted with syngeneic bone marrow (S-HSCT) following myeloablative irradiation showed disease remission, with normalization of peripheral blood parameters and marked decrease in circulating leukocytes derived from the MDS clone. Despite the disease remission and improved survival compared to non-transplanted NHD13 controls, all mice eventually relapsed, indicating persistence of a long-lived radio-resistant MDS clone. In an effort to induce a graft versus leukemia (GVL) effect, A-HSCT with donor bone marrow that was mismatched at minor histocompatibility loci was compared to S-HSCT. Although recipients in the A-HSCT showed a lower early relapse rate than in S-HSCT, all mice in both groups eventually relapsed and died by 54 weeks post-transplant. To obtain a more significant GVL effect, donor splenocytes containing reactive T-cells were transplanted with allogeneic bone marrow. Although the relapse rate was only 20% at post-transplantation week 38, suggesting a GVL effect, this was accompanied by a severe graft versus host disease (GVHD) Taken together, these findings indicate that a myeloablative dose of ionizing radiation is insufficient to eradicate the MDS initiating cell, and that transplantation of donor splenocytes leads to decreased relapse rates, at the cost of severe GVHD. We suggest that NHD13 mice represent a feasible pre-clinical model for the study of HSCT for MDS.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes/therapy , Animals , Disease Models, Animal , Graft vs Host Disease/etiology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Mice, Inbred C57BL , Recurrence , Remission Induction , Survival Analysis , T-Lymphocytes, Regulatory/immunology , Transplantation Conditioning/adverse effects , Transplantation, Homologous
11.
Blood Adv ; 1(20): 1749-1759, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29296821

ABSTRACT

B-1 and B-2 lymphocytes are derived from distinct developmental pathways and represent layered arms of the innate and adaptive immune systems, respectively. In contrast to a majority of murine B-cell malignancies, which stain positive with the B220 antibody, we discovered a novel form of B-cell leukemia in NUP98-PHF23 (NP23) transgenic mice. The immunophenotype (Lin- B220- CD19+ AA4.1+) was identical to that of progenitor (pro) B-1 cells, and VH gene usage was skewed toward 3' V regions, similar to murine fetal liver B cells. Moreover, the gene expression profile of these leukemias was most similar to that of fetal liver pro-B fraction BC, a known source of B-1 B cells, further supporting a pro-B-1 origin of these leukemias. The NP23 pro-B-1 acute lymphoblastic leukemias (ALLs) acquired spontaneous mutations in both Bcor and Janus kinase (Jak) pathway (Jak1/2/3 and Stat5a) genes, supporting a hypothesis that mutations in 3 critical pathways (stem-cell self-renewal, B-cell differentiation, and cytokine signaling) collaborate to induce B-cell precursor (BCP) ALL. Finally, the thymic stromal lymphopoietin (Tslp) cytokine is required for murine B-1 development, and chromosomal rearrangements resulting in overexpression of the TSLP receptor (CRLF2) are present in some patients with high-risk BCP-ALL (referred to as CRLF2r ALL). Gene expression profiles of NP23 pro-B-1 ALL were more similar to that of CRLF2r ALL than non-CRLF2r ALL, and analysis of VH gene usage from patients with CRLF2r ALL demonstrated preferential usage of VH regions used by human B-1 B cells, leading to the suggestion that this subset of patients with BCP-ALL has a malignancy of B-1, rather than B-2, B-cell origin.

12.
Am J Pathol ; 186(3): 701-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774856

ABSTRACT

The transcription factor far upstream element binding protein (FBP) binds and activates the MYC promoter when far upstream element is via TFIIH helicase activity early in the transcription cycle. The fundamental biology and pathology of FBP are complex. In some tumors FBP seems pro-oncogenic, whereas in others it is a tumor suppressor. We generated an FBP knockout (Fubp1(-/-)) mouse to study FBP deficiency. FBP is embryo lethal from embryonic day 10.5 to birth. A spectrum of pathology is associated with FBP loss; besides cerebral hyperplasia and pulmonary hypoplasia, pale livers, hypoplastic spleen, thymus, and bone marrow, cardiac hypertrophy, placental distress, and small size were all indicative of anemia. Immunophenotyping of hematopoietic cells in wild-type versus knockout livers revealed irregular trilineage anemia, with deficits in colony formation. Despite normal numbers of hematopoietic stem cells, transplantation of Fubp1(-/-) hematopoietic stem cells into irradiated mice entirely failed to reconstitute hematopoiesis. In competitive transplantation assays against wild-type donor bone marrow, Fubp1(-/-) hematopoietic stem cells functioned only sporadically at a low level. Although cultures of wild-type mouse embryo fibroblasts set Myc levels precisely, Myc levels of mouse varied wildly between fibroblasts harvested from different Fubp1(-/-) embryos, suggesting that FBP contributes to Myc set point fixation. FBP helps to hold multiple physiologic processes to close tolerances, at least in part by constraining Myc expression.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation , Hematopoiesis/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Fibroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Pregnancy , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/genetics
13.
Cancer Discov ; 4(5): 564-77, 2014 May.
Article in English | MEDLINE | ID: mdl-24535671

