Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Eur J Haematol ; 112(5): 731-742, 2024 May.
Article in English | MEDLINE | ID: mdl-38192186

ABSTRACT

BACKGROUND: B-cell acute lymphoblastic leukemia (B-ALL) is classified into subgroups based on known driver oncogenes and molecular lesions, including translocations and recurrent mutations. However, the current diagnostic tests do not identify subtypes or oncogenic lesions for all B-ALL samples, creating a heterogeneous B-ALL group of unknown subtypes. METHODS: We sorted primary adult B-ALL cells and performed transcriptome analysis by bulk RNA sequencing (RNA-seq). RESULTS: Transcriptomic analysis of an adult B-ALL cohort allowed the classification of four patient samples with subtypes that were not previously revealed by standard gene panels. The leukemia of two patients were of the DUX4 subtype and two were CRLF2+ Ph-like B-ALL. Furthermore, single nucleotide variant analysis detected the oncogenic NRAS-G12D, KRAS-G12D, and KRAS-G13D mutations in three of the patient samples, presenting targetable mutations. Additional oncogenic variants and gene fusions were uncovered, as well as multiple variants in the PDE4DIP gene across five of the patient samples. CONCLUSION: We demonstrate that RNA-seq is an effective tool for precision medicine in B-ALL by providing comprehensive molecular profiling of leukemia cells, identifying subtype and oncogenic lesions, and stratifying patients for appropriate therapy.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Humans , Cell Lineage , Proto-Oncogene Proteins p21(ras)/genetics , Transcriptome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Gene Expression Profiling , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Gene Fusion
2.
Sci Immunol ; 8(88): eabq3109, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37889983

ABSTRACT

Mutations in the gene encoding the zinc-finger transcription factor Ikaros (IKZF1) are found in patients with immunodeficiency, leukemia, and autoimmunity. Although Ikaros has a well-established function in modulating gene expression programs important for hematopoietic development, its role in other cell types is less well defined. Here, we uncover functions for Ikaros in thymic epithelial lineage development in mice and show that Ikzf1 expression in medullary thymic epithelial cells (mTECs) is required for both autoimmune regulator-positive (Aire+) mTEC development and tissue-specific antigen (TSA) gene expression. Accordingly, TEC-specific deletion of Ikzf1 in mice results in a profound decrease in Aire+ mTECs, a global loss of TSA gene expression, and the development of autoimmunity. Moreover, Ikaros shapes thymic mimetic cell diversity, and its deletion results in a marked expansion of thymic tuft cells and muscle-like mTECs and a loss of other Aire-dependent mimetic populations. Single-cell analysis reveals that Ikaros modulates core transcriptional programs in TECs that correlate with the observed cellular changes. Our findings highlight a previously undescribed role for Ikaros in regulating epithelial lineage development and function and suggest that failed thymic central tolerance could contribute to the autoimmunity seen in humans with IKZF1 mutations.


Subject(s)
Central Tolerance , Thymus Gland , Humans , Mice , Animals , Cell Differentiation , Transcription Factors , Gene Expression Regulation
3.
bioRxiv ; 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36993619

