Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 30(10): 2140-2159, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38376926

ABSTRACT

PURPOSE: The multi-kinase inhibitor (mKi) regorafenib has demonstrated efficacy in chemorefractory patients with metastatic colorectal cancer (mCRC). However, lack of predictive biomarkers and concerns over significant toxicities hamper the use of regorafenib in clinical practice. EXPERIMENTAL DESIGN: Serial liquid biopsies were obtained at baseline and monthly until disease progression in chemorefractory patients with mCRC treated with regorafenib in a phase II clinical trial (PROSPECT-R n = 40; NCT03010722) and in a multicentric validation cohort (n = 241). Tissue biopsies collected at baseline, after 2 months and at progression in the PROSPECT-R trial were used to establish patient-derived organoids (PDO) and for molecular analyses. MicroRNA profiling was performed on baseline bloods using the NanoString nCounter platform and results were validated by digital-droplet PCR and/or ISH in paired liquid and tissue biopsies. PDOs co-cultures and PDO-xenotransplants were generated for functional analyses. RESULTS: Large-scale microRNA expression analysis in longitudinal matched liquid and tissue biopsies from the PROSPECT-R trial identified MIR652-3p as a biomarker of clinical benefit to regorafenib. These findings were confirmed in an independent validation cohort and in a "control" group of 100 patients treated with lonsurf. Using ex vivo co-culture assays paired with single-cell RNA-sequencing of PDO established pre- and post-treatment, we modeled regorafenib response observed in vivo and in patients, and showed that MIR652-3p controls resistance to regorafenib by impairing regorafenib-induced lethal autophagy and by orchestrating the switch from neo-angiogenesis to vessel co-option. CONCLUSIONS: Our results identify MIR652-3p as a potential biomarker and as a driver of cell and non-cell-autonomous mechanisms of resistance to regorafenib.


Subject(s)
Biomarkers, Tumor , Circulating MicroRNA , Colorectal Neoplasms , Drug Resistance, Neoplasm , Phenylurea Compounds , Pyridines , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/blood , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Pyridines/therapeutic use , Pyridines/pharmacology , Drug Resistance, Neoplasm/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Animals , Female , Prospective Studies , Male , Mice , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic/drug effects , Aged , Liquid Biopsy/methods , Middle Aged , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/blood
2.
Cancer Gene Ther ; 29(7): 940-950, 2022 07.
Article in English | MEDLINE | ID: mdl-34522028

ABSTRACT

Deregulated polyamine biosynthesis is emerging as a common feature of neuroblastoma and drugs targeting this metabolic pathway such as DFMO are in clinical and preclinical development. The polyamine analog verlindamycin inhibits the polyamine biosynthesis pathway enzymes SMOX and PAOX, as well as the histone demethylase LSD1. Based on our previous research in acute myeloid leukemia (AML), we reasoned verlindamycin may also unblock neuroblastoma differentiation when combined with all-trans-retinoic acid (ATRA). Indeed, co-treatment with verlindamycin and ATRA strongly induced differentiation regardless of MYCN status, but in MYCN-expressing cells, protein levels were strongly diminished. This process was not transcriptionally regulated but was due to increased degradation of MYCN protein, at least in part via ubiquitin-independent, proteasome-dependent destruction. Here we report that verlindamycin effectively induces the expression of functional tumor suppressor-antizyme via ribosomal frameshifting. Consistent with previous results describing the function of antizyme, we found that verlindamycin treatment led to the selective targeting of ornithine decarboxylase (the rate-limiting enzyme for polyamine biosynthesis) as well as key oncoproteins, such as cyclin D and Aurora A kinase. Retinoid-based multimodal differentiation therapy is one of the few interventions that extends relapse-free survival in MYCN-associated high-risk neuroblastoma and these results point toward the potential use of verlindamycin in this regimen.


