Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Haematologica ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38841778

ABSTRACT

IKZF1-deletions occur in 10-15% of patients with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) and predict a poor outcome. However, the impact of IKZF1-loss on sensitivity to drugs used in contemporary treatment protocols has remained underexplored. Here we show in experimental models and in patients that loss of IKZF1 promotes resistance to AraC, a key component of both upfront and relapsed treatment protocols. We attribute this resistance, in part, to diminished import and incorporation of cytarabine (AraC) due to reduced expression of the solute carrier hENT1. Moreover, we find elevated mRNA expression of Evi1, a known driver of therapy resistance in myeloid malignancies. Finally, a kinase directed CRISPR/Cas9-screen identified that inhibition of either mediator kinases CDK8/19 or casein kinase 2 can restore response to AraC. We conclude that this high-risk patient group could benefit from alternative antimetabolites, or targeted therapies that resensitize the cells to AraC.

2.
Haematologica ; 109(6): 1755-1765, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38124624

ABSTRACT

In pediatric acute lymphoblastic leukemia (ALL), mutations/deletions affecting the TP53 gene are rare at diagnosis. However, at relapse about 12% of patients show TP53 aberrations, which are predictive of a very poor outcome. Since p53-mediated apoptosis is an endpoint for many cytotoxic drugs, loss of p53 function frequently leads to therapy failure. In this study we show that CRISPR/Cas9-induced loss of TP53 drives resistance to a large majority of drugs used to treat relapsed ALL, including novel agents such as inotuzumab ozogamicin. Using a high-throughput drug screen, we identified the histone deacetylase inhibitor romidepsin as a potent sensitizer of drug responsiveness, improving sensitivity to all chemotherapies tested. In addition, romidepsin improved the response to cytarabine in TP53-deleted ALL cells in vivo. Together, these results indicate that the histone deacetylase inhibitor romidepsin can improve the efficacy of salvage therapies for relapsed TP53-mutated leukemia. Since romidepsin has been approved for clinical use in some adult malignancies, these findings may be rapidly translated to clinical practice.


Subject(s)
Depsipeptides , Histone Deacetylase Inhibitors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Tumor Suppressor Protein p53 , Humans , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Tumor Suppressor Protein p53/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Depsipeptides/pharmacology , Depsipeptides/therapeutic use , Mice , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CRISPR-Cas Systems , Xenograft Model Antitumor Assays , Drug Synergism
3.
Front Oncol ; 12: 905665, 2022.
Article in English | MEDLINE | ID: mdl-36119546

ABSTRACT

Although long-term survival in pediatric acute lymphoblastic leukemia (ALL) currently exceeds 90%, some subgroups, defined by specific genomic aberrations, respond poorly to treatment. We previously reported that leukemias harboring deletions or mutations affecting the B-cell transcription factor IKZF1 exhibit a tumor cell intrinsic resistance to glucocorticoids (GCs), one of the cornerstone drugs used in the treatment of ALL. Here, we identified increased activation of both AKT and ERK signaling pathways as drivers of GC resistance in IKZF1-deficient leukemic cells. Indeed, combined pharmacological inhibition of AKT and ERK signaling effectively reversed GC resistance in IKZF1-deficient leukemias. As inhibitors for both pathways are under clinical investigation, their combined use may enhance the efficacy of prednisolone-based therapy in this high-risk patient group.

4.
Blood ; 138(23): 2383-2395, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34280258

ABSTRACT

Asparaginase (ASNase) therapy has been a mainstay of acute lymphoblastic leukemia (ALL) protocols for decades and shows promise in the treatment of a variety of other cancers. To improve the efficacy of ASNase treatment, we used a CRISPR/Cas9-based screen to identify actionable signaling intermediates that improve the response to ASNase. Both genetic inactivation of Bruton's tyrosine kinase (BTK) and pharmacological inhibition by the BTK inhibitor ibrutinib strongly synergize with ASNase by inhibiting the amino acid response pathway, a mechanism involving c-Myc-mediated suppression of GCN2 activity. This synthetic lethal interaction was observed in 90% of patient-derived xenografts, regardless of the genomic subtype. Moreover, ibrutinib substantially improved ASNase treatment response in a murine PDX model. Hence, ibrutinib may be used to enhance the clinical efficacy of ASNase in ALL. This trial was registered at www.clinicaltrials.gov as # NCT02884453.


