Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Eur J Pharmacol ; 891: 173700, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33137331

ABSTRACT

Pharmacologically induced production of fetal hemoglobin (HbF) is a pragmatic therapeutic strategy for the reduction of globin chain imbalance and improving the clinical severities of patients with ß-hemoglobinopathies. To identify highly desirable new therapeutic HbF-inducing agents, we screened functionally diverse ten monoterpenes, as molecular entities for their potent induction and erythroid differentiation ability in human erythroleukemia cell line (K562) and transgenic mice. Benzidine hemoglobin staining demonstrated six compounds to have significantly induced erythroid differentiation of K562 cells in a dose and time-dependent manner. This induction paralleled well with the optimal accumulated quantity of total hemoglobin in treated cultures. The cytotoxic studies revealed that three (carvacrol, 3-carene, and 1,4-cineole) of the six compounds with their maximal erythroid expansion ability did not affect cell proliferation and were found non-toxic. Four compounds were found to have high potency, with 4-8-fold induction of HbF at both transcriptional and protein levels in vitro. Subsequently, an in vivo study with the three active non-cytotoxic compounds showed significant overexpression of the γ-globin gene and HbF production. Carvacrol emerged as a lead HbF regulator suggested by the increase in expression of γ-globin mRNA content (5.762 ± 0.54-fold in K562 cells and 5.59 ± 0.20-fold increase in transgenic mice), accompanied by an increase in fetal hemoglobin (F-cells) levels (83.47% in K562 cells and 79.6% in mice model). This study implicates monoterpenes as new HbF inducing candidates but warrants mechanistic elucidation to develop them into potential therapeutic drugs in ß-thalassemia and sickle cell anemia.


Subject(s)
Erythrocytes/drug effects , Erythropoiesis/drug effects , Fetal Hemoglobin/biosynthesis , Hematinics/pharmacology , Monoterpenes/pharmacology , gamma-Globulins/biosynthesis , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Chromosomes, Artificial, Yeast , Cymenes/pharmacology , Erythrocytes/metabolism , Fetal Hemoglobin/genetics , Gene Expression Regulation, Developmental , Humans , K562 Cells , Mice, Transgenic , Up-Regulation , beta-Thalassemia/blood , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , gamma-Globulins/genetics
2.
Antioxid Redox Signal ; 25(17): 921-935, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27393735

ABSTRACT

AIMS: Sickle retinopathy (SR) is a major cause of blindness in sickle cell disease (SCD). The genetic mutation responsible for SCD is known, however; oxidative stress and inflammation also figure prominently in the development and progression of pathology. Development of therapies for SR is hampered by the lack of (a) animal models that accurately recapitulate human SR and (b) strategies for noninvasive yet effective retinal drug delivery. This study addressed both issues by validating the Townes humanized SCD mouse as a model of SR and demonstrating the efficacy of oral administration of the antioxidant fumaric acid ester monomethyl fumarate (MMF) in the disease. RESULTS: In vivo ophthalmic imaging, electroretinography, and postmortem histological RNA and protein analyses were used to monitor retinal health and function in normal (HbAA) and sickle (HbSS) hemoglobin-producing mice over a one-year period and in additional HbAA and HbSS mice treated with MMF (15 mg/ml) for 5 months. Functional and morphological abnormalities and molecular hallmarks of oxidative stress/inflammation were evident early in HbSS retinas and increased in number and severity with age. Treatment with MMF, a known inducer of Nrf2, induced γ-globin expression and fetal hemoglobin production, improved hematological profiles, and ameliorated SR-related pathology. Innovation and Conclusion: United States Food and Drug Administration-approved formulations in which MMF is the primary bioactive ingredient are currently available to treat multiple sclerosis; such drugs may be effective for treatment of ocular and systemic complications of SCD, and given the pleiotropic effects, other nonsickle-related diseases in which oxidative stress, inflammation, and retinal vascular pathology figure prominently. Antioxid. Redox Signal. 25, 921-935.