Subject(s)
Biguanides , Neuroblastoma , Biguanides/therapeutic use , Humans , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/therapeutic use , Polyamines/metabolism , Polyamines/therapeutic use
3.
Cancer Discov ; 10(7): 942-963, 2020 07.
Article in English | MEDLINE | ID: mdl-32238360

ABSTRACT

Infant high-grade gliomas appear clinically distinct from their counterparts in older children, indicating that histopathologic grading may not accurately reflect the biology of these tumors. We have collected 241 cases under 4 years of age, and carried out histologic review, methylation profiling, and custom panel, genome, or exome sequencing. After excluding tumors representing other established entities or subgroups, we identified 130 cases to be part of an "intrinsic" spectrum of disease specific to the infant population. These included those with targetable MAPK alterations, and a large proportion of remaining cases harboring gene fusions targeting ALK (n = 31), NTRK1/2/3 (n = 21), ROS1 (n = 9), and MET (n = 4) as their driving alterations, with evidence of efficacy of targeted agents in the clinic. These data strongly support the concept that infant gliomas require a change in diagnostic practice and management. SIGNIFICANCE: Infant high-grade gliomas in the cerebral hemispheres comprise novel subgroups, with a prevalence of ALK, NTRK1/2/3, ROS1, or MET gene fusions. Kinase fusion-positive tumors have better outcome and respond to targeted therapy clinically. Other subgroups have poor outcome, with fusion-negative cases possibly representing an epigenetically driven pluripotent stem cell phenotype.See related commentary by Szulzewsky and Cimino, p. 904.This article is highlighted in the In This Issue feature, p. 890.


Subject(s)
Gene Fusion/genetics , Glioma/genetics , Humans , Infant , Neoplasm Grading , Prognosis , Treatment Outcome
4.
Autophagy ; 16(6): 1044-1060, 2020 06.
Article in English | MEDLINE | ID: mdl-31517566

ABSTRACT

Macroautophagy/autophagy can enable cancer cells to withstand cellular stress and maintain bioenergetic homeostasis by sequestering cellular components into newly formed double-membrane vesicles destined for lysosomal degradation, potentially affecting the efficacy of anti-cancer treatments. Using 13C-labeled choline and 13C-magnetic resonance spectroscopy and western blotting, we show increased de novo choline phospholipid (ChoPL) production and activation of PCYT1A (phosphate cytidylyltransferase 1, choline, alpha), the rate-limiting enzyme of phosphatidylcholine (PtdCho) synthesis, during autophagy. We also discovered that the loss of PCYT1A activity results in compromised autophagosome formation and maintenance in autophagic cells. Direct tracing of ChoPLs with fluorescence and immunogold labeling imaging revealed the incorporation of newly synthesized ChoPLs into autophagosomal membranes, endoplasmic reticulum (ER) and mitochondria during anticancer drug-induced autophagy. Significant increase in the colocalization of fluorescence signals from the newly synthesized ChoPLs and mCherry-MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) was also found on autophagosomes accumulating in cells treated with autophagy-modulating compounds. Interestingly, cells undergoing active autophagy had an altered ChoPL profile, with longer and more unsaturated fatty acid/alcohol chains detected. Our data suggest that de novo synthesis may be required to increase autophagosomal ChoPL content and alter its composition, together with replacing phospholipids consumed from other organelles during autophagosome formation and turnover. This addiction to de novo ChoPL synthesis and the critical role of PCYT1A may lead to development of agents targeting autophagy-induced drug resistance. In addition, fluorescence imaging of choline phospholipids could provide a useful way to visualize autophagosomes in cells and tissues. ABBREVIATIONS: AKT: AKT serine/threonine kinase; BAX: BCL2 associated X, apoptosis regulator; BECN1: beclin 1; ChoPL: choline phospholipid; CHKA: choline kinase alpha; CHPT1: choline phosphotransferase 1; CTCF: corrected total cell fluorescence; CTP: cytidine-5'-triphosphate; DCA: dichloroacetate; DMEM: dulbeccos modified Eagles medium; DMSO: dimethyl sulfoxide; EDTA: ethylenediaminetetraacetic acid; ER: endoplasmic reticulum; GDPD5: glycerophosphodiester phosphodiesterase domain containing 5; GFP: green fluorescent protein; GPC: glycerophosphorylcholine; HBSS: hanks balances salt solution; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; LPCAT1: lysophosphatidylcholine acyltransferase 1; LysoPtdCho: lysophosphatidylcholine; MRS: magnetic resonance spectroscopy; MTORC1: mechanistic target of rapamycin kinase complex 1; PCho: phosphocholine; PCYT: choline phosphate cytidylyltransferase; PLA2: phospholipase A2; PLB: phospholipase B; PLC: phospholipase C; PLD: phospholipase D; PCYT1A: phosphate cytidylyltransferase 1, choline, alpha; PI3K: phosphoinositide-3-kinase; pMAFs: pancreatic mouse adult fibroblasts; PNPLA6: patatin like phospholipase domain containing 6; Pro-Cho: propargylcholine; Pro-ChoPLs: propargylcholine phospholipids; PtdCho: phosphatidylcholine; PtdEth: phosphatidylethanolamine; PtdIns3P: phosphatidylinositol-3-phosphate; RPS6: ribosomal protein S6; SCD: stearoyl-CoA desaturase; SEM: standard error of the mean; SM: sphingomyelin; SMPD1/SMase: sphingomyelin phosphodiesterase 1, acid lysosomal; SGMS: sphingomyelin synthase; WT: wild-type.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagosomes/enzymology , Autophagosomes/metabolism , Choline-Phosphate Cytidylyltransferase/metabolism , Furans/pharmacology , Macroautophagy , Phosphatidylcholines/biosynthesis , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Autophagosomes/drug effects , Autophagosomes/ultrastructure , CHO Cells , Cell Line, Tumor , Choline/metabolism , Choline-Phosphate Cytidylyltransferase/genetics , Cricetulus , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Macroautophagy/drug effects , Magnetic Resonance Spectroscopy , Mass Spectrometry , Metabolomics , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Vacuoles/drug effects , Vacuoles/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
Dis Model Mech ; 9(12): 1483-1495, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27799148

