Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Blood ; 142(20): 1740-1751, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37738562

ABSTRACT

Histiocytoses are inflammatory myeloid neoplasms often driven by somatic activating mutations in mitogen-activated protein kinase (MAPK) cascade genes. H syndrome is an inflammatory genetic disorder caused by germ line loss-of-function mutations in SLC29A3, encoding the lysosomal equilibrative nucleoside transporter 3 (ENT3). Patients with H syndrome are predisposed to develop histiocytosis, yet the mechanism is unclear. Here, through phenotypic, molecular, and functional analysis of primary cells from a cohort of patients with H syndrome, we reveal the molecular pathway leading to histiocytosis and inflammation in this genetic disorder. We show that loss of function of ENT3 activates nucleoside-sensing toll-like receptors (TLR) and downstream MAPK signaling, inducing cytokine secretion and inflammation. Importantly, MEK inhibitor therapy led to resolution of histiocytosis and inflammation in a patient with H syndrome. These results demonstrate a yet-unrecognized link between a defect in a lysosomal transporter and pathological activation of MAPK signaling, establishing a novel pathway leading to histiocytosis and inflammation.


Subject(s)
Histiocytosis , Mitogen-Activated Protein Kinases , Humans , Histiocytosis/genetics , Histiocytosis/pathology , Mutation , Toll-Like Receptors , Inflammation/genetics , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism
2.
Nat Commun ; 14(1): 5871, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37735473

ABSTRACT

The ERG (ETS-related gene) transcription factor is linked to various types of cancer, including leukemia. However, the specific ERG domains and co-factors contributing to leukemogenesis are poorly understood. Drug targeting a transcription factor such as ERG is challenging. Our study reveals the critical role of a conserved amino acid, proline, at position 199, located at the 3' end of the PNT (pointed) domain, in ERG's ability to induce leukemia. P199 is necessary for ERG to promote self-renewal, prevent myeloid differentiation in hematopoietic progenitor cells, and initiate leukemia in mouse models. Here we show that P199 facilitates ERG's interaction with the NCoR-HDAC3 co-repressor complex. Inhibiting HDAC3 reduces the growth of ERG-dependent leukemic and prostate cancer cells, indicating that the interaction between ERG and the NCoR-HDAC3 co-repressor complex is crucial for its oncogenic activity. Thus, targeting this interaction may offer a potential therapeutic intervention.


Subject(s)
Leukemia , Transcription Factors , Animals , Male , Mice , Co-Repressor Proteins , Gene Expression Regulation , Genes, Regulator
3.
Haematologica ; 108(9): 2316-2330, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36475518

ABSTRACT

Mono-allelic germline disruptions of the transcription factor GATA2 result in a propensity for developing myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), affecting more than 85% of carriers. How a partial loss of GATA2 functionality enables leukemic transformation years later is unclear. This question has remained unsolved mainly due to the lack of informative models, as Gata2 heterozygote mice do not develop hematologic malignancies. Here we show that two different germline Gata2 mutations (TgErg/Gata2het and TgErg/Gata2L359V) accelerate AML in mice expressing the human hematopoietic stem cell regulator ERG. Analysis of Erg/Gata2het fetal liver and bone marrow-derived hematopoietic cells revealed a distinct pre-leukemic phenotype. This was characterized by enhanced transition from stem to progenitor state, increased proliferation, and a striking mitochondrial phenotype, consisting of highly expressed oxidative-phosphorylation-related gene sets, elevated oxygen consumption rates, and notably, markedly distorted mitochondrial morphology. Importantly, the same mitochondrial gene-expression signature was observed in human AML harboring GATA2 aberrations. Similar to the observations in mice, non-leukemic bone marrows from children with germline GATA2 mutation demonstrated marked mitochondrial abnormalities. Thus, we observed the tumor suppressive effects of GATA2 in two germline Gata2 genetic mouse models. As oncogenic mutations often accumulate with age, GATA2 deficiency-mediated priming of hematopoietic cells for oncogenic transformation may explain the earlier occurrence of MDS/AML in patients with GATA2 germline mutation. The mitochondrial phenotype is a potential therapeutic opportunity for the prevention of leukemic transformation in these patients.


