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










Publication year range
1.
Leukemia ; 30(10): 2064-2073, 2016 10.
Article in English | MEDLINE | ID: mdl-27125308

ABSTRACT

Although NOD-SCID IL2Rγ-/- (NSG) xenograft mice are currently the most frequently used model to study human leukemia in vivo, the absence of a human niche severely hampers faithful recapitulation of the disease. We used NSG mice in which ceramic scaffolds seeded with human mesenchymal stromal cells were implanted to generate a human bone marrow (huBM-sc)-like niche. We observed that, in contrast to the murine bone marrow (mBM) niche, the expression of BCR-ABL or MLL-AF9 was sufficient to induce both primary acute myeloid leukemia (AML) and acute lymphocytic leukemia (ALL). Stemness was preserved within the human niches as demonstrated by serial transplantation assays. Efficient engraftment of AML MLL-AF9 and blast-crisis chronic myeloid leukemia patient cells was also observed, whereby the immature blast-like phenotype was maintained in the huBM-sc niche but to a much lesser extent in mBM niches. We compared transcriptomes of leukemias derived from mBM niches versus leukemias from huBM-like scaffold-based niches, which revealed striking differences in the expression of genes associated with hypoxia, mitochondria and metabolism. Finally, we utilized the huBM-sc MLL-AF9 B-ALL model to evaluate the efficacy of the I-BET151 inhibitor in vivo. In conclusion, we have established human niche models in which the myeloid and lymphoid features of BCR-ABL+ and MLL-AF9+ leukemias can be studied in detail.


Subject(s)
Bone Marrow/pathology , Disease Models, Animal , Fusion Proteins, bcr-abl , Leukemia, Myeloid, Acute/pathology , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Humans , Mice , Transplantation, Heterologous
2.
Leukemia ; 29(3): 625-35, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25184385

ABSTRACT

As the transcriptional coactivator CITED2 (CBP/p300-interacting-transactivator-with-an ED-rich-tail 2) can be overexpressed in acute myeloid leukemia (AML) cells, we analyzed the consequences of high CITED2 expression in normal and AML cells. CITED2 overexpression in normal CD34(+) cells resulted in enhanced hematopoietic stem and progenitor cell (HSPC) output in vitro, as well as in better hematopoietic stem cell (HSC) engraftability in NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice. This was because of an enhanced quiescence and maintenance of CD34(+)CD38(-) HSCs, due in part to an increased expression of the cyclin-dependent kinase inhibitor CDKN1A. We demonstrated that PU.1 is a critical regulator of CITED2, as PU.1 repressed CITED2 expression in a DNA methyltransferase 3A/B (DNMT3A/B)-dependent manner in normal CD34(+) cells. CD34(+) cells from a subset of AML patients displayed higher expression levels of CITED2 as compared with normal CD34(+) HSPCs, and knockdown of CITED2 in AML CD34(+) cells led to a loss of long-term expansion, both in vitro and in vivo. The higher CITED2 expression resulted from reduced PU.1 activity and/or dysfunction of mutated DNMT3A/B. Collectively, our data demonstrate that increased CITED2 expression results in better HSC maintenance. In concert with low PU.1 levels, this could result in a perturbed myeloid differentiation program that contributes to leukemia maintenance.


Subject(s)
Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mutation , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transplantation, Heterologous , DNA Methyltransferase 3B
3.
Leukemia ; 27(7): 1461-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23403462

ABSTRACT

The most frequent chromosomal translocations in pediatric acute myeloid leukemia affect the 11q23 locus and give rise to mixed lineage leukemia (MLL) fusion genes, MLL-AF9 being the most prevalent. The MLL-AF9 fusion gene has been shown to induce leukemia in both mouse and human models. In this study, we demonstrate that leukemogenic activity of MLL-AF9 requires RUVBL2 (RuvB-like 2), an AAA+ ATPase family member that functions in a wide range of cellular processes, including chromatin remodeling and transcriptional regulation. Expression of RUVBL2 was dependent on MLL-AF9, as it increased upon immortalization of human cord blood-derived hematopoietic progenitor cells with the fusion gene and decreased following loss of fusion gene expression in conditionally immortalized mouse cells. Short hairpin RNA-mediated silencing experiments demonstrated that both the immortalized human cells and the MLL-AF9-expressing human leukemia cell line THP-1 required RUVBL2 expression for proliferation and survival. Furthermore, inhibition of RUVBL2 expression in THP-1 cells led to reduced telomerase activity and clonogenic potential. These data were confirmed with a dominant-negative Walker B-mutated RUVBL2 construct. Taken together, these data suggest the possibility of targeting RUVBL2 as a potential therapeutic strategy for MLL-AF9-associated leukemia.


