Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunol ; 204(7): 1982-1987, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32122998

ABSTRACT

GFP is frequently used as a marker for tracking donor cells adoptively transplanted into recipient animals. The human ubiquitin C promoter (UBC)-driven-GFP transgenic mouse is a commonly used source of donor cells for this purpose. This mouse was initially generated in the C57BL/6 inbred strain and has been backcrossed into the BALB/cBy strain for over 11 generations. Both the C57BL/6 inbred and BALB/cBy congenic UBC-GFP lines are commercially available and have been widely distributed. These UBC-GFP lines can be a convenient resource for tracking donor cells in both syngenic MHC-matched and in allogenic MHC-mismatched studies as C57BL/6 (H-2b) and BALB/cBy (H-2d) have disparate MHC haplotypes. In this report, we surprisingly discover that the UBC-GFP BALB/cBy congenic mice still retain the H-2b MHC haplotype of their original C57BL/6 founder, suggesting that the UBC-GFP transgene integration site is closely linked to the MHC locus on chromosome 17. Using linear amplification-mediated PCR, we successfully map the UBC-GFP transgene to the MHC locus. This study highlights the importance and urgency of mapping the transgene integration site of transgenic mouse strains used in biomedical research. Furthermore, this study raises the possibility of alternative interpretations of previous studies using congenic UBC-GFP mice and focuses attention on the necessity for rigor and reproducibility in scientific research.


Subject(s)
Chromosomes/genetics , Green Fluorescent Proteins/genetics , Major Histocompatibility Complex/genetics , Mutagenesis, Insertional/genetics , Transgenes/genetics , Ubiquitin C/genetics , Animals , Haplotypes/genetics , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Reproducibility of Results
3.
Sci Rep ; 9(1): 11592, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406195

ABSTRACT

In utero gene therapy (IUGT) to the fetal hematopoietic compartment could be used to treat congenital blood disorders such as ß-thalassemia. A humanised mouse model of ß-thalassemia was used, in which heterozygous animals are anaemic with splenomegaly and extramedullary hematopoiesis. Intrahepatic in utero injections of a ß globin-expressing lentiviral vector (GLOBE), were performed in fetuses at E13.5 of gestation. We analysed animals at 12 and 32 weeks of age, for vector copy number in bone marrow, peripheral blood liver and spleen and we performed integration site analysis. Compared to noninjected heterozygous animals IUGT normalised blood haemoglobin levels and spleen weight. Integration site analysis showed polyclonality. The left ventricular ejection fraction measured using magnetic resonance imaging (MRI) in treated heterozygous animals was similar to that of normal non-ß-thalassemic mice but significantly higher than untreated heterozygous thalassemia mice suggesting that IUGT ameliorated poor cardiac function. GLOBE LV-mediated IUGT normalised the haematological and anatomical phenotype in a heterozygous humanised model of ß-thalassemia.


Subject(s)
Genetic Therapy , Heterozygote , Magnetic Resonance Imaging/methods , Animals , Female , Humans , Mice , Phenotype , Pregnancy , beta-Thalassemia/genetics
4.
Sci Rep ; 7: 43439, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240317

ABSTRACT

ß-thalassemia (ßT) is a genetic blood disorder causing profound and life threatening anemia. Current clinical management of ßT is a lifelong dependence on regular blood transfusions, a consequence of which is systemic iron overload leading to acute heart failure. Recent developments in gene and chelation therapy give hope of better prognosis for patients, but successful translation to clinical practice is hindered by the lack of thorough preclinical testing using representative animal models and clinically relevant quantitative biomarkers. Here we demonstrate a quantitative and non-invasive preclinical Magnetic Resonance Imaging (MRI) platform for the assessment of ßT in the γß0/γßA humanized mouse model of ßT. Changes in the quantitative MRI relaxation times as well as severe splenomegaly were observed in the heart, liver and spleen in ßT. These data showed high sensitivity to iron overload and a strong relationship between quantitative MRI relaxation times and hepatic iron content. Importantly these changes preceded the onset of iron overload cardiomyopathy, providing an early biomarker of disease progression. This work demonstrates that multiparametric MRI is a powerful tool for the assessment of preclinical ßT, providing sensitive and quantitative monitoring of tissue iron sequestration and cardiac dysfunction- parameters essential for the preclinical development of new therapeutics.