Subject(s)
GATA2 Deficiency , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Child , Humans , Mice , Animals , GATA2 Deficiency/genetics , Myelodysplastic Syndromes/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Bone Marrow/pathology , Hematopoietic Stem Cells/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism
4.
Nat Commun ; 13(1): 659, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115489

ABSTRACT

Kinase signaling fuels growth of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Yet its role in leukemia initiation is unclear and has not been shown in primary human hematopoietic cells. We previously described activating mutations in interleukin-7 receptor alpha (IL7RA) in poor-prognosis "ph-like" BCP-ALL. Here we show that expression of activated mutant IL7RA in human CD34+ hematopoietic stem and progenitor cells induces a preleukemic state in transplanted immunodeficient NOD/LtSz-scid IL2Rγnull mice, characterized by persistence of self-renewing Pro-B cells with non-productive V(D)J gene rearrangements. Preleukemic CD34+CD10highCD19+ cells evolve into BCP-ALL with spontaneously acquired Cyclin Dependent Kinase Inhibitor 2 A (CDKN2A) deletions, as commonly observed in primary human BCP-ALL. CRISPR mediated gene silencing of CDKN2A in primary human CD34+ cells transduced with activated IL7RA results in robust development of BCP-ALLs in-vivo. Thus, we demonstrate that constitutive activation of IL7RA can initiate preleukemia in primary human hematopoietic progenitors and cooperates with CDKN2A silencing in progression into BCP-ALL.


Subject(s)
Interleukin-7 Receptor alpha Subunit/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cells, B-Lymphoid/immunology , Signal Transduction/immunology , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Antigens, CD34/metabolism , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/immunology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression/immunology , Humans , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , RNA-Seq/methods , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Receptors, Cytokine/metabolism , Signal Transduction/genetics , Single-Cell Analysis/methods , Transplantation, Heterologous
5.
Blood ; 139(3): 399-412, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34624096

ABSTRACT

Mixed-phenotype acute leukemia is a rare subtype of leukemia in which both myeloid and lymphoid markers are co-expressed on the same malignant cells. The pathogenesis is largely unknown, and the treatment is challenging. We previously reported the specific association of the recurrent t(8;12)(q13;p13) chromosomal translocation that creates the ETV6-NCOA2 fusion with T/myeloid leukemias. Here we report that ETV6-NCOA2 initiates T/myeloid leukemia in preclinical models; ectopic expression of ETV6-NCOA2 in mouse bone marrow hematopoietic progenitors induced T/myeloid lymphoma accompanied by spontaneous Notch1-activating mutations. Similarly, cotransduction of human cord blood CD34+ progenitors with ETV6-NCOA2 and a nontransforming NOTCH1 mutant induced T/myeloid leukemia in immunodeficient mice; the immunophenotype and gene expression pattern were similar to those of patient-derived ETV6-NCOA2 leukemias. Mechanistically, we show that ETV6-NCOA2 forms a transcriptional complex with ETV6 and the histone acetyltransferase p300, leading to derepression of ETV6 target genes. The expression of ETV6-NCOA2 in human and mouse nonthymic hematopoietic progenitor cells induces transcriptional dysregulation, which activates a lymphoid program while failing to repress the expression of myeloid genes such as CSF1 and MEF2C. The ETV6-NCOA2 induced arrest at an early immature T-cell developmental stage. The additional acquisition of activating NOTCH1 mutations transforms the early immature ETV6-NCOA2 cells into T/myeloid leukemias. Here, we describe the first preclinical model to depict the initiation of T/myeloid leukemia by a specific somatic genetic aberration.


Subject(s)
Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid/genetics , Nuclear Receptor Coactivator 2/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Female , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , ETS Translocation Variant 6 Protein
6.
Pediatr Blood Cancer ; 68(10): e29138, 2021 10.
Article in English | MEDLINE | ID: mdl-34019335

