Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114227, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38735044

ABSTRACT

CUX1 is a homeodomain-containing transcription factor that is essential for the development and differentiation of multiple tissues. CUX1 is recurrently mutated or deleted in cancer, particularly in myeloid malignancies. However, the mechanism by which CUX1 regulates gene expression and differentiation remains poorly understood, creating a barrier to understanding the tumor-suppressive functions of CUX1. Here, we demonstrate that CUX1 directs the BAF chromatin remodeling complex to DNA to increase chromatin accessibility in hematopoietic cells. CUX1 preferentially regulates lineage-specific enhancers, and CUX1 target genes are predictive of cell fate in vivo. These data indicate that CUX1 regulates hematopoietic lineage commitment and homeostasis via pioneer factor activity, and CUX1 deficiency disrupts these processes in stem and progenitor cells, facilitating transformation.

2.
Exp Hematol ; 115: 54-67, 2022 11.
Article in English | MEDLINE | ID: mdl-35995095

ABSTRACT

Therapy-related myeloid neoplasms (t-MNs) share many clinical and molecular characteristics with AML de novo in the elderly. One common factor is that they arise in the setting of chronic inflammation, likely because of advanced age or chemotherapy-induced senescence. Here, we examined the effect of haploinsufficient loss of the del(5q) tumor suppressor gene, EGR1, commonly deleted in high-risk MNs. In mice, under the exogenous stress of either serial transplant or successive doses of the alkylating agent N-ethyl-nitrosourea (ENU), Egr1-haploinsufficient hematopoietic stem cells (HSCs) exhibit a clonal advantage. Complete loss of EGR1 function is incompatible with transformation; mutations of EGR1 are rare and are not observed in the remaining allele in del(5q) patients, and complete knockout of Egr1 in mice leads to HSC exhaustion. Using chromatin immunoprecipitation sequencing (ChIP-seq), we identified EGR1 binding sites in human CD34+ cord blood-derived stem and progenitor cells (HSPCs) and found that EGR1 binds genes critical for stem cell differentiation, inflammatory signaling, and the DNA damage response. Notably, in the chromosome 5 sequences frequently deleted in patients, there is a significant enrichment of innate and inflammatory genes, which may confer a fitness advantage in an inflammatory environment. Short hairpin RNA (shRNA)-mediated silencing of EGR1 biases HSPCs toward a self-renewal transcriptional signature. In the absence of EGR1, HSPCs are characterized by upregulated MYC-driven proliferative signals, downregulated CDKN1A (p21), disrupted DNA damage response, and downregulated inflammation-adaptations anticipated to confer a relative fitness advantage for stem cells especially in an environment of chronic inflammation.


Subject(s)
Haploinsufficiency , Hematopoietic Stem Cells , Humans , Mice , Animals , Aged , Hematopoietic Stem Cells/metabolism , Antigens, CD34/metabolism , Ethylnitrosourea/metabolism , Inflammation/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism
3.
Blood Cancer Discov ; 1(1): 32-47, 2020 07.
Article in English | MEDLINE | ID: mdl-32924016

ABSTRACT

Therapy-related myeloid neoplasms (t-MNs) following treatment with alkylating agents are characterized by a del(5q), complex karyotypes, alterations of TP53, and a dismal prognosis. To decipher the molecular pathway(s) leading to the pathogenesis of del(5q) t-MN and the effect(s) of cytotoxic therapy on the marrow microenvironment, we developed a mouse model with loss of two key del(5q) genes, EGR1 and APC, in hematopoietic cells. We used the well-characterized drug, N-ethyl-N-nitrosurea (ENU) to demonstrate that alkylating agent exposure of stromal cells in the microenvironment increases the incidence of myeloid disease. In addition, loss of Trp53 with Egr1 and Apc was required to drive the development of a transplantable leukemia, and accompanied by the acquisition of somatic mutations in DNA damage response genes. ENU treatment of mesenchymal stromal cells induced cellular senescence, and led to the acquisition of a senescence-associated secretory phenotype, which may be a critical microenvironmental alteration in the pathogenesis of myeloid neoplasms.