ABSTRACT

Histone deacetylase 9 (HDAC9) is expressed in B cells, and its overexpression has been observed in B-lymphoproliferative disorders, including B-cell non-Hodgkin lymphoma (B-NHL). We examined HDAC9 protein expression and copy number alterations in primary B-NHL samples, identifying high HDAC9 expression among various lymphoma entities and HDAC9 copy number gains in 50% of diffuse large B-cell lymphoma (DLBCL). To study the role of HDAC9 in lymphomagenesis, we generated a genetically engineered mouse (GEM) model that constitutively expressed an HDAC9 transgene throughout B-cell development under the control of the immunoglobulin heavy chain (IgH) enhancer (Eµ). Here, we report that the Eµ-HDAC9 GEM model develops splenic marginal zone lymphoma and lymphoproliferative disease (LPD) with progression towards aggressive DLBCL, with gene expression profiling supporting a germinal center cell origin, as is also seen in human B-NHL tumors. Analysis of Eµ-HDAC9 tumors suggested that HDAC9 might contribute to lymphomagenesis by altering pathways involved in growth and survival, as well as modulating BCL6 activity and p53 tumor suppressor function. Epigenetic modifications play an important role in the germinal center response, and deregulation of the B-cell epigenome as a consequence of mutations and other genomic aberrations are being increasingly recognized as important steps in the pathogenesis of a variety of B-cell lymphomas. A thorough mechanistic understanding of these alterations will inform the use of targeted therapies for these malignancies. These findings strongly suggest a role for HDAC9 in B-NHL and establish a novel GEM model for the study of lymphomagenesis and, potentially, preclinical testing of therapeutic approaches based on histone deacetylase inhibitors.


Subject(s)
B-Lymphocytes/enzymology , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/enzymology , Lymphoproliferative Disorders/genetics , Repressor Proteins/genetics , Acetylation , Animals , B-Lymphocytes/pathology , Cell Cycle/genetics , Gene Expression Profiling , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , HeLa Cells , Histone Deacetylases/metabolism , Humans , Lymphoma, B-Cell/pathology , Lymphoproliferative Disorders/pathology , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Oncotarget ; 6(33): 34087-105, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460951

