Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biotechnol J ; 19(5): e2400090, 2024 May.
Article in English | MEDLINE | ID: mdl-38719592

ABSTRACT

The production of lentiviral vectors (LVs) pseudotyped with the vesicular stomatitis virus envelope glycoprotein (VSV-G) is limited by the associated cytotoxicity of the envelope and by the production methods used, such as transient transfection of adherent cell lines. In this study, we established stable suspension producer cell lines for scalable and serum-free LV production derived from two stable, inducible packaging cell lines, named GPRG and GPRTG. The established polyclonal producer cell lines produce self-inactivating (SIN) LVs carrying a WAS-T2A-GFP construct at an average infectious titer of up to 4.64 × 107 TU mL-1 in a semi-perfusion process in a shake flask and can be generated in less than two months. The derived monoclonal cell lines are functionally stable in continuous culture and produce an average infectious titer of up to 9.38 × 107 TU mL-1 in a semi-perfusion shake flask process. The producer clones are able to maintain a productivity of >1 × 107 TU mL-1 day-1 for up to 29 consecutive days in a non-optimized 5 L stirred-tank bioreactor perfusion process, representing a major milestone in the field of LV manufacturing. As the producer cell lines are based on an inducible Tet-off expression system, the established process allows LV production in the absence of inducers such as antibiotics. The purified LVs efficiently transduce human CD34+ cells, reducing the LV quantities required for gene and cell therapy applications.


Subject(s)
Bioreactors , Genetic Vectors , Lentivirus , Lentivirus/genetics , Humans , Genetic Vectors/genetics , Culture Media, Serum-Free , Cell Line , Cell Culture Techniques/methods , Virus Cultivation/methods , HEK293 Cells , Transfection/methods
2.
Biochem J ; 480(2): 161-176, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36719792

ABSTRACT

Chronic myeloid leukemia (CML) was considered for a long time one of the most hostile leukemia that was incurable for most of the patients, predominantly due to the extreme resistance to chemotherapy. Part of the resistance to cell death (apoptosis) is the result of increased levels of anti-apoptotic and decreased levels of pro-apoptotic member of the BCL-2 family induced by the BCR-ABL1 oncoprotein. BCR-ABL1 is a constitutively active tyrosine kinase responsible for initiating multiple and oncogenic signaling pathways. With the development of specific BCR-ABL1 tyrosine kinase inhibitors (TKIs) CML became a much more tractable disease. Nevertheless, TKIs do not cure CML patients and a substantial number of them develop intolerance or become resistant to the treatment. Therefore, novel anti-cancer strategies must be developed to treat CML patients independently or in combination with TKIs. Here, we will discuss the mechanisms of BCR-ABL1-dependent and -independent resistance to TKIs and the use of BH3-mimetics as a potential tool to fight CML.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein Kinase Inhibitors , Humans , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
3.
Pharmaceutics ; 14(1)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35057108

ABSTRACT

The constitutively active BCR-ABL1 tyrosine kinase, found in t(9;22)(q34;q11) chromosomal translocation-derived leukemia, initiates an extremely complex signaling transduction cascade that induces a strong state of resistance to chemotherapy. Targeted therapies based on tyrosine kinase inhibitors (TKIs), such as imatinib, dasatinib, nilotinib, bosutinib, and ponatinib, have revolutionized the treatment of BCR-ABL1-driven leukemia, particularly chronic myeloid leukemia (CML). However, TKIs do not cure CML patients, as some develop TKI resistance and the majority relapse upon withdrawal from treatment. Importantly, although BCR-ABL1 tyrosine kinase is necessary to initiate and establish the malignant phenotype of Ph-related leukemia, in the later advanced phase of the disease, BCR-ABL1-independent mechanisms are also in place. Here, we present an overview of the signaling pathways initiated by BCR-ABL1 and discuss the major challenges regarding immunologic/pharmacologic combined therapies.

