Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Viruses ; 16(1)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38257782

ABSTRACT

Coagulation disorders are described in COVID-19 and long COVID patients. In particular, SARS-CoV-2 infection in megakaryocytes, which are precursors of platelets involved in thrombotic events in COVID-19, long COVID and, in rare cases, in vaccinated individuals, requires further investigation, particularly with the emergence of new SARS-CoV-2 variants. CD147, involved in the regulation of inflammation and required to fight virus infection, can facilitate SARS-CoV-2 entry into megakaryocytes. MCT4, a co-binding protein of CD147 and a key player in the glycolytic metabolism, could also play a role in SARS-CoV-2 infection. Here, we investigated the susceptibility of megakaryocytes to SARS-CoV-2 infection via CD147 and MCT4. We performed infection of Dami cells and human CD34+ hematopoietic progenitor cells induced to megakaryocytic differentiation with SARS-CoV-2 pseudovirus in the presence of AC-73 and syrosingopine, respective inhibitors of CD147 and MCT4 and inducers of autophagy, a process essential in megakaryocyte differentiation. Both AC-73 and syrosingopine enhance autophagy during differentiation but only AC-73 enhances megakaryocytic maturation. Importantly, we found that AC-73 or syrosingopine significantly inhibits SARS-CoV-2 infection of megakaryocytes. Altogether, our data indicate AC-73 and syrosingopine as inhibitors of SARS-CoV-2 infection via CD147 and MCT4 that can be used to prevent SARS-CoV-2 binding and entry into megakaryocytes, which are precursors of platelets involved in COVID-19-associated coagulopathy.


Subject(s)
Megakaryocytes , Phenols , Reserpine , SARS-CoV-2 , Humans , COVID-19 , Megakaryocytes/virology , Phenols/pharmacology , Post-Acute COVID-19 Syndrome , Reserpine/analogs & derivatives , Reserpine/pharmacology , SARS-CoV-2/drug effects , Virus Internalization/drug effects
2.
PLoS One ; 16(7): e0254878, 2021.
Article in English | MEDLINE | ID: mdl-34293019

ABSTRACT

Musculoskeletal injuries, a public health priority also in the military context, are ascribed to several risk factors, including: increased reaction forces; low/reduced muscle strength, endurance, body mass, Vitamin D level, and bone density; inadequate lifestyles and environment. The MOVIDA Project-funded by the Italian Ministry of Defence-aims at developing a transportable toolkit (assessment instrumentation, assessment protocols and reference/risk thresholds) which integrates motor function assessment with biological, environmental and behavioural factors to help characterizing the risk of stress fracture, stress injury or muscle fatigue due to mechanical overload. The MOVIDA study has been designed following the STROBE guidelines for observational cross-sectional studies addressing healthy adults, both militaries and civilians, with varying levels of physical fitness (sedentary people, recreational athletes, and competitive athletes). The protocol of the study has been designed and validated and is hereby reported. It allows to collect and analyse anamnestic, diagnostic and lifestyle-related data, environmental parameters, and functional parameters measured through portable and wearable instrumentation during adapted 6 minutes walking test. The t-test, one and two-way ANOVA with post-hoc corrections, and ANCOVA tests will be used to investigate relevant differences among the groups with respect to biomechanical parameters; non-parametric statistics will be rather used for non-normal continuous variables and for quantitative discrete variables. Generalized linear models will be used to account for risk and confounding factors.


Subject(s)
Athletes , Athletic Performance , Physical Fitness , Vitamin D/blood , Adolescent , Adult , Cross-Sectional Studies , Female , Humans , Male , Risk Assessment , Risk Factors
4.
Front Immunol ; 8: 1946, 2017.
Article in English | MEDLINE | ID: mdl-29375570

ABSTRACT

Platelets (PLTs) are the major source of high-mobility group box 1 (HMGB1), a protein that is involved in sterile inflammation of blood vessels and thrombosis. Megakaryocytes (MKs) synthesize HMGB1 and transfer both protein and mRNA into PLTs and PLT-derived microvesicles (MV). Free HMGB1 found in supernatants of in vitro differentiated MKs and in a megakaryoblastic cell line (DAMI cells). Aspirin "in vivo" and "in vitro" not only reduces HMGB1 and receptor for advanced glycation end products expression on MKs and PLTs but also drives the movement of HMGB1 from MKs into PLTs and PLT-derived MV. These findings suggest that consumption of low doses of aspirin reduces the risk of atherosclerosis complications as well as reducing PLT aggregation by the inhibition of COX-1.