ABSTRACT

Triple negative breast cancer (TNBC) is characterized by a poorly differentiated phenotype and limited treatment options. Aberrant epigenetics in this subtype represent a potential therapeutic opportunity, but a better understanding of the mechanisms contributing to the TNBC pathogenesis is required. The SIN3 molecular scaffold performs a critical role in multiple cellular processes, including epigenetic regulation, and has been identified as a potential therapeutic target. Using a competitive peptide corresponding to the SIN3 interaction domain of MAD (Tat-SID), we investigated the functional consequences of selectively blocking the paired amphipathic α-helix (PAH2) domain of SIN3. Here, we report the identification of the SID-containing adaptor PF1 as a factor required for maintenance of the TNBC stem cell phenotype and epithelial-to-mesenchymal transition (EMT). Tat-SID peptide blocked the interaction between SIN3A and PF1, leading to epigenetic modulation and transcriptional downregulation of TNBC stem cell and EMT markers. Importantly, Tat-SID treatment also led to a reduction in primary tumor growth and disseminated metastatic disease in vivo. In support of these findings, knockdown of PF1 expression phenocopied treatment with Tat-SID both in vitro and in vivo. These results demonstrate a critical role for a complex containing SIN3A and PF1 in TNBC and provide a rational for its therapeutic targeting.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Epithelial-Mesenchymal Transition/physiology , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/pathology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Female , Homeodomain Proteins/genetics , Humans , Mice , Protein Structure, Tertiary , Sin3 Histone Deacetylase and Corepressor Complex , Spheroids, Cellular , Transcription Factors/genetics , Tumor Cells, Cultured
7.
Mol Cancer Ther ; 14(8): 1824-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26078298

ABSTRACT

Triple-negative breast cancers (TNBC) lacking estrogen, progesterone, and HER2 receptors account for 10% to 20% of breast cancer and are indicative of poor prognosis. The development of effective treatment strategies therefore represents a pressing unmet clinical need. We previously identified a molecularly targeted approach to target aberrant epigenetics of TNBC using a peptide corresponding to the SIN3 interaction domain (SID) of MAD. SID peptide selectively blocked binding of SID-containing proteins to the paired α-helix (PAH2) domain of SIN3, resulting in epigenetic and transcriptional modulation of genes associated with epithelial-mesenchymal transition (EMT). To find small molecule inhibitor (SMI) mimetics of SID peptide, we performed an in silico screen for PAH2 domain-binding compounds. This led to the identification of the avermectin macrocyclic lactone derivatives selamectin and ivermectin (Mectizan) as candidate compounds. Both selamectin and ivermectin phenocopied the effects of SID peptide to block SIN3-PAH2 interaction with MAD, induce expression of CDH1 and ESR1, and restore tamoxifen sensitivity in MDA-MB-231 human and MMTV-Myc mouse TNBC cells in vitro. Treatment with selamectin or ivermectin led to transcriptional modulation of genes associated with EMT and maintenance of a cancer stem cell phenotype in TNBC cells. This resulted in impairment of clonogenic self-renewal in vitro and inhibition of tumor growth and metastasis in vivo. Underlining the potential of avermectins in TNBC, pathway analysis revealed that selamectin also modulated the expression of therapeutically targetable genes. Consistent with this, an unbiased drug screen in TNBC cells identified selamectin-induced sensitization to a number of drugs, including those targeting modulated genes.


Subject(s)
Ivermectin/analogs & derivatives , Repressor Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Animals , Antigens, CD , Antiparasitic Agents/pharmacology , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ivermectin/chemistry , Ivermectin/pharmacology , Mice , Models, Molecular , Molecular Conformation , Protein Interaction Domains and Motifs , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
PLoS One ; 10(3): e0119834, 2015.
Article in English | MEDLINE | ID: mdl-25785590

ABSTRACT

Medulloblastoma (MB) is the most common malignant pediatric brain tumor. While the pathways that are deregulated in MB remain to be fully characterized, amplification and/or overexpression of the MYCN gene, which is has a critical role in cerebellar development as a regulator of neural progenitor cell fate, has been identified in several MB subgroups. Phenotypically, aberrant expression of MYCN is associated with the large-cell/anaplastic MB variant, which accounts for 5-15% of cases and is associated with aggressive disease and poor clinical outcome. To better understand the role of MYCN in MB in vitro and in vivo and to aid the development of MYCN-targeted therapeutics we established tumor-derived neurosphere cell lines from the GTML (Glt1-tTA/TRE-MYCN-Luc) genetically engineered mouse model. A fraction of GTML neurospheres were found to be growth factor independent, expressed CD133 (a marker of neural stem cells), failed to differentiate upon MYCN withdrawal and were highly tumorigenic when orthotopically implanted into the cerebellum. Principal component analyzes using single cell RNA assay data suggested that the clinical candidate aurora-A kinase inhibitor MLN8237 converts GTML neurospheres to resemble non-MYCN expressors. Correlating with this, MLN8237 significantly extended the survival of mice bearing GTML MB allografts. In summary, our results demonstrate that MYCN plays a critical role in expansion and survival of aggressive MB-propagating cells, and establish GTML neurospheres as an important resource for the development of novel therapeutic strategies.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellum/pathology , Medulloblastoma/pathology , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , Proto-Oncogene Proteins/genetics , Allografts , Animals , Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Azepines/pharmacology , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/mortality , Cerebellum/drug effects , Cerebellum/metabolism , Female , Gene Expression , Humans , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/mortality , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Principal Component Analysis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis
9.
Cancer Cell ; 27(1): 72-84, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25533335

