Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Viruses ; 12(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33353185

ABSTRACT

Human parvovirus B19 (B19V) causes various human diseases, ranging from childhood benign infection to arthropathies, severe anemia and fetal hydrops, depending on the health state and hematological status of the patient. To counteract B19V blood-borne contamination, evaluation of B19 DNA in plasma pools and viral inactivation/removal steps are performed, but nucleic acid testing does not correctly reflect B19V infectivity. There is currently no appropriate cellular model for detection of infectious units of B19V. We describe here an improved cell-based method for detecting B19V infectious units by evaluating its host transcription. We evaluated the ability of various cell lines to support B19V infection. Of all tested, UT7/Epo cell line, UT7/Epo-STI, showed the greatest sensitivity to B19 infection combined with ease of performance. We generated stable clones by limiting dilution on the UT7/Epo-STI cell line with graduated permissiveness for B19V and demonstrated a direct correlation between infectivity and S/G2/M cell cycle stage. Two of the clones tested, B12 and E2, reached sensitivity levels higher than those of UT7/Epo-S1 and CD36+ erythroid progenitor cells. These findings highlight the importance of cell cycle status for sensitivity to B19V, and we propose a promising new straightforward cell-based method for quantifying B19V infectious units.


Subject(s)
Cell Cycle , Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology , Parvovirus B19, Human/physiology , Biomarkers , Cell Line , Erythroid Cells/metabolism , Erythroid Cells/virology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/virology , Gene Expression , Gene Expression Regulation, Viral , Humans , Molecular Diagnostic Techniques , Parvoviridae Infections/metabolism , Sensitivity and Specificity , Viral Tropism
2.
Exp Physiol ; 104(7): 1074-1089, 2019 07.
Article in English | MEDLINE | ID: mdl-31012180

ABSTRACT

NEW FINDINGS: What is the central question of this study? Do Fog2Rb-/Rb- mice present a defect of small intestine homeostasis? What is the main finding and its importance? The importance of interactions between FOG-2 and pRb in adipose tissue physiology has previously been demonstrated. Here it is shown that this interaction is also intrinsic to small intestine homeostasis and exerts extrinsic control over mouse metabolism. Thus, this association is involved in maintaining small intestine morphology, and regulating crypt proliferation and lineage differentiation. It therefore affects mouse growth and adaptation to a high-fat diet. ABSTRACT: GATA transcription factors and their FOG cofactors play a key role in tissue-specific development and differentiation, from worms to humans. We have shown that GATA-1 and FOG-2 contain an LXCXE pRb-binding motif. Interactions between retinoblastoma protein (pRb) and GATA-1 are crucial for erythroid proliferation and differentiation, whereas the LXCXE pRb-binding site of FOG-2 is involved in adipogenesis. Fog2-knock-in mice have defective pRb binding and are resistant to obesity, due to efficient white-into-brown fat conversion. Our aim was to investigate the pathophysiological impact of FOG-2-pRb interaction on the small intestine and mouse growth. Histological analysis of the small intestine revealed architectural changes in Fog2Rb-/Rb- mice, including villus shortening, with crypt expansion and a change in muscularis propria thickness. These differences were more marked in the proximo-distal part of the small intestine and were associated with an increase in crypt cell proliferation and disruption of the goblet and Paneth cell lineage. The small intestine of the mutants was unable to adapt to a high-fat diet, and had significantly lower plasma lipid levels on such a diet. Fog2Rb-/Rb- mice displayed higher levels of glucose-dependent insulinotropic peptide release, and lower levels of insulin-like growth factor I release on a regular diet. Their intestinal lipid absorption was impaired, resulting in restricted weight gain. In addition to the intrinsic effects of the mutation on adipose tissue, we show here an extrinsic relationship between the intestine and the effect of FOG-2 mutation on mouse metabolism. In conclusion, the interaction of FOG-2 with pRb coordinates the crypt-villus axis and controls small intestine homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Homeostasis/physiology , Intestine, Small/metabolism , Proline-Rich Protein Domains/physiology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Intestine, Small/cytology , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Protein Binding/physiology , Random Allocation , Transcription Factors/genetics
3.
N Engl J Med ; 378(16): 1479-1493, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29669226