6.
Cancer Cell Int ; 13(1): 101, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24148231

ABSTRACT

BACKGROUND: Homeobox (HOX) genes deregulation has been largely implicated in the development of human leukemia. Among the HOXB cluster, HOXB1 was silent in a number of analyzed acute myeloid leukemia (AML) primary cells and cell lines, whereas it was expressed in normal terminally differentiated peripheral blood cells. METHODS: We evaluated the biological effects and the transcriptome changes determined by the retroviral transduction of HOXB1 in the human promyelocytic cell line HL60. RESULTS: Our results suggest that the enforced expression of HOXB1 reduces cell growth proliferation, inducing apoptosis and cell differentiation along the monocytic and granulocytic lineages. Accordingly, gene expression analysis showed the HOXB1-dependent down-regulation of some tumor promoting genes, paralleled by the up-regulation of apoptosis- and differentiation-related genes, thus supporting a tumor suppressor role for HOXB1 in AML. Finally, we indicated HOXB1 promoter hypermethylation as a mechanism responsible for HOXB1 silencing. CONCLUSIONS: We propose HOXB1 as an additional member of the HOX family with tumour suppressor properties suggesting a HOXB1/ATRA combination as a possible future therapeutic strategy in AML.

7.
PLoS One ; 8(4): e60436, 2013.
Article in English | MEDLINE | ID: mdl-23593217

ABSTRACT

MicroRNAs (miRNAs) play key roles in modulating a variety of cellular processes through repression of mRNAs target. The functional relevance of microRNAs has been proven in normal and malignant hematopoiesis. While analyzing miRNAs expression profile in unilineage serum-free liquid suspension unilineage cultures of peripheral blood CD34(+) hematopoietic progenitor cells (HPCs) through the erythroid, megakaryocytic, granulocytic and monocytic pathways, we identified miR-486-3p as mainly expressed within the erythroid lineage. We showed that miR-486-3p regulates BCL11A expression by binding to the extra-long isoform of BCL11A 3'UTR. Overexpression of miR-486-3p in erythroid cells resulted in reduced BCL11A protein levels, associated to increased expression of γ-globin gene, whereas inhibition of physiological miR-486-3p levels increased BCL11A and, consequently, reduced γ-globin expression. Thus, miR-486-3p regulating BCL11A expression might contributes to fetal hemoglobin (HbF) modulation and arise the question as to what extent this miRNA might contribute to different HbF levels observed among ß-thalassemia patients. Erythroid cells, differentiated from PB CD34(+) cells of a small cohort of patients affected by major or intermedia ß-thalassemia, showed miR-486-3p levels significantly higher than those observed in normal counterpart. Importantly, in these patients, miR-486-3p expression correlates with increased HbF synthesis. Thus, our data indicate that miR-486-3p might contribute to different HbF levels observed among thalassemic patients and, possibly, to the clinical severity of the disease.


Subject(s)
Carrier Proteins/genetics , Erythroid Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Nuclear Proteins/genetics , gamma-Globins/genetics , Alternative Splicing , Antigens, CD34/metabolism , Base Pairing , Base Sequence , Cell Line , Cell Lineage/genetics , Cells, Cultured , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Knockout Techniques , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Models, Biological , RNA Isoforms , Repressor Proteins , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
8.
PLoS One ; 7(7): e39796, 2012.
Article in English | MEDLINE | ID: mdl-22792187