ABSTRACT

In most cell types, nuclear ß-catenin functions as prominent oncogenic driver and pairs with TCF7-family factors for transcriptional activation of MYC. Surprisingly, B-lymphoid malignancies not only lacked expression and activating lesions of ß-catenin but critically depended on GSK3ß for effective ß-catenin degradation. Our interactome studies in B-lymphoid tumors revealed that ß-catenin formed repressive complexes with lymphoid-specific Ikaros factors at the expense of TCF7. Instead of MYC-activation, ß-catenin was essential to enable Ikaros-mediated recruitment of nucleosome remodeling and deacetylation (NuRD) complexes for transcriptional repression of MYC. To leverage this previously unrecognized vulnerability of B-cell-specific repressive ß-catenin-Ikaros-complexes in refractory B-cell malignancies, we examined GSK3ß small molecule inhibitors to subvert ß-catenin degradation. Clinically approved GSK3ß-inhibitors that achieved favorable safety prof les at micromolar concentrations in clinical trials for neurological disorders and solid tumors were effective at low nanomolar concentrations in B-cell malignancies, induced massive accumulation of ß-catenin, repression of MYC and acute cell death. Preclinical in vivo treatment experiments in patient-derived xenografts validated small molecule GSK3ß-inhibitors for targeted engagement of lymphoid-specific ß-catenin-Ikaros complexes as a novel strategy to overcome conventional mechanisms of drug-resistance in refractory malignancies. HIGHLIGHTS: Unlike other cell lineages, B-cells express nuclear ß-catenin protein at low baseline levels and depend on GSK3ß for its degradation.In B-cells, ß-catenin forms unique complexes with lymphoid-specific Ikaros factors and is required for Ikaros-mediated tumor suppression and assembly of repressive NuRD complexes. CRISPR-based knockin mutation of a single Ikaros-binding motif in a lymphoid MYC superenhancer region reversed ß-catenin-dependent Myc repression and induction of cell death. The discovery of GSK3ß-dependent degradation of ß-catenin as unique B-lymphoid vulnerability provides a rationale to repurpose clinically approved GSK3ß-inhibitors for the treatment of refractory B-cell malignancies. GRAPHICAL ABSTRACT: Abundant nuclear ß-cateninß-catenin pairs with TCF7 factors for transcriptional activation of MYCB-cells rely on efficient degradation of ß-catenin by GSK3ßB-cell-specific expression of Ikaros factors Unique vulnerability in B-cell tumors: GSK3ß-inhibitors induce nuclear accumulation of ß-catenin.ß-catenin pairs with B-cell-specific Ikaros factors for transcriptional repression of MYC.

4.
Epigenomes ; 6(4)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36278683

ABSTRACT

The hematopoietic transcription factor Ikaros (IKZF1) regulates normal B cell development and functions as a tumor suppressor in precursor B cell acute lymphoblastic leukemia (B-ALL). MicroRNAs (miRNAs) are small regulatory RNAs that through post-transcriptional gene regulation play critical roles in intracellular processes including cell growth in cancer. However, the role of Ikaros in the regulation of miRNA expression in developing B cells is unknown. In this study, we examined the Ikaros-regulated miRNA targets using human IKZF1-mutated Ph+ B-ALL cell lines. Inducible expression of wild-type Ikaros (the Ik1 isoform) caused B-ALL growth arrest and exit from the cell cycle. Global miRNA expression analysis revealed a total of 31 miRNAs regulated by IK1, and ChIP-seq analysis showed that Ikaros bound to several Ik1-responsive miRNA genes. Examination of the prognostic significance of miRNA expression in B-ALL indicate that the IK1-regulated miRNAs hsa-miR-26b, hsa-miR-130b and hsa-miR-4649 are significantly associated with outcome in B-ALL. Our findings establish a potential regulatory circuit between the tumor-suppressor Ikaros and the oncogenic miRNA networks in IKZF1-mutated B-ALL. These results indicate that Ikaros regulates the expression of a subset of miRNAs, of which several may contribute to B-ALL growth.

5.
BMC Res Notes ; 14(1): 366, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34544495

ABSTRACT

OBJECTIVE: Among the different methods to profile the genome-wide patterns of transcription factor binding and histone modifications in cells and tissues, CUT&RUN has emerged as a more efficient approach that allows for a higher signal-to-noise ratio using fewer number of cells compared to ChIP-seq. The results from CUT&RUN and other related sequence enrichment assays requires comprehensive quality control (QC) and comparative analysis of data quality across replicates. While several computational tools currently exist for read mapping and analysis, a systematic reporting of data quality is lacking. Our aims were to (1) compare methods for using frozen versus fresh cells for CUT&RUN and (2) to develop an easy-to-use pipeline for assessing data quality. RESULTS: We compared a workflow for CUT&RUN with fresh and frozen samples, and present an R package called ssvQC for quality control and comparison of data quality derived from CUT&RUN and other enrichment-based sequence data. Using ssvQC, we evaluate results from different CUT&RUN protocols for transcription factors and histone modifications from fresh and frozen tissue samples. Overall, this process facilitates evaluation of data quality across datasets and permits inspection of peak calling analysis, replicate analysis of different data types. The package ssvQC is readily available at https://github.com/FrietzeLabUVM/ssvQC .