Subject(s)
Carrier Proteins/genetics , DNA Helicases/genetics , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Line, Transformed , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Helicases/metabolism , Gene Expression Regulation, Leukemic/physiology , Humans , Leukemia, Biphenotypic, Acute/metabolism , Leukemia, Biphenotypic, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , RNA Interference , Telomerase/metabolism
4.
Leukemia ; 27(5): 1116-26, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23178754

ABSTRACT

The MLL-AF9 fusion gene is associated with aggressive leukemias of both the myeloid and lymphoid lineage in infants, whereas in adults, this translocation is mainly associated with acute myeloid leukemia. These observations suggest that differences exist between fetal and adult tissues in terms of the 'cell of origin' from which the leukemia develops. Here we show that depending on extrinsic cues, human neonatal CD34(+) cells are readily immortalized along either the myeloid or lymphoid lineage upon MLL-AF9 expression and give rise to mainly lymphoid leukemia in immunocompromised mice. In contrast, immortalization of adult bone marrow CD34(+) cells is more difficult to achieve and is myeloid-biased, even when MLL-AF9 is expressed in purified hematopoietic stem cells (HSCs). Transcriptome analysis identified enrichment of HSC but not progenitor gene signatures in MLL-AF9-expressing cells. Although not observed in adult cells, neonatal cells expressing MLL-AF9 were enriched for gene signatures associated with poor prognosis, resistance to chemotherapeutic agents and MYC signaling. These results indicate that neonatal cells are inherently more prone to MLL-AF9-mediated immortalization than adult cells and suggest that intrinsic properties of the cell of origin, in addition to extrinsic cues, dictate lineage of the immortalized cell.


Subject(s)
Cell Lineage , Cell Transformation, Neoplastic , Hematopoietic Stem Cells/pathology , Myeloid-Lymphoid Leukemia Protein/physiology , Oncogene Proteins, Fusion/physiology , Animals , Antigens, CD19/analysis , Female , Humans , Infant, Newborn , Leukemia, Myeloid, Acute/etiology , Lewis X Antigen/analysis , Lipopolysaccharide Receptors/analysis , Mice , Mice, SCID , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology
6.
Leukemia ; 25(12): 1825-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21760593

ABSTRACT

In order to identify acute myeloid leukemia (AML) CD34(+)-specific gene expression profiles, mononuclear cells from AML patients (n=46) were sorted into CD34(+) and CD34(-) subfractions, and genome-wide expression analysis was performed using Illumina BeadChip Arrays. AML CD34(+) and CD34(-) gene expression was compared with a large group of normal CD34(+) bone marrow (BM) cells (n=31). Unsupervised hierarchical clustering analysis showed that CD34(+) AML samples belonged to a distinct cluster compared with normal BM and that in 61% of the cases the AML CD34(+) transcriptome did not cluster together with the paired CD34(-) transcriptome. The top 50 of AML CD34(+)-specific genes was selected by comparing the AML CD34(+) transcriptome with the AML CD34(-) and CD34(+) normal BM transcriptomes. Interestingly, for three of these genes, that is, ankyrin repeat domain 28 (ANKRD28), guanine nucleotide binding protein, alpha 15 (GNA15) and UDP-glucose pyrophosphorylase 2 (UGP2), a high transcript level was associated with a significant poorer overall survival (OS) in two independent cohorts (n=163 and n=218) of normal karyotype AML. Importantly, the prognostic value of the continuous transcript levels of ANKRD28 (OS hazard ratio (HR): 1.32, P=0.008), GNA15 (OS HR: 1.22, P=0.033) and UGP2 (OS HR: 1.86, P=0.009) was shown to be independent from the well-known risk factors FLT3-ITD, NPM1c(+) and CEBPA mutation status.