ABSTRACT

The tyrosine kinase Tie-2 and its ligands Angiopoietins (Angs) transduce critical signals for angiogenesis in endothelial cells. This receptor and Ang-1 are coexpressed in hematopoietic stem cells and in a subset of megakaryocytes, though a possible role of angiopoietins in megakaryocytic differentiation/proliferation remains to be demonstrated. To investigate a possible effect of Ang-1/Ang-2 on megakaryocytic proliferation/differentiation we have used both normal CD34(+) cells induced to megakaryocytic differentiation and the UT7 cells engineered to express the thrombopoietin receptor (TPOR, also known as c-mpl, UT7/mpl). Our results indicate that Ang-1/Ang-2 may have a role in megakaryopoiesis. Particularly, Ang-2 is predominantly produced and released by immature normal megakaryocytic cells and by undifferentiated UT7/mpl cells and slightly stimulated TPO-induced cell proliferation. Ang-1 production is markedly induced during megakaryocytic differentiation/maturation and potentiated TPO-driven megakaryocytic differentiation. Blocking endogenously released angiopoietins partially inhibited megakaryocytic differentiation, particularly for that concerns the process of polyploidization. According to these data it is suggested that an autocrine angiopoietin/Tie-2 loop controls megakaryocytic proliferation and differentiation.


Subject(s)
Angiopoietins/metabolism , Autocrine Communication , Cell Differentiation , Megakaryocytes/cytology , Megakaryocytes/metabolism , Angiopoietins/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/drug effects , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Signal Transduction/drug effects , Thrombopoietin/pharmacology
9.
Haematologica ; 95(8): 1253-60, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20305142

ABSTRACT

BACKGROUND: The human hemoglobin switch (HbF-->HbA) takes place in the peri/post-natal period. In adult life, however, the residual HbF (<1%) may be partially reactivated by chemical inducers and/or cytokines such as the kit ligand (KL). MicroRNAs (miRs) play a pivotal role in normal hematopoiesis: downmodulation of miR-221/222 stimulates human erythropoietic proliferation through upmodulation of the kit receptor. DESIGN AND METHODS: We have explored the possible role of kit/KL in perinatal Hb switching by evaluating: i) the expression levels of both kit and kit ligand on CD34(+) cells and in plasma isolated from pre-, mid- and full-term cord blood samples; ii) the reactivation of HbF synthesis in KL-treated unilineage erythroid cell cultures; iii) the functional role of miR-221/222 in HbF production. RESULTS: In perinatal life, kit expression showed a gradual decline directly correlated to the decrease of HbF (from 80-90% to <30%). Moreover, in full-term cord blood erythroid cultures, kit ligand induced a marked increase of HbF (up to 80%) specifically abrogated by addition of the kit inhibitor imatinib, thus reversing the Hb switch. MiR-221/222 expression exhibited rising levels during peri/post-natal development. In functional studies, overexpression of these miRs in cord blood progenitors caused a remarkable decrease in kit expression, erythroblast proliferation and HbF content, whereas their suppression induced opposite effects. CONCLUSIONS: Our studies indicate that human perinatal Hb switching is under control of the kit receptor/miR 221-222 complex. We do not exclude, however, that other mechanisms (i.e. glucocorticoids and the HbF inhibitor BCL11A) may also contribute to the peri/post-natal Hb switch.


Subject(s)
Fetal Hemoglobin/metabolism , Hemoglobin A/metabolism , MicroRNAs/physiology , Stem Cell Factor/physiology , Adult , Antigens, CD34/blood , Benzamides , Cell Cycle , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Erythropoiesis/drug effects , Erythropoiesis/genetics , Fetal Blood/cytology , Fetal Blood/metabolism , Flow Cytometry , Gene Expression , Humans , Imatinib Mesylate , Infant, Newborn , MicroRNAs/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/blood , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Factor/blood , Stem Cell Factor/genetics , Time Factors
10.
Blood ; 114(9): 1753-63, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19542302