Subject(s)
Histone Code , Transcription Factors , Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , Quality Control , Workflow
6.
Int J Mol Sci ; 22(5)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33799946

ABSTRACT

Non-coding RNAs (ncRNAs) comprise a diverse class of non-protein coding transcripts that regulate critical cellular processes associated with cancer. Advances in RNA-sequencing (RNA-Seq) have led to the characterization of non-coding RNA expression across different types of human cancers. Through comprehensive RNA-Seq profiling, a growing number of studies demonstrate that ncRNAs, including long non-coding RNA (lncRNAs) and microRNAs (miRNA), play central roles in progenitor B-cell acute lymphoblastic leukemia (B-ALL) pathogenesis. Furthermore, due to their central roles in cellular homeostasis and their potential as biomarkers, the study of ncRNAs continues to provide new insight into the molecular mechanisms of B-ALL. This article reviews the ncRNA signatures reported for all B-ALL subtypes, focusing on technological developments in transcriptome profiling and recently discovered examples of ncRNAs with biologic and therapeutic relevance in B-ALL.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Untranslated/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Glucocorticoids/pharmacology , Humans , MicroRNAs/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Long Noncoding/genetics
7.
Sci Transl Med ; 12(526)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941829

ABSTRACT

Overcoming drug resistance remains a key challenge to cure patients with acute and chronic B cell malignancies. Here, we describe a stromal cell-autonomous signaling pathway, which contributes to drug resistance of malignant B cells. We show that protein kinase C (PKC)-ß-dependent signals from bone marrow-derived stromal cells markedly decrease the efficacy of cytotoxic therapies. Conversely, small-molecule PKC-ß inhibitors antagonize prosurvival signals from stromal cells and sensitize tumor cells to targeted and nontargeted chemotherapy, resulting in enhanced cytotoxicity and prolonged survival in vivo. Mechanistically, stromal PKC-ß controls the expression of adhesion and matrix proteins, required for activation of phosphoinositide 3-kinases (PI3Ks) and the extracellular signal-regulated kinase (ERK)-mediated stabilization of B cell lymphoma-extra large (BCL-XL) in tumor cells. Central to the stroma-mediated drug resistance is the PKC-ß-dependent activation of transcription factor EB, regulating lysosome biogenesis and plasma membrane integrity. Stroma-directed therapies, enabled by direct inhibition of PKC-ß, enhance the effectiveness of many antileukemic therapies.


Subject(s)
Protein Kinase C beta/metabolism , Apoptosis/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Phosphorylation/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured
8.
J Exp Med ; 215(10): 2586-2599, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30158115

ABSTRACT

B cell progenitors require paracrine signals such as interleukin-7 (IL-7) provided by bone marrow stromal cells for proliferation and survival. Yet, how B cells regulate access to these signals in vivo remains unclear. Here we show that proB and IL-7+ cells form a cell circuit wired by IL-7R signaling, which controls CXCR4 and focal adhesion kinase (FAK) expression and restricts proB cell movement due to increased adhesion to IL-7+CXCL12Hi cells. PreBCR signaling breaks this circuit by switching the preB cell behavior into a fast-moving and lower-adhesion state via increased CXCR4 and reduced FAK/α4ß1 expression. This behavioral change reduces preB cell exposure to IL-7, thereby attenuating IL-7R signaling in vivo. Remarkably, IL-7 production is downregulated by signals provided by preB cells with unrepaired double-stranded DNA breaks and by preB acute lymphoblastic leukemic cells. Combined, these studies revealed that distinct cell circuits control the quality and homeostasis of B cell progenitors.


Subject(s)
Interleukin-7/immunology , Mesenchymal Stem Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Receptors, Interleukin-7/immunology , Signal Transduction/immunology , Animals , Chemokine CXCL12/genetics , Interleukin-7/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/cytology , Receptors, Interleukin-7/genetics , Signal Transduction/genetics
9.
Nature ; 558(7711): E5, 2018 06.
Article in English | MEDLINE | ID: mdl-29849140

ABSTRACT

In Fig. 3c of this Letter, the the effects of CRISPR-Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. For curves depicting NR3C1 (left graph), data s for TXNIP (middle graph) were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendment for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1-4 and Extended Data Figs. 1-10 of the original Letter.

