Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Blood ; 132(3): 307-320, 2018 07 19.
Article in English | MEDLINE | ID: mdl-29724897

ABSTRACT

Heat shock protein 90 (HSP90) stabilizes many client proteins, including the BCR-ABL1 oncoprotein. BCR-ABL1 is the hallmark of chronic myeloid leukemia (CML) in which treatment-free remission (TFR) is limited, with clinical and economic consequences. Thus, there is an urgent need for novel therapeutics that synergize with current treatment approaches. Several inhibitors targeting the N-terminal domain of HSP90 are under investigation, but side effects such as induction of the heat shock response (HSR) and toxicity have so far precluded their US Food and Drug Administration approval. We have developed a novel inhibitor (aminoxyrone [AX]) of HSP90 function by targeting HSP90 dimerization via the C-terminal domain. This was achieved by structure-based molecular design, chemical synthesis, and functional preclinical in vitro and in vivo validation using CML cell lines and patient-derived CML cells. AX is a promising potential candidate that induces apoptosis in the leukemic stem cell fraction (CD34+CD38-) as well as the leukemic bulk (CD34+CD38+) of primary CML and in tyrosine kinase inhibitor (TKI)-resistant cells. Furthermore, BCR-ABL1 oncoprotein and related pro-oncogenic cellular responses are downregulated, and targeting the HSP90 C terminus by AX does not induce the HSR in vitro and in vivo. We also probed the potential of AX in other therapy-refractory leukemias. Therefore, AX is the first peptidomimetic C-terminal HSP90 inhibitor with the potential to increase TFR in TKI-sensitive and refractory CML patients and also offers a novel therapeutic option for patients with other types of therapy-refractory leukemia because of its low toxicity profile and lack of HSR.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Heat-Shock Response/drug effects , Imatinib Mesylate/pharmacology , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Animals , Antineoplastic Agents/chemistry , Binding Sites , Biomarkers, Tumor , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , Imatinib Mesylate/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Multimerization/drug effects , Spectrum Analysis , Structure-Activity Relationship , Xenograft Model Antitumor Assays
2.
Hum Gene Ther Clin Dev ; 29(2): 69-79, 2018 06.
Article in English | MEDLINE | ID: mdl-29664709

ABSTRACT

Chronic granulomatous disease (CGD) is a debilitating primary immunodeficiency affecting phagocyte function due to the absence of nicotinamide dinucleotide phosphate (NADPH) oxidase activity. The vast majority of CGD patients in the Western world have mutations within the X-linked CYBB gene encoding for gp91phox (NOX2), the redox center of the NADPH oxidase complex (XCGD). Current treatments of XCGD are not entirely satisfactory, and prior attempts at autologous gene therapy using gammaretrovirus vectors did not provide long-term curative effects. A new strategy was developed based on the use of the lentiviral vector G1XCGD expressing high levels of the gp91phox transgene in myeloid cells. As a requisite for a clinical trial approval, standardized non-clinical studies were conducted in vitro and in mice in order to evaluate the pharmacodynamics and biosafety of the vector and the biodistribution of G1XCGD-transduced cells. Transduced CD34+ cells derived from XCGD patients engrafted and differentiated similarly to their non-transduced counterparts in xenograft mouse models and generated therapeutically relevant levels of NADPH activity in myeloid cells expressing gp91phox. Expression of functional gp91phox in hematopoietic cells did not affect their homing properties, which engrafted at high levels in mice. Extensive in vitro and in vivo genotoxicity studies found no evidence for adverse mutagenesis related to vector treatment. These studies paved the way for the approval of clinical trials in Europe and in the United States for the treatment of XCGD patients with G1XCGD gene-modified autologous hematopoietic cells.


Subject(s)
Genetic Diseases, X-Linked/genetics , Granulomatous Disease, Chronic/genetics , NADPH Oxidase 2/genetics , NADPH Oxidases/genetics , Animals , Clinical Trials as Topic , Gene Expression Regulation/drug effects , Genetic Diseases, X-Linked/pathology , Genetic Diseases, X-Linked/therapy , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Granulomatous Disease, Chronic/pathology , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/drug effects , Heterografts , Humans , Lentivirus/genetics , Mice , NADPH Oxidase 2/administration & dosage
3.
Oncotarget ; 8(16): 26169-26184, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28412732