Subject(s)
Anemia, Sickle Cell/complications , Fumarates/administration & dosage , Retinal Diseases/etiology , Retinal Diseases/pathology , Administration, Oral , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/diagnosis , Animals , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins , Disease Models, Animal , Electroretinography , Gene Expression , Gene Expression Regulation , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Neovascularization, Pathologic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Repressor Proteins , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Retinal Neurons/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , gamma-Globulins/genetics , gamma-Globulins/metabolism
3.
Cell Cycle ; 15(12): 1545-51, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27124741

ABSTRACT

Silent information regulator type-1 (SIRT1) is the best-studied member of the Sirtuin (Sir2) family of nicotinamide dinucleotide (NAD)-dependent class III histone deacetylases (HDACs). Rrecently, it is suggested that SIRT1 may be involved in the development of malignant tumors including mouse lymphoma, but has not yet been explored in Angioimmunoblastic T-cell lymphoma (AITL). Therefore, we investigated the prevalence and the prognostic impact of SIRT1 expression in AITL. Immunohistochemical expression of SIRT1, p53 were evaluated by using a 2 mm core from 45 AITL patients. Positive expression of SIRT1 was seen in 71.11% (32 of 45) of patients and p53 expression were seen in 53.33% (24 of 45). SIRT1 and p53 expression were significantly associated with shorter PFS by univariate analysis (P=0.009 and P < 0.001, respectively), multivariate analysis also shows that SIRT1 expression relate to worse prognosis. We also suggest inferior survival in AITL with the combined expression of SIRT1 and clinical characteristics of high IPI scores, high clinical stage, increased serum LDH, decreased HGB and increased γ-Globulin. In conclusion, our results indicate that SIRT1 is strongly expressed in AITL and it act as a clinically significant prognostic indicator for AITL patients, may also serve as a therapeutic target in AITL.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell, Peripheral/genetics , Sirtuin 1/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cyclophosphamide/therapeutic use , Doxorubicin/therapeutic use , Female , Humans , Immunoblastic Lymphadenopathy/diagnosis , Immunoblastic Lymphadenopathy/drug therapy , Immunoblastic Lymphadenopathy/mortality , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/genetics , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/mortality , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prednisone/therapeutic use , Prognosis , Sirtuin 1/metabolism , Survival Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vincristine/therapeutic use , gamma-Globulins/genetics , gamma-Globulins/metabolism
4.
Cancer Chemother Pharmacol ; 75(3): 537-46, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25567350

ABSTRACT

PURPOSE: Inhibitors of DNA (cytosine-5)-methyltransferases (DNMT) are active antineoplastic agents. We conducted the first-in-human phase I trial of 5-fluoro-2'-deoxycytidine (FdCyd), a DNMT inhibitor stable in aqueous solution, in patients with advanced solid tumors. Objectives were to establish the safety, maximum tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of FdCyd + tetrahydrouridine (THU). METHODS: FdCyd + THU were administered by 3 h IV infusion on days 1-5 every 3 weeks, or days 1-5 and 8-12 every 4 weeks. FdCyd was administered IV with a fixed 350 mg/m(2)/day dose of THU to inhibit deamination of FdCyd. Pharmacokinetics of FdCyd, downstream metabolites and THU were assessed by LC-MS/MS. RBC γ-globin expression was evaluated as a pharmacodynamics biomarker. RESULTS: Patients were enrolled on the 3-week schedule at doses up to 80 mg/m(2)/day without dose-limiting toxicity (DLT) prior to transitioning to the 4-week schedule, which resulted in an MTD of 134 mg/m(2)/day; one of six patients had a first-cycle DLT (grade 3 colitis). FdCyd ≥40 mg/m(2)/day produced peak plasma concentrations >1 µM. Although there was inter-patient variability, γ-globin mRNA increased during the first two treatment cycles. One refractory breast cancer patient experienced a partial response (PR) of >90 % decrease in tumor size, lasting over a year. CONCLUSIONS: The MTD was established at 134 mg/m(2) FdCyd + 350 mg/m(2) THU days 1-5 and 8-12 every 4 weeks. Based on toxicities observed over multiple cycles, good plasma exposures, and the sustained PR observed at 67 mg/m(2)/day, the phase II dose for our ongoing multi-histology trial is 100 mg/m(2)/day FdCyd with 350 mg/m(2)/day THU.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Neoplasms/drug therapy , gamma-Globulins/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chromatography, Liquid , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Tandem Mass Spectrometry , Tetrahydrouridine/administration & dosage , Treatment Outcome
5.
Blood ; 122(17): 3030-3, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-23940281