Subject(s)
Antigens, CD34/metabolism , Biomarkers, Tumor/genetics , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , Female , Gene Expression Regulation, Leukemic , Humans , Karyotyping , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Mutation/genetics , Prognosis , Risk Factors , Young Adult
7.
Bone Marrow Transplant ; 46(1): 110-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20383220

ABSTRACT

Autologous SCT (auto-SCT) introduces a reduced tolerance to chemotherapy even in patients with adequate engraftment, suggesting long-term effects of the transplantation procedure on the BM capacity. To study the hematopoietic cell compartment after auto-SCT, CD34(+) BM cells (n = 16) from patients at 6-9 months after auto-SCT were studied with regard to the progenitor subsets, colony frequency and cell cycle status. The BM compartments were studied in vivo using PET tracer 3-fluoro-3-deoxy-L-thymidine (¹8F-FLT PET). BM CD34(+) cells after auto-SCT were compared with normal CD34(+) cells and showed a phenotypic shift from common myeloid progenitor (CMP mean percentage 3.7 vs 19.4%, P=0.001) to granulocyte-macrophage progenitor (GMP mean percentage 51.8 vs 27.6%, P=0.01). In addition, a reduced clonogenic potential and higher cycling activity especially of the GMP fraction (41% ± 4 in G2/S phase vs 19% ± 2, P = 0.03) were observed in BM after auto-SCT compared with normal. The enhanced cycling activity was confirmed in vivo by showing a significantly higher uptake of the ¹8F-FLT PET tracer by the BM compartment. This study shows that auto-SCT results in defects of the hematopoietic compartment at least 6 months after auto-SCT, characterized by changes in the composition of progenitor subsets and enhanced in vitro and in vivo cycling activity.


Subject(s)
Cell Cycle , Hematopoietic Stem Cell Transplantation , Molecular Imaging/methods , Myeloid Progenitor Cells/metabolism , Phenotype , Adult , Antigens, CD34/metabolism , Bone Marrow/metabolism , Colony-Forming Units Assay , Dideoxynucleosides/pharmacokinetics , Follow-Up Studies , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Humans , Lymphoma/therapy , Middle Aged , Myeloid Progenitor Cells/cytology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Transplantation, Autologous , Whole Body Imaging
8.
Ann N Y Acad Sci ; 1106: 114-42, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442773

ABSTRACT

Nucleoporin 98 (NUP98) is a component of the nuclear pore complex that facilitates mRNA export from the nucleus. It is mapped to 11p15.5 and is fused to a number of distinct partners, including nine members of the homeobox family as a consequence of leukemia-associated chromosomal translocations. NUP98-HOXA9 is associated with the t(7;11)(p15;p15) translocation in acute myeloid leukemia (AML), myelodysplastic syndrome, and blastic crisis of chronic myeloid leukemia. Expression of NUP98-HOXA9 in murine bone marrow resulted in a myeloproliferative disease progressing to AML by 7-8 months. Transduction of NUP98 fusion genes into human CD34(+) cells confers a proliferative advantage in long-term cytokine-stimulated and stromal cocultures and in NOD-SCID engrafted mice, associated with a five- to eight-fold increase in hematopoietic stem cells. NUP98-HOXA9 expression inhibited erythroid and myeloid differentiation but enhanced serial progenitor replating. NUP98-HOXA9 upregulated a number of homeobox genes of the A and B cluster as well as MEIS1 and Pim-1, and downmodulated globin genes and C/EBPalpha. The HOXA9 component of the NUP98-HOXA9 fusion protein was protected from cullin-4A-mediated ubiquitination and subsequent proteasome-dependent degradation. In NUP98-HOX-transduced CD34(+) cells and cells from AML patients with t(7;11)(p15;p15) NUP98 was no longer associated with the nuclear pore complex but formed intranuclear aggregation bodies. Analysis of NUP98 allelic expression in AML and myelodysplastic syndrome showed loss of heterozygosity observed in 29% of the former and 8% of the latter. This was associated with poor prognosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Pore Complex Proteins/physiology , Alleles , Animals , Antigens, CD34/biosynthesis , Cell Nucleus/metabolism , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 7 , Humans , Loss of Heterozygosity , Mice , Mice, Inbred NOD , Mice, SCID , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism
9.
Leukemia ; 16(4): 675-82, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11960349