ABSTRACT

It is generally conceded that selective combinations of transcription factors determine hematopoietic lineage commitment and differentiation. Here we show that in normal human hematopoiesis the transcription factor nuclear factor I-A (NFI-A) exhibits a marked lineage-specific expression pattern: it is upmodulated in the erythroid (E) lineage while fully suppressed in the granulopoietic (G) series. In unilineage E culture of hematopoietic progenitor cells (HPCs), NFI-A overexpression or knockdown accelerates or blocks erythropoiesis, respectively: notably, NFI-A overexpression restores E differentiation in the presence of low or minimal erythropoietin stimulus. Conversely, NFI-A ectopic expression in unilineage G culture induces a sharp inhibition of granulopoiesis. Finally, in bilineage E + G culture, NFI-A overexpression or suppression drives HPCs into the E or G differentiation pathways, respectively. These NFI-A actions are mediated, at least in part, by a dual and opposite transcriptional action: direct binding and activation or repression of the promoters of the beta-globin and G-CSF receptor gene, respectively. Altogether, these results indicate that, in early hematopoiesis, the NFI-A expression level acts as a novel factor channeling HPCs into either the E or G lineage.


Subject(s)
Erythrocytes/metabolism , Gene Expression Regulation , Granulocytes/metabolism , Hematopoietic Stem Cells/cytology , NFI Transcription Factors/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , beta-Globins/metabolism , Antigens, CD34/biosynthesis , Cell Differentiation , Cell Lineage , Erythropoietin/metabolism , Fetal Blood/metabolism , Humans , Models, Biological , Promoter Regions, Genetic
11.
Haematologica ; 94(4): 479-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19278969

ABSTRACT

BACKGROUND: MicroRNAs are small non-coding RNAs that regulate gene expression through mRNA degradation or translational inhibition. MicroRNAs are emerging as key regulators of normal hematopoiesis and hematologic malignancies. Several miRNAs are differentially expressed during hematopoiesis and their specific expression regulates key functional proteins involved in hematopoietic lineage differentiation. This study focused on the functional role of microRNA-223 (miR-223) on erythroid differentiation. DESIGN AND METHODS: Purified cord blood CD34+ hematopoietic progenitor cells were grown in strictly controlled conditions in the presence of saturating dosage of erythropoietin to selectively induce erythroid differentiation. The effects of enforced expression of miR-223 in unilin-eage erythroid cultures were evaluated in liquid phase culture experiments and clonogenic studies. RESULTS: In unilineage erythroid culture of cord blood CD34+ hematopoietic progenitor cells miR-223 is down-regulated, whereas LMO2, an essential protein for erythroid differentiation, is up-regulated. Functional studies showed that enforced expression of miR-223 reduces the mRNA and protein levels of LMO2, by binding to LMO2 3' UTR, and impairs differentiation of erythroid cells. Accordingly, knockdown of LMO2 by short interfering RNA mimics the action of miR-223. Furthermore, hematopoietic progenitor cells transduced with miR-223 showed a significant reduction of their erythroid clonogenic capacity, suggesting that downmodulation of this miRNA is required for erythroid progenitor recruitment and commitment. CONCLUSIONS: These results show that the decline of miR-223 is an important event for erythroid differentiation that leads to the expansion of erythroblast cells at least partially mediated by unblocking LMO2 protein expression.


Subject(s)
DNA-Binding Proteins/genetics , Erythropoiesis , Metalloproteins/genetics , MicroRNAs/physiology , Adaptor Proteins, Signal Transducing , Cell Differentiation , Erythroid Cells , Fetal Blood , Gene Expression Regulation , Humans , LIM Domain Proteins , Proto-Oncogene Proteins
12.
Blood ; 111(1): 421-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17951528