Subject(s)
Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Amino Acids/metabolism , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Piperidines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adenine/pharmacology , Adenine/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Asparaginase/pharmacology , Cell Line, Tumor , Humans , Mice , Piperidines/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction/drug effects
5.
Trends Endocrinol Metab ; 32(6): 367-381, 2021 06.
Article in English | MEDLINE | ID: mdl-33795176

ABSTRACT

Targeting tumor cell metabolism is an attractive form of therapy, as it may enhance treatment response in therapy resistant cancers as well as mitigate treatment-related toxicities by reducing the need for genotoxic agents. To meet their increased demand for biomass accumulation and energy production and to maintain redox homeostasis, tumor cells undergo profound changes in their metabolism. In addition to the diversion of glucose metabolism, this is achieved by upregulation of amino acid metabolism. Interfering with amino acid availability can be selectively lethal to tumor cells and has proven to be a cancer specific Achilles' heel. Here we review the biology behind such cancer specific amino acid dependencies and discuss how these vulnerabilities can be exploited to improve cancer therapies.


Subject(s)
Amino Acids , Neoplasms , Amino Acids/metabolism , Humans , Neoplasms/therapy , Oxidation-Reduction
6.
J Cell Physiol ; 234(5): 5379-5389, 2019 05.
Article in English | MEDLINE | ID: mdl-30350856

ABSTRACT

Since the identification of B-cell translocation gene 1 (BTG1) and BTG2 as antiproliferation genes more than two decades ago, their protein products have been implicated in a variety of cellular processes including cell division, DNA repair, transcriptional regulation and messenger RNA stability. In addition to affecting differentiation during development and in the adult, BTG proteins play an important role in maintaining homeostasis under conditions of cellular stress. Genomic profiling of B-cell leukemia and lymphoma has put BTG1 and BTG2 in the spotlight, since both genes are frequently deleted or mutated in these malignancies, pointing towards a role as tumor suppressors. Moreover, in solid tumors, reduced expression of BTG1 or BTG2 is often correlated with malignant cell behavior and poor treatment outcome. Recent studies have uncovered novel roles for BTG1 and BTG2 in genotoxic and integrated stress responses, as well as during hematopoiesis. This review summarizes what is currently known about the roles of BTG1 and BTG2 in these and other cellular processes. In addition, we will highlight the molecular mechanisms and biological consequences of BTG1 and BTG2 deregulation during cancer progression and elaborate on the potential clinical implications of these findings.


Subject(s)
Cell Proliferation , Immediate-Early Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle , Cell Differentiation , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Transcription, Genetic , Tumor Suppressor Proteins/genetics
7.
Exp Hematol ; 60: 57-62.e3, 2018 04.
Article in English | MEDLINE | ID: mdl-29408281

ABSTRACT

Translocation t(12;21) (p13;q22), giving rise to the ETV6-RUNX1 fusion gene, is the most common genetic abnormality in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). This translocation usually arises in utero, but its expression is insufficient to induce leukemia and requires other cooperating genetic lesions for BCP-ALL to develop. Deletions affecting the transcriptional coregulator BTG1 are frequently observed in ETV6-RUNX1-positive leukemia. Here we report that Btg1 deficiency enhances the self-renewal capacity of ETV6-RUNX1-positive mouse fetal liver-derived hematopoietic progenitors (FL-HPCs). Combined expression of the fusion protein and a loss of BTG1 drive upregulation of the proto-oncogene Bcl6 and downregulation of BCL6 target genes, such as p19Arf and Tp53. Similarly, ectopic expression of BCL6 promotes the self-renewal and clonogenic replating capacity of FL-HPCs, by suppressing the expression of p19Arf and Tp53. Together these results identify BCL6 as a potential driver of ETV6-RUNX1-mediated leukemogenesis, which could involve loss of BTG1-dependent suppression of ETV6-RUNX1 function.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Leukemic , Leukemia/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-bcl-6/biosynthesis , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Leukemia/genetics , Leukemia/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53 , Tumor Suppressor Proteins/genetics , ETS Translocation Variant 6 Protein
8.
Trends Endocrinol Metab ; 28(11): 794-806, 2017 11.
Article in English | MEDLINE | ID: mdl-28797581