ABSTRACT

A high prevalence of monoclonal gammopathy (MG) has been observed in HIV-infected patients. We explored the conditions associated with long-term persistence of serum monoclonal protein (M protein) in HIV-infected patients on antiretroviral therapy (ART). Of 21 patients with MG, M protein disappeared in 12 patients (58%) over 5 years of ART. Higher level of serum γ-globulin and higher percentages of circulating plasmablasts and plasma cells were observed in patients with persistent MG compared with patients with transient MG. MG persistence was associated with the cumulative time of detectable plasma HIV RNA after ART initiation, detection of Epstein-Barr virus (EBV) DNA in plasma, and a high level of EBV DNA in B cells. Poor control of HIV replication and detectable EBV replication in plasma were both associated with long-term MG persistence in patients on ART. In the case of viral control, MG associated with HIV infection is usually transient.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active , DNA, Viral/antagonists & inhibitors , Epstein-Barr Virus Infections/drug therapy , HIV Infections/drug therapy , Paraproteinemias/drug therapy , Virus Replication/drug effects , Adult , Coinfection , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Female , Glycoproteins/blood , Glycoproteins/genetics , Glycoproteins/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Herpesvirus 4, Human/immunology , Humans , Male , Middle Aged , Paraproteinemias/immunology , Paraproteinemias/virology , Plasma Cells/drug effects , Plasma Cells/immunology , Plasma Cells/virology , Time Factors , Viral Load/drug effects , gamma-Globulins/genetics , gamma-Globulins/immunology
6.
Nucleic Acids Res ; 39(9): 3505-19, 2011 May.
Article in English | MEDLINE | ID: mdl-21245044

ABSTRACT

Ikaros is associated with both gene transcriptional activation and repression in lymphocytes. Ikaros acts also as repressor of human γ-globin (huγ-) gene transcription in fetal and adult erythroid cells. Whether and eventually, how Ikaros can function as a transcriptional activator in erythroid cells remains poorly understood. Results presented herein demonstrate that Ikaros is a developmental-specific activator of huγ-gene expression in yolk sac erythroid cells. Molecular analysis in primary cells revealed that Ikaros interacts with Gata-1 and favors Brg1 recruitment to the human ß-globin Locus Control Region and the huγ-promoters, supporting long-range chromatin interactions between these regions. Additionally, we demonstrate that Ikaros contributes to transcription initiation and elongation of the huγ-genes, since it is not only required for TBP and RNA Polymerase II (Pol II) assembly at the huγ-promoters but also for conversion of Pol II into the elongation-competent phosphorylated form. In agreement with the latter, we show that Ikaros interacts with Cyclin-dependent kinase 9 (Cdk9), which contributes to efficient transcription elongation by phosphorylating the C-terminal domain of the large subunit of Pol II on Serine 2, and favours Cdk9 recruitment to huγ-promoters. Our results show that Ikaros exerts dual functionality during gene activation, by promoting efficient transcription initiation and elongation.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , Ikaros Transcription Factor/metabolism , Transcriptional Activation , Animals , Cell Line , DNA Helicases/metabolism , Humans , Ikaros Transcription Factor/genetics , Locus Control Region , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Yolk Sac/anatomy & histology , Yolk Sac/growth & development , gamma-Globulins/genetics , gamma-Globulins/metabolism
7.
J Ethnopharmacol ; 120(3): 437-41, 2008 Dec 08.
Article in English | MEDLINE | ID: mdl-18951967