ABSTRACT

BACKGROUND: Donor availability and transplantation-related risks limit the broad use of allogeneic hematopoietic-cell transplantation in patients with transfusion-dependent ß-thalassemia. After previously establishing that lentiviral transfer of a marked ß-globin (ßA-T87Q) gene could substitute for long-term red-cell transfusions in a patient with ß-thalassemia, we wanted to evaluate the safety and efficacy of such gene therapy in patients with transfusion-dependent ß-thalassemia. METHODS: In two phase 1-2 studies, we obtained mobilized autologous CD34+ cells from 22 patients (12 to 35 years of age) with transfusion-dependent ß-thalassemia and transduced the cells ex vivo with LentiGlobin BB305 vector, which encodes adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q). The cells were then reinfused after the patients had undergone myeloablative busulfan conditioning. We subsequently monitored adverse events, vector integration, and levels of replication-competent lentivirus. Efficacy assessments included levels of total hemoglobin and HbAT87Q, transfusion requirements, and average vector copy number. RESULTS: At a median of 26 months (range, 15 to 42) after infusion of the gene-modified cells, all but 1 of the 13 patients who had a non-ß0/ß0 genotype had stopped receiving red-cell transfusions; the levels of HbAT87Q ranged from 3.4 to 10.0 g per deciliter, and the levels of total hemoglobin ranged from 8.2 to 13.7 g per deciliter. Correction of biologic markers of dyserythropoiesis was achieved in evaluated patients with hemoglobin levels near normal ranges. In 9 patients with a ß0/ß0 genotype or two copies of the IVS1-110 mutation, the median annualized transfusion volume was decreased by 73%, and red-cell transfusions were discontinued in 3 patients. Treatment-related adverse events were typical of those associated with autologous stem-cell transplantation. No clonal dominance related to vector integration was observed. CONCLUSIONS: Gene therapy with autologous CD34+ cells transduced with the BB305 vector reduced or eliminated the need for long-term red-cell transfusions in 22 patients with severe ß-thalassemia without serious adverse events related to the drug product. (Funded by Bluebird Bio and others; HGB-204 and HGB-205 ClinicalTrials.gov numbers, NCT01745120 and NCT02151526 .).


Subject(s)
Genetic Therapy , beta-Globins/genetics , beta-Thalassemia/therapy , Adolescent , Adult , Antigens, CD34 , Child , Erythrocyte Transfusion/statistics & numerical data , Female , Gene Transfer Techniques , Genetic Vectors , Hemoglobins/analysis , Hemoglobins/genetics , Humans , Lentivirus/genetics , Male , Mutation , Transplantation, Autologous , Young Adult , beta-Thalassemia/genetics
4.
Cell Rep ; 21(12): 3524-3535, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262331

ABSTRACT

GATA transcription factors and their FOG cofactors play a key role in tissue-specific development and differentiation, from worms to humans. Mammals have six GATA and two FOG factors. We recently demonstrated that interactions between retinoblastoma protein (pRb) and GATA-1 are crucial for erythroid proliferation and differentiation. We show here that the LXCXE pRb-binding site of FOG-2 is involved in adipogenesis. Unlike GATA-1, which inhibits cell division, FOG-2 promotes proliferation. Mice with a knockin of a Fog2 gene bearing a mutated LXCXE pRb-binding site are resistant to obesity and display higher rates of white-to-brown fat conversion. Thus, each component of the GATA/FOG complex (GATA-1 and FOG-2) is involved in pRb/E2F regulation, but these molecules have markedly different roles in the control of tissue homeostasis.


Subject(s)
Adipogenesis , DNA-Binding Proteins/metabolism , Obesity/genetics , Transcription Factors/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Amino Acid Motifs , Animals , Cell Line , Cell Proliferation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Mice , Mutation , Obesity/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics
5.
N Engl J Med ; 376(9): 848-855, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28249145

ABSTRACT

Sickle cell disease results from a homozygous missense mutation in the ß-globin gene that causes polymerization of hemoglobin S. Gene therapy for patients with this disorder is complicated by the complex cellular abnormalities and challenges in achieving effective, persistent inhibition of polymerization of hemoglobin S. We describe our first patient treated with lentiviral vector-mediated addition of an antisickling ß-globin gene into autologous hematopoietic stem cells. Adverse events were consistent with busulfan conditioning. Fifteen months after treatment, the level of therapeutic antisickling ß-globin remained high (approximately 50% of ß-like-globin chains) without recurrence of sickle crises and with correction of the biologic hallmarks of the disease. (Funded by Bluebird Bio and others; HGB-205 ClinicalTrials.gov number, NCT02151526 .).