ABSTRACT

BACKGROUND: Inflammatory manifestations (IM) are well described in adult patients with myelodysplastic syndrome (MDS), but the presentation is highly variable and no standardized treatment exists. This phenomenon is rarely reported in children. As more pediatric patients are hematopoietic stem cell transplantation (HSCT) candidates, the role of anti-inflammatory treatment in relation to HSCT should be defined. PROCEDURE: Here, we report a series of five children from a tertiary center. We describe the clinical presentation, molecular findings, and treatment options. RESULTS: All patients presented with advanced MDS with blast percentages ranging 10-30%, all had severe IM. One patient had MDS secondary to severe congenital neutropenia, the other four patients had presumably primary MDS. All four were found to harbor a PTPN11 gene driver mutation, which is found in 35% of cases of juvenile myelomonocytic leukemia (JMML). The mutation was present in the myeloid lineage but not in T lymphocytes. Three had symptoms of Behcet's-like disease with trisomy 8 in their bone marrow. All patients were treated with anti-inflammatory medications (mainly systemic steroids) in an attempt to bring them to allogeneic HSCT in a better clinical condition. All demonstrated clinical improvement as well as regression in their MDS status post anti-inflammatory treatment. All have recovered from both MDS and their inflammatory symptoms post HSCT. CONCLUSION: Primary pediatric MDS with IM is driven in some cases by PTPN11 mutations, and might be on the clinical spectrum of JMML. Anti-inflammatory treatment may reverse MDS progression and improve the outcome of subsequent HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Juvenile , Myelodysplastic Syndromes , Child , Humans , Leukemia, Myelomonocytic, Juvenile/diagnosis , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/therapy , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/therapy , Treatment Outcome , Trisomy
7.
Nat Cancer ; 1(10): 998-1009, 2020 10.
Article in English | MEDLINE | ID: mdl-33479702

ABSTRACT

Metabolic reprogramming is a key hallmark of cancer, but less is known about metabolic plasticity of the same tumor at different sites. Here, we investigated the metabolic adaptation of leukemia in two different microenvironments, the bone marrow and the central nervous system (CNS). We identified a metabolic signature of fatty-acid synthesis in CNS leukemia, highlighting Stearoyl-CoA desaturase (SCD1) as a key player. In vivo SCD1 overexpression increases CNS disease, whilst genetic or pharmacological inhibition of SCD1 decreases CNS load. Overall, we demonstrated that leukemic cells dynamically rewire metabolic pathways to suit local conditions and that targeting these adaptations can be exploited therapeutically.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Stearoyl-CoA Desaturase , Central Nervous System/metabolism , Humans , Lipogenesis , Stearoyl-CoA Desaturase/genetics , Tumor Microenvironment
8.
Proc Natl Acad Sci U S A ; 114(20): E4030-E4039, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28461505

ABSTRACT

Children with Down syndrome (DS) are prone to development of high-risk B-cell precursor ALL (DS-ALL), which differs genetically from most sporadic pediatric ALLs. Increased expression of cytokine receptor-like factor 2 (CRLF2), the receptor to thymic stromal lymphopoietin (TSLP), characterizes about half of DS-ALLs and also a subgroup of sporadic "Philadelphia-like" ALLs. To understand the pathogenesis of relapsed DS-ALL, we performed integrative genomic analysis of 25 matched diagnosis-remission and -relapse DS-ALLs. We found that the CRLF2 rearrangements are early events during DS-ALL evolution and generally stable between diagnoses and relapse. Secondary activating signaling events in the JAK-STAT/RAS pathway were ubiquitous but highly redundant between diagnosis and relapse, suggesting that signaling is essential but that no specific mutations are "relapse driving." We further found that activated JAK2 may be naturally suppressed in 25% of CRLF2pos DS-ALLs by loss-of-function aberrations in USP9X, a deubiquitinase previously shown to stabilize the activated phosphorylated JAK2. Interrogation of large ALL genomic databases extended our findings up to 25% of CRLF2pos, Philadelphia-like ALLs. Pharmacological or genetic inhibition of USP9X, as well as treatment with low-dose ruxolitinib, enhanced the survival of pre-B ALL cells overexpressing mutated JAK2. Thus, somehow counterintuitive, we found that suppression of JAK-STAT "hypersignaling" may be beneficial to leukemic B-cell precursors. This finding and the reduction of JAK mutated clones at relapse suggest that the therapeutic effect of JAK specific inhibitors may be limited. Rather, combined signaling inhibitors or direct targeting of the TSLP receptor may be a useful therapeutic strategy for DS-ALL.