ABSTRACT

OBJECTIVES: To study the molecular pharmacological basis of the YiSui ShenXu Granule, a complex prescription of the Chinese traditional medicine used to treat beta-thalassemia. METHODS: Real-time quantitative PCR method had been applied to analyze the genes expression: gamma-globin, Ckit, EpoR, Spi, FKLF, GATA1 and GATA2 in K562 cell treated and untreated with this complex prescription and its each single herbal medicine. RESULTS: The results showed that this complex prescription increased the gamma-globin, EpoR, Spi and FKLF expression and decreased the Ckit, GATA1 and GATA2 expression. And all single herbal medicines of this complex prescription could change some of those gene expressions, but not the same as the complex prescription. Even that, this study results indicated that the YiSui ShenXu Granule has its molecular pharmacological basis in treating beta-thalassemia.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression/drug effects , beta-Thalassemia/drug therapy , Angelica sinensis , Apoptosis Regulatory Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Primers , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/genetics , Humans , K562 Cells , Medicine, Chinese Traditional , Polymerase Chain Reaction , Prescriptions , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/genetics , Receptors, Erythropoietin/genetics , Repressor Proteins/genetics , gamma-Globulins/genetics
8.
Blood ; 110(8): 2864-71, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17620452

ABSTRACT

Although thalidomide has been shown to improve anemia in some patients with myelodysplastic syndromes and stimulates erythropoietin in patients with multiple myeloma, thalidomide's specific effects on gamma-globin gene expression during erythroid differentiation have not been studied. Here, we investigated the effects of thalidomide on gamma-globin gene expression and the involved signaling pathway using an ex vivo culture system of primary human CD34+ cells. We found that thalidomide induced gamma-globin mRNA expression in a dose-dependent manner, but had no effect on beta-globin expression. We also demonstrated that intracellular reactive oxygen species (ROS) levels were increased by treatment with thalidomide for 48 hours (from day 3 to day 5). Western blot analysis demonstrated that thalidomide activated the p38 mitogen-activated protein kinase (MAPK) signaling pathway in a time- and dose-dependent manner and increased histone H4 acetylation. Pretreatment of cells with the antioxidant enzyme catalase and the intracellular hydroxyl scavenger dimethylthiourea (DMTU) abrogated the thalidomide-induced p38 MAPK activation and histone H4 acetylation. Moreover, pretreatment with catalase and DMTU diminished thalidomide-induced gamma-globin gene expression. These data indicate that thalidomide induces increased expression of the gamma-globin gene via ROS-dependent activation of the p38 MAPK signaling pathway and histone H4 acetylation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Erythropoiesis/drug effects , Gene Expression/drug effects , Signal Transduction/drug effects , Thalidomide/pharmacology , gamma-Globulins/drug effects , Acetylation , Antigens, CD34/metabolism , Antioxidants/pharmacology , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Histones/drug effects , Histones/metabolism , Humans , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , gamma-Globulins/genetics , p38 Mitogen-Activated Protein Kinases/drug effects
9.
Blood ; 108(10): 3590-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16896160

ABSTRACT

The histone deacetylase inhibitors (HDA-CIs) butyrate and trichostatin A activate gamma-globin expression via a p38 mitogen-activating protein kinase (MAPK)-dependent mechanism. We hypothesized that down-stream effectors of p38 MAPK, namely activating transcription factor-2 (ATF-2) and cyclic AMP response element (CRE) binding protein (CREB), are intimately involved in fetal hemoglobin induction by these agents. In this study, we observed increased ATF-2 and CREB1 phosphorylation mediated by the HDACIs in K562 cells, in conjunction with histone H4 hyperacetylation. Moreover, enhanced DNA-protein interactions occurred in the CRE in the (G)gamma-globin promoter (G-CRE) in vitro after drug treatments; subsequent chromatin immunoprecipitation assay confirmed ATF-2 and CREB1 binding to the G-CRE in vivo. Enforced expression of ATF-2 and CREB produced (G)gamma-promoter trans-activation which was abolished by a 2-base pair mutation in the putative G-CRE. The data presented herein demonstrate that gamma-gene induction by butyrate and trichostatin A involves ATF-2 and CREB1 activation via p38 MAPK signaling.