5.
Blood Adv ; 4(20): 5062-5077, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33080008

ABSTRACT

The specific targeting of inhibitor of apoptosis (IAP) proteins by Smac-mimetic (SM) drugs, such as birinapant, has been tested in clinical trials of acute myeloid leukemia (AML) and certain solid cancers. Despite their promising safety profile, SMs have had variable and limited success. Using a library of more than 5700 bioactive compounds, we screened for approaches that could sensitize AML cells to birinapant and identified multidrug resistance protein 1 inhibitors (MDR1i) as a class of clinically approved drugs that can enhance the efficacy of SM therapy. Genetic or pharmacological inhibition of MDR1 increased intracellular levels of birinapant and sensitized AML cells from leukemia murine models, human leukemia cell lines, and primary AML samples to killing by birinapant. The combination of clinical MDR1 and IAP inhibitors was well tolerated in vivo and more effective against leukemic cells, compared with normal hematopoietic progenitors. Importantly, birinapant combined with third-generation MDR1i effectively killed murine leukemic stem cells (LSCs) and prolonged survival of AML-burdened mice, suggesting a therapeutic opportunity for AML. This study identified a drug combination strategy that, by efficiently killing LSCs, may have the potential to improve outcomes in patients with AML.


Subject(s)
Leukemia, Myeloid, Acute , Animals , Biological Availability , Dipeptides , Humans , Indoles , Inhibitor of Apoptosis Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Mice
6.
Viruses ; 12(8)2020 08 17.
Article in English | MEDLINE | ID: mdl-32824616

ABSTRACT

Chronic hepatitis B virus (HBV) infection remains a global health threat and affects hundreds of millions worldwide. Small molecule compounds that mimic natural antagonists of inhibitor of apoptosis (IAP) proteins, known as Smac-mimetics (second mitochondria-derived activator of caspases-mimetics), can promote the death of HBV-replicating liver cells and promote clearance of infection in preclinical models of HBV infection. The Smac-mimetic birinapant is a substrate of the multidrug resistance protein 1 (MDR1) efflux pump, and therefore inhibitors of MDR1 increase intracellular concentration of birinapant in MDR1 expressing cells. Liver cells are known to express MDR1 and other drug pump proteins. In this study, we investigated whether combining the clinical drugs, birinapant and the MDR1 inhibitor zosuquidar, increases the efficacy of birinapant in killing HBV expressing liver cells. We showed that this combination treatment is well tolerated and, compared to birinapant single agent, was more efficient at inducing death of HBV-positive liver cells and improving HBV-DNA and HBV surface antigen (HBsAg) control kinetics in an immunocompetent mouse model of HBV infection. Thus, this study identifies a novel and safe combinatorial treatment strategy to potentiate substantial reduction of HBV replication using an IAP antagonist.


Subject(s)
Antiviral Agents/therapeutic use , Dibenzocycloheptenes/therapeutic use , Dipeptides/therapeutic use , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Indoles/therapeutic use , Quinolines/therapeutic use , Virus Replication/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Disease Models, Animal , Drug Therapy, Combination , Hep G2 Cells , Hepatitis B virus/physiology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Liver/cytology , Liver/drug effects , Liver/virology , Male , Mice , Mice, Inbred C57BL
7.
Nat Commun ; 11(1): 3150, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561755

ABSTRACT

MLKL is the essential effector of necroptosis, a form of programmed lytic cell death. We have isolated a mouse strain with a single missense mutation, MlklD139V, that alters the two-helix 'brace' that connects the killer four-helix bundle and regulatory pseudokinase domains. This confers constitutive, RIPK3 independent killing activity to MLKL. Homozygous mutant mice develop lethal postnatal inflammation of the salivary glands and mediastinum. The normal embryonic development of MlklD139V homozygotes until birth, and the absence of any overt phenotype in heterozygotes provides important in vivo precedent for the capacity of cells to clear activated MLKL. These observations offer an important insight into the potential disease-modulating roles of three common human MLKL polymorphisms that encode amino acid substitutions within or adjacent to the brace region. Compound heterozygosity of these variants is found at up to 12-fold the expected frequency in patients that suffer from a pediatric autoinflammatory disease, chronic recurrent multifocal osteomyelitis (CRMO).