Subject(s)
Down Syndrome/complications , Janus Kinases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , STAT Transcription Factors/metabolism , Adolescent , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Male , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Cytokine/genetics , Recurrence , Signal Transduction , Ubiquitin Thiolesterase/genetics , Young Adult
9.
Cell Stem Cell ; 19(2): 177-191, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27292188

ABSTRACT

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1's effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Subject(s)
Adenosine Deaminase/metabolism , Cell Self Renewal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Animals , Base Sequence , Cell Self Renewal/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Janus Kinase 2/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA Editing/genetics , RNA-Binding Proteins/genetics , Signal Transduction/genetics
10.
J Transl Med ; 13: 98, 2015 Mar 21.
Article in English | MEDLINE | ID: mdl-25889765

ABSTRACT

BACKGROUND: Dormant leukemia stem cells (LSC) promote therapeutic resistance and leukemic progression as a result of unbridled activation of stem cell gene expression programs. Thus, we hypothesized that 1) deregulation of the hedgehog (Hh) stem cell self-renewal and cell cycle regulatory pathway would promote dormant human LSC generation and 2) that PF-04449913, a clinical antagonist of the GLI2 transcriptional activator, smoothened (SMO), would enhance dormant human LSC eradication. METHODS: To test these postulates, whole transcriptome RNA sequencing (RNA-seq), microarray, qRT-PCR, stromal co-culture, confocal fluorescence microscopic, nanoproteomic, serial transplantation and cell cycle analyses were performed on FACS purified normal, chronic phase (CP) chronic myeloid leukemia (CML), blast crisis (BC) phase CML progenitors with or without PF-04449913 treatment. RESULTS: Notably, RNA-seq analyses revealed that Hh pathway and cell cycle regulatory gene overexpression correlated with leukemic progression. While lentivirally enforced GLI2 expression enhanced leukemic progenitor dormancy in stromal co-cultures, this was not observed with a mutant GLI2 lacking a transactivation domain, suggesting that GLI2 expression prevented cell cycle transit. Selective SMO inhibition with PF-04449913 in humanized stromal co-cultures and LSC xenografts reduced downstream GLI2 protein and cell cycle regulatory gene expression. Moreover, SMO inhibition enhanced cell cycle transit and sensitized BC LSC to tyrosine kinase inhibition in vivo at doses that spare normal HSC. CONCLUSION: In summary, while GLI2, forms part of a core HH pathway transcriptional regulatory network that promotes human myeloid leukemic progression and dormant LSC generation, selective inhibition with PF-04449913 reduces the dormant LSC burden thereby providing a strong rationale for clinical trials predicated on SMO inhibition in combination with TKIs or chemotherapeutic agents with the ultimate aim of obviating leukemic therapeutic resistance, persistence and progression.


Subject(s)
Kruppel-Like Transcription Factors/antagonists & inhibitors , Leukemia/pathology , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Animals , Base Sequence , Coculture Techniques , DNA Primers , Fetal Blood/cytology , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Zinc Finger Protein Gli2
11.
Blood ; 125(8): 1292-301, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25533034

ABSTRACT

Children with Down syndrome (DS) are at increased risk for acute myeloid leukemias (ML-DS) characterized by mixed megakaryocytic and erythroid phenotype and by acquired mutations in the GATA1 gene resulting in a short GATA1s isoform. The chromosome 21 microRNA (miR)-125b cluster has been previously shown to cooperate with GATA1s in transformation of fetal hematopoietic progenitors. In this study, we report that the expression of miR-486-5p is increased in ML-DS compared with non-DS acute megakaryocytic leukemias (AMKLs). miR-486-5p is regulated by GATA1 and GATA1s that bind to the promoter of its host gene ANK1. miR-486-5p is highly expressed in mouse erythroid precursors and knockdown (KD) in ML-DS cells reduced their erythroid phenotype. Ectopic expression and KD of miR-486-5p in primary fetal liver hematopoietic progenitors demonstrated that miR-486-5p cooperates with Gata1s to enhance their self renewal. Consistent with its activation of AKT, overexpression and KD experiments showed its importance for growth and survival of human leukemic cells. Thus, miR-486-5p cooperates with GATA1s in supporting the growth and survival, and the aberrant erythroid phenotype of the megakaryocytic leukemias of DS.