Subject(s)
Activating Transcription Factor 2/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , gamma-Globulins/metabolism , Activating Transcription Factor 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors/pharmacology , Histones/metabolism , Humans , K562 Cells , Phosphorylation/drug effects , Transcriptional Activation , gamma-Globulins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Blood Cells Mol Dis ; 34(3): 235-47, 2005.
Article in English | MEDLINE | ID: mdl-15885608

ABSTRACT

Several lines of evidence indicate that in vivo drug selection can be used to overcome the low rates of gene transfer and engraftment encountered in many hematopoietic stem cell gene therapy settings. However, whether selection imposed on one transcription cassette effects the likelihood of expression from a second, independent transcription cassette within the same vector has been less well studied. In order to address this issue, we engineered an oncoretrovirus vector to express two separate transcription units: (i) a bicistronic cassette encoding both GFP and a pharmacologically regulated cell growth switch based on the thrombopoietin receptor Mpl; and (ii) a highly position-dependent second cassette encoding human gamma-globin. Studies in cell cultures and in mice transplanted with transduced marrow indicated that selective expansion increased by more than 9-fold the fraction of erythroid cells expressing the linked but separate expression cassette for gamma-globin. This increase was far greater then that observed for the bicistronic GFP gene, and cannot be explained by a simple increase in the fraction of cells containing provirus. These results suggest that selective expansion favors erythroid stem/progenitor cells with provirus integrated at chromosomal sites which are relatively resistant to silencing position effects.


Subject(s)
Cell Growth Processes/genetics , Erythroid Cells/cytology , Transcription, Genetic , gamma-Globulins/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Line, Tumor , Cell Proliferation , Erythroid Cells/metabolism , Genetic Engineering/methods , Genetic Vectors , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Receptors, Cytokine/genetics , Receptors, Thrombopoietin , Retroviridae/genetics , Transduction, Genetic , Virus Integration
11.
Blood ; 105(1): 387-93, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15367428

ABSTRACT

We systematically compared cytokine-mediated increases or decreases in proliferation with globin gene and protein expression in adult human erythroblasts. Despite their opposite effects on growth, stem cell factor (SCF) and transforming growth factor beta (TGF-B) had synergistic effects with respect to fetal hemoglobin (HbF): average HbF/HbF + adult hemoglobin (HbA) ratio in erythropoietin (EPO) = 1.4 +/- 1.0%; EPO + TGF-B = 10.8 +/- 1.9%; EPO + SCF = 19.1 +/- 6.2%; and EPO + SCF + TGF-B (EST) = 39.3 +/- 6.3%. Polymerase chain reaction (PCR) revealed significant increases in gamma-globin transcripts that were balanced by reduced beta-globin transcripts. Single-cell quantitative PCR demonstrated a complete reversal of gamma-globin gene silencing with detectable gamma-globin mRNA in more than 95% of the cells. Immunostaining with HbF antibodies also showed a pancellular distribution in EST (96.2 +/- 0.01% HbF positive) compared with a heterocellular distribution in EPO (42.9 +/- 0.01% HbF positive). As shown here for the first time, a robust and pancellular reversal of gamma-globin gene silencing among hemoglobinized erythroblasts from adult humans may be achieved in the absence of hereditary mutation or direct genomic manipulation.


Subject(s)
Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Gene Silencing , gamma-Globulins/genetics , Adult , Carbonic Anhydrases/biosynthesis , Carbonic Anhydrases/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Fetal Blood/metabolism , Hemoglobins/analysis , Hemoglobins/biosynthesis , Hemoglobins/genetics , Humans , Kinetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein , Stem Cell Factor/pharmacology , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology
13.
Blood ; 98(9): 2664-72, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675336

ABSTRACT

Use of oncoretroviral vectors in gene therapy for hemoglobinopathies has been impeded by low titer vectors, genetic instability, and poor expression. Fifteen self- inactivating (SIN) lentiviral vectors using 4 erythroid promoters in combination with 4 erythroid enhancers with or without the woodchuck hepatitis virus postregulatory element (WPRE) were generated using the enhanced green fluorescent protein as a reporter gene. Vectors with high erythroid-specific expression in cell lines were tested in primary human CD34(+) cells and in vivo in the murine bone marrow (BM) transplantation model. Vectors containing the ankyrin-1 promoter showed high-level expression and stable proviral transmission. Two vectors containing the ankyrin-1 promoter and 2 erythroid enhancers (HS-40 plus GATA-1 or HS-40 plus 5-aminolevulinate synthase intron 8 [I8] enhancers) and WPRE expressed at levels higher than the HS2/beta-promoter vector in bulk unilineage erythroid cultures and individual erythroid blast-forming units derived from human BM CD34(+) cells. Sca1(+)/lineage(-) Ly5.1 mouse hematopoietic cells, transduced with these 2 ankyrin-1 promoter vectors, were injected into lethally irradiated Ly5.2 recipients. Eleven weeks after transplantation, high-level expression was seen from both vectors in blood (63%-89% of red blood cells) and erythroid cells in BM (70%-86% engraftment), compared with negligible expression in myeloid and lymphoid lineages in blood, BM, spleen, and thymus (0%-4%). The I8/HS-40-containing vector encoding a hybrid human beta/gamma-globin gene led to 43% to 113% human gamma-globin expression/copy of the mouse alpha-globin gene. Thus, modular use of erythroid-specific enhancers/promoters and WPRE in SIN-lentiviral vectors led to identification of high-titer, stably transmitted vectors with high-level erythroid-specific expression for gene therapy of red cell diseases.