Subject(s)
Heart/diagnostic imaging , Iron Overload/diagnostic imaging , Liver/diagnostic imaging , Spleen/diagnostic imaging , Splenomegaly/diagnostic imaging , beta-Thalassemia/diagnostic imaging , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Female , Heart/physiopathology , Humans , Iron/analysis , Iron/metabolism , Iron Overload/metabolism , Iron Overload/pathology , Liver/metabolism , Liver/pathology , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Transgenic , Spleen/metabolism , Spleen/pathology , Splenomegaly/metabolism , Splenomegaly/pathology , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
5.
Blood Adv ; 1(25): 2421-2432, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29296892

ABSTRACT

ß-thalassemia is a group of inherited blood disorders that result in defects in ß-globin chain production. Cooley anemia (CA), or ß-thalassemia major, is the most severe form of the disease and occurs when an individual has mutations in both copies of the adult ß-globin gene. Patients with CA fail to make adult hemoglobin, exhibit ineffective erythropoiesis, experience severe anemia, and are transfusion dependent for life. Currently, allogeneic bone marrow transplantation (BMT) is the only cure; however, few patients have suitable donors for this procedure, which has significant morbidity and mortality. In this study, a novel humanized murine model of CA is rescued from lethal anemia by allogeneic BMT in the absence of cytoreductive conditioning. A single intravenous postnatal injection of allogeneic bone marrow results in stable, mixed hematopoietic chimerism. Five months after transplantation, donor cells accounted for approximately 90% of circulating erythrocytes and up to 15% of hematopoietic stem and progenitor cells. Transplanted mice are transfusion independent, have marked improvement of hematological indices, exhibit no growth retardation or signs of graft-versus-host disease, and are fertile. This study describes a method for the consistent engraftment of allogeneic donor hematopoietic cells that rescues a humanized mouse model of CA from lethal anemia, all in the absence of toxic cytoreductive conditioning.

6.
Proc Natl Acad Sci U S A ; 113(1): E51-60, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26699484

ABSTRACT

Epigenetic mechanisms play important regulatory roles in hematopoiesis and hematopoietic stem cell (HSC) function. Subunits of polycomb repressive complex 1 (PRC1), the major histone H2A ubiquitin ligase, are critical for both normal and pathological hematopoiesis; however, it is unclear which of the several counteracting H2A deubiquitinases functions along with PRC1 to control H2A ubiquitination (ubH2A) level and regulates hematopoiesis in vivo. Here we investigated the function of Usp16 in mouse hematopoiesis. Conditional deletion of Usp16 in bone marrow resulted in a significant increase of global ubH2A level and lethality. Usp16 deletion did not change HSC number but was associated with a dramatic reduction of mature and progenitor cell populations, revealing a role in governing HSC lineage commitment. ChIP- and RNA-sequencing studies in HSC and progenitor cells revealed that Usp16 bound to many important hematopoietic regulators and that Usp16 deletion altered the expression of genes in transcription/chromosome organization, immune response, hematopoietic/lymphoid organ development, and myeloid/leukocyte differentiation. The altered gene expression was partly rescued by knockdown of PRC1 subunits, suggesting that Usp16 and PRC1 counterbalance each other to regulate cellular ubH2A level and gene expression in the hematopoietic system. We further discovered that knocking down Cdkn1a (p21cip1), a Usp16 target and regulated gene, rescued the altered cell cycle profile and differentiation defect of Usp16-deleted HSCs. Collectively, these studies identified Usp16 as one of the histone H2A deubiquitinases, which coordinates with the H2A ubiquitin ligase PRC1 to regulate hematopoiesis, and revealed cell cycle regulation by Usp16 as key for HSC differentiation.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Ubiquitin Thiolesterase/physiology , Ubiquitin-Specific Proteases/physiology , Animals , Cell Count , Cyclin-Dependent Kinase Inhibitor p21/genetics , Endopeptidases/genetics , Endopeptidases/physiology , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Lethal , Hematopoiesis/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/physiology , Trans-Activators , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases/genetics
7.
Mol Cell Biol ; 33(11): 2241-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530053