ABSTRACT

Gallbladder cancer (GBC) is a lethal cancer with poor prognosis associated with high invasiveness and poor response to chemotherapy and radiotherapy. New therapeutic approaches are urgently needed in order to improve survival and response rates of GBC patients. We screened 130 small molecule inhibitors on a panel of seven GBC cell lines and identified the HSP90 inhibitor 17-AAG as one of the most potent inhibitory drugs across the different lines. We tested the antitumor efficacy of 17-AAG and geldanamycin (GA) in vitro and in a subcutaneous preclinical tumor model NOD-SCID mice. We also evaluated the expression of HSP90 by immunohistochemistry in human GBC tumors.In vitro assays showed that 17-AAG and GA significantly reduced the expression of HSP90 target proteins, including EGFR, AKT, phospho-AKT, Cyclin B1, phospho-ERK and Cyclin D1. These molecular changes were consistent with reduced cell viability and cell migration and promotion of G2/M cell cycle arrest and apoptosis observed in our in vitro studies.In vivo, 17-AAG showed efficacy in reducing subcutaneous tumors size, exhibiting a 69.6% reduction in tumor size in the treatment group compared to control mice (p < 0.05).The HSP90 immunohistochemical staining was seen in 182/209 cases of GBC (87%) and it was strongly expressed in 70 cases (33%), moderately in 58 cases (28%), and weakly in 54 cases (26%).Our pre-clinical observations strongly suggest that the inhibition of HSP90 function by HSP90 inhibitors is a promising therapeutic strategy for gallbladder cancer that may benefit from new HSP90 inhibitors currently in development.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Drug Discovery , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Gallbladder Neoplasms , High-Throughput Screening Assays , Humans , Mice , Small Molecule Libraries , Xenograft Model Antitumor Assays
5.
Oncotarget ; 6(31): 31877-88, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26397134

ABSTRACT

Gallbladder cancer (GBC) is a highly malignant tumor characterized by a poor response to chemotherapy and radiotherapy. We evaluated the in vitro and in vivo antitumor efficacy of mTOR inhibitors, rapamycin and WYE-354. In vitro assays showed WYE-354 significantly reduced cell viability, migration and invasion and phospho-P70S6K expression in GBC cells. Mice harboring subcutaneous gallbladder tumors, treated with WYE-354 or rapamycin, exhibited a significant reduction in tumor mass. A short-term treatment with a higher dose of WYE-354 decreased the tumor size by 68.6% and 52.4%, in mice harboring G-415 or TGBC-2TKB tumors, respectively, compared to the control group. By contrast, treatment with a prolonged-low-dose regime of rapamycin almost abrogated tumor growth, exhibiting 92.7% and 97.1% reduction in tumor size, respectively, compared to control mice. These results were accompanied by a greater decrease in the phosphorylation status of P70S6K and a lower cell proliferation Ki67 index, compared to WYE-354 treated mice, suggesting a more effective mTOR pathway inhibition. These findings provide a proof of concept for the use of rapamycin or WYE-354 as potentially good candidates to be studied in clinical trials in GBC patients.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Gallbladder Neoplasms/drug therapy , Guanine/analogs & derivatives , Sirolimus/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Therapy, Combination , Gallbladder Neoplasms/pathology , Guanine/pharmacology , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation/drug effects , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Nucleic Acids Res ; 43(3): 1577-92, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25605798

ABSTRACT

Epigenetic silencing of transgene expression represents a major obstacle for the efficient genetic modification of multipotent and pluripotent stem cells. We and others have demonstrated that a 1.5 kb methylation-free CpG island from the human HNRPA2B1-CBX3 housekeeping genes (A2UCOE) effectively prevents transgene silencing and variegation in cell lines, multipotent and pluripotent stem cells, and their differentiated progeny. However, the bidirectional promoter activity of this element may disturb expression of neighboring genes. Furthermore, the epigenetic basis underlying the anti-silencing effect of the UCOE on juxtaposed promoters has been only partially explored. In this study we removed the HNRPA2B1 moiety from the A2UCOE and demonstrate efficient anti-silencing properties also for a minimal 0.7 kb element containing merely the CBX3 promoter. This DNA element largely prevents silencing of viral and tissue-specific promoters in multipotent and pluripotent stem cells. The protective activity of CBX3 was associated with reduced promoter CpG-methylation, decreased levels of repressive and increased levels of active histone marks. Moreover, the anti-silencing effect of CBX3 was locally restricted and when linked to tissue-specific promoters did not activate transcription in off target cells. Thus, CBX3 is a highly attractive element for sustained, tissue-specific and copy-number dependent transgene expression in vitro and in vivo.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Gene Silencing , Multipotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Differentiation , Cell Line, Tumor , Cell Separation , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Transgenes
7.
Mol Ther ; 23(1): 63-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25189742