Subject(s)
Down Syndrome/genetics , Erythropoiesis/genetics , Leukemia, Myeloid, Acute/genetics , MicroRNAs/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Child, Preschool , Down Syndrome/complications , Down Syndrome/physiopathology , Erythroid Cells/metabolism , HEK293 Cells , Humans , K562 Cells , Leukemia, Myeloid, Acute/pathology , Megakaryocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Tumor Cells, Cultured
13.
Cancer Lett ; 352(1): 15-20, 2014 Sep 28.
Article in English | MEDLINE | ID: mdl-24569093

ABSTRACT

The successful therapy of childhood leukemia has been characterized by careful personalized adaptation of therapy by risk stratification. Yet almost all drugs are relatively non-specific. To achieve greater precision in therapy, druggable targets and specific targeting drugs are necessary. Here we review the recent discoveries of cytokine receptors and their signaling components in high risk leukemias and the potential approaches to target them.


Subject(s)
Precision Medicine/trends , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Cytokine/genetics , Genetic Predisposition to Disease , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction
14.
Cell Mol Life Sci ; 71(3): 365-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23625073

ABSTRACT

Cancer is often caused by deregulation of normal developmental processes. Here, we review recent research on the aberrant activation of two hematopoietic cytokine receptors in acute lymphoid leukemias. Somatic events in the genes for thymic stromal lymphopoietin and Interleukin 7 receptors as well as in their downstream JAK kinases result in constitutive ligand-independent activation of survival and proliferation in B and T lymphoid precursors. Drugs targeting these receptors or the signaling pathways might provide effective therapies of these leukemias.


Subject(s)
Cytokines/immunology , Interleukin-7/immunology , Models, Biological , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Cytokine/metabolism , Receptors, Interleukin-7/metabolism , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing , Humans , Intracellular Signaling Peptides and Proteins , Janus Kinases/genetics , Janus Kinases/metabolism , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, T-Lymphoid/immunology , Proteins/genetics , Proteins/metabolism , Receptors, Cytokine/genetics , Receptors, Interleukin-7/genetics , Signal Transduction/genetics , Thymic Stromal Lymphopoietin
15.
Cell Stem Cell ; 12(3): 316-28, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23333150

ABSTRACT

Leukemia stem cells (LSCs) play a pivotal role in the resistance of chronic myeloid leukemia (CML) to tyrosine kinase inhibitors (TKIs) and its progression to blast crisis (BC), in part, through the alternative splicing of self-renewal and survival genes. To elucidate splice-isoform regulators of human BC LSC maintenance, we performed whole-transcriptome RNA sequencing, splice-isoform-specific quantitative RT-PCR (qRT-PCR), nanoproteomics, stromal coculture, and BC LSC xenotransplantation analyses. Cumulatively, these studies show that the alternative splicing of multiple prosurvival BCL2 family genes promotes malignant transformation of myeloid progenitors into BC LSCS that are quiescent in the marrow niche and that contribute to therapeutic resistance. Notably, sabutoclax, a pan-BCL2 inhibitor, renders marrow-niche-resident BC LSCs sensitive to TKIs at doses that spare normal progenitors. These findings underscore the importance of alternative BCL2 family splice-isoform expression in BC LSC maintenance and suggest that the combinatorial inhibition of prosurvival BCL2 family proteins and BCR-ABL may eliminate dormant LSCs and obviate resistance.


Subject(s)
Leukemia/pathology , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Blast Crisis/metabolism , Blast Crisis/pathology , Gossypol/analogs & derivatives , Gossypol/pharmacology , Humans , Leukemia/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 7(6): e39725, 2012.
Article in English | MEDLINE | ID: mdl-22768113