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy , beta-Globins/genetics , Adolescent , Anemia, Sickle Cell/blood , Clinical Trials as Topic , Gene Expression , Genetic Therapy/adverse effects , Genetic Vectors , Hemoglobin A/metabolism , Humans , Lentivirus , Male
6.
Biochem Biophys Res Commun ; 472(4): 624-30, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26972250

ABSTRACT

The bromodomain and extraterminal (BET) domain family proteins are epigenetic modulators involved in the reading of acetylated lysine residues. The first BET protein inhibitor to be identified, (+)-JQ1, a thienotriazolo-1, 4-diazapine, binds selectively to the acetyl lysine-binding pocket of BET proteins. We evaluated the impact on adipogenesis of this druggable targeting of chromatin epigenetic readers, by investigating the physiological consequences of epigenetic modifications through targeting proteins binding to chromatin. JQ1 significantly inhibited the differentiation of 3T3-L1 preadipocytes into white and brown adipocytes by down-regulating the expression of genes involved in adipogenesis, particularly those encoding the peroxisome proliferator-activated receptor (PPAR-γ), the CCAAT/enhancer-binding protein (C/EBPα) and, STAT5A and B. The expression of a constitutively activated STAT5B mutant did not prevent inhibition by JQ1. Thus, the association of BET/STAT5 is required for adipogenesis but STAT5 transcription activity is not the only target of JQ1. Treatment with JQ1 did not lead to the conversion of white adipose tissue into brown adipose tissue (BAT). BET protein inhibition thus interferes with generation of adipose tissue from progenitors, confirming the importance of the connections between epigenetic mechanisms and specific adipogenic transcription factors.


Subject(s)
Adipogenesis/drug effects , Azepines/pharmacology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Histone Acetyltransferases/antagonists & inhibitors , Lysine/metabolism , Triazoles/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Binding Sites/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Down-Regulation/drug effects , Histone Acetyltransferases/metabolism , Humans , Lipid Metabolism/drug effects , Mice , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Transcription Factors
7.
Genes Dev ; 29(24): 2603-16, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26680303

ABSTRACT

Tight coordination of cell proliferation and differentiation is central to red blood cell formation. Erythropoietin controls the proliferation and survival of red blood cell precursors, while variations in GATA-1/FOG-1 complex composition and concentrations drive their maturation. However, clear evidence of cross-talk between molecular pathways is lacking. Here, we show that erythropoietin activates AKT, which phosphorylates GATA-1 at Ser310, thereby increasing GATA-1 affinity for FOG-1. In turn, FOG-1 displaces pRb/E2F-2 from GATA-1, ultimately releasing free, proproliferative E2F-2. Mice bearing a Gata-1(S310A) mutation suffer from fatal anemia when a compensatory pathway for E2F-2 production involving insulin-like growth factor-1 (IGF-1) signaling is simultaneously abolished. In the context of the GATA-1(V205G) mutation resulting in lethal anemia, we show that the Ser310 cannot be phosphorylated and that constitutive phosphorylation at this position restores partial erythroid differentiation. This study sheds light on the GATA-1 pathways that synchronize cell proliferation and differentiation for tissue homeostasis.


Subject(s)
Cell Differentiation/genetics , Erythroid Cells/cytology , Erythropoiesis/physiology , Erythropoietin/metabolism , Insulin-Like Growth Factor I/metabolism , Signal Transduction , Anemia, Hemolytic/genetics , Animals , Cell Proliferation/genetics , Enzyme Activation/genetics , Erythropoiesis/genetics , Erythropoietin/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Knock-In Techniques , Mice , Mutation , Nuclear Proteins/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Protein Binding/genetics , Transcription Factors/metabolism
8.
Nature ; 525(7569): 380-3, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26331539