11.
Cell Rep ; 21(8): 2277-2290, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166616

ABSTRACT

The local environment may affect the development and function of tissue-resident T regulatory cells (Tregs), which are crucial for controlling inflammation. Although the aryl hydrocarbon receptor (Ahr), an environmental sensor, is expressed by Tregs, its role in Treg cell development and/or function remains elusive. Here, we generated mouse genetic models to ablate or activate Ahr expression specifically in Tregs. We showed that Ahr was expressed more abundantly by peripherally induced Tregs (pTregs) in the gut and that its expression was independent of microbiota. Ahr was important for Treg gut homing and function. Ahr inhibited pro-inflammatory cytokines produced by Tregs but was dispensable for Treg stability. Furthermore, Ahr-expressing Tregs had enhanced in vivo suppressive activity compared with Tregs lacking Ahr expression in a T cell transfer model of colitis. Our data suggest that Ahr signaling in Tregs may be important for gut immune homeostasis.


Subject(s)
Colitis/metabolism , Forkhead Transcription Factors/metabolism , Receptors, Aryl Hydrocarbon/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/metabolism , Inflammation/immunology , Mice, Transgenic , Signal Transduction/immunology , Th17 Cells/immunology
12.
Nat Immunol ; 18(6): 694-704, 2017 06.
Article in English | MEDLINE | ID: mdl-28369050

ABSTRACT

The transcription factor STAT5 has a critical role in B cell acute lymphoblastic leukemia (B-ALL). How STAT5 mediates this effect is unclear. Here we found that activation of STAT5 worked together with defects in signaling components of the precursor to the B cell antigen receptor (pre-BCR), including defects in BLNK, BTK, PKCß, NF-κB1 and IKAROS, to initiate B-ALL. STAT5 antagonized the transcription factors NF-κB and IKAROS by opposing regulation of shared target genes. Super-enhancers showed enrichment for STAT5 binding and were associated with an opposing network of transcription factors, including PAX5, EBF1, PU.1, IRF4 and IKAROS. Patients with a high ratio of active STAT5 to NF-κB or IKAROS had more-aggressive disease. Our studies indicate that an imbalance of two opposing transcriptional programs drives B-ALL and suggest that restoring the balance of these pathways might inhibit B-ALL.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , B-Lymphocytes , Gene Expression Regulation, Neoplastic , Ikaros Transcription Factor/genetics , Pre-B Cell Receptors/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , STAT5 Transcription Factor/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Interferon Regulatory Factors/genetics , Mice , Multiplex Polymerase Chain Reaction , NF-kappa B p50 Subunit/genetics , PAX5 Transcription Factor/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Protein Kinase C beta/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Survival Rate , Trans-Activators/genetics
13.
J Exp Med ; 214(3): 793-814, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28190001

ABSTRACT

Inactivation of the tumor suppressor gene encoding the transcriptional regulator Ikaros (IKZF1) is a hallmark of BCR-ABL1+ precursor B cell acute lymphoblastic leukemia (pre-B ALL). However, the mechanisms by which Ikaros functions as a tumor suppressor in pre-B ALL remain poorly understood. Here, we analyzed a mouse model of BCR-ABL1+ pre-B ALL together with a new model of inducible expression of wild-type Ikaros in IKZF1 mutant human BCR-ABL1+ pre-B ALL. We performed integrated genome-wide chromatin and expression analyses and identified Ikaros target genes in mouse and human BCR-ABL1+ pre-B ALL, revealing novel conserved gene pathways associated with Ikaros tumor suppressor function. Notably, genetic depletion of different Ikaros targets, including CTNND1 and the early hematopoietic cell surface marker CD34, resulted in reduced leukemic growth. Our results suggest that Ikaros mediates tumor suppressor function by enforcing proper developmental stage-specific expression of multiple genes through chromatin compaction at its target genes.