ABSTRACT

BACKGROUND: Leukemia initiating cells (LIC) contribute to therapeutic resistance through acquisition of mutations in signaling pathways, such as NOTCH1, that promote self-renewal and survival within supportive niches. Activating mutations in NOTCH1 occur commonly in T cell acute lymphoblastic leukemia (T-ALL) and have been implicated in therapeutic resistance. However, the cell type and context specific consequences of NOTCH1 activation, its role in human LIC regeneration, and sensitivity to NOTCH1 inhibition in hematopoietic microenvironments had not been elucidated. METHODOLOGY AND PRINCIPAL FINDINGS: We established humanized bioluminescent T-ALL LIC mouse models transplanted with pediatric T-ALL samples that were sequenced for NOTCH1 and other common T-ALL mutations. In this study, CD34(+) cells from NOTCH1(Mutated) T-ALL samples had higher leukemic engraftment and serial transplantation capacity than NOTCH1(Wild-type) CD34(+) cells in hematopoietic niches, suggesting that self-renewing LIC were enriched within the NOTCH1(Mutated) CD34(+) fraction. Humanized NOTCH1 monoclonal antibody treatment reduced LIC survival and self-renewal in NOTCH1(Mutated) T-ALL LIC-engrafted mice and resulted in depletion of CD34(+)CD2(+)CD7(+) cells that harbor serial transplantation capacity. CONCLUSIONS: These results reveal a functional hierarchy within the LIC population based on NOTCH1 activation, which renders LIC susceptible to targeted NOTCH1 inhibition and highlights the utility of NOTCH1 antibody targeting as a key component of malignant stem cell eradication strategies.


Subject(s)
Neoplastic Stem Cells/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/metabolism , Regeneration , Signal Transduction , Stem Cell Niche , Adolescent , Animals , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Child , Child, Preschool , Humans , Mice , Mutation/genetics , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/transplantation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Young Adult
17.
J Biomol Screen ; 15(6): 663-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20547533

ABSTRACT

A number of diabetogenic stimuli interact to influence insulin promoter activity, making it an attractive target for both mechanistic studies and therapeutic interventions. High-throughput screening (HTS) for insulin promoter modulators has the potential to reveal novel inputs into the control of that central element of the pancreatic beta-cell. A cell line from human islets in which the expression of insulin and other beta-cell-restricted genes are modulated by an inducible form of the bHLH transcription factor E47 was developed. This cell line, T6PNE, was adapted for HTS by transduction with a vector expressing green fluorescent protein under the control of the human insulin promoter. The resulting cell line was screened against a library of known drugs for those that increase insulin promoter activity. Members of the phenothiazine class of neuroleptics increased insulin gene expression upon short-term exposure. Chronic treatment, however, resulted in suppression of insulin promoter activity, consistent with the effect of phenothiazines observed clinically to induce diabetes in chronically treated patients. In addition to providing insights into previously unrecognized targets and mechanisms of action of phenothiazines, the novel cell line described here provides a broadly applicable platform for mining new molecular drug targets and central regulators of beta-cell differentiated function.


Subject(s)
Antipsychotic Agents/pharmacology , High-Throughput Screening Assays/methods , Insulin/genetics , Phenothiazines/pharmacology , Promoter Regions, Genetic , Signal Transduction/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Gene Expression Regulation/drug effects , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Structure-Activity Relationship , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 1 Protein
18.
Transplantation ; 87(7): 983-91, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19352116

ABSTRACT

BACKGROUND: Islet transplantation is limited by the need for chronic immunosuppression and the paucity of donor tissue. As new sources of human beta-cells are developed (e.g., stem cell-derived tissue), transplanting them in a durable device could obviate the need for immunosuppression, while also protecting the patient from any risk of tumorigenicity. Here, we studied (1) the survival and function of encapsulated human beta-cells and their progenitors and (2) the engraftment of encapsulated murine beta-cells in allo- and autoimmune settings. METHODS: Human islets and human fetal pancreatic islet-like cell clusters were encapsulated in polytetrafluorethylene devices (TheraCyte) and transplanted into immunodeficient mice. Graft survival and function was measured by immunohistochemistry, circulating human C-peptide levels, and blood glucose levels. Bioluminescent imaging was used to monitor encapsulated neonatal murine islets. RESULTS: Encapsulated human islet-like cell clusters survived, replicated, and acquired a level of glucose responsive insulin secretion sufficient to ameliorate hyperglycemia in diabetic mice. Bioluminescent imaging of encapsulated murine neonatal islets revealed a dynamic process of cell death followed by regrowth, resulting in robust long-term allograft survival. Further, in the non-obese diabetic (NOD) mouse model of type I diabetes, encapsulated primary beta-cells ameliorated diabetes without stimulating a detectable T-cell response. CONCLUSIONS: We demonstrate for the first time that human beta-cells function is compatible with encapsulation in a durable, immunoprotective device. Moreover, our study suggests that encapsulation of beta-cells before terminal differentiation will be a successful approach for new cell-based therapies for diabetes, such as those derived from stem cells.