ABSTRACT

In human beta-thalassemia, the imbalance between alpha- and non-alpha-globin chains causes ineffective erythropoiesis, hemolysis, and anemia: this condition is effectively treated by an enhanced level of fetal hemoglobin (HbF). In spite of extensive studies on pharmacologic induction of HbF synthesis, clinical trials based on HbF reactivation in beta-thalassemia produced inconsistent results. Here, we investigated the in vitro response of beta-thalassemic erythroid progenitors to kit ligand (KL) in terms of HbF reactivation, stimulation of effective erythropoiesis, and inhibition of apoptosis. In unilineage erythroid cultures of 20 patients with intermedia or major beta-thalassemia, addition of KL, alone or combined with dexamethasone (Dex), remarkably stimulated cell proliferation (3-4 logs more than control cultures), while decreasing the percentage of apoptotic and dyserythropoietic cells (<5%). More important, in both thalassemic groups, addition of KL or KL plus Dex induced a marked increase of gamma-globin synthesis, thus reaching HbF levels 3-fold higher than in con-trol cultures (eg, from 27% to 75% or 81%, respectively, in beta-thalassemia major). These studies indicate that in beta-thalassemia, KL, alone or combined with Dex, induces an expansion of effective erythropoiesis and the reactivation of gamma-globin genes up to fetal levels and may hence be considered as a potential therapeutic agent for this disease.


Subject(s)
Erythropoiesis/drug effects , Fetal Hemoglobin/physiology , Stem Cell Factor/pharmacology , beta-Thalassemia/blood , beta-Thalassemia/drug therapy , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Dexamethasone/pharmacology , Drug Therapy, Combination , Erythrocytes/cytology , Erythrocytes/physiology , Glucocorticoids/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans
13.
Haematologica ; 90(12): 1595-606, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16330432

ABSTRACT

BACKGROUND AND OBJECTIVES: Ferroportin-1 (FPN1) is expressed in various types of cells that play critical roles in mammalian iron metabolism and appears to act as an iron exporter in these tissues. The aim of this study was to investigate whether erythroid cells possess specific mechanisms for iron export. DESIGN AND METHODS: The expression of FPN1 during human erythroid differentiation, the characterization of alternative transcripts, the modulation by iron and the subcellular localization of this protein were studied. RESULTS: FPN1 mRNA and protein are highly expressed during human erythroid differentiation. The iron-responsive element (IRE) in the 5'- untranslated region (UTR) of FPN1 mRNA is functional but, in spite of that, FPN1 protein expression, as well as mRNA level and half-life, seem not to be affected by iron. To explain these apparenthy discordant results we searched for alternative transcripts of FPN1 and found at least three different types of transcripts, displaying alternative 5' ends. Most of the FPN1 transcripts code for the canonical protein, but only half of them contain an IRE in the 5'-UTR and have the potential to be translationally regulated by iron. Expression analysis shows that alternative FPN1 transcripts are differentially expressed during erythroid differentiation. Finally, sustained expression of alternative FPN1 transcripts is apparently observed only in erythroid cells. INTERPRETATION AND CONCLUSIONS: This is the first report describing the presence of FPN1 in erythroid cells at all stages of differentiation, providing evidence that erythroid cells possess specific mechanisms of iron export. The existence of multiple FPN1 transcripts indicates a complex regulation of the FPN1 gene in erythroid cells.


Subject(s)
Alternative Splicing , Cation Transport Proteins/biosynthesis , Erythroid Cells/metabolism , Gene Expression Regulation , Iron/blood , 5' Untranslated Regions/genetics , Amino Acid Sequence , Base Sequence , Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , Cell Differentiation/genetics , Cells, Cultured/cytology , Cells, Cultured/metabolism , Deferoxamine/pharmacology , Erythroid Cells/cytology , Erythropoiesis , Exons/genetics , Gene Expression Regulation, Neoplastic , Genes, Reporter , Half-Life , Humans , Iron Chelating Agents/pharmacology , K562 Cells/cytology , K562 Cells/metabolism , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Organ Specificity , Protein Biosynthesis , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Subcellular Fractions/chemistry , Subcellular Fractions/ultrastructure , Transcription, Genetic , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism
14.
Proc Natl Acad Sci U S A ; 102(50): 18081-6, 2005 Dec 13.
Article in English | MEDLINE | ID: mdl-16330772