ABSTRACT

In this report, we show that expression of a NUP98-PHF23 (NP23) fusion, associated with acute myeloid leukemia (AML) in humans, leads to myeloid, erythroid, T-cell, and B-cell leukemia in mice. The leukemic and preleukemic tissues display a stem cell-like expression signature, including Hoxa, Hoxb, and Meis1 genes. The PHF23 plant homeodomain (PHD) motif is known to bind to H3K4me3 residues, and chromatin immunoprecipitation experiments demonstrated that the NP23 protein binds to chromatin at a specific subset of H3K4me3 sites, including at Hoxa, Hoxb, and Meis1. Treatment of NP23 cells with disulfiram, which inhibits the binding of PHD motifs to H3K4me3, rapidly and selectively killed NP23-expressing myeloblasts; cell death was preceded by decreased expression of Hoxa, Hoxb, and Meis1. Furthermore, AML driven by a related fusion gene, NUP98-JARID1A (NJL), was also sensitive to disulfiram. Thus, the NP23 mouse provides a platform to evaluate compounds that disrupt binding of oncogenic PHD proteins to H3K4me3.


Subject(s)
Binding Sites/drug effects , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Disulfiram/pharmacology , Histones/metabolism , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/pathology , Nuclear Pore Complex Proteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Leukemia, Experimental/drug therapy , Mice , Mice, Transgenic , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
14.
Leuk Res ; 38(1): 95-102, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23958061

ABSTRACT

The myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, dysplasia, and transformation to acute myeloid leukemia (AML). Although it has been suggested that additional mutations lead to progression of MDS to AML, the causative agent(s) for such mutations remains unclear. Oxidative stress is a potential cause, therefore, we evaluated levels of reactive oxygen species (ROS) in NUP98-HOXD13 (NHD13) transgenic mice, a murine model for MDS. Increased levels of ROS were detected in bone marrow nucleated cells (BMNC) that express CD71, a marker for cell proliferation, as well as immature, lineage negative bone marrow nucleated cells from NHD13 mice. In addition to the increase in ROS, increased DNA double strand breaks and activation of a G2/M phase cell cycle checkpoint were noted in NHD13 BMNC. Finally, using an in vivo assay for mutation frequency, we detected an increased mutation frequency in NHD13 BMNC. These results suggest that oxidative stress may contribute to disease progression of MDS to AML through ineffective repair of DNA damage and acquisition of oncogenic mutations.


Subject(s)
Disease Models, Animal , Mutation Rate , Myelodysplastic Syndromes/genetics , Oncogene Proteins, Fusion/genetics , Oxidative Stress , Acute Disease , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Blotting, Western , Bone Marrow Cells/metabolism , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , DNA Breaks, Double-Stranded , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelodysplastic Syndromes/metabolism , Oncogene Proteins, Fusion/metabolism , Reactive Oxygen Species/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Exp Hematol ; 41(6): 518-529.e5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23435313

ABSTRACT

Hoxa9 is expressed in hematopoietic stem and progenitor cells, although this expression is usually diminished as these cells undergo differentiation. In addition, aberrant expression of Hoxa9 is strongly associated with both T cell and myeloid leukemia in mice and humans. Despite this strong association, enforced expression of Hoxa9 in murine bone marrow or thymus has only shown a modest ability to transform cells. To investigate this question, we used Vav regulatory elements to generate a transgenic mouse that targets Hoxa9 overexpression to all hematopoietic tissues. High-level expression of the Hoxa9 transgene in the hematopoietic compartment was associated with embryonic lethality, as no pups from founders that expressed high levels of the transgene were born live. However, offspring of an additional founder line, which expressed lower levels of Hoxa9, developed a precursor T cell lymphoblastic leukemia/lymphoma, accompanied by spontaneous Notch1 mutations. In contrast to most murine models of leukemia associated with Hoxa9 overexpression, the Vav-Hoxa9 mice did not overexpress other Hoxa cluster genes, mir196b (a microRNA that is embedded in the Hoxa locus), Meis1, or Pbx3. The Hoxa9 transgenic mouse reported in this study provides a suitable system for the study of Hoxa9 collaborators that drive myeloid and lymphoid malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, Homeobox , Homeodomain Proteins/physiology , Leukemia, Prolymphocytic, T-Cell/genetics , Animals , Bone Marrow Cells/pathology , Cells, Cultured/pathology , Female , Gene Expression Regulation, Leukemic , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, Lethal , Hematopoietic System/metabolism , Hematopoietic System/pathology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Mutation , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-vav/genetics , Receptor, Notch1/genetics , Recombinant Fusion Proteins/physiology
16.
Blood ; 120(5): 1048-59, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22723554