ABSTRACT

To explore the possible cross-talk between the IL-6 and TGF-beta1 pathways in AML blast cells, the effect of TGF-beta1 pretreatment on IL-6-induced STAT3 tyrosine phosphorylation was studied. A reduction of STAT3 tyrosine phosphorylation after TGF-beta1 pretreatment was observed in four out of 40 AML cases (10%), although all of the AML cases responded to TGF-beta1 by means of SMAD3 translocation. The reduced IL-6-mediated STAT3 tyrosine phosphorylation after pre-treatment with TGF-beta1 was associated with apoptosis and coincided with the degradation of certain cellular proteins, including JAK1 and -2 and Tyk2, without affecting the ERK expression and phosphorylation. Furthermore, treatment of AML blasts with the cytostatic agent VP16, as an alternative way to induce apoptosis, resulted in a similar degree of degradation of JAK kinases and concomitant reduction of IL-6-mediated STAT3 tyrosine phosphorylation. Although degradation of JAK kinases could be rescued by incubating the cells with the pan-caspase inhibitor Z-VAD-fmk, the attenuating effect of TGF-beta1 treatment on the STAT3 tyrosine phosphorylation was still partly present. It was shown that in AML cells cultured in the presence of Z-VAD-fmk, TGF-beta1 pretreatment resulted in a reduction of JAK1 phosphorylation upon IL-6 stimulation. Expression of SOCS1 and -3 could be ruled out as a possible cause of reduced JAK1 phosphorylation levels in the investigated AML case.


Subject(s)
Caspases/metabolism , DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid, Acute/metabolism , Repressor Proteins , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Annexin A5/metabolism , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3 , Caspase Inhibitors , Cysteine Proteinase Inhibitors , Down-Regulation , Electrophoretic Mobility Shift Assay , Epithelial Cells/drug effects , Etoposide/metabolism , Etoposide/pharmacology , Humans , Janus Kinase 1 , Leukemia, Myeloid, Acute/pathology , Phosphorylation , Protein Transport , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Signal Transduction/drug effects , Smad3 Protein , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Transforming Growth Factor beta1 , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tyrosine/metabolism
10.
Oncogene ; 20(38): 5350-8, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11536047

ABSTRACT

The MEN2A oncogene encodes for a constitutive active member of the RET receptor tyrosine kinase family. Here, we report that MEN2A-RET activates Signal Transducer and Activator of Transcription 3 (STAT3) via two YxxV/Q STAT3 docking sites, Tyr752 and Tyr928. MEN2A-RET induces both Tyr705 and Ser727 phosphorylation of STAT3, and STAT3 serine phosphorylation is required for its maximal transcriptional activity. Stable NIH3T3 cell lines expressing both MEN2A-RET and STAT3alpha but not STAT3beta, are characterized by enhanced proliferation and cyclin-D1 promoter activity, and enhanced growth in soft agar. These data indicate that malignant cell growth induced by MEN2A-RET involves its activation of STAT3.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Drosophila Proteins , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Binding Sites , COS Cells , Cell Division , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Genes, Reporter , Humans , Mice , Oncogenes/genetics , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret , STAT3 Transcription Factor , Serine/chemistry , Time Factors , Transcriptional Activation , Transfection , Tyrosine/chemistry , Up-Regulation
11.
Cytokine ; 14(2): 78-87, 2001 Apr 21.
Article in English | MEDLINE | ID: mdl-11356008

ABSTRACT

Transcriptional activation of eukaryotic genes often requires the cooperative action of many proteins. The interleukin 6 (IL-6) response element (IRE) is activated by signal transducer and activator of transcription 3 (STAT3), and stimulation with IL-6 leads to STAT3 tyr705 phosphorylation, dimerization, translocation to the nucleus and transactivation of target gene promoters containing IREs. Here, we report that IL-6 and 12-O-tetradecanoylphorbol-13-acetate (TPA) synergistically transactivate the IRE in HepG2 cells, which is coupled to a strong upregulation of c-Jun and c-Fos expression by TPA via the mitogen-activated protein kinase (MAPK) pathway. Overexpression of c-Jun and c-Fos strongly enhanced STAT3-driven IRE transactivation as well as transactivation of the human intercellular adhesion molecule (ICAM)-1 promoter. In contrast, c-Jun mutants lacking the transactivation domain, the DNA-binding domain, or mutants in which the serine residues 63 and 73 were replaced by alanine, did not cooperate with STAT3. In immunoprecipitation experiments, a direct association of STAT3 with c-Jun and c-Fos was observed in response to IL-6. Furthermore, c-Jun/STAT3 and c-Fos/STAT3 complexes were detected on IRE probes in electrophoretic mobility shift assay (EMSA) experiments, but did not bind nor transactivate the TPA response element (TRE). These results demonstrate that activator protein-1 (AP-1) transcription factors can cooperate with STAT3 in IRE transactivation in the absence of direct AP-1 DNA binding.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Response Elements/genetics , Trans-Activators/metabolism , Transcriptional Activation/drug effects , Blotting, Western , DNA/genetics , DNA/metabolism , Genes, Reporter , Humans , Macromolecular Substances , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , STAT3 Transcription Factor , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Up-Regulation/drug effects
12.
J Biol Chem ; 276(29): 27709-15, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11335711