ABSTRACT

Activating transcription factor 4 (ATF4) is a stress-induced transcription factor that is frequently upregulated in cancer cells. ATF4 controls the expression of a wide range of adaptive genes that allow cells to endure periods of stress, such as hypoxia or amino acid limitation. However, under persistent stress conditions, ATF4 promotes the induction of apoptosis. Recent advances point to a role for post-translational modifications (PTMs) and epigenetic mechanisms in balancing these pro- and anti-survival effects of ATF4. We review here how PTMs and epigenetic modifiers associated with ATF4 may be exploited by cancer cells to cope with cellular stress conditions that are intrinsically associated with tumor growth. Identification of mechanisms that modulate ATF4-mediated transcription and its effects on cellular metabolism may uncover new targets for cancer treatment.


Subject(s)
Activating Transcription Factor 4/physiology , Adaptation, Physiological , Apoptosis/genetics , Neoplasms/pathology , Stress, Physiological/physiology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adaptation, Physiological/genetics , Amino Acids/deficiency , Animals , Cell Survival/genetics , Humans , Hypoxia/genetics , Hypoxia/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Protein Processing, Post-Translational
9.
Haematologica ; 102(3): 541-551, 2017 03.
Article in English | MEDLINE | ID: mdl-27979924

ABSTRACT

Deletions and mutations affecting lymphoid transcription factor IKZF1 (IKAROS) are associated with an increased relapse risk and poor outcome in B-cell precursor acute lymphoblastic leukemia. However, additional genetic events may either enhance or negate the effects of IKZF1 deletions on prognosis. In a large discovery cohort of 533 childhood B-cell precursor acute lymphoblastic leukemia patients, we observed that single-copy losses of BTG1 were significantly enriched in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia (P=0.007). While BTG1 deletions alone had no impact on prognosis, the combined presence of BTG1 and IKZF1 deletions was associated with a significantly lower 5-year event-free survival (P=0.0003) and a higher 5-year cumulative incidence of relapse (P=0.005), when compared with IKZF1-deleted cases without BTG1 aberrations. In contrast, other copy number losses commonly observed in B-cell precursor acute lymphoblastic leukemia, such as CDKN2A/B, PAX5, EBF1 or RB1, did not affect the outcome of IKZF1-deleted acute lymphoblastic leukemia patients. To establish whether the combined loss of IKZF1 and BTG1 function cooperate in leukemogenesis, Btg1-deficient mice were crossed onto an Ikzf1 heterozygous background. We observed that loss of Btg1 increased the tumor incidence of Ikzf1+/- mice in a dose-dependent manner. Moreover, murine B cells deficient for Btg1 and Ikzf1+/- displayed increased resistance to glucocorticoids, but not to other chemotherapeutic drugs. Together, our results identify BTG1 as a tumor suppressor in leukemia that, when deleted, strongly enhances the risk of relapse in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia, and augments the glucocorticoid resistance phenotype mediated by the loss of IKZF1 function.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epistasis, Genetic , Ikaros Transcription Factor/genetics , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Animals , Biomarkers, Tumor , Cell Transformation, Neoplastic/metabolism , Child , Child, Preschool , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Deletion , Genetic Predisposition to Disease , Humans , Ikaros Transcription Factor/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Patient Outcome Assessment , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Recurrence , Tumor Suppressor Proteins/metabolism
10.
J Nucl Med ; 58(2): 214-220, 2017 02.
Article in English | MEDLINE | ID: mdl-27493268