ABSTRACT

LIN28A and LIN28B, the mammalian homologs of lin-28, are implicated in malignant transformation in part because of their ability to promote degradation of the let-7 family of miRs. In the present study, we show that overexpression of Lin28b in vivo leads to an aggressive peripheral T-cell lymphoma (PTCL) characterized by widespread infiltration of parenchymal organs with malignant CD4(+) cells. Similar to patients with PTCL, Lin28b-transgenic mice show signs of inflammation such as eosinophilia, increased C-reactive protein, release of inflammatory cytokines, and pleural effusion. The PTCLs that develop in Lin28b mice are derived from activated T cells and show decreased let-7 expression, increased Il6 expression, activation of NF-κB, and infiltration of B cells, all resulting in an inflammatory microenvironment. In addition, LIN28B is overexpressed 7.5-fold in PTCL patient samples compared with activated CD4(+) cells. The results of the present study demonstrate for the first time that Lin28b can transform primary cells in vivo, identify a previously unsuspected link between Lin28b and PTCL, and provide a unique animal model for the study of PTCL biology and therapy.


Subject(s)
Cell Differentiation/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Inflammation Mediators/metabolism , Lymphoma, T-Cell, Peripheral/genetics , T-Lymphocytes/physiology , Animals , Cell Differentiation/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/physiology , Female , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Proteins , T-Lymphocytes/metabolism , Transfection , Transgenes/genetics
17.
PLoS One ; 7(5): e36876, 2012.
Article in English | MEDLINE | ID: mdl-22606303

ABSTRACT

Myelodysplastic syndrome (MDS) and aplastic anemia (AA) patients both present with symptoms of bone marrow failure. In many AA patients, these features are thought to result from an oligoclonal expansion of cytotoxic T-cells that destroy haematopoietic stem or progenitor cells. This notion is supported by the observation that AA patients respond to immunosuppressive therapy. A fraction of MDS patients also respond well to immunosuppressive therapy suggesting a similar role for cytotoxic T-cells in the etiology of MDS, however the role of cytotoxic T-cells in MDS remains unclear. Mice that express a NUP98-HOXD13 (NHD13) transgene develop a MDS that closely mimics the human condition in terms of dysplasia, ineffective hematopoiesis, and transformation to acute myeloid leukemia (AML). We followed a cohort of NHD13 mice lacking the Rag1 protein (NHD13/Rag1KO) to determine if the absence of lymphocytes might 1) delay the onset and/or diminish the severity of the MDS, or 2) effect malignant transformation and survival of the NHD13 mice. No difference was seen in the onset or severity of MDS between the NHD13 and NHD13/Rag1KO mice. However, NHD13/Rag1KO mice had decreased survival and showed a trend toward increased incidence of transformation to AML compared to the NHD13 mice, suggesting protection from AML transformation by a modest immuno-surveillance effect. In the absence of functional Tcrb signaling in the NHD13/Rag1KO T-cell tumors, Pak7 was identified as a potential Tcrb surrogate survival signal.


Subject(s)
Immunologic Surveillance , Myelodysplastic Syndromes/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Survival/genetics , Cell Survival/immunology , Disease Models, Animal , Genes, T-Cell Receptor beta , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Leukemia, Experimental/etiology , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/therapy , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , p21-Activated Kinases/immunology
18.
J Immunol ; 183(10): 6227-35, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841179