ABSTRACT

Gene therapy for hematological disorders relies on the genetic modification of CD34(+) cells, a heterogeneous cell population containing about 0.01% long-term repopulating cells. Here, we show that the lentiviral vector CD133-LV, which uses a surface marker on human primitive hematopoietic stem cells (HSCs) as entry receptor, transfers genes preferentially into cells with high engraftment capability. Transduction of unstimulated CD34(+) cells with CD133-LV resulted in gene marking of cells with competitive proliferative advantage in vitro and in immunodeficient mice. The CD133-LV-transduced population contained significantly more cells with repopulating capacity than cells transduced with vesicular stomatitis virus (VSV)-LV, a lentiviral vector pseudotyped with the vesicular stomatitis virus G protein. Upon transfer of a barcode library, CD133-LV-transduced cells sustained gene marking in vivo for a prolonged period of time with a 6.7-fold higher recovery of barcodes compared to transduced control cells. Moreover, CD133-LV-transduced cells were capable of repopulating secondary recipients. Lastly, we show that this targeting strategy can be used for transfer of a therapeutic gene into CD34(+) cells obtained from patients suffering of X-linked chronic granulomatous disease. In conclusion, direct gene transfer into CD133(+) cells allows for sustained long-term engraftment of gene corrected cells.


Subject(s)
Antigens, CD/genetics , Genetic Therapy/methods , Glycoproteins/genetics , Hematopoietic Stem Cells/immunology , Lentivirus/genetics , Peptides/genetics , AC133 Antigen , Animals , Antigens, CD/immunology , Antigens, CD34/genetics , Antigens, CD34/immunology , Gene Expression , Genetic Vectors , Glycoproteins/immunology , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/pathology , Granulomatous Disease, Chronic/therapy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Peptides/immunology , Primary Cell Culture , Transduction, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
8.
Biochem Pharmacol ; 87(4): 625-35, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24355567

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a T cell-mediated inflammatory autoimmune disease model of multiple sclerosis (MS). The inflammatory process is initiated by activation and proliferation of T cells and monocytes and by their subsequent migration into the central nervous system (CNS), where they induce demyelination and neurodegeneration. Prostaglandin E2 (PGE2) - synthesized by cyclooxygenase 2 (COX-2) - has both pro- and anti-inflammatory potential, which is translated via four different EP receptors. We hypothesized that PGE2 synthesized in the preclinical phase by peripheral immune cells exerts pro-inflammatory properties in the EAE model. To investigate this, we used a bone marrow transplantation model, which enables PGE2 synthesis or EP receptor expression to be blocked specifically in peripheral murine immune cells. Our results reveal that deletion of COX-2 or its EP4 receptor in bone marrow-derived cells leads to a significant delay in the onset of EAE. This effect is due to an impaired preclinical inflammatory process indicated by a reduced level of the T cell activating interleukin-6 (IL-6), reduced numbers of T cells and of the T cell secreted interleukin-17 (IL-17) in the blood of mice lacking COX-2 or EP4 in peripheral immune cells. Moreover, mice lacking COX-2 or EP4 in bone marrow-derived cells show a reduced expression of matrix metalloproteinase 9 (MMP9), which results in decreased infiltration of monocytes and T cells into the CNS. In conclusion, our data demonstrate that PGE2 synthesized by monocytes in the early preclinical phase promotes the development of EAE in an EP4 receptor dependent manner.


Subject(s)
Bone Marrow Cells/immunology , Dinoprostone/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Monocytes/immunology , Receptors, Prostaglandin E, EP4 Subtype/physiology , Signal Transduction/immunology , Animals , Bone Marrow Cells/pathology , Dinoprostone/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Monocytes/pathology , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis
9.
Pain ; 155(3): 545-555, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333781