ABSTRACT

Whether cancer is maintained by a small number of stem cells or is composed of proliferating cells with approximate phenotypic equivalency is a central question in cancer biology. In the stem cell hypothesis, relapse after treatment may occur by failure to eradicate cancer stem cells. Chronic myeloid leukaemia (CML) is quintessential to this hypothesis. CML is a myeloproliferative disorder that results from dysregulated tyrosine kinase activity of the fusion oncoprotein BCR-ABL. During the chronic phase, this sole genetic abnormality (chromosomal translocation Ph(+): t(9;22)(q34;q11)) at the stem cell level causes increased proliferation of myeloid cells without loss of their capacity to differentiate. Without treatment, most patients progress to the blast phase when additional oncogenic mutations result in a fatal acute leukaemia made of proliferating immature cells. Imatinib mesylate and other tyrosine kinase inhibitors (TKIs) that target the kinase activity of BCR-ABL have improved patient survival markedly. However, fewer than 10% of patients reach the stage of complete molecular response (CMR), defined as the point when BCR-ABL transcripts become undetectable in blood cells. Failure to reach CMR results from the inability of TKIs to eradicate quiescent CML leukaemia stem cells (LSCs). Here we show that the residual CML LSC pool can be gradually purged by the glitazones, antidiabetic drugs that are agonists of peroxisome proliferator-activated receptor-γ (PPARγ). We found that activation of PPARγ by the glitazones decreases expression of STAT5 and its downstream targets HIF2α and CITED2, which are key guardians of the quiescence and stemness of CML LSCs. When pioglitazone was given temporarily to three CML patients in chronic residual disease in spite of continuous treatment with imatinib, all of them achieved sustained CMR, up to 4.7 years after withdrawal of pioglitazone. This suggests that clinically relevant cancer eradication may become a generally attainable goal by combination therapy that erodes the cancer stem cell pool.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/drug effects , PPAR gamma/agonists , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Thiazolidinediones/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , PPAR gamma/metabolism , Pioglitazone , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Repressor Proteins/metabolism , STAT5 Transcription Factor/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Trans-Activators/metabolism
10.
Nature ; 514(7521): 242-6, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25156257

ABSTRACT

ß-Thalassaemia major (ß-TM) is an inherited haemoglobinopathy caused by a quantitative defect in the synthesis of ß-globin chains of haemoglobin, leading to the accumulation of free α-globin chains that form toxic aggregates. Despite extensive knowledge of the molecular defects causing ß-TM, little is known of the mechanisms responsible for the ineffective erythropoiesis observed in the condition, which is characterized by accelerated erythroid differentiation, maturation arrest and apoptosis at the polychromatophilic stage. We have previously demonstrated that normal human erythroid maturation requires a transient activation of caspase-3 at the later stages of maturation. Although erythroid transcription factor GATA-1, the master transcriptional factor of erythropoiesis, is a caspase-3 target, it is not cleaved during erythroid differentiation. We have shown that, in human erythroblasts, the chaperone heat shock protein70 (HSP70) is constitutively expressed and, at later stages of maturation, translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. The primary role of this ubiquitous chaperone is to participate in the refolding of proteins denatured by cytoplasmic stress, thus preventing their aggregation. Here we show in vitro that during the maturation of human ß-TM erythroblasts, HSP70 interacts directly with free α-globin chains. As a consequence, HSP70 is sequestrated in the cytoplasm and GATA-1 is no longer protected, resulting in end-stage maturation arrest and apoptosis. Transduction of a nuclear-targeted HSP70 mutant or a caspase-3-uncleavable GATA-1 mutant restores terminal maturation of ß-TM erythroblasts, which may provide a rationale for new targeted therapies of ß-TM.


Subject(s)
Erythroblasts/metabolism , Erythropoiesis , HSP70 Heat-Shock Proteins/metabolism , alpha-Globins/metabolism , beta-Thalassemia/blood , beta-Thalassemia/metabolism , Apoptosis , Bone Marrow/metabolism , Caspase 3/metabolism , Cell Nucleus/metabolism , Cell Survival/genetics , Cells, Cultured , Cytoplasm/metabolism , Enzyme Activation , Erythroblasts/cytology , Erythroblasts/pathology , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , Humans , Kinetics , Molecular Targeted Therapy , Protein Binding , Protein Refolding , beta-Thalassemia/pathology
11.
Stem Cells ; 31(9): 1785-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712774