ABSTRACT

We undertook a comprehensive clinical and biological investigation of serial medulloblastoma biopsies obtained at diagnosis and relapse. Combined MYC family amplifications and P53 pathway defects commonly emerged at relapse, and all patients in this group died of rapidly progressive disease postrelapse. To study this interaction, we investigated a transgenic model of MYCN-driven medulloblastoma and found spontaneous development of Trp53 inactivating mutations. Abrogation of p53 function in this model produced aggressive tumors that mimicked characteristics of relapsed human tumors with combined P53-MYC dysfunction. Restoration of p53 activity and genetic and therapeutic suppression of MYCN all reduced tumor growth and prolonged survival. Our findings identify P53-MYC interactions at medulloblastoma relapse as biomarkers of clinically aggressive disease that may be targeted therapeutically.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Animals , Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Gene Amplification , Humans , Infant , Male , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Mice , Molecular Sequence Data , Mutation , N-Myc Proto-Oncogene Protein , Neoplasm Recurrence, Local/drug therapy , Neoplasms, Experimental , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Young Adult
10.
Mol Cancer Res ; 12(12): 1796-806, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25061103

ABSTRACT

UNLABELLED: Although the ETV6-RUNX1 fusion is a frequent initiating event in childhood leukemia, its role in leukemogenesis is only partly understood. The main impact of the fusion itself is to generate and sustain a clone of clinically silent preleukemic B-cell progenitors (BCP). Additional oncogenic hits, occurring even several years later, are required for overt disease. The understanding of the features and interactions of ETV6-RUNX1-positive cells during this "latency" period may explain how these silent cells can persist and whether they could be prone to additional genetic changes. In this study, two in vitro murine models were used to investigate whether ETV6-RUNX1 alters the cellular adhesion and migration properties of BCP. ETV6-RUNX1-expressing cells showed a significant defect in the chemotactic response to CXCL12, caused by a block in CXCR4 signaling, as demonstrated by inhibition of CXCL12-associated calcium flux and lack of ERK phosphorylation. Moreover, the induction of ETV6-RUNX1 caused changes in the expression of cell-surface adhesion molecules. The expression of genes regulating the cytoskeleton was also affected, resulting in a block of CDC42 signaling. The abnormalities described here could alter the interaction of ETV6-RUNX1 preleukemic BCP with the microenvironment and contribute to the pathogenesis of the disease. IMPLICATIONS: Alterations in the expression of cytoskeletal regulatory genes and migration properties of BCP represent early events in the evolution of the disease, from the preleukemic phase to the clinical onset, and suggest new strategies for effective eradication of leukemia.


Subject(s)
Chemokine CXCL12/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Oncogene Proteins, Fusion/metabolism , Precursor Cells, B-Lymphoid/cytology , Receptors, CXCR4/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Mice , Models, Biological , Oncogene Proteins, Fusion/genetics , Precursor Cells, B-Lymphoid/metabolism , Signal Transduction
11.
Nat Med ; 18(4): 605-11, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406747