Subject(s)
Antineoplastic Agents, Alkylating , Bone Marrow , Leukemia, Myeloid , Neoplasms, Second Primary , Animals , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Chromosome Deletion , Genes, p53 , Leukemia, Myeloid/chemically induced , Leukemia, Myeloid/genetics , Mice , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/genetics , Stromal Cells , Tumor Microenvironment/genetics
4.
Blood ; 129(22): 2959-2970, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28348148

ABSTRACT

There is accumulating evidence that functional alteration(s) of the bone marrow (BM) microenvironment contribute to the development of some myeloid disorders, such as myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). In addition to a cell-intrinsic role of WNT activation in leukemia stem cells, WNT activation in the BM niche is also thought to contribute to the pathogenesis of MDS and AML. We previously showed that the Apc-haploinsufficient mice (Apcdel/+ ) model MDS induced by an aberrant BM microenvironment. We sought to determine whether Apc, a multifunctional protein and key negative regulator of the canonical ß-catenin (Ctnnb1)/WNT-signaling pathway, mediates this disease through modulating WNT signaling, and whether inhibition of WNT signaling prevents the development of MDS in Apcdel/+ mice. Here, we demonstrate that loss of 1 copy of Ctnnb1 is sufficient to prevent the development of MDS in Apcdel/+ mice and that altered canonical WNT signaling in the microenvironment is responsible for the disease. Furthermore, the US Food and Drug Administration (FDA)-approved drug pyrvinium delays and/or inhibits disease in Apcdel/+ mice, even when it is administered after the presentation of anemia. Other groups have observed increased nuclear CTNNB1 in stromal cells from a high frequency of MDS/AML patients, a finding that together with our results highlights a potential new strategy for treating some myeloid disorders.


Subject(s)
Genes, APC , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/prevention & control , Stem Cell Niche/genetics , Wnt Signaling Pathway , Animals , Disease Models, Animal , Haploinsufficiency , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/pathology , Pyrvinium Compounds/pharmacology , Wnt Signaling Pathway/genetics , beta Catenin/genetics
7.
Haematologica ; 99(6): 1032-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532040

ABSTRACT

The control of mRNA stability plays a central role in orchestrating gene-regulatory networks in hematopoietic cell growth, differentiation and tumorigenesis. HNRNPA0, which encodes an RNA-binding protein shown to regulate transcript stability via binding to the AU-rich elements of mRNAs, is located within the commonly deleted segment of 5q31.2 in myeloid neoplasms with a del(5q), and is expressed at haploinsufficient levels in these patients. We show that HNRNPA0 is normally highly expressed in hematopoietic stem cells and exhibits dynamic changes in expression during the course of differentiation. To model HNRNPA0 haploinsufficiency, we used RNAi interference in primary murine cells and an experimental cell system, and found that reduced Hnrnpa0 expression leads to a shift from monocytic towards granulocytic differentiation. Microarray-based global expression profiling revealed that Hnrnpa0 knockdown disproportionally impacts AU-rich containing transcripts and alters expression of myeloid specification genes. In therapy-related myeloid neoplasms with a del(5q), AU-rich containing mRNAs are enriched in transcripts that encode proteins associated with increased growth and proliferation. Our findings implicate haploinsufficiency of HNRNPA0 as one of the key initiating mutations in the pathogenesis of myeloid neoplasms with a del(5q), and suggest that therapies that target AU-rich elements warrant consideration in efforts to develop new mechanism-based treatment strategies.


Subject(s)
AT Rich Sequence , Chromosome Deletion , Chromosomes, Human, Pair 5 , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Myeloid Cells/metabolism , Transcription, Genetic , Animals , Cell Line , Cell Transdifferentiation/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Granulocytes/cytology , Granulocytes/metabolism , Hematopoiesis/genetics , Humans , Leukemia, Myeloid/genetics , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Neoplasms, Second Primary/genetics
8.
Blood ; 123(7): 1069-78, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24381225

ABSTRACT

An interstitial deletion of chromosome 5, del(5q), is the most common structural abnormality in primary myelodysplastic syndromes (MDS) and therapy-related myeloid neoplasms (t-MNs) after cytotoxic therapy. Loss of TP53 activity, through mutation or deletion, is highly associated with t-MNs with a del(5q). We previously demonstrated that haploinsufficiency of Egr1 and Apc, 2 genes lost in the 5q deletion, are key players in the progression of MDS with a del(5q). Using genetically engineered mice, we now show that reduction or loss of Tp53 expression, in combination with Egr1 haploinsufficiency, increased the rate of development of hematologic neoplasms and influenced the disease spectrum, but did not lead to overt myeloid leukemia, suggesting that altered function of additional gene(s) on 5q are likely required for myeloid leukemia development. Next, we demonstrated that cell intrinsic loss of Tp53 in hematopoietic stem and progenitor cells haploinsufficient for both Egr1 and Apc led to the development of acute myeloid leukemia (AML) in 17% of mice. The long latency (234-299 days) and clonal chromosomal abnormalities in the AMLs suggest that additional genetic changes may be required for full transformation. Thus, loss of Tp53 activity in cooperation with Egr1 and Apc haploinsufficiency creates an environment that is permissive for malignant transformation and the development of AML.