ABSTRACT

During erythropoiesis, hemoglobin (Hb) synthesis increases from early progenitors to mature enucleated erythrocytes. Although Hb is one of the most extensively studied proteins, the role of Hb in erythroid lineage commitment, differentiation, and maturation remains unclear. In this study, we generate mouse embryos and embryonic stem (ES) cells with all of the adult α and ß globin genes deleted (Hb Null). While Hb Null embryos die in midgestation, adult globin genes are not required for primitive or definitive erythroid lineage commitment. In vitro differentiation of Hb Null ES cells generates viable definitive proerythroblasts that undergo apoptosis upon terminal differentiation. Surprisingly, all stages of Hb Null-derived definitive erythroblasts develop normally in vivo in chimeric mice, and Hb Null erythroid cells undergo enucleation to form reticulocytes. Free heme toxicity is not observed in Hb Null-derived erythroblasts. Transplantation of Hb Null-derived bone marrow cells provides short-term radioprotection of lethally irradiated recipients, whose progressive anemia results in an erythroid hyperplasia composed entirely of Hb Null-derived erythroblasts. This novel experimental model system enables the role played by Hb in erythroid cell enucleation, cytoskeleton maturation, and heme and iron regulation to be studied.


Subject(s)
Embryonic Stem Cells/physiology , Erythroid Cells/metabolism , Erythropoiesis/physiology , Hemoglobins/genetics , Animals , Bone Marrow Transplantation , Cell Differentiation , Embryonic Stem Cells/cytology , Fetal Death/genetics , Gestational Age , Heme/metabolism , Hemoglobins/metabolism , Liver/cytology , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reticulocytes/cytology , Reticulocytes/metabolism , alpha-Globins/genetics , beta-Globins/genetics
8.
Mol Cell Biol ; 31(4): 876-83, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21173165

ABSTRACT

Elevated levels of fetal γ-globin can cure disorders caused by mutations in the adult ß-globin gene. This clinical finding has motivated studies to improve our understanding of hemoglobin switching. Unlike humans, mice do not express a distinct fetal globin. Transgenic mice that contain the human ß-globin locus complete their fetal-to-adult hemoglobin switch prior to birth, with human γ-globin predominantly restricted to primitive erythroid cells. We established humanized (100% human hemoglobin) knock-in mice that demonstrate a distinct fetal hemoglobin (HbF) stage, where γ-globin is the dominant globin chain produced during mid- to late gestation. Human γ- and ß-globin gene competition is evident around the time of birth, and γ-globin chain production diminishes in postnatal life, with transient production of HbF reticulocytes. Following completion of the γ- to-ß-globin switch, adult erythroid cells synthesize low levels of HbF. We conclude that the knock-in globin genes are expressed in a pattern strikingly similar to that in human development, most notably with postnatal resolution of the fetal-to-adult hemoglobin switch. Our findings are consistent with the importance of BCL11A in hemoglobin switching, since removal of intergenic binding sites for BCL11A results in human γ-globin expression in mouse definitive erythroid cells.


Subject(s)
Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Animals , Animals, Newborn , Binding Sites/genetics , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Female , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genes, Switch , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
Ann N Y Acad Sci ; 1202: 45-51, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20712771

ABSTRACT

beta thalassemia major or Cooley's Anemia (CA) has been difficult to model in mice due to their lack of a fetal hemoglobin gene equivalent. This summary describes novel preclinical humanized mouse models of CA that survive on human fetal hemoglobin at birth and are blood-transfusion dependent for life upon completion of their human fetal-to-adult hemoglobin switch after birth. These CA models are the first to recapitulate the temporal onset of the disease in human patients. These novel humanized CA disease models are useful for the study of the regulation of globin gene expression, synthesis, and switching; examining the onset of disease pathology; development of transfusion and iron chelation therapies; induction of fetal hemoglobin synthesis; and the testing of novel genetic and cell-based therapies for the correction of thalassemia.


Subject(s)
Disease Models, Animal , Fetal Hemoglobin/genetics , Hemoglobins/genetics , beta-Thalassemia/pathology , beta-Thalassemia/physiopathology , Age of Onset , Animals , Blood Transfusion , Genetic Therapy , Humans , Mice , beta-Thalassemia/genetics , beta-Thalassemia/therapy
10.
Blood ; 113(19): 4763-70, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19258591