Subject(s)
Erythroid Precursor Cells/metabolism , Genetic Vectors/metabolism , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic/methods , Animals , Antigens, CD34 , Bone Marrow Cells , Bone Marrow Transplantation , Enhancer Elements, Genetic , Gene Expression , Gene Expression Regulation, Viral , Genetic Vectors/standards , Green Fluorescent Proteins , Hepatitis B Virus, Woodchuck/genetics , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Models, Animal , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional/genetics , Transduction, Genetic/standards , gamma-Globulins/genetics , gamma-Globulins/metabolism
14.
Ann N Y Acad Sci ; 938: 246-61, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11458514

ABSTRACT

Gene therapy for patients with hemoglobin disorders such has been hampered by the inability of retrovirus vectors to transfer globin genes and the locus control region (LCR) into hematopoietic stem cells without rearrangement. In addition, the expression from intact globin gene vectors has been variable in red blood cells as a result of position effects and retrovirus silencing. We hypothesized that by substituting the globin gene promoter for the promoter of another gene expressed in red blood cells, we could generate stable retrovirus vectors that would express globin at sufficient levels to treat hemoglobinopathies. Transgenic mice containing the human ankyrin (Ank) gene promoter fused to the human gamma-globin gene showed position-independent, copy number-dependent expression of a linked gamma-globin mRNA. We generated a "double-copy" Ank/A gamma-globin retrovirus vector that transferred two copies of the Ank/A gamma-globin gene into target cells. Stable gene transfer was observed in primary primary mouse progenitor cells and long-term repopulating hematopoietic stem cells. Expression of Ank/A gamma-globin mRNA in mature red blood cells was approximately 8% of the level of mouse alpha-globin mRNA. We conclude that this novel retrovirus vector may be valuable for treating a variety of hemoglobinopathies by gene therapy if the level of expression can be further increased.


Subject(s)
Erythrocytes/metabolism , Genetic Vectors/genetics , RNA, Messenger/biosynthesis , Retroviridae/genetics , gamma-Globulins/genetics , 3T3 Cells , Anemia/genetics , Anemia/therapy , Animals , Ankyrins/genetics , Flow Cytometry , Gene Expression , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
15.
Mamm Genome ; 11(11): 1024-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11063260

ABSTRACT

Genetic factors affecting postnatal gamma-globin expression--a major modifier of the severity of both beta-thalassemia and sickle cell anemia--have been difficult to study. This is especially so in mice, an organism lacking a globin gene with an expression pattern equivalent to that of human gamma-globin. To model the human beta-cluster in mice, with the goal of screening for loci affecting human gamma-globin expression in vivo, we introduced a human beta-globin cluster YAC transgene into the genome of FVB/N mice. The beta-cluster contained a Greek hereditary persistence of fetal hemoglobin (HPFH) gamma allele, resulting in postnatal expression of human gamma-globin in transgenic mice. The level of human gamma-globin for various F1 hybrids derived from crosses between the FVB/N transgenics and other inbred mouse strains was assessed. The gamma-globin level of the (C3HeB/FeJ x FVB/N)F1 transgenic mice was noted to be significantly elevated. To map genes affecting postnatal y-globin expression, we performed a 20-centiMorgan (cM) genome scan of a (C3HeB/FeJ x FVB/N)F1 transgenics x FVB/N backcross, followed by high-resolution marker analysis of promising loci. From this analysis we mapped a locus within an 18-cM interval of mouse Chromosome (Chr) 1 (LOD = 4.3) that contributes 10.9% of variation in gamma-globin level. Combining transgenic modeling of the human beta-globin gene cluster with quantitative trait analysis, we have identified and mapped a murine locus that impacts on human gamma-globin level in vivo.