Subject(s)
Chromosome Deletion , Early Growth Response Protein 1/genetics , Genes, APC , Genes, p53/physiology , Haploinsufficiency , Leukemia, Myeloid, Acute/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Blood ; 123(2): 228-38, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24264229

ABSTRACT

Therapy-related myeloid neoplasms (t-MN) are a late complication of the successful use of cytotoxic therapy for patients with cancer. A heterozygous deletions of the long arm of chromosome 5 [del(5q)], observed in 40% of patients, is associated with prior exposure to alkylating agents, and a high frequency of TP53 loss or mutation. In previous studies, we demonstrated that haploinsufficiency of 2 del(5q) genes, Egr1, and Apc, individually play a role in the pathogenesis of hematologic disease in mice. We now show that loss of one copy of Egr1 or Tp53 in an Apc haploinsufficient background (Apc (del/+)) accelerated the development of a macrocytic anemia with monocytosis, early features of t-MN. The development of anemia was significantly accelerated by treatment of mice with the alkylating agent, N-ethyl-N-nitrosourea (ENU), regardless of the levels of expression of Egr1 and Tp53. Transplantation of either wild type; Egr1(+/-); Tp53(+/-); Apc(del/+); or Egr1(+/-), Apc(del/+) bone marrow cells into lethally irradiated Apc(del/+) recipients resulted in rapid development of anemia that was further accelerated by administration of ENU to recipients, demonstrating that the Apc(del/+)-induced anemia was cell extrinsic and potentiated by ENU mutagenesis. These data emphasize the synergistic role of cell intrinsic and cell extrinsic (microenvironment) factors in the pathogenesis of t-MN, and raise awareness of the deleterious effects of cytotoxic therapy on the stromal microenvironment.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5 , Early Growth Response Protein 1/genetics , Haploinsufficiency , Tumor Suppressor Protein p53/genetics , Alleles , Anemia, Macrocytic/chemically induced , Anemia, Macrocytic/genetics , Anemia, Macrocytic/mortality , Animals , Apoptosis/genetics , Bone Marrow/drug effects , Cellular Microenvironment/drug effects , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis/genetics , Ethylnitrosourea/adverse effects , Genes, Lethal , Genotype , Heterozygote , Humans , Mice , Mice, Transgenic , Spleen/metabolism , Spleen/pathology
10.
Mediterr J Hematol Infect Dis ; 3(1): e2011019, 2011.
Article in English | MEDLINE | ID: mdl-21713073

ABSTRACT

Therapy-related myeloid neoplasm (t-MN) is a distinctive clinical syndrome occurring after exposure to chemotherapy or radiotherapy. t-MN arises in most cases from a multipotential hematopoietic stem cell or, less commonly, in a lineage committed progenitor cell. The prognosis for patients with t-MN is poor, as current forms of therapy are largely ineffective. Cytogenetic analysis, molecular analysis and gene expression profiling analysis of t-MN has revealed that there are distinct subtypes of the disease; however, our understanding of the genetic basis of t-MN is incomplete. Elucidating the genetic pathways and molecular networks that are perturbed in t-MNs, may facilitate the identification of therapeutic targets that can be exploited for the development of urgently-needed targeted therapies.

11.
Chem Biol Interact ; 184(1-2): 50-7, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-19958752

ABSTRACT

Therapy-related myelodysplastic syndrome and acute myeloid leukemia (t-MDS/t-AML) are late complications of cytotoxic therapy used in the treatment of malignant diseases. The most common subtype of t-AML ( approximately 75% of cases) develops after exposure to alkylating agents, and is characterized by loss or deletion of chromosome 5 and/or 7 [-5/del(5q), -7/del(7q)], and a poor outcome (median survival 8 months). In the University of Chicago's series of 386 patients with t-MDS/t-AML, 79 (20%) patients had abnormalities of chromosome 5, 95 (25%) patients had abnormalities of chromosome 7, and 85 (22%) patients had abnormalities of both chromosomes 5 and 7. t-MDS/t-AML with a -5/del(5q) is associated with a complex karyotype, characterized by trisomy 8, as well as loss of 12p, 13q, 16q22, 17p (TP53 locus), chromosome 18, and 20q. In addition, this subtype of t-AML is characterized by a unique expression profile (higher expression of genes) involved in cell cycle control (CCNA2, CCNE2, CDC2), checkpoints (BUB1), or growth (MYC), loss of expression of IRF8, and overexpression of FHL2. Haploinsufficiency of the RPS14, EGR1, APC, NPM1, and CTNNA1 genes on 5q has been implicated in the pathogenesis of MDS/AML. In previous studies, we determined that Egr1 acts by haploinsufficiency and cooperates with mutations induced by alkylating agents to induce myeloid leukemias in the mouse. To identify mutations that cooperate with Egr1 haploinsufficiency, we used retroviral insertional mutagenesis. To date, we have identified two common integration sites involving genes encoding transcription factors that play a critical role in hematopoiesis (Evi1 and Gfi1b loci). Of note is that the EVI1 transcription factor gene is deregulated in human AMLs, particularly those with -7, and abnormalities of 3q. Identifying the genetic pathways leading to t-AML will provide new insights into the underlying biology of this disease, and may facilitate the identification of new therapeutic targets.