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic System/pathology , Necroptosis/genetics , Protein Kinases/genetics , Animals , Animals, Newborn , Hereditary Autoinflammatory Diseases , Humans , Inflammation/genetics , Mice , Mutation, Missense , Osteomyelitis/genetics , Protein Kinases/metabolism
8.
Cells ; 9(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-32053868

ABSTRACT

It is well accepted that the ability of cancer cells to circumvent the cell death program that untransformed cells are subject to helps promote tumor growth. Strategies designed to reinstate the cell death program in cancer cells have therefore been investigated for decades. Overexpression of members of the Inhibitor of APoptosis (IAP) protein family is one possible mechanism hindering the death of cancer cells. To promote cell death, drugs that mimic natural IAP antagonists, such as second mitochondria-derived activator of caspases (Smac/DIABLO) were developed. Smac-Mimetics (SMs) have entered clinical trials for hematological and solid cancers, unfortunately with variable and limited results so far. This review explores the use of SMs for the treatment of cancer, their potential to synergize with up-coming treatments and, finally, discusses the challenges and optimism facing this strategy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Inhibitor of Apoptosis Proteins/genetics , Mitochondrial Proteins/genetics , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/therapeutic use , Biomimetics , Cell Proliferation/drug effects , Clinical Trials as Topic , Humans , Mitochondrial Proteins/therapeutic use , Neoplasms/genetics , Neoplasms/pathology , Protein Binding
9.
Mol Cell ; 66(5): 698-710.e5, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28506461

ABSTRACT

TNF is an inflammatory cytokine that upon binding to its receptor, TNFR1, can drive cytokine production, cell survival, or cell death. TNFR1 stimulation causes activation of NF-κB, p38α, and its downstream effector kinase MK2, thereby promoting transcription, mRNA stabilization, and translation of target genes. Here we show that TNF-induced activation of MK2 results in global RIPK1 phosphorylation. MK2 directly phosphorylates RIPK1 at residue S321, which inhibits its ability to bind FADD/caspase-8 and induce RIPK1-kinase-dependent apoptosis and necroptosis. Consistently, a phospho-mimetic S321D RIPK1 mutation limits TNF-induced death. Mechanistically, we find that phosphorylation of S321 inhibits RIPK1 kinase activation. We further show that cytosolic RIPK1 contributes to complex-II-mediated cell death, independent of its recruitment to complex-I, suggesting that complex-II originates from both RIPK1 in complex-I and cytosolic RIPK1. Thus, MK2-mediated phosphorylation of RIPK1 serves as a checkpoint within the TNF signaling pathway that integrates cell survival and cytokine production.


Subject(s)
Apoptosis/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspase 8/metabolism , Dose-Response Relationship, Drug , Fas-Associated Death Domain Protein/metabolism , HT29 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 14/metabolism , Multiprotein Complexes , NF-kappa B/metabolism , Necrosis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Transfection
10.
Proteomics Clin Appl ; 11(9-10)2017 09.
Article in English | MEDLINE | ID: mdl-28447382

ABSTRACT

PURPOSE: Chromosomal translocation of the mixed lineage leukemia (MLL) locus generates fusion proteins that drive acute myeloid leukemia (AML) resulting in atypical histone methyltransferase activity and alterations in the epigenetic regulation of gene expression. Targeting histone regulators, such as Enhancer of Zeste Homologue 2 (EZH2), has shown promise in AML. Profiling differential protein expression following inhibition of epigenetic regulators in AML may help to identify novel targets for therapeutics. EXPERIMENTAL DESIGN: Murine models of AML combined with quantitative SILAC analysis were used to identify differentially expressed proteins following inhibition of EZH2 activity using 3-Deazaneplanocin A (DZnep). Western blotting and flow cytometry were used to validate a subset of differentially expressed proteins. Gene set analysis was used to determine changes to reported EZH2 target genes. RESULTS: Our quantitative proteomic analysis and subsequent validation of protein changes identified that epigenetic therapy leads to cell death preceded by the induction of differentiation with concurrent p53 up-regulation and cell cycle arrest. Gene set analysis revealed a specific subset of EZH2 target genes that were regulated by DZnep in AML. CONCLUSION AND CLINICAL RELEVANCE: These discoveries highlight how this new class of drugs affects AML cell biology and cell survival, and may help identify novel targets and strategies to increase treatment efficacy.