ABSTRACT

MicroRNAs (miRs) are small noncoding RNAs that regulate gene expression primarily through translational repression. In erythropoietic (E) culture of cord blood CD34+ progenitor cells, the level of miR 221 and 222 is gradually and sharply down-modulated. Hypothetically, this decline could promote erythropoiesis by unblocking expression of key functional proteins. Indeed, (i) bioinformatic analysis suggested that miR 221 and 222 target the 3' UTR of kit mRNA; (ii) the luciferase assay confirmed that both miRs directly interact with the kit mRNA target site; and (iii) in E culture undergoing exponential cell growth, miR down-modulation is inversely related to increasing kit protein expression, whereas the kit mRNA level is relatively stable. Functional studies show that treatment of CD34+ progenitors with miR 221 and 222, via oligonucleotide transfection or lentiviral vector infection, causes impaired proliferation and accelerated differentiation of E cells, coupled with down-modulation of kit protein: this phenomenon, observed in E culture releasing endogenous kit ligand, is magnified in E culture supplemented with kit ligand. Furthermore, transplantation experiments in NOD-SCID mice reveal that miR 221 and 222 treatment of CD34+ cells impairs their engraftment capacity and stem cell activity. Finally, miR 221 and 222 gene transfer impairs proliferation of the kit+ TF-1 erythroleukemic cell line. Altogether, our studies indicate that the decline of miR 221 and 222 during exponential E growth unblocks kit protein production at mRNA level, thus leading to expansion of early erythroblasts. Furthermore, the results on kit+ erythroleukemic cells suggest a potential role of these miRs in cancer therapy.


Subject(s)
Erythropoiesis/physiology , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Animals , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Proliferation , Computational Biology , Erythropoiesis/genetics , Fetal Blood/cytology , Gene Expression Profiling , Humans , Luciferases , Mice , Mice, Inbred NOD , Mice, SCID
15.
Biochem J ; 381(Pt 3): 629-34, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15084147

ABSTRACT

Human TFR2 (transferrin receptor 2) is a membrane-bound protein homologous with TFR1. High levels of TFR2 mRNA were found mainly in the liver and, to a lesser extent, in erythroid precursors. However, although the presence of the TFR2 protein in hepatic cells has been confirmed in several studies, evidence is lacking about the presence of the TFR2 protein in normal erythroid cells. Using two anti-TFR2 monoclonal antibodies, G/14C2 and G/14E8, we have provided evidence that TFR2 protein is not expressed in normal erythroid cells at any stage of differentiation, from undifferentiated CD34+ cells to mature orthochromatic erythroblasts. In contrast, erythroleukaemic cells (K562 cells) exhibited a high level of expression of TFR2 at both the mRNA and the protein level. We can therefore conclude that an elevated expression of TFR2 protein is observed in leukaemic cells, but not in normal erythroblasts. The implications of this observation for the understanding of the phenotypic features of haemochromatosis due to mutation of the TFR2 gene are discussed.


Subject(s)
Erythroid Cells/chemistry , Erythroid Cells/metabolism , Receptors, Transferrin/biosynthesis , Antibodies/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Erythroid Cells/cytology , Erythroid Cells/pathology , Erythroid Precursor Cells/chemistry , Erythroid Precursor Cells/metabolism , Humans , K562 Cells , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Molecular Weight , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/immunology , RNA, Messenger/biosynthesis , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Subcellular Fractions/chemistry
16.
Blood ; 101(7): 2826-32, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12424200