ABSTRACT

The antileukemic drug asparaginase, a key component in the treatment of acute lymphoblastic leukemia, acts by depleting asparagine from the blood. However, little is known about its pharmacokinetics, and mechanisms of therapy resistance are poorly understood. Here, we explored the in vivo biodistribution of radiolabeled asparaginase, using a combination of imaging and biochemical techniques, and provide evidence for tissue-specific clearance mechanisms, which could reduce the effectiveness of the drug at these specific sites. METHODS: In vivo localization of 111In-labeled Escherichia coli asparaginase was performed in C57BL/6 mice by both small-animal SPECT/CT and ex vivo biodistribution studies. Mice were treated with liposomal clodronate to investigate the effect of macrophage depletion on tracer localization and drug clearance in vivo. Moreover, macrophage cell line models RAW264.7 and THP-1, as well as knockout mice, were used to identify the cellular and molecular components controlling asparaginase pharmacokinetics. RESULTS: In vivo imaging and biodistribution studies showed a rapid accumulation of asparaginase in macrophage-rich tissues such as the liver, spleen, and in particular bone marrow. Clodronate-mediated depletion of phagocytic cells markedly prolonged the serum half-life of asparaginase in vivo and decreased drug uptake in these macrophage-rich organs. Immunohistochemistry and in vitro binding assays confirmed the involvement of macrophagelike cells in the uptake of asparaginase. We identified the activity of the lysosomal protease cathepsin B in macrophages as a rate-limiting factor in degrading asparaginase both in vitro and in vivo. CONCLUSION: We showed that asparaginase is rapidly cleared from the serum by liver-, spleen-, and bone marrow-resident phagocytic cells. As a consequence of this efficient uptake and protease-mediated degradation, particularly bone marrow-resident macrophages may provide a protective niche to leukemic cells.


Subject(s)
Asparaginase/pharmacokinetics , Bone Marrow/enzymology , Macrophages/enzymology , Molecular Imaging/methods , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Antineoplastic Agents/pharmacokinetics , Bone Marrow/diagnostic imaging , Cell Line , Female , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Reproducibility of Results , Sensitivity and Specificity
11.
Oncotarget ; 7(3): 3128-43, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657730

ABSTRACT

Cancer cells are frequently exposed to physiological stress conditions such as hypoxia and nutrient limitation. Escape from stress-induced apoptosis is one of the mechanisms used by malignant cells to survive unfavorable conditions. B-cell Translocation Gene 1 (BTG1) is a tumor suppressor that is frequently deleted in acute lymphoblastic leukemia and recurrently mutated in diffuse large B cell lymphoma. Moreover, low BTG1 expression levels have been linked to poor outcome in several solid tumors. How loss of BTG1 function contributes to tumor progression is not well understood. Here, using Btg1 knockout mice, we demonstrate that loss of Btg1 provides a survival advantage to primary mouse embryonic fibroblasts (MEFs) under stress conditions. This pro-survival effect involves regulation of Activating Transcription Factor 4 (ATF4), a key mediator of cellular stress responses. We show that BTG1 interacts with ATF4 and positively modulates its activity by recruiting the protein arginine methyl transferase PRMT1 to methylate ATF4 on arginine residue 239. We further extend these findings to B-cell progenitors, by showing that loss of Btg1 expression enhances stress adaptation of mouse bone marrow-derived B cell progenitors. In conclusion, we have identified the BTG1/PRMT1 complex as a new modifier of ATF4 mediated stress responses.


Subject(s)
Activating Transcription Factor 4/metabolism , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Stress, Physiological/physiology , Animals , Apoptosis/physiology , B-Lymphocytes/cytology , Cell Line, Tumor , Fibroblasts , Humans , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
13.
PLoS Genet ; 8(2): e1002533, 2012.
Article in English | MEDLINE | ID: mdl-22359517

ABSTRACT

Recurrent submicroscopic deletions in genes affecting key cellular pathways are a hallmark of pediatric acute lymphoblastic leukemia (ALL). To gain more insight into the mechanism underlying these deletions, we have studied the occurrence and nature of abnormalities in one of these genes, the B-cell translocation gene 1 (BTG1), in a large cohort of pediatric ALL cases. BTG1 was found to be exclusively affected by genomic deletions, which were detected in 65 out of 722 B-cell precursor ALL (BCP-ALL) patient samples (9%), but not in 109 T-ALL cases. Eight different deletion sizes were identified, which all clustered at the telomeric site in a hotspot region within the second (and last) exon of the BTG1 gene, resulting in the expression of truncated BTG1 read-through transcripts. The presence of V(D)J recombination signal sequences at both sites of virtually all deletions strongly suggests illegitimate RAG1/RAG2-mediated recombination as the responsible mechanism. Moreover, high levels of histone H3 lysine 4 trimethylation (H3K4me3), which is known to tether the RAG enzyme complex to DNA, were found within the BTG1 gene body in BCP-ALL cells, but not T-ALL cells. BTG1 deletions were rarely found in hyperdiploid BCP-ALLs, but were predominant in other cytogenetic subgroups, including the ETV6-RUNX1 and BCR-ABL1 positive BCP-ALL subgroups. Through sensitive PCR-based screening, we identified multiple additional BTG1 deletions at the subclonal level in BCP-ALL, with equal cytogenetic distribution which, in some cases, grew out into the major clone at relapse. Taken together, our results indicate that BTG1 deletions may act as "drivers" of leukemogenesis in specific BCP-ALL subgroups, in which they can arise independently in multiple subclones at sites that are prone to aberrant RAG1/RAG2-mediated recombination events. These findings provide further evidence for a complex and multiclonal evolution of ALL.