ABSTRACT

A patient with ß(E)/ß(0) -thalassemia major was converted to transfusion-independence 4.5 years ago by lentiviral gene transfer in hematopoietic stem cells while showing a myeloid-biased cell clone. Induced pluripotent stem cells (iPSCs) are a potential alternative source of hematopoietic stem cells. If fetal to adult globin class, switching does not occur in vivo in iPSC-derived erythroid cells, ß-globin gene transfer would be unnecessary. To investigate both vector integration skewing and the potential use of iPSCs for the treatment of thalassemia, we derived iPSCs from the thalassemia gene therapy patient and compared iPSC-derived hematopoietic cells to their natural isogenic somatic counterparts. In NSG immunodeficient mice, embryonic to fetal and a partial fetal to adult globin class switching were observed, indicating that the gene transfer is likely necessary for iPSC-based therapy of the ß-hemoglobinopathies. Lentivector integration occurred in regions of low and high genotoxicity. Surprisingly, common integration sites (CIS) were identified across those iPSCs and cells retrieved from isogenic and nonisogenic gene therapy patients with ß-thalassemia and adrenoleukodystrophy, respectively. This suggests that CIS observed in the absence of overt tumorigenesis result from nonrandom lentiviral integration rather than oncogenic in vivo selection. These findings bring the use of iPSCs closer to practicality and further clarify our interpretation of genome-wide lentivector integration.


Subject(s)
Globins/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Lentivirus/metabolism , Transduction, Genetic , beta-Thalassemia/pathology , Adult , Animals , Cell Differentiation/drug effects , Erythroid Cells/cytology , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Gene Expression Regulation/drug effects , Genetic Vectors/metabolism , Globins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Mutagens/toxicity , Regeneration/drug effects , Virus Integration/drug effects
12.
Stem Cells ; 31(10): 2162-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23554255

ABSTRACT

Our understanding of system dynamics of mixed cell populations in whole organisms has benefited from the advent of individual cell marking by nonarrayed DNA barcodes subsequently analyzed by high-throughput DNA sequencing. However, key limitations include statistical biases compromising quantification and the lack of applicability to deconvolute individual cell fate in vivo after pooling single cells differentially exposed to different conditions ex vivo. Here, we have derived an arrayed lentiviral library of DNA barcodes and obtained a proof-of-concept of its resolving capacity by quantifying hematopoietic regeneration after engraftment of mice with genetically modified autologous cells. This method has helped clarify and bridge the seemingly opposed clonal-succession and continuous-recruitment models of hematopoietic stem cell behavior and revealed that myeloid-lymphoid biases are common occurrences in steady-state hematopoiesis. Arrayed lentiviral barcoding should prove a versatile and powerful approach to deconvolute cell dynamics in vivo with applications in hematology, embryology, and cancer biology.


Subject(s)
Hematopoietic Stem Cells/physiology , Lentivirus/genetics , Animals , Cell Tracking/methods , DNA Barcoding, Taxonomic , Genetic Vectors , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
13.
Biochem Biophys Res Commun ; 429(1-2): 1-5, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23137537

ABSTRACT

Malignant transformation is a multistep process requiring oncogenic activation, promoting cellular proliferation, frequently coupled to inhibition of terminal differentiation. Consequently, forcing the reengagement of terminal differentiation of transformed cells coupled or not with an inhibition of their proliferation is a putative therapeutic approach to counteracting tumorigenicity. UT7 is a human leukemic cell line able to grow in the presence of IL3, GM-CSF and Epo. This cell line has been widely used to study Epo-R/Epo signaling pathways but is a poor model for erythroid differentiation. We used the BET bromodomain inhibition drug JQ1 to target gene expression, including that of c-Myc. We have shown that only 2 days of JQ1 treatment was required to transitory inhibit Epo-induced UT7 proliferation and to restore terminal erythroid differentiation. This study highlights the importance of a cellular erythroid cycle break mediated by c-Myc inhibition before initiation of the erythropoiesis program and describes a new model for BET bromodomain inhibitor drug application.


Subject(s)
Azepines/pharmacology , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Leukemia, Erythroblastic, Acute/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Triazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Humans , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism
14.
Nature ; 467(7313): 318-22, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20844535

ABSTRACT

The ß-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of ß-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound ß(E)/ß(0)-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The ß(E)-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated ß(E)-globin with partial instability. When this is compounded with a non-functional ß(0) allele, a profound decrease in ß-globin synthesis results, and approximately half of ß(E)/ß(0)-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral ß-globin gene transfer, an adult patient with severe ß(E)/ß(0)-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl(-1), of which one-third contains vector-encoded ß-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Subject(s)
Blood Transfusion , Genetic Therapy , HMGA2 Protein/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Adolescent , Blood Cells/cytology , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Child, Preschool , Clone Cells/metabolism , Gene Expression , Genetic Vectors/genetics , HMGA2 Protein/genetics , Homeostasis , Humans , Lentivirus/genetics , Male , MicroRNAs/genetics , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/genetics , Time Factors , Transcriptional Activation , Young Adult , beta-Thalassemia/metabolism
15.
PLoS Biol ; 7(6): e1000123, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19513100