ABSTRACT

Prostacyclin is an important mediator of peripheral pain sensation. Here, we investigated its potential participation in mediating neuropathic pain and found that prostacyclin receptor (IP) knockout mice exhibited markedly decreased pain behavior. Application of an IP antagonist to the injury site or selective IP deficiency in myeloid cells mimicked the antinociceptive effect observed in IP knockout mice. At the site of nerve injury, IP was expressed in interleukin (IL) 1ß-containing resident macrophages, which were less common in IP knockout mice. Local administration of the IP agonist cicaprost inhibited macrophage migration in vitro and promoted accumulation of IP- and IL1ß-expressing cells as well as an increase of IL1ß concentrations at the application site in vivo. Fittingly, the IL1-receptor antagonist anakinra (IL-1ra) decreased neuropathic pain behavior in wild-type mice but not in IP knockout mice. Finally, continuous, but not single administration, of the cyclooxygenase inhibitor meloxicam early after nerve injury decreased pain behavior and the number of resident macrophages. Thus, early synthesis of prostacyclin at the site of injury causes accumulation of IL1ß-expressing macrophages as a key step in neuropathic pain after traumatic injury.


Subject(s)
Epoprostenol/physiology , Gene Expression Regulation , Interleukin-1beta/biosynthesis , Macrophages/metabolism , Neuralgia/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/pathology
10.
Hum Gene Ther Clin Dev ; 24(2): 86-98, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23845071

ABSTRACT

Chronic granulomatous disease (CGD) is a primary immunodeficiency characterized by impaired antimicrobial activity in phagocytic cells. As a monogenic disease affecting the hematopoietic system, CGD is amenable to gene therapy. Indeed in a phase I/II clinical trial, we demonstrated a transient resolution of bacterial and fungal infections. However, the therapeutic benefit was compromised by the occurrence of clonal dominance and malignant transformation demanding alternative vectors with equal efficacy but safety-improved features. In this work we have developed and tested a self-inactivating (SIN) gammaretroviral vector (SINfes.gp91s) containing a codon-optimized transgene (gp91(phox)) under the transcriptional control of a myeloid promoter for the gene therapy of the X-linked form of CGD (X-CGD). Gene-corrected cells protected X-CGD mice from Aspergillus fumigatus challenge at low vector copy numbers. Moreover, the SINfes.gp91s vector generates substantial amounts of superoxide in human cells transplanted into immunodeficient mice. In vitro genotoxicity assays and longitudinal high-throughput integration site analysis in transplanted mice comprising primary and secondary animals for 11 months revealed a safe integration site profile with no signs of clonal dominance.


Subject(s)
Gammaretrovirus/genetics , Genetic Vectors/metabolism , Granulomatous Disease, Chronic/therapy , Animals , Aspergillus fumigatus/pathogenicity , Cells, Cultured , DNA Methylation , Disease Models, Animal , Drug Evaluation, Preclinical , Genetic Therapy , Genetic Vectors/genetics , Humans , Lung Diseases/microbiology , Lung Diseases/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fes/genetics , Superoxides/metabolism
11.
Mol Ther ; 21(3): 648-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23207695

ABSTRACT

Comparative integrome analysis has revealed that the most neutral integration pattern among retroviruses is attributed to alpharetroviruses. We chose X-linked chronic granulomatous disease (X-CGD) as model to evaluate the potential of self-inactivating (SIN) alpharetroviral vectors for gene therapy of monogenic diseases. Therefore, we combined the alpharetroviral vector backbone with the elongation factor-1α short promoter, both considered to possess a low genotoxic profile, to drive transgene (gp91(phox)) expression. Following efficient transduction transgene expression was sustained and provided functional correction of the CGD phenotype in a cell line model at low vector copy number. Further analysis in a murine X-CGD transplantation model revealed gene-marking of bone marrow cells and oxidase positive granulocytes in peripheral blood. Transduction of human X-CGD CD34+ cells provided functional correction up to wild-type levels and long-term expression upon transplantation into a humanized mouse model. In contrast to lentiviral vectors, no aberrantly spliced transcripts containing cellular exons fused to alpharetroviral sequences were found in transduced cells, implying that the safety profile of alpharetroviral vectors may extend beyond their neutral integration profile. Taken together, this highlights the potential of this SIN alpharetroviral system as a platform for new candidate vectors for future gene therapy of hematopoietic disorders.