ABSTRACT

Activation of signal transducer and activator of transcription 3 (STAT3) by interleukin-6 (IL-6) involves phosphorylation of Tyr-705 and Ser-727, both of which are critical for STAT3 transactivation. Here, we demonstrate that IL-6 activates Rac-1 and SEK-1/MKK-4 of the stress-activated protein kinase pathway, as well as protein kinase Cdelta (PKCdelta), as indicated by PKCdelta Thr-505 phosphorylation. However, JNK-1, the end point kinase of the stress-activated protein kinase pathway signal transduction cascade, is not activated by IL-6. PKCdelta was found to be associated with SEK-1/MKK-4 in unstimulated HepG2 cells but rapidly dissociates from SEK-1/MKK-4 upon IL-6 stimulation to become associated with STAT3. Inhibition of PKCdelta using rottlerin (6 microm) or by overexpression of dominant negative PKCdelta demonstrates that PKCdelta kinase activity is required for STAT3 Ser-727 phosphorylation and transactivation but not for STAT3 Tyr-705 phosphorylation or nuclear import. PKCdelta signals downstream of Rac-1 and SEK-1/MKK-4, because enhanced STAT3 transactivation induced by overexpression of constitutive active RacV12 was strongly abrogated by rottlerin, whereas IL-6-induced SEK-1/MKK-4 Thr-223 phosphorylation was not affected under these conditions. Studying the kinetics of STAT3 and PKCdelta phosphorylation in cytoplasmic and nuclear fractions revealed that STAT3 Tyr-705 phosphorylation and nuclear translocation precedes PKCdelta Thr-505 and STAT3 Ser-727 phosphorylation. Furthermore, the IL-6-induced PKCdelta Thr-505 and STAT3 Ser-727 phosphorylation were only observed in nuclear fractions of HepG2 cells. These results demonstrate that IL-6-induced STAT3 transactivation involves the sequential activation of Rac-1 and SEK-1/MKK-4, which leads to nuclear translocation of PKCdelta by release from a SEK-1/MKK-4-containing complex. Our results further indicate that PKCdelta-mediated STAT3 Ser-727 phosphorylation is mainly a nuclear event.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Isoenzymes/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase C/metabolism , Serine/metabolism , Trans-Activators/metabolism , Transcriptional Activation , rac1 GTP-Binding Protein/metabolism , Base Sequence , DNA Primers , DNA-Binding Proteins/chemistry , Humans , Phosphorylation , Protein Kinase C-delta , STAT3 Transcription Factor , Trans-Activators/chemistry , Tumor Cells, Cultured
13.
FEBS Lett ; 495(1-2): 71-6, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11322950

ABSTRACT

Activation of the signal transducer and activator of transcription 3 (STAT3) in response to interleukin-6 (IL-6) type cytokines involves both phosphorylation of Tyr705, which enables dimerization, nuclear translocation and DNA binding, as well as ser727 phosphorylation. Here, we describe that the 65 C-terminal amino acids of STAT3 can function as an independent transcription activation domain (TAD), particularly when a negative charge is introduced at position 727 by mutation of the serine residue into aspartate. The strong transcriptional activity of the C-terminal STAT3 Ser727Asp TAD is coupled to a constitutive association with the co-activator p300. In HepG2 cells, p300 associates with STAT3 upon IL-6 stimulation, and overexpression of p300 enhances the transcriptional activity of STAT3alpha, but not of STAT3beta or STAT3 Ser727Ala. We conclude that Ser727 phosphorylation in the C-terminal region of STAT3 is required for transactivation by association with p300.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Nuclear Proteins/metabolism , Serine/metabolism , Trans-Activators/metabolism , Transcriptional Activation/drug effects , Amino Acid Substitution , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , DNA-Binding Proteins/genetics , Humans , Mutagenesis, Site-Directed , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Protein Structure, Tertiary/physiology , STAT3 Transcription Factor , Trans-Activators/genetics , Transcriptional Activation/physiology
14.
Blood ; 95(12): 3765-70, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10845908