Subject(s)
Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/transplantation , Insulin/metabolism , Animals , Cell Survival , Diabetes Mellitus, Experimental/surgery , Friend murine leukemia virus/genetics , Genes, Reporter , Glucose/metabolism , Humans , Hyperglycemia/prevention & control , Insulin Secretion , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Luciferases/genetics , Mice , Mice, SCID , Mice, Transgenic , Transplantation, Heterologous
19.
Proc Natl Acad Sci U S A ; 106(10): 3925-9, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19237556

ABSTRACT

Recent evidence suggests that a rare population of self-renewing cancer stem cells (CSC) is responsible for cancer progression and therapeutic resistance. Chronic myeloid leukemia (CML) represents an important paradigm for understanding the genetic and epigenetic events involved in CSC production. CML progresses from a chronic phase (CP) in hematopoietic stem cells (HSC) that harbor the BCR-ABL translocation, to blast crisis (BC), characterized by aberrant activation of beta-catenin within granulocyte-macrophage progenitors (GMP). A major barrier to predicting and inhibiting blast crisis transformation has been the identification of mechanisms driving beta-catenin activation. Here we show that BC CML myeloid progenitors, in particular GMP, serially transplant leukemia in immunocompromised mice and thus are enriched for leukemia stem cells (LSC). Notably, cDNA sequencing of Wnt/beta-catenin pathway regulatory genes, including adenomatous polyposis coli, GSK3beta, axin 1, beta-catenin, lymphoid enhancer factor-1, cyclin D1, and c-myc, revealed a novel in-frame splice deletion of the GSK3beta kinase domain in the GMP of BC samples that was not detectable by sequencing in blasts or normal progenitors. Moreover, BC CML progenitors with misspliced GSK3beta have enhanced beta-catenin expression as well as serial engraftment potential while reintroduction of full-length GSK3beta reduces both in vitro replating and leukemic engraftment. We propose that CP CML is initiated by BCR-ABL expression in an HSC clone but that progression to BC may include missplicing of GSK3beta in GMP LSC, enabling unphosphorylated beta-catenin to participate in LSC self-renewal. Missplicing of GSK3beta represents a unique mechanism for the emergence of BC CML LSC and might provide a novel diagnostic and therapeutic target.


Subject(s)
Alternative Splicing/genetics , Glycogen Synthase Kinase 3/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/enzymology , Animals , Base Sequence , Blast Crisis/enzymology , Blast Crisis/pathology , Glycogen Synthase Kinase 3 beta , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Molecular Sequence Data , Stem Cell Transplantation
20.
Cancer Cell ; 13(4): 321-30, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18394555

ABSTRACT

Polycythemia Vera (PV) is a myeloproliferative disorder (MPD) that is commonly characterized by mutant JAK2 (JAK2V617F) signaling, erythrocyte overproduction, and a propensity for thrombosis, progression to myelofibrosis, or acute leukemia. In this study, JAK2V617F expression by human hematopoietic progenitors promoted erythroid colony formation and erythroid engraftment in a bioluminescent xenogeneic immunocompromised mouse transplantation model. A selective JAK2 inhibitor, TG101348 (300 nM), significantly inhibited JAK2V617F+ progenitor-derived colony formation as well as engraftment (120 mg/kg) in xenogeneic transplantation studies. TG101348 treatment decreased GATA-1 expression, which is associated with erythroid-skewing of JAK2V617F+ progenitor differentiation, and inhibited STAT5 as well as GATA S310 phosphorylation. Thus, TG101348 may be an effective inhibitor of JAK2V617F+ MPDs in clinical trials.


Subject(s)
Cell Differentiation/drug effects , Erythroid Precursor Cells/enzymology , Erythroid Precursor Cells/pathology , Janus Kinase 2/antagonists & inhibitors , Polycythemia Vera/enzymology , Polycythemia Vera/pathology , Protein Kinase Inhibitors/pharmacology , Adult , Aged , Amino Acid Substitution , Animals , Base Sequence , Erythroid Precursor Cells/drug effects , Female , Humans , Janus Kinase 2/genetics , Male , Mice , Middle Aged , Molecular Sequence Data , Phenylalanine/genetics , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Stem Cell Transplantation , Valine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...