ABSTRACT

Expression of a NUP98-HOXD13 (NHD13) fusion gene leads to myelodysplastic syndrome in mice. In addition to ineffective hematopoiesis, we observed that NHD13 mice were lymphopenic; the lymphopenia was due to a decrease in both T and B lymphocytes. Although the pro-B cell (B220(+)/CD43(+)) populations from the NHD13 and wild-type mice were similar, the NHD13 mice showed decreased pre-B cells (B220(+)/CD43(-)), indicating impaired differentiation at the pro-B to pre-B stage. Thymi from NHD13 mice were smaller and overexpressed Hoxa cluster genes, including Hoxa7, Hoxa9, and Hoxa10. In addition, the NHD13 thymi contained fewer thymocytes, with an increased percentage of CD4(-)/CD8(-) (double-negative (DN)) cells and a decreased percentage of CD4(+)/CD8(+) (double-positive) cells; the DN1/DN2 population was increased and the DN3/DN4 population was decreased, suggesting a partial block at the DN2 to DN3 transition. To determine clonality of the thymocytes, we used degenerate RT-PCR to identify clonal Tcrb gene rearrangements. Five of six NHD13 thymi showed an unusual Tcrb gene rearrangement pattern with common, clonal DJ rearrangements, but distinct V-D junctions, suggesting a marked clonal expansion of thymocytes that had undergone a DJ rearrangement, but not completed a VDJ rearrangement. Taken together, these findings demonstrate that expression of the NHD13 transgene inhibits lymphoid as well as myeloid and erythroid differentiation, results in overexpression of Hoxa cluster genes, and leads to a precursor T cell lymphoblastic leukemia/lymphoma.


Subject(s)
B-Lymphocytes/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Homeodomain Proteins/metabolism , Myelodysplastic Syndromes/immunology , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression/genetics , Gene Expression/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/immunology , Oncogene Proteins, Fusion/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , Transcription Factors/genetics , Transcription Factors/immunology
19.
Blood ; 114(12): 2401-10, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19633199

ABSTRACT

Members of the tristetraprolin family of tandem CCCH finger proteins can bind to AU-rich elements in the 3'-untranslated region of mRNAs, leading to their deadenylation and subsequent degradation. Partial deficiency of 1 of the 4 mouse tristetraprolin family members, Zfp36l2, resulted in complete female infertility because of early embryo death. We have now generated mice completely deficient in the ZFP36L2 protein. Homozygous Zfp36l2 knockout (KO) mice died within approximately 2 weeks of birth, apparently from intestinal or other hemorrhage. Analysis of peripheral blood from KO mice showed a decrease in red and white cells, hemoglobin, hematocrit, and platelets. Yolk sacs from embryonic day 11.5 (E11.5) Zfp36l2 KO mice and fetal livers from E14.5 KO mice gave rise to markedly reduced numbers of definitive multilineage and lineage-committed hematopoietic progenitors. Competitive reconstitution experiments demonstrated that Zfp36l2 KO fetal liver hematopoietic stem cells were unable to adequately reconstitute the hematopoietic system of lethally irradiated recipients. These data establish Zfp36l2 as a critical modulator of definitive hematopoiesis and suggest a novel regulatory pathway involving control of mRNA stability in the life cycle of hematopoietic stem and progenitor cells.


Subject(s)
Embryo, Mammalian/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , Pancytopenia/genetics , RNA Stability/genetics , RNA-Binding Proteins/genetics , Tristetraprolin/physiology , Animals , Blotting, Northern , Blotting, Southern , Bone Marrow/metabolism , Embryo, Mammalian/cytology , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunoenzyme Techniques , In Situ Hybridization , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Pancytopenia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/metabolism , Tissue Distribution
20.
Proc Natl Acad Sci U S A ; 105(37): 14088-93, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18768819

ABSTRACT

The myelodysplastic syndromes (MDS) comprise a group of premalignant hematologic disorders characterized by ineffective hematopoiesis, dysplasia, and transformation to acute myeloid leukemia (AML). Although it is well established that many malignancies can be transplanted, there is little evidence to demonstrate that a premalignant disease entity, such as MDS or colonic polyps, can be transplanted and subsequently undergo malignant transformation in vivo. Using mice that express a NUP98-HOXD13 (NHD13) transgene in hematopoietic tissues, we show that a MDS can be transplanted to WT recipients. Recipients of the MDS bone marrow displayed all of the critical features of MDS, including peripheral blood cytopenias, dysplasia, and transformation to AML. Even when transplanted with a 10-fold excess of WT cells, the NHD13 cells outcompeted the WT cells over a 38-week period. Limiting-dilution experiments demonstrated that the frequency of the cell that could transmit the disease was approximately 1/6,000-1/16,000 and that the MDS was also transferable to secondary recipients as a premalignant condition. Transformation to AML in primary transplant recipients was generally delayed (46-49 weeks after transplant); however, 6 of 10 secondary transplant recipients developed AML. These findings demonstrate that MDS originates in a transplantable, premalignant, long-term repopulating, MDS-initiating cell.


Subject(s)
Cell Lineage , Hematopoietic Stem Cell Transplantation , Hematopoietic System/cytology , Myelodysplastic Syndromes/pathology , Animals , Bone Marrow/immunology , Cell Separation , Leukemia/pathology , Mice , Mice, Inbred C57BL , Myelodysplastic Syndromes/surgery , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...