Subject(s)
Fusion Proteins, bcr-abl/analysis , Ikaros Transcription Factor/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Proteins/physiology , Animals , Antigens, CD34/analysis , Cell Cycle , Cell Line, Tumor , Gene Expression Regulation, Leukemic , Humans , Ikaros Transcription Factor/genetics , Leukosialin/analysis , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/genetics
14.
Nature ; 542(7642): 479-483, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28192788

ABSTRACT

B-lymphoid transcription factors, such as PAX5 and IKZF1, are critical for early B-cell development, yet lesions of the genes encoding these transcription factors occur in over 80% of cases of pre-B-cell acute lymphoblastic leukaemia (ALL). The importance of these lesions in ALL has, until now, remained unclear. Here, by combining studies using chromatin immunoprecipitation with sequencing and RNA sequencing, we identify a novel B-lymphoid program for transcriptional repression of glucose and energy supply. Our metabolic analyses revealed that PAX5 and IKZF1 enforce a state of chronic energy deprivation, resulting in constitutive activation of the energy-stress sensor AMPK. Dominant-negative mutants of PAX5 and IKZF1, however, relieved this glucose and energy restriction. In a transgenic pre-B ALL mouse model, the heterozygous deletion of Pax5 increased glucose uptake and ATP levels by more than 25-fold. Reconstitution of PAX5 and IKZF1 in samples from patients with pre-B ALL restored a non-permissive state and induced energy crisis and cell death. A CRISPR/Cas9-based screen of PAX5 and IKZF1 transcriptional targets identified the products of NR3C1 (encoding the glucocorticoid receptor), TXNIP (encoding a glucose-feedback sensor) and CNR2 (encoding a cannabinoid receptor) as central effectors of B-lymphoid restriction of glucose and energy supply. Notably, transport-independent lipophilic methyl-conjugates of pyruvate and tricarboxylic acid cycle metabolites bypassed the gatekeeper function of PAX5 and IKZF1 and readily enabled leukaemic transformation. Conversely, pharmacological TXNIP and CNR2 agonists and a small-molecule AMPK inhibitor strongly synergized with glucocorticoids, identifying TXNIP, CNR2 and AMPK as potential therapeutic targets. Furthermore, our results provide a mechanistic explanation for the empirical finding that glucocorticoids are effective in the treatment of B-lymphoid but not myeloid malignancies. Thus, B-lymphoid transcription factors function as metabolic gatekeepers by limiting the amount of cellular ATP to levels that are insufficient for malignant transformation.


Subject(s)
B-Lymphocytes/metabolism , Energy Metabolism/genetics , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/drug effects , Carcinogenesis/genetics , Carrier Proteins/agonists , Carrier Proteins/metabolism , Cell Death , Chromatin Immunoprecipitation , Citric Acid Cycle , Disease Models, Animal , Female , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Ikaros Transcription Factor/metabolism , Mice , Mice, Transgenic , PAX5 Transcription Factor/deficiency , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Serine-Threonine Kinases/metabolism , Pyruvic Acid/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Receptors, Glucocorticoid/metabolism , Sequence Analysis, RNA
15.
Immunity ; 45(1): 185-97, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27438771

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) expressing the transcription factor (TF) RORγt are important for the defense and homeostasis of host intestinal tissues. The zinc finger TF Ikaros, encoded by Ikzf1, is essential for the development of RORγt(+) fetal lymphoid tissue inducer (LTi) cells and lymphoid organogenesis, but its role in postnatal ILC3s is unknown. Here, we show that small-intestinal ILC3s had lower Ikaros expression than ILC precursors and other ILC subsets. Ikaros inhibited ILC3s in a cell-intrinsic manner through zinc-finger-dependent inhibition of transcriptional activity of the aryl hydrocarbon receptor, a key regulator of ILC3 maintenance and function. Ablation of Ikzf1 in RORγt(+) ILC3s resulted in increased expansion and cytokine production of intestinal ILC3s and protection against infection and colitis. Therefore, in contrast to being required for LTi development, Ikaros inhibits postnatal ILC3 development and function to regulate gut immune responses at steady state and in disease.