Subject(s)
Chromosomes, Artificial, Yeast , Genomics/methods , Mice, Transgenic/genetics , gamma-Globulins/genetics , Animals , Animals, Newborn , Chromosome Mapping , Chromosomes, Human, Pair 1 , Fetal Hemoglobin/genetics , Gene Expression Regulation, Developmental , Humans , Lod Score , Male , Mice , Mice, Inbred Strains
16.
Blood Cells Mol Dis ; 25(3-4): 255-69, 1999.
Article in English | MEDLINE | ID: mdl-10575551

ABSTRACT

Pharmacologic agents such as hydroxyurea (HU), N, 3-4 trihydroxybenzamide (didox), and isobutyramide (ISB) can elevate gamma-globin as a potential treatment for the beta-hemoglobinopathies. In these experiments, transgenic mice with 5'HS2 from the human beta-globin locus control region, the fetal (A gamma), and adult (beta s) globin genes were used. Mice were treated with HU, didox, or ISB individually, or with combinations of HU or didox with ISB. The aim was to determine whether these drugs have synergistic effects on the induction of fetal hemoglobin (HbF) and whether the combination regimens are more hematotoxic. In the combination regimens, injections of HU or didox for five weeks were concomitant with ISB treatment every other day for the final three weeks of treatment. The combination of HU + ISB was more hematotoxic than the individual drugs based on significantly increased percentages of reticulocytes and reduced hemoglobin, indicating that caution should be taken in treatments involving combinations of these types of drugs. The didox + ISB combination was not more hematotoxic than the individual drugs. HbF was not induced in the groups treated with the combinations of HU or didox with ISB compared to the individual agents. There was a negligible effect on the percentage of HbF and an unexpected negative effect on the percentage of F cells. The results also have implications for future testing of HbF-inducing drugs in mouse models. In control mice that were phlebotomized but not treated with any drugs, increased percentages of F cells were observed, indicating that blood sampling can cause this effect. In addition, increases in the percentage of F cells did not correlate with increases in the percentage of HbF, indicating that monitoring F cells alone is not a sufficient measure of HbF induction.


Subject(s)
Amides/toxicity , Amides/therapeutic use , Fetal Hemoglobin/metabolism , Hydroxyurea/toxicity , Hydroxyurea/therapeutic use , Animals , Disease Models, Animal , Drug Therapy, Combination , Fetal Hemoglobin/analysis , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/drug effects , Hemoglobinopathies/drug therapy , Humans , Mice , Mice, Transgenic , RNA, Messenger/analysis , Reticulocytes/chemistry , Reticulocytes/metabolism , gamma-Globulins/drug effects , gamma-Globulins/genetics
17.
Eur J Haematol ; 58(5): 320-5, 1997 May.
Article in English | MEDLINE | ID: mdl-9222287

ABSTRACT

The -175 (T-->C) G gamma hereditary persistence of fetal haemoglobin is a very rare promoter mutation occurring in Caucasians as well as in African-Americans. Heterozygotes for this non-deletional HPFH show 20% HbF, mostly of G gamma type. We describe here a healthy Sardinian man who coinherited -175 (T-->C) G gamma HPFH with the beta-thalassaemia codon 39 nonsense mutation in trans; he showed 64% HbF, 100% of G gamma type. Although the beta-globin haplotype pattern (II/II) was indicative of the presence of the A gamma T allele on both chromosomes, the A gamma T expression was undetectable by HPLC even in red cell populations separated by age. The proband was, moreover, homozygous for the -4 bp deletion at position -225 to -222 of A gamma promoter which has recently been associated with decreased A gamma T globin expression. These findings suggest that this maximal overexpression of G gamma-globin probably reflects intensified stimulation of the mutated G gamma promoter in this hitherto undescribed genetic condition.