ABSTRACT

Acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML), characterized by the t(15;17)-associated PML-RARA fusion, has been successfully treated with therapy utilizing all-trans-retinoic acid (ATRA) to differentiate leukemic blasts. However, among patients with non-APL AML, ATRA-based treatment has not been effective. Here we show that, through epigenetic reprogramming, inhibitors of lysine-specific demethylase 1 (LSD1, also called KDM1A), including tranylcypromine (TCP), unlocked the ATRA-driven therapeutic response in non-APL AML. LSD1 inhibition did not lead to a large-scale increase in histone 3 Lys4 dimethylation (H3K4(me2)) across the genome, but it did increase H3K4(me2) and expression of myeloid-differentiation-associated genes. Notably, treatment with ATRA plus TCP markedly diminished the engraftment of primary human AML cells in vivo in nonobese diabetic (NOD)-severe combined immunodeficient (SCID) mice, suggesting that ATRA in combination with TCP may target leukemia-initiating cells. Furthermore, initiation of ATRA plus TCP treatment 15 d after engraftment of human AML cells in NOD-SCID γ (with interleukin-2 (IL-2) receptor γ chain deficiency) mice also revealed the ATRA plus TCP drug combination to have a potent anti-leukemic effect that was superior to treatment with either drug alone. These data identify LSD1 as a therapeutic target and strongly suggest that it may contribute to AML pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML.


Subject(s)
Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Tretinoin/therapeutic use , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Apoptosis/physiology , CD11b Antigen/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Drug Interactions , Enzyme Inhibitors/therapeutic use , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Histone Demethylases/metabolism , Humans , Lysine/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/metabolism , Stem Cell Factor/metabolism , Time Factors , Transplants , Tranylcypromine/therapeutic use , Tretinoin/pharmacology
12.
Genes Dev ; 23(17): 2076-87, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19723763

ABSTRACT

A major question in hematopoiesis is how the system maintains long-term homeostasis whereby the generation of large numbers of differentiated cells is balanced with the requirement for maintenance of progenitor pools, while remaining sufficiently flexible to respond to periods of perturbed cellular output during infection or stress. We focused on the development of the myeloid lineage and present evidence that promyelocytic leukemia zinc finger (PLZF) provides a novel function that is critical for both normal and stress-induced myelopoiesis. During homeostasis, PLZF restricts proliferation and differentiation of human cord blood-derived myeloid progenitors to maintain a balance between the progenitor and mature cell compartments. Analysis of PLZF promoter-binding sites revealed that it represses transcription factors involved in normal myeloid differentiation, including GFI-1, C/EBPalpha, and LEF-1, and induces negative regulators DUSP6 and ID2. Loss of ID2 relieves PLZF-mediated repression of differentiation identifying it as a functional target of PLZF in myelopoiesis. Furthermore, induction of ERK1/2 by myeloid cytokines, reflective of a stress response, leads to nuclear export and inactivation of PLZF, which augments mature cell production. Thus, negative regulators of differentiation can serve to maintain developmental systems in a primed state, so that their inactivation by extrinsic signals can induce proliferation and differentiation to rapidly satisfy increased demand for mature cells.


Subject(s)
Cell Differentiation , Cytokines/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis/physiology , Myelopoiesis/physiology , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Fetal Blood/cytology , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Transcription Factors/metabolism
13.
Mol Cell Biol ; 25(13): 5552-66, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15964811

ABSTRACT

Histone acetyltransferase (HAT) activities of proteins such as p300, CBP, and P/CAF play important roles in activation of gene expression. We now show that the HAT activity of p300 can also be required for down-regulation of transcription by a DNA binding repressor protein. Promyelocytic leukemia zinc finger (PLZF), originally identified as a fusion with retinoic acid receptor alpha in rare cases of all-trans-retinoic acid-resistant acute promyelocytic leukemia, is a transcriptional repressor that recruits histone deacetylase-containing corepressor complexes to specific DNA binding sites. PLZF associates with p300 in vivo, and its ability to repress transcription is specifically dependent on HAT activity of p300 and acetylation of lysines in its C-terminal C2-H2 zinc finger motif. An acetylation site mutant of PLZF does not repress transcription and is functionally deficient in a colony suppression assay despite retaining its abilities to interact with corepressor/histone deacetylase complexes. This is due to the fact that acetylation of PLZF activates its ability to bind specific DNA sequences both in vitro and in vivo. Taken together, our results indicate that a histone deacetylase-dependent transcriptional repressor can be positively regulated through acetylation and point to an unexpected role of a coactivator protein in transcriptional repression.