ABSTRACT

To explore the activation patterns of signal transducer and activator of transcription 3 (Stat3) in acute myeloid leukemia (AML), we examined whether the phosphorylation of tyrosine705 (Tyr705) and serine727 (Ser727) residues was abnormally regulated in cells from patients with AML. In 5 of 20 (25%) patients with AML, Stat3 was constitutively phosphorylated on Tyr705 and Ser727, which were not further up-regulated by treatment with IL-6. Furthermore, Stat3 was constitutively bound to the IRE response element in these cells as determined by electrophoretic mobility shift assay, and stimulation with IL-6 did not result in increased DNA binding. Interestingly, AML cells with constitutive Stat3 activation also secreted high levels of IL-6 protein. Treating these AML cells with anti-IL-6 resulted in restored IL-6-inducible Stat3 phosphorylation on both Tyr705 and Ser727 with low or undetectable basal phosphorylation levels in unstimulated cells. In contrast, treatment with anti-IL-1 did not result in altered Stat3 phosphorylation patterns. The constitutive IL-6 expression was associated with elevated levels of suppressor of cytokine signaling-1 (SOCS-1) and SOCS-3 mRNA expression, which were not down-regulated by anti-IL-6. These data indicate that the constitutive Stat3 activation in the investigated AML blasts is caused by high IL-6 secretion levels, thus stimulating the Jak/Stat pathway in an autocrine manner, a paracrine manner, or both. (Blood. 2000;95:3765-3770)


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/metabolism , Interleukin-6/blood , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid/blood , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Proteins/genetics , Repressor Proteins , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic , Acute Disease , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/genetics , Leukemia, Myeloid/classification , Leukemia, Myeloid/immunology , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Tumor Cells, Cultured
15.
Biochem J ; 347 Pt 1: 89-96, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10727406

ABSTRACT

In the present study, signal transducer and activator of transcription 3 (STAT3) Ser(727) phosphorylation and transactivation was investigated in relation to activation of mitogen-activated protein (MAP) kinase family members including extracellular-signal-regulated protein kinase (ERK)-1, c-Jun N-terminal kinase (JNK)-1 and p38 ('reactivating kinase') in response to interleukin (IL)-6 stimulation. Although IL-6 can activate ERK-1 in HepG2 cells, STAT3 transactivation and Ser(727) phosphorylation were not reduced by using the MAP kinase/ERK kinase (MEK) inhibitor PD98059 or by overexpression of dominant-negative Raf. IL-6 did not activate JNK-1 in HepG2 cells and STAT3 was a poor substrate for JNK-1 activated by anisomycin, excluding a role for JNK1 in IL-6-induced STAT3 activation. However, SEK-1/MKK-4 [where SEK-1 stands for stress-activated protein kinase (SAPK)/ERK kinase 1, and MKK-4 stands for MAP kinase kinase 4] was activated in response to IL-6 and overexpression of dominant-negative SEK-1/MKK-4(A-L) reduced both IL-6-induced STAT3 Ser(727) phosphorylation as well as STAT3 transactivation. Subsequently, the SEK-1/MKK-4 upstream components Vav, Rac-1 and MEKK were identified as components of a signal transduction cascade that leads to STAT3 transactivation in response to IL-6 stimulation. Furthermore, inhibition of p38 kinase activity with the inhibitor SB203580 did not block STAT3 Ser(727) phosphorylation but rather increased both basal as well as IL-6-induced STAT3 transactivation, indicating that p38 may act as a negative regulator of IL-6-induced STAT3 transactivation through a presently unknown mechanism. In conclusion, these data indicate that IL-6-induced STAT3 transactivation and Ser(727) phosphorylation is independent of ERK-1 or JNK-1 activity, but involves a gp130 receptor-signalling cascade that includes Vav, Rac-1, MEKK and SEK-1/MKK-4 as signal transduction components.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Serine , Signal Transduction/physiology , Trans-Activators/metabolism , rac GTP-Binding Proteins/metabolism , Amino Acid Substitution , Carcinoma, Hepatocellular , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/pharmacology , Genes, Reporter , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Liver Neoplasms , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Proto-Oncogene Proteins c-vav , Pyridines/pharmacology , Recombinant Proteins/metabolism , STAT3 Transcription Factor , Trans-Activators/chemistry , Transcriptional Activation , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...