Subject(s)
Alpharetrovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Granulomatous Disease, Chronic/therapy , RNA Splicing , Animals , Bone Marrow Cells , Cell Line, Tumor , DNA Copy Number Variations , Disease Models, Animal , Granulocytes , Granulomatous Disease, Chronic/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic , Transgenes
12.
Cell ; 144(4): 566-76, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21335238

ABSTRACT

TAp63α, a homolog of the p53 tumor suppressor, is a quality control factor in the female germline. Remarkably, already undamaged oocytes express high levels of the protein, suggesting that TAp63α's activity is under tight control of an inhibitory mechanism. Biochemical studies have proposed that inhibition requires the C-terminal transactivation inhibitory domain. However, the structural mechanism of TAp63α inhibition remains unknown. Here, we show that TAp63α is kept in an inactive dimeric state. We reveal that relief of inhibition leads to tetramer formation with ∼20-fold higher DNA affinity. In vivo, phosphorylation-triggered tetramerization of TAp63α is not reversible by dephosphorylation. Furthermore, we show that a helix in the oligomerization domain of p63 is crucial for tetramer stabilization and competes with the transactivation domain for the same binding site. Our results demonstrate how TAp63α is inhibited by complex domain-domain interactions that provide the basis for regulating quality control in oocytes.


Subject(s)
Oocytes/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Animals , DNA/metabolism , Dimerization , Female , Gamma Rays , Mice , Models, Molecular , Phosphorylation , Protein Multimerization , Tumor Suppressor Protein p53/metabolism
13.
Nat Med ; 12(4): 401-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16582916

ABSTRACT

Gene transfer into hematopoietic stem cells has been used successfully for correcting lymphoid but not myeloid immunodeficiencies. Here we report on two adults who received gene therapy after nonmyeloablative bone marrow conditioning for the treatment of X-linked chronic granulomatous disease (X-CGD), a primary immunodeficiency caused by a defect in the oxidative antimicrobial activity of phagocytes resulting from mutations in gp91(phox). We detected substantial gene transfer in both individuals' neutrophils that lead to a large number of functionally corrected phagocytes and notable clinical improvement. Large-scale retroviral integration site-distribution analysis showed activating insertions in MDS1-EVI1, PRDM16 or SETBP1 that had influenced regulation of long-term hematopoiesis by expanding gene-corrected myelopoiesis three- to four-fold in both individuals. Although insertional influences have probably reinforced the therapeutic efficacy in this trial, our results suggest that gene therapy in combination with bone marrow conditioning can be successfully used to treat inherited diseases affecting the myeloid compartment such as CGD.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/physiology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Chromosomes, Human, X , Clinical Trials as Topic , Gene Transfer Techniques , Genetic Linkage , Genetic Markers , Genetic Vectors , Granulomatous Disease, Chronic/blood , Granulomatous Disease, Chronic/etiology , Granulomatous Disease, Chronic/genetics , Humans , MDS1 and EVI1 Complex Locus Protein , Mutagenesis, Insertional , Neutrophils/physiology , Proto-Oncogenes , RNA, Messenger/analysis , Retroviridae/genetics , Treatment Outcome
14.
Blood ; 99(8): 2647-52, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11929749

ABSTRACT

The transcription factor Stat5 mediates the cellular response to activation of multiple cytokine receptors involved in the regulation of proliferation and differentiation of hematopoietic cells. Recently, the human Stat5 gene was found to be translocated to the RARalpha gene in a patient with acute promyelocytic leukemia indicating that Stat5 might also play a role in cellular transformation. We investigated the mechanism by which Stat5 might exert this function and studied the biochemical and cellular functions of fusion proteins comprising Stat5 and RARalpha. The expression of Stat5-RARalpha causes the transcriptional repression of gene transcription, a process that requires the coiled-coil domain of Stat5 (amino acid positions 133-333). Oligomerization of this domain in the Stat5-RARalpha fusion protein leads to stable binding of the corepressor SMRT independent of all-trans retinoic acid (ATRA) stimulation and is accompanied by an impaired response to differentiation signals in hematopoietic cells. This inhibitory effect on myeloid differentiation cannot be overcome by simultaneous coexpression of RARalpha. We conclude that Stat5 is capable of interacting with a corepressor complex that alters the pattern of corepressor binding to RARalpha and its dissociation in response to ATRA stimulation, leading to enhanced repressor activity and a block of hematopoietic differentiation.


Subject(s)
DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Milk Proteins , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/pharmacology , Receptors, Retinoic Acid/metabolism , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Trans-Activators/genetics , Cell Differentiation/drug effects , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Leukemia, Promyelocytic, Acute/etiology , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Receptor Co-Repressor 2 , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Repressor Proteins/physiology , Retinoic Acid Receptor alpha , STAT5 Transcription Factor , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcription, Genetic/drug effects , Transfection , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...