Subject(s)
Cytogenetic Analysis , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/radiation effects , Leukemia, Myeloid, Acute/genetics , Neoplasms, Second Primary/genetics , Animals , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/radiotherapy , Nucleophosmin
12.
Mol Biol Cell ; 16(5): 2339-48, 2005 May.
Article in English | MEDLINE | ID: mdl-15716350

ABSTRACT

B cell antigen receptor (BCR) association with lipid rafts, the actin cytoskeleton, and clathrin-coated pits influences B cell signaling and antigen presentation. Although all three cellular structures have been separately implicated in BCR internalization, the relationship between them has not been clearly defined. In this study, internalization pathways were characterized by specifically blocking each potential mechanism of internalization. BCR uptake was reduced by approximately 70% in B cells conditionally deficient in clathrin heavy chain expression. Actin or raft antagonists were both able to block the residual, clathrin-independent BCR internalization. These agents also affected clathrin-dependent internalization, indicating that clathrin-coated pits, in concert with mechanisms dependent on rafts and actin, mediate the majority of BCR internalization. Clustering G(M1) gangliosides enhanced clathrin-independent BCR internalization, and this required actin. Thus, although rafts or actin independently did not mediate BCR internalization, they apparently cooperate to promote some internalization even in the absence of clathrin. Simultaneous inhibition of all BCR uptake pathways resulted in sustained tyrosine phosphorylation and activation of the extracellular signal-regulated kinase (ERK), strongly suggesting that downstream BCR signaling can occur without receptor translocation to endosomes and that internalization leads to signal attenuation.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Clathrin/metabolism , Receptors, Antigen, B-Cell/metabolism , Actins/metabolism , Animals , Biological Transport, Active , Cell Line , Chickens , Clathrin/deficiency , Clathrin/genetics , Endocytosis , Humans , Membrane Microdomains/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
J Immunol ; 173(9): 5601-9, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494510

ABSTRACT

The B lymphocyte adaptor molecule of 32 kDa (Bam32) is an adaptor that plays an indispensable role in BCR signaling. In this study, we found that upon BCR ligation, Bam32 is recruited to the plasma membrane where it associates with BCR complexes and redistributes and internalizes with BCRs. BCR ligation induced colocalization of Bam32 with lipid rafts, clathrin, and actin filaments. An inhibitor of Src family protein tyrosine kinases (PTKs) blocked both BCR-induced tyrosine phosphorylation of Bam32 and BCR internalization. Moreover, BCR internalization is impaired in Bam32-/- and Lyn-/- cells, and expression of Bam32 with a mutation of its tyrosine phosphorylation site (Y139F) inhibited BCR internalization. These data suggest that Bam32 functions downstream of Src family PTKs to regulate BCR internalization. Bam32 deficiency does not affect tyrosine phosphorylation of clathrin or the association of clathrin with lipid rafts upon BCR cross-linking. However, BCR-induced actin polymerization is impaired in Bam32-/- cells. Collectively, these findings indicate a novel role of Bam32 in connecting Src family PTKs to BCR internalization by an actin-dependent mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Lipoproteins/physiology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/immunology , Actins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Antigen Presentation , B-Lymphocytes/enzymology , Cell Line, Tumor , Clathrin/metabolism , Cross-Linking Reagents/metabolism , Humans , Immune Sera/metabolism , Ligands , Lipoproteins/deficiency , Lipoproteins/metabolism , Membrane Microdomains/enzymology , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Transport/immunology , Receptors, Antigen, B-Cell/immunology , Transferrin/metabolism , src-Family Kinases/metabolism
14.
Immunity ; 17(4): 451-62, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12387739

ABSTRACT

A major function of the B cell is the internalization of antigen through the BCR for processing and presentation to T cells. While there is evidence suggesting that lipid raft signaling may regulate internalization, the molecular machinery coordinating these two processes remains to be defined. Here we present a link between the B cell signaling and internalization machinery and show that Src-family kinase activity is required for inducible clathrin heavy chain phosphorylation, BCR colocalization with clathrin, and regulated internalization. An analysis of different B cell lines shows that BCR uptake occurs only when clathrin is associated with rafts and is tyrosine phosphorylated following BCR crosslinking. We therefore propose that lipid rafts spatially organize signaling cascades with clathrin to regulate BCR internalization.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Clathrin/metabolism , Membrane Microdomains/metabolism , Receptors, Antigen, B-Cell/metabolism , Antigen Presentation , Biological Transport, Active , Cell Line , Clathrin/chemistry , Humans , Ligands , Membrane Microdomains/chemistry , Phosphorylation , Signal Transduction , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...