Subject(s)
Adenosine/analogs & derivatives , Cell Death/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Proteomics , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Carcinogenesis/drug effects , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mice
11.
Trends Mol Med ; 23(3): 264-281, 2017 03.
Article in English | MEDLINE | ID: mdl-28196625

ABSTRACT

Malignant cells must circumvent endogenous cell death pathways to survive and develop into cancers. Acquired cell death resistance also sets up malignant cells to survive anticancer therapies. Acute Myeloid Leukemia (AML) is an aggressive blood cancer characterized by high relapse rate and resistance to cytotoxic therapies. Recent collaborative profiling projects have led to a greater understanding of the 'fearful symmetry' of the genomic landscape of AML, and point to the development of novel potential therapies that can overcome factors linked to chemoresistance. We review here the most recent research in the genetics of AML and how these discoveries have led, or might lead, to therapies that specifically activate cell death pathways to substantially challenge this 'fearful' disease.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Leukemia, Myeloid, Acute/drug therapy , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , DNA Modification Methylases/antagonists & inhibitors , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy/methods , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
12.
Immunol Cell Biol ; 95(2): 137-145, 2017 02.
Article in English | MEDLINE | ID: mdl-27922620

ABSTRACT

Resistance to caspase-dependent apoptosis is often responsible for treatment failures in cancer. Finding novel therapeutic strategies that can activate alternative cell death programs appears to be appealing. Necroptosis is a form of programmed necrosis that occurs under caspase-deficient conditions. This alternative form of cell death has recently emerged as a potential anticancer therapy that could overcome apoptosis resistance. A growing understanding of the molecular events triggering necroptosis helped to examine its implication in cancer development and to define new therapeutic strategies. Genetic and proteomic analysis suggest that necroptosis is deregulated in many cancers. Various preclinical and clinical compounds induced necroptosis and have demonstrated significant therapeutic efficacy. Moreover, accumulating evidence has shown that necroptosis promotes anticancer immune response. A better knowledge of the cascade of events regulating necroptosis is expected to assess the feasibility of its therapeutic exploitation for cancer therapy.


Subject(s)
Apoptosis , Neoplasms/pathology , Animals , Carcinogenesis/pathology , Humans , Immunologic Surveillance , Models, Biological , Necrosis , Neoplasms/immunology , Neoplasms/therapy
13.
Blood Adv ; 1(15): 1067-1079, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-29296749

ABSTRACT

High expression of the α chain of the interleukin-3 receptor (IL-3Rα; CD123) is a hallmark of acute myeloid leukemia (AML) leukemic stem cells (LSCs). Elevated CD123 expression is part of the diagnostic immunophenotyping of myeloid leukemia, and higher expression is associated with poor prognosis. However, the biological basis of the poorer prognosis is unclear, and may include heightened IL-3 signaling and non-cell autonomous interactions with the bone marrow (BM) microenvironment. We used TF-1 cells expressing different levels of CD123 and found elevated CD123 levels amplified the proliferative response to exogenous IL-3 and maintained viability in reducing IL-3 concentrations. This was associated with stronger activation of STAT5, Akt, and extracellular signal-regulated kinase 1/2 in vitro. Surprisingly, in vivo e14.5 fetal liver cells transduced with retroviral constructs to express high CD123 failed to engraft in syngeneic recipients. In exploring the underlying mechanism for this, we found that CXCR4, a key molecule involved in LSC/BM interactions, was specifically downregulated in CD123 overexpressing cells in a manner dependent on IL-3 signaling. CXCR4 downregulation was sufficient to alter the chemotactic response of hematopoietic cells to stromal derived factor-1 (SDF-1). Thus, we propose that the overexpression of CD123 in AML LSC dictates their location by altering CXCR4/SDF-1 interaction in the BM, raising the possibility that this mechanism underpins the egress of BM AML LSC and more mature cells into the circulation.