ABSTRACT

How cell proliferation subsides as cells terminally differentiate remains largely enigmatic, although this phenomenon is central to the existence of multicellular organisms. Here, we show that GATA-1, the master transcription factor of erythropoiesis, forms a tricomplex with the retinoblastoma protein (pRb) and E2F-2. This interaction requires a LXCXE motif that is evolutionary conserved among GATA-1 orthologs yet absent from the other GATA family members. GATA-1/pRb/E2F-2 complex formation stalls cell proliferation and steers erythroid precursors towards terminal differentiation. This process can be disrupted in vitro by FOG-1, which displaces pRb/E2F-2 from GATA-1. A GATA-1 mutant unable to bind pRb fails to inhibit cell proliferation and results in mouse embryonic lethality by anemia. These findings clarify the previously suspected cell-autonomous role of pRb during erythropoiesis and may provide a unifying molecular mechanism for several mouse phenotypes and human diseases associated with GATA-1 mutations.


Subject(s)
E2F2 Transcription Factor/metabolism , Erythropoiesis , GATA1 Transcription Factor/metabolism , Retinoblastoma Protein/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Division , Cell Proliferation , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/deficiency , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Proteins/metabolism , Protein Binding , Retinoblastoma Protein/deficiency , Transcription Factors/metabolism
16.
Mol Cell Biol ; 25(17): 7412-22, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16107690

ABSTRACT

The contribution of erythropoietin to the differentiation of the red blood cell lineage remains elusive, and the demonstration of a molecular link between erythropoietin and the transcription of genes associated with erythroid differentiation is lacking. In erythroid cells, expression of the tissue inhibitor of matrix metalloproteinase (TIMP-1) is strictly dependent on erythropoietin. We report here that erythropoietin regulates the transcription of the TIMP-1 gene upon binding to its receptor in erythroid cells by triggering the activation of phosphatidylinositol 3-kinase (PI3K)/Akt. We found that Akt directly phosphorylates the transcription factor GATA-1 at serine 310 and that this site-specific phosphorylation is required for the transcriptional activation of the TIMP-1 promoter. This chain of events can be recapitulated in nonerythroid cells by transfection of the implicated molecular partners, resulting in the expression of the normally silent endogenous TIMP-1 gene. Conversely, TIMP-1 secretion is profoundly decreased in erythroid cells from fetal livers of transgenic knock-in mice homozygous for a GATA(S310A) gene, which encodes a GATA-1 mutant that cannot be phosphorylated at Ser(310). Furthermore, retrovirus-mediated expression of GATA(S310A) into GATA-1(null)-derived embryonic stem cells decreases the rate of hemoglobinization by more than 50% compared to expressed wild-type GATA-1. These findings provide the first example of a chain of coupling mechanisms between the binding of erythropoietin to its receptor and GATA-1-dependent gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Cells/metabolism , Erythropoietin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/chemistry , Erythroid Cells/cytology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Erythropoietin/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/deficiency , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transcription Factors/chemistry , Transcription, Genetic/genetics
17.
Blood ; 105(10): 4035-42, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15677568