Subject(s)
Clonal Evolution , Gene Deletion , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Acute Disease , Child , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology
14.
Blood ; 110(9): 3128-35, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17646546

ABSTRACT

Growth factor independence 1 (Gfi1) is a transcriptional repressor essential for the function and development of many different hematopoietic lineages. The Gfi1 protein expression is regulated by the ubiquitin-proteasome system. In granulocytes, Gfi1 is rapidly degraded by the proteasome, while it is more stable in monocytes. How the ubiquitination and degradation of Gfi1 is regulated is unclear. Here, we show that the ubiquitin ligase Triad1 interacts with the DNA-binding domain of Gfi1. Unexpectedly, we found that Triad1 inhibited Gfi1 ubiquitination, resulting in a prolonged half-life. Down-regulation of endogenous Triad1 by siRNAs resulted in increased Gfi1 ubiquitination. In U937 cells, Triad1 caused an increase in endogenous Gfi1 protein levels and slowed cell proliferation in a similar manner when Gfi1 itself was expressed. A Triad1 mutant that lacks the Gfi1-binding domain did not affect Gfi1 levels and proliferation. Because neither proteasome-ubiquitin nor Triad1 ubiquitin ligase activity was required for the inhibition of Gfi1 ubiquitination, these data suggest that Triad1 competes for Gfi1 binding with as yet to be identified E3 ubiquitin ligases that do mark Gfi1 for proteasomal degradation. The fine-tuning of Gfi1 protein levels regulated by Triad1 defines an unexpected role for this protein in hematopoiesis.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Animals , COS Cells , Chlorocebus aethiops , Hematopoiesis/physiology , Humans , Protein Binding , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Zinc Fingers
15.
Blood ; 109(1): 100-8, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16888099

ABSTRACT

Gfi1 is a transcriptional repressor essential during myeloid differentiation. Gfi1-/- mice exhibit a block in myeloid differentiation resulting in the accumulation of an immature myelo-monocytic cell population and the complete absence of mature neutrophils. Even though mRNA levels of Gfi1 appear to be very low in monocytes, Gfi1 might play a role in the monocytic lineage as Gfi1-/- mice exhibit diminished monocyte-derived dendritic cells and disturbed cytokine production by macrophages in response to LPS. We show here that Gfi1 protein levels are mainly regulated by the ubiquitin-proteasome system. Upon forced monocytic differentiation of U937 cells, Gfi1 mRNA levels dropped but protein levels increased due to diminished proteasomal turnover. Similarly, Gfi1 mRNA levels are low in primary monocytes whereas the protein is clearly detectable. Conversely, Gfi1 mRNA levels are high in granulocytes but the protein is swiftly degraded by the proteasome in these cells. Chromatin immunoprecipitation experiments showed that Gfi1 binds to the promoter of several granulocyte-specific genes in primary monocytes, including C/EBPalpha, neutrophil elastase, and Gfi1 itself. The binding of the repressor Gfi1 to these promoters correlated with low expression of these genes in monocytes compared with granulocytes. Our data fit a model in which Gfi1 protein levels are induced in primary monocytes, due to diminished proteasomal degradation, to repress genes that play a role in granulocytic differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Monocytes/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , CCAAT-Enhancer-Binding Protein-alpha/genetics , COS Cells , Cell Differentiation/drug effects , Cell Line , Cell Lineage , Chlorocebus aethiops , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Granulocytes/metabolism , HL-60 Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Kidney , Leukocyte Elastase/biosynthesis , Leukocyte Elastase/genetics , Mice , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , U937 Cells/drug effects , U937 Cells/metabolism , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...