15.
Sci Transl Med ; 8(339): 339ra69, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194727

ABSTRACT

Resistance to chemotherapy is a major problem in cancer treatment, and it is frequently associated with failure of tumor cells to undergo apoptosis. Birinapant, a clinical SMAC mimetic, had been designed to mimic the interaction between inhibitor of apoptosis proteins (IAPs) and SMAC/Diablo, thereby relieving IAP-mediated caspase inhibition and promoting apoptosis of cancer cells. We show that acute myeloid leukemia (AML) cells are sensitive to birinapant-induced death and that the clinical caspase inhibitor emricasan/IDN-6556 augments, rather than prevents, killing by birinapant. Deletion of caspase-8 sensitized AML to birinapant, whereas combined loss of caspase-8 and the necroptosis effector MLKL (mixed lineage kinase domain-like) prevented birinapant/IDN-6556-induced death, showing that inhibition of caspase-8 sensitizes AML cells to birinapant-induced necroptosis. However, loss of MLKL alone did not prevent a caspase-dependent birinapant/IDN-6556-induced death, implying that AML will be less likely to acquire resistance to this drug combination. A therapeutic breakthrough in AML has eluded researchers for decades. Demonstrated antileukemic efficacy and safety of the birinapant/emricasan combination in vivo suggest that induction of necroptosis warrants clinical investigation as a therapeutic opportunity in AML.


Subject(s)
Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Dipeptides/pharmacology , Indoles/pharmacology , Pentanoic Acids/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid, Acute/metabolism , Necrosis/metabolism , Tumor Cells, Cultured
16.
Methods Mol Biol ; 1419: 265-76, 2016.
Article in English | MEDLINE | ID: mdl-27108445

ABSTRACT

Ectopic expression of proteins involved in cell death pathways is an important tool to analyze their role during apoptosis or other forms of cell death. Lentiviral vectors offer the advantages of high rate of transduction and stable integration of donor DNA into the genome of the host cell, leading to reproducible and relevant readouts compared to classical overexpression by transfection of naked plasmid DNA.Here, we describe the production and application of lentiviral vectors to express cell death proteins in eukaryotic cells. A packaging cell line, usually HEK293T cells, is transfected with viral packaging plasmids and your gene of interest, which is flanked by long terminal repeat sites with an internal ribosome entry site in the 5'UTR (Un translated region). Virions are harvested from the supernatant and can be directly used to transduce target cells. Varied selection markers as well as a variety of promoters that regulate expression of the gene of interest make this system attractive for a wide range of application in many cell lines or in whole organisms.


Subject(s)
Cell Death/genetics , Genetic Vectors , Lentivirus/genetics , Transduction, Genetic/methods , Transfection/methods , Virion/physiology , HEK293 Cells , Humans , Viral Envelope Proteins/metabolism , Virus Assembly
17.
Cancer Cell ; 29(2): 145-58, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26859455

ABSTRACT

Birinapant is a smac-mimetic (SM) in clinical trials for treating cancer. SM antagonize inhibitor of apoptosis (IAP) proteins and simultaneously induce tumor necrosis factor (TNF) secretion to render cancers sensitive to TNF-induced killing. To enhance SM efficacy, we screened kinase inhibitors for their ability to increase TNF production of SM-treated cells. We showed that p38 inhibitors increased TNF induced by SM. Unexpectedly, even though p38 is required for Toll-like receptors to induce TNF, loss of p38 or its downstream kinase MK2 increased induction of TNF by SM. Hence, we show that the p38/MK2 axis can inhibit or promote TNF production, depending on the stimulus. Importantly, clinical p38 inhibitors overcame resistance of primary acute myeloid leukemia to birinapant.