ABSTRACT

Myelodysplastic syndromes (MDSs) are characterized by peripheral blood cytopenia including anemia. We have investigated the implication of the extrinsic pathway of apoptosis in MDS-ineffective erythropoiesis by in vitro expansion of erythroid precursors from early stage (low and intermediate-1 International Prognosis Scoring System [IPSS]) MDS, advanced stage (intermediate-2 IPSS) MDS, and control bone marrow samples. We have previously shown that Fas and its ligand were overexpressed in early stage MDS erythroid cells. Here, we show that caspase-8 activity is significantly increased, whereas the expression of death receptors other than Fas, including the type 1 receptor for tumor necrosis factor alpha (TNF-alpha) and the receptors for the TNF-related apoptosis-inducing ligand (TRAIL), DR4 and DR5, was normal. We also observed that the adapter Fas-associated death domain (FADD) was overexpressed in early stage MDS erythroid cells. Transduction of early stage MDS-derived CD34+ progenitors with a FADD-encoding construct increased apoptosis of erythroid cells and dramatically reduced erythroid burst-forming unit (BFU-E) growth. Transduction of a dominant-negative (dn) mutant of FADD inhibited caspase-8 activity and cell death and rescued BFU-E growth without abrogating erythroid differentiation. These results extend the observation that Fas-dependent activation of caspase-8 accounts for apoptosis of early stage MDS erythroid cells and demonstrate for the first time that FADD is a valuable target to correct ineffective erythropoiesis in these syndromes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Erythroid Cells/metabolism , Erythroid Cells/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/therapy , Adult , Aged , Aged, 80 and over , Apoptosis , Apoptosis Regulatory Proteins , Caspase 8 , Caspases/metabolism , Cells, Cultured , Fas-Associated Death Domain Protein , Female , Gene Expression Regulation, Neoplastic , Genes, Dominant/genetics , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasm Staging , Stem Cells/cytology , Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism
18.
Blood ; 105(2): 600-8, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15358619

ABSTRACT

Activation of the erythropoietin receptor (EpoR) after Epo binding is very transient because of the rapid activation of strong down-regulation mechanisms that quickly decrease Epo sensitivity of the cells. Among these down-regulation mechanisms, receptor internalization and degradation are probably the most efficient. Here, we show that the Epo receptor was rapidly ubiquitinated after ligand stimulation and that the C-terminal part of the Epo receptor was degraded by the proteasomes. Both ubiquitination and receptor degradation by the proteasomes occurred at the cell surface and required Janus kinase 2 (Jak2) activation. Moreover, Epo-EpoR complexes were rapidly internalized and targeted to the lysosomes for degradation. Neither Jak2 nor proteasome activities were required for internalization. In contrast, Jak2 activation was necessary for lysosome targeting of the Epo-EpoR complexes. Blocking Jak2 with the tyrphostin AG490 led to some recycling of internalized Epo-Epo receptor complexes to the cell surface. Thus, activated Epo receptors appear to be quickly degraded after ubiquitination by 2 proteolytic systems that proceed successively: the proteasomes remove part of the intracellular domain at the cell surface, and the lysosomes degrade the remaining part of the receptor-hormone complex. The efficiency of these processes probably explains the short duration of intracellular signaling activated by Epo.


Subject(s)
Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Erythropoietin/metabolism , Cells, Cultured , Erythropoietin/metabolism , Erythropoietin/pharmacology , Humans , Iodine Radioisotopes , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism , Receptors, Erythropoietin/chemistry , Ubiquitin/metabolism
20.
J Biol Chem ; 278(13): 11050-6, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12538595

ABSTRACT

We have recently shown that a heterotrimeric G(i) protein is coupled to the erythropoietin (Epo) receptor. The G(i) protein constitutively associates in its heterotrimeric form with the intracellular domain of Epo receptor (EpoR). After Epo stimulation G(i) is released from the receptor and activated. In the present study we have investigated the functional role of the heterotrimeric G(i) protein bound to EpoR. In Chinese hamster ovary cells expressing EpoR, the G(i) inhibitor pertussis toxin blocked mitogen-activated protein kinase (MAPK) Erk1/2 activation induced by Epo. Epo-dependent MAPK activation was also sensitive to the G beta gamma competitive inhibitor beta ARK1-ct (C-terminal fragment of the beta-adrenergic receptor kinase), to the Ras dominant negative mutant RasN17, and to the phosphoinositide 3-kinase (PI3K) inhibitor LY 294002. A region of 7 amino acids (469-475) in the C-terminal end of EpoR was shown to be required for G(i) binding to EpoR in vivo. Deletion of this region in EpoR abolished both MAPK and PI3K activation in response to Epo. We conclude that in Chinese hamster ovary cells, Epo activates MAPK via a novel pathway dependent on G(i) association to EpoR, G beta gamma subunit, Ras, and PI3K. The tyrosine kinase Jak2 also contributes to this new pathway, more likely downstream of beta gamma and upstream of Ras and PI3K. This pathway is similar to the best characterized pathway used by seven transmembrane receptors coupled to G(i) to activate MAPK and may cooperate with other described Epo-dependent MAPK activation pathways in hematopoietic cells.


Subject(s)
GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Erythropoietin/physiology , Animals , CHO Cells , Cricetinae , Enzyme Activation , Mitogen-Activated Protein Kinase 3 , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...