Subject(s)
Acetyltransferases/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Acetylation , Acetyltransferases/analysis , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/genetics , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Direct , Fluorescent Dyes , Gene Expression Regulation, Neoplastic , HeLa Cells , Histone Acetyltransferases , Humans , Kruppel-Like Transcription Factors , Leukemia, Promyelocytic, Acute/genetics , Microscopy, Confocal , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promyelocytic Leukemia Zinc Finger Protein , Repressor Proteins/chemistry , Repressor Proteins/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Zinc Fingers
14.
Oncogene ; 22(24): 3685-97, 2003 Jun 12.
Article in English | MEDLINE | ID: mdl-12802276

ABSTRACT

The PLZF gene is translocated in a subset of all-trans-retinoic acid resistant acute promyelocytic leukaemia (APL) cases, encodes a DNA binding transcription factor and is expressed highly in haematopoietic progenitor cells as well-developing central nervous system (CNS). The spatially restricted and temporally dynamic pattern of PLZF expression in the developing CNS suggested that it might play a role in the circuitry regulating hindbrain segmentation. We have now identified a PLZF binding site (PLZF-RE) in an enhancer region of Hoxb2 that itself is required for directing high-level expression in rhombomers 3 and 5 of the developing hindbrain. The wild-type r3/r5 enhancer linked to a heterologous promoter was responsive to regulation by PLZF, and this activity was lost in variants containing a mutated PLZF-RE. Compared with the wild-type protein, the binding of the APL-associated reciprocal RARalpha-PLZF fusion to PLZF-RE was much stronger, suggesting that the N-terminal PLZF sequences missing from the fusion may play a role in the regulation of DNA binding. Consistent with this, the N-terminal POZ domain was required for cooperative binding of PLZF to a multimerized PLZF-RE. In the context of the r3/r5 enhancer, the PLZF-RE cooperated for PLZF binding with an additional A/T-rich motif positioned downstream of the PLZF-RE. This A/T motif was previously shown to be essential for the regulation of Hoxb2 expression in r3 and r5 in cooperation with another Krüppel-like zinc finger protein Krox 20. The presence of both the PLZF-RE and the A/T-rich motif was required for a maximal effect of PLZF on a heterologous promoter and was essential in vivo to direct the expression of a lacZ reporter in the chick neural tube. Hence, both PLZF and Krox20 cooperate with a common A/T motif in mediating in vivo activity of the Hoxb2 enhancer. Our findings indicate that Hoxb2 is a direct target for regulation by PLZF in the developing CNS and suggest that deregulation of Hox gene expression may contribute to APL pathogenesis.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Binding Sites , Chick Embryo , DNA/metabolism , Enhancer Elements, Genetic , Hematopoiesis , Humans , Kruppel-Like Transcription Factors , Leukemia, Promyelocytic, Acute/etiology , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Retinoic Acid/physiology , Repressor Proteins/physiology , Retinoic Acid Receptor alpha , Rhombencephalon/embryology
15.
J Biol Chem ; 278(18): 16059-72, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12590135

ABSTRACT

Histone deacetylases (HDACs) perform an important function in transcriptional regulation by modifying the core histones of the nucleosome. We have now fully characterized a new member of the Class II HDAC family, HDAC9. The enzyme contains a conserved deacetylase domain, represses reporter activity when recruited to a promoter, and utilizes histones H3 and H4 as substrates in vitro and in vivo. HDAC9 is expressed in a tissue-specific pattern that partially overlaps that of HDAC4. Within the human hematopoietic system, expression of HDAC9 is biased toward cells of monocytic and lymphoid lineages. The HDAC9 gene encodes multiple protein isoforms, some of which display distinct cellular localization patterns. For example, full-length HDAC9 is localized in the nucleus, but the isoform lacking the region encoded by exon 7 is in the cytoplasm. HDAC9 interacts and co-localizes in vivo with a number of transcriptional repressors and co-repressors, including TEL and N-CoR, whose functions have been implicated in the pathogenesis of hematological malignancies. These results suggest that HDAC9 plays a role in hematopoiesis; its deregulated expression may be associated with some human cancers.


Subject(s)
Histone Deacetylases/genetics , Isoenzymes/genetics , Repressor Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Exons , Gene Expression Regulation, Enzymologic , Histone Deacetylases/physiology , Humans , Molecular Sequence Data , Neoplasms/enzymology , Organ Specificity , RNA, Messenger/analysis , Repressor Proteins/physiology , SUMO-1 Protein/metabolism , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...