Subject(s)
Colitis/immunology , Ikaros Transcription Factor/metabolism , Intestinal Mucosa/immunology , Lymphocytes/physiology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Differentiation , Cells, Cultured , Colitis/chemically induced , Dextran Sulfate , Homeostasis , Ikaros Transcription Factor/genetics , Immunity, Innate , Intestinal Mucosa/microbiology , Lymphocyte Activation , Lymphocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Transcriptional Activation
16.
Genes Dev ; 29(17): 1801-16, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26314708

ABSTRACT

The DNA-binding protein Ikaros is a potent tumor suppressor and hematopoietic regulator. However, the mechanisms by which Ikaros functions remain poorly understood, due in part to its atypical DNA-binding properties and partnership with the poorly understood Mi-2/NuRD complex. In this study, we analyzed five sequential stages of thymocyte development in a mouse strain containing a targeted deletion of Ikaros zinc finger 4, which exhibits a select subset of abnormalities observed in Ikaros-null mice. By examining thymopoiesis in vivo and in vitro, diverse abnormalities were observed at each developmental stage. RNA sequencing revealed that each stage is characterized by the misregulation of a limited number of genes, with a strong preference for stage-specific rather than lineage-specific genes. Strikingly, individual genes rarely exhibited Ikaros dependence at all stages. Instead, a consistent feature of the aberrantly expressed genes was a reduced magnitude of expression level change during developmental transitions. These results, combined with analyses of the interplay between Ikaros loss of function and Notch signaling, suggest that Ikaros may not be a conventional activator or repressor of defined sets of genes. Instead, a primary function may be to sharpen the dynamic range of gene expression changes during developmental transitions via atypical molecular mechanisms that remain undefined.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Nerve Tissue Proteins/metabolism , Thymocytes/cytology , Animals , Carrier Proteins/genetics , Cell Cycle Checkpoints , Cells, Cultured , DNA-Binding Proteins , Female , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Sequence Analysis, RNA
17.
Blood ; 126(15): 1813-22, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26219304

ABSTRACT

Ikaros (IKZF1) is a tumor suppressor that binds DNA and regulates expression of its target genes. The mechanism of Ikaros activity as a tumor suppressor and the regulation of Ikaros function in leukemia are unknown. Here, we demonstrate that Ikaros controls cellular proliferation by repressing expression of genes that promote cell cycle progression and the phosphatidylinositol-3 kinase (PI3K) pathway. We show that Ikaros function is impaired by the pro-oncogenic casein kinase II (CK2), and that CK2 is overexpressed in leukemia. CK2 inhibition restores Ikaros function as transcriptional repressor of cell cycle and PI3K pathway genes, resulting in an antileukemia effect. In high-risk leukemia where one IKZF1 allele has been deleted, CK2 inhibition restores the transcriptional repressor function of the remaining wild-type IKZF1 allele. CK2 inhibition demonstrated a potent therapeutic effect in a panel of patient-derived primary high-risk B-cell acute lymphoblastic leukemia xenografts as indicated by prolonged survival and a reduction of leukemia burden. We demonstrate the efficacy of a novel therapeutic approach for high-risk leukemia: restoration of Ikaros tumor suppressor activity via inhibition of CK2. These results provide a rationale for the use of CK2 inhibitors in clinical trials for high-risk leukemia, including cases with deletion of one IKZF1 allele.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Genes, Tumor Suppressor , Ikaros Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Apoptosis/drug effects , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Enzyme Inhibitors/pharmacology , Female , Humans , Ikaros Transcription Factor/genetics , Mice , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Nat Commun ; 6: 6471, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25753524