Subject(s)
Antineoplastic Agents/therapeutic use , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Leukemia/drug therapy , Mitochondrial Proteins/physiology , Molecular Mimicry , Protein Serine-Threonine Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins , Humans , Mice , Tumor Necrosis Factor-alpha/biosynthesis
18.
J Leukoc Biol ; 96(1): 83-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24598054

ABSTRACT

Aberrant activation of ß-catenin is a common event in AML and is an independent predictor of poor prognosis. Although increased ß-catenin signaling in AML has been associated with oncogenic translocation products and activating mutations in the FLT3R, the mechanisms that activate ß-catenin in AML more broadly are still unclear. Here, we describe a novel link between IL-3 signaling and the regulation of ß-catenin in myeloid transformation and AML. In a murine model of HoxB8 and IL-3 cooperation, we show that ß-catenin protein levels are modulated by IL-3 and that Cre-induced deletion of ß-catenin abolishes IL-3-dependent growth and colony formation. In IL-3-dependent leukemic TF-1.8 cells, we observed increased ß-catenin protein levels and nuclear localization in response to IL-3, and this correlated with transcriptional induction of ß-catenin target genes. Furthermore, IL-3 promoted ß-catenin accumulation in a subset of AML patient samples, and gene-expression profiling of these cells revealed induction of WNT/ß-catenin and TCF4 gene signatures in an IL-3-dependent manner. This study is the first to link ß-catenin activation to IL-3 and suggests that targeting IL-3 signaling may be an effective approach for the inhibition of ß-catenin activity in some patients with AML.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Interleukin-3/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Leukemic/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interleukin-3/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Transcription Factor 4 , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics
19.
Oncotarget ; 4(11): 1933-47, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24177192

ABSTRACT

Deregulated expression of Hox genes such as HoxA9 is associated with development of myeloproliferative disorders and leukemia and indicates a poor prognosis. To investigate the molecular mechanisms by which HoxA9 promotes immortalization of hematopoietic cells, we generated growth factor dependent myeloid cells in which HoxA9 expression is regulated by administration of 4-hydroxy-tamoxifen. Maintenance of HoxA9 overexpression is required for continued cell survival and proliferation, even in the presence of growth factors. We show for the first time that maintenance of Bcl-2 expression is critical for HoxA9-dependent immortalization and influences the latency of HoxA9-dependent leukemia. Hematopoietic cells lacking Bcl-2 were not immortalized by HoxA9 in vitro. Furthermore, deletion of Bcl-2 delayed the onset and reduced the severity of HoxA9/Meis1 and MLL-AF9 leukemias. This is the first description of a molecular link between HoxA9 and the regulation of Bcl-2 family members in acute myeloid leukemia.


Subject(s)
Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid Progenitor Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Animals , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Survival/physiology , Gene Expression Regulation, Leukemic , Genes, bcl-2 , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Interleukin-3/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
20.
Genomics ; 102(1): 38-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23639479

ABSTRACT

Illumina Infinium Human Methylation (HM) BeadChips are widely used for measuring genome-scale DNA methylation, particularly in relation to epigenome-wide association studies (EWAS) studies. The methylation profile of human samples can be assessed accurately and reproducibly using the HM27 BeadChip (27,578 CpG sites) or its successor, the HM450 BeadChip (482,421 CpG sites). To date no mouse equivalent has been developed, greatly hindering the application of this methodology to the wide range of valuable murine models of disease and development currently in existence. We found 1308 and 13,715 probes from HM27 and HM450 BeadChip respectively, uniquely matched the bisulfite converted reference mouse genome (mm9). We demonstrate reproducible measurements of DNA methylation at these probes in a range of mouse tissue samples and in a murine cell line model of acute myeloid leukaemia. In the absence of a mouse counterpart, the Infinium Human Methylation BeadChip arrays have utility for methylation profiling in non-human species.


Subject(s)
CpG Islands/genetics , DNA Fingerprinting , DNA Methylation/genetics , DNA/genetics , Animals , Genome, Human , Humans , Mice , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...