ABSTRACT

Mechanisms underlying fetal hemoglobin (HbF) reactivation in stress erythropoiesis have not been fully elucidated. We suggested that a key role is played by kit ligand (KL). Because glucocorticoids (GCs) mediate stress erythropoiesis, we explored their capacity to potentiate the stimulatory effect of KL on HbF reactivation, as evaluated in unilineage erythropoietic culture of purified adult progenitors (erythroid burst-forming units [BFU-Es]). The GC derivative dexamethasone (Dex) was tested in minibulk cultures at graded dosages within the therapeutical range (10(-6) to 10(-9) M). Dex did not exert significant effects alone, but synergistically it potentiated the action of KL in a dose-dependent fashion. Specifically, Dex induced delayed erythroid maturation coupled with a 2-log increased number of generated erythroblasts and enhanced HbF synthesis up to 85% F cells and 55% gamma-globin content at terminal maturation (ie, in more than 80%-90% mature erythroblasts). Equivalent results were obtained in unicellular erythroid cultures of sibling BFU-Es treated with KL alone or combined with graded amounts of Dex. These results indicate that the stimulatory effect of KL + Dex is related to the modulation of gamma-globin expression rather than to recruitment of BFU-Es with elevated HbF synthetic potential. At the molecular level, Id2 expression is totally suppressed in control erythroid culture but is sustained in KL + Dex culture. Hypothetically, Id2 may mediate the expansion of early erythroid cells, which correlates with HbF reactivation. These studies indicate that GCs play an important role in HbF reactivation. Because Dex acts at dosages used in immunologic disease therapy, KL + Dex administration may be considered to develop preclinical models for beta-hemoglobinopathy treatment.


Subject(s)
Dexamethasone/pharmacology , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Stem Cell Factor/pharmacology , Cell Culture Techniques , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Fetal Hemoglobin/drug effects , Globins/analysis , Humans , Hypoxia , Kinetics , Male , Transcription Factors/analysis
17.
Blood ; 101(4): 1316-23, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12406876

ABSTRACT

The expression/function of vascular endothelial growth factor (VEGF) receptors (VEGFR1/Flt1 and VEGFR2/KDR/Flk1) in hematopoiesis is under scrutiny. We have investigated the expression of Flt1 and kinase domain receptor (KDR) on hematopoietic precursors, as evaluated in liquid culture of CD34(+) hematopoietic progenitor cells (HPCs) induced to unilineage differentiation/maturation through the erythroid (E), megakaryocytic (Mk), granulocytic (G), or monocytic (Mo) lineage. KDR, expressed on 0.5% to 1.5% CD34(+) cells, is rapidly downmodulated on induction of differentiation. Similarly, Flt1 is present at very low levels in HPCs and is downmodulated in E and G lineages; however, Flt1 is induced in the precursors of both Mo and Mk series; ie, its level progressively increases during Mo maturation, and it peaks at the initial-intermediate culture stages in the Mk lineage. Functional experiments indicate that Mk and E, but not G and Mo, precursors release significant amounts of VEGF in the culture medium, particularly at low O(2) levels. The functional role of VEGF release on Mk maturation is indicated by 2 series of observations. (1) Molecules preventing the VEGF-Flt1 interaction on the precursor membrane (eg, soluble Flt1 receptors) significantly inhibit Mk polyploidization. (2) Addition of exogenous VEGF or placenta growth factor (PlGF) markedly potentiates Mk maturation. Conversely, VEGF does not modify Mo differentiation/maturation. Altogether, our results suggest that in the hematopoietic microenvironment an autocrine VEGF loop contributes to optimal Mk maturation through Flt1. A paracrine loop involving VEGF release by E precursors may also operate. Similarly, recent studies indicate that an autocrine loop involving VEGF and Flt1/Flk1 receptors mediates hematopoietic stem cell survival and differentiation.


Subject(s)
Cell Differentiation/physiology , Endothelial Growth Factors/physiology , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/physiology , Lymphokines/physiology , Megakaryocytes/cytology , Vascular Endothelial Growth Factor Receptor-1/physiology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Differentiation/drug effects , Cell Membrane/chemistry , Cells, Cultured , DNA/analysis , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/pharmacology , Erythroid Precursor Cells/chemistry , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Flow Cytometry , Gene Expression , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/biosynthesis , Lymphokines/pharmacology , Megakaryocytes/chemistry , Megakaryocytes/metabolism , Monocytes/chemistry , Monocytes/cytology , Monocytes/metabolism , Oxygen/administration & dosage , Polyploidy , RNA, Messenger/analysis , Thrombopoietin/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/analysis , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...