ABSTRACT

A preclinical humanized mouse model of beta thalassemia major or Cooley anemia (CA) was generated by targeted gene replacement of the mouse adult globin genes in embryonic stem cells. The mouse adult alpha and beta globin genes were replaced with adult human alpha globin genes (alpha2alpha1) and a human fetal to adult hemoglobin (Hb)-switching cassette (gamma(HPFH)deltabeta(0)), respectively. Similar to human infants with CA, fully humanized mice survived postnatally by synthesizing predominantly human fetal Hb, HbF (alpha(2)gamma(2)), with a small amount of human minor adult Hb, HbA2 (alpha(2)delta(2)). Completion of the human fetal to adult Hb switch after birth resulted in severe anemia marked by erythroid hyperplasia, ineffective erythropoiesis, hemolysis, and death. Similar to human patients, CA mice were rescued from lethal anemia by regular blood transfusion. Transfusion corrected the anemia and effectively suppressed the ineffective erythropoiesis, but led to iron overload. This preclinical humanized animal model of CA will be useful for the development of new transfusion and iron chelation regimens, the study of iron homeostasis in disease, and testing of cellular and genetic therapies for the correction of thalassemia.


Subject(s)
Disease Models, Animal , Erythropoiesis/physiology , Globins/genetics , Globins/metabolism , beta-Thalassemia/therapy , Anemia/etiology , Anemia/prevention & control , Animals , Blood Transfusion , Chromatography, High Pressure Liquid , Embryonic Stem Cells/metabolism , Erythroid Precursor Cells , Fetal Hemoglobin/metabolism , Flow Cytometry , Genetic Therapy , Hemoglobins/metabolism , Hemolysis , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Iron Overload/etiology , Iron Overload/prevention & control , Mice , Phenotype , beta-Thalassemia/blood , beta-Thalassemia/genetics
11.
J Biol Chem ; 284(8): 4889-96, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19098001

ABSTRACT

A novel humanized mouse model of Cooley's Anemia (CA) was generated by targeted gene replacement in embryonic stem (ES) cells. Because the mouse does not have a true fetal hemoglobin, a delayed switching human gamma to beta(0) globin gene cassette (gammabeta(0)) was inserted directly into the murine beta globin locus replacing both adult mouse beta globin genes. The inserted human beta(0) globin allele has a mutation in the splice donor site that produces the same aberrant transcripts in mice as described in human cells. No functional human beta globin polypeptide chains are produced. Heterozygous gammabeta(0) mice suffer from microcytic anemia. Unlike previously described animal models of beta thalassemia major, homozygous gammabeta(0) mice switch from mouse embryonic globin chains to human fetal gamma globin during fetal life. When bred with human alpha globin knockin mice, homozygous CA mice survive solely upon human fetal hemoglobin at birth. This preclinical animal model of CA can be utilized to study the regulation of globin gene expression, synthesis, and switching; the reactivation of human fetal globin gene expression; and the testing of genetic and cell-based therapies for the correction of thalassemia.


Subject(s)
Disease Models, Animal , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Quantitative Trait Loci , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , Animals , Embryonic Stem Cells/metabolism , Heterozygote , Homozygote , Humans , Mice , Mice, Transgenic
12.
J Biol Chem ; 282(9): 6875-86, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17204476

ABSTRACT

Heme oxygenase-1 is a highly inducible gene, the product of which catalyzes breakdown of the prooxidant heme. The purpose of this study was to investigate the regulation of the human heme oxygenase-1 gene in renal epithelial cells. DNase I hyper-sensitivity studies identified three distal sites (HS-2, -3, and -4) corresponding to approximately -4.0, -7.2, and -9.2 kb, respectively, of the heme oxygenase-1 promoter in addition to one proximal region, HS-1, which we have shown previously to be an E box. In vivo dimethyl sulfate footprinting of the HS-2 region revealed six individual protected guanines. Two mutations within HS-2 combined with a third mutation of the proximal E box abolished hemin- and cadmium-driven heme oxygenase-1 promoter activation, suggesting that these three sites synergized for maximal heme oxygenase-1 induction. Jun proteins bound to the antioxidant response element in the HS-2 region in vitro and associated with the heme oxygenase-1 promoter in vivo. JunB and JunD contribute opposing effects; JunB activated whereas JunD repressed heme oxygenase-1 expression in human renal epithelial cells, results that were corroborated in junB(-)(/)(-) and junD(-)(/)(-) cells. We propose that heme oxygenase-1 induction is controlled by a dynamic interplay of regulatory proteins, and we provide new insights into the molecular control of the human heme oxygenase-1 gene.


Subject(s)
Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Kidney/enzymology , Proto-Oncogene Proteins c-jun/physiology , Animals , Binding Sites , Cell Line , E-Box Elements , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , Kidney