ABSTRACT

Despite recent advances in the cure rate of acute lymphoblastic leukaemia (ALL), the prognosis for patients with relapsed ALL remains poor. Here we identify FOXM1 as a candidate responsible for an aggressive clinical course. We show that FOXM1 levels peak at the pre-B-cell receptor checkpoint but are dispensable for normal B-cell development. Compared with normal B-cell populations, FOXM1 levels are 2- to 60-fold higher in ALL cells and are predictive of poor outcome in ALL patients. FOXM1 is negatively regulated by FOXO3A, supports cell survival, drug resistance, colony formation and proliferation in vitro, and promotes leukemogenesis in vivo. Two complementary approaches of pharmacological FOXM1 inhibition-(i) FOXM1 transcriptional inactivation using the thiazole antibiotic thiostrepton and (ii) an FOXM1 inhibiting ARF-derived peptide-recapitulate the findings of genetic FOXM1 deletion. Taken together, our data identify FOXM1 as a novel therapeutic target, and demonstrate feasibility of FOXM1 inhibition in ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Transcription Factors/antagonists & inhibitors , Gene Expression Regulation, Leukemic , Peptides/chemical synthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Thiostrepton/pharmacology , Adult , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Child , Clinical Trials as Topic , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein M1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Mice , Peptides/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
19.
J Immunol ; 193(8): 3934-46, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25194055

ABSTRACT

Proper immune responses are needed to control pathogen infection at mucosal surfaces. IL-22-producing CD4(+) T cells play an important role in controlling bacterial infection in the gut; however, transcriptional regulation of these cells remains elusive. In this study, we show that mice with targeted deletion of the fourth DNA-binding zinc finger of the transcription factor Ikaros had increased IL-22-producing, but not IL-17-producing, CD4(+) T cells in the gut. Adoptive transfer of CD4(+) T cells from these Ikaros-mutant mice conferred enhanced mucosal immunity against Citrobacter rodentium infection. Despite an intact in vivo thymic-derived regulatory T cell (Treg) compartment in these Ikaros-mutant mice, TGF-ß, a cytokine well known for induction of Tregs, failed to induce Foxp3 expression in Ikaros-mutant CD4(+) T cells in vitro and, instead, promoted IL-22. Aberrant upregulation of IL-21 in CD4(+) T cells expressing mutant Ikaros was responsible, at least in part, for the enhanced IL-22 expression in a Stat3-dependent manner. Genetic analysis using compound mutations further demonstrated that the aryl hydrocarbon receptor, but not RORγt, was required for aberrant IL-22 expression by Ikaros-mutant CD4(+) T cells, whereas forced expression of Foxp3 was sufficient to inhibit this aberrant cytokine production. Together, our data identified new functions for Ikaros in maintaining mucosal immune homeostasis by restricting IL-22 production by CD4(+) T cells.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections/immunology , Ikaros Transcription Factor/metabolism , Interleukins/biosynthesis , T-Lymphocytes, Regulatory/immunology , Animals , Enterobacteriaceae Infections/genetics , Forkhead Transcription Factors/biosynthesis , Ikaros Transcription Factor/genetics , Immunity, Mucosal , Interleukin-17/biosynthesis , Interleukins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Aryl Hydrocarbon , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/transplantation , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Interleukin-22
20.
Nat Immunol ; 14(10): 1073-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24013668

ABSTRACT

C2H2 zinc fingers are found in several key transcriptional regulators in the immune system. However, these proteins usually contain more fingers than are needed for sequence-specific DNA binding, which suggests that different fingers regulate different genes and functions. Here we found that mice lacking finger 1 or finger 4 of Ikaros exhibited distinct subsets of the hematological defects of Ikaros-null mice. Most notably, the two fingers controlled different stages of lymphopoiesis, and finger 4 was selectively required for tumor suppression. The distinct defects support the hypothesis that only a small number of genes that are targets of Ikaros are critical for each of its biological functions. The subcategorization of functions and target genes by mutagenesis of individual zinc fingers will facilitate efforts to understand how zinc-finger transcription factors regulate development, immunity and disease.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation , Ikaros Transcription Factor/genetics , Leukemia/genetics , Lymphopoiesis/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Base Sequence , Binding Sites , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin Immunoprecipitation , Cluster Analysis , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Ikaros Transcription Factor/metabolism , Immunophenotyping , Leukemia/metabolism , Leukemia/mortality , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/mortality , Mice , Mice, Knockout , Molecular Sequence Data , Nucleotide Motifs , Phenotype , Position-Specific Scoring Matrices , Protein Binding , Thymocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...