Subject(s)
Fetal Hemoglobin/genetics , Hemoglobinopathies/genetics , beta-Thalassemia/genetics , gamma-Globulins/genetics , Adult , Family Health , Fetal Hemoglobin/chemistry , Gene Expression , Heterozygote , Humans , Italy/epidemiology , Male , Mutation , Prevalence , beta-Thalassemia/epidemiology , gamma-Globulins/chemistry
18.
Leuk Lymphoma ; 23(5-6): 545-50, 1996 Nov.
Article in English | MEDLINE | ID: mdl-9031085

ABSTRACT

Currently available data indicate that erythroid and megakaryocytic differentiation pathways are closely related to each other, and there may exist progenitor cells common to those two lineages may exist. Acute megakaryoblastic leukemia (AML-M7) and transient myeloproliferative disorder in Down's syndrome (TMD) are characterized by rapid growth of abnormal blast cells which express megakaryocytic markers. These blast cells express lineage-specific transcription factors such as GATA-1 common to these lineages and frequently express erythroid-specific mRNAs such as gamma-globin and erythroid delta-aminolevulinate synthase (ALAS-E), indicating that most of the blasts in M7 and TMD cases have erythroid and megakaryocytic phenotypes. These results suggest that blasts in M7 and TMD may correspond to progenitors of both erythroid and megakaryocytic lineages.


Subject(s)
DNA-Binding Proteins/biosynthesis , Down Syndrome/complications , Erythroid Precursor Cells/metabolism , Leukemia, Megakaryoblastic, Acute/metabolism , Transcription Factors/biosynthesis , gamma-Globulins/biosynthesis , DNA-Binding Proteins/genetics , Down Syndrome/genetics , Erythroid Precursor Cells/physiology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , GATA2 Transcription Factor , Gene Expression , Humans , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Sensitivity and Specificity , Transcription Factors/genetics , gamma-Globulins/genetics
20.
J Clin Endocrinol Metab ; 80(8): 2291-7, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7543111

ABSTRACT

To determine why blacks have a higher bone mineral density (BMD) and lower incidence of osteoporosis and fractures than whites, we investigated whether the secretion of GH is higher in black than in white men. Measurements of GH were obtained at 20-min intervals over 24 h and analyzed by deconvolution. BMD was determined by dual energy x-ray absorptiometry in 16 normal black and 17 normal white men, aged 20-40 yr. The 24-h integrated GH concentration 942 +/- 174 vs. 602 +/- 104 micrograms/L; P = 0.0495) and GH secretory burst amplitude (0.499 +/- 0.163 vs. 0.169 +/- 0.027 micrograms/L.min; P = 0.0482) were higher in black than in white men. GH burst frequency, half-duration, mass, and half-life were not different in the 2 groups. The serum 17 beta-estradiol level (162 +/- 12 vs. 108 +/- 11 pmol/L; P = 0.0011) was higher, and the serum insulin-like growth factor-binding protein 3 level (2.2 +/- 0.1 vs. 2.8 +/- 0.1 microgram/mL; P = 0.0001) was lower in black than in white men. BMD values for total body (1.22 +/- 0.02 vs. 1.14 +/- 0.02 g/cm2; P = 0.0041), forearm (0.69 +/- 0.01 vs. 0.66 +/- 0.01 g/cm2; P = 0.0211), trochanter (0.91 +/- 0.03 vs. 0.77 +/- 0.03 g/cm2; P = 0.0003), and femoral neck (1.08 +/- 0.03 vs. 0.93 +/- 0.03 g/cm2; P = 0.0007) were higher in black than in white men. Thus, serum 17 beta-estradiol level, GH secretion, and BMD values for the total body, forearm, trochanter, and femoral neck are greater in black than in white men. As estrogen is known to increase GH secretion and GH to increase bone mass, increases in circulating 17 beta-estradiol may contribute to the higher GH secretion and bone mass in black men.


Subject(s)
Black People , Bone Density , Growth Hormone/metabolism , White People , Absorptiometry, Photon/methods , Adult , Body Composition , Carrier Proteins/blood , Circadian Rhythm , Estradiol/blood , Fractures, Bone/epidemiology , Growth Hormone/blood , Half-Life , Humans , Insulin-Like Growth Factor Binding Proteins , Male , Osteoporosis/epidemiology , Regression Analysis , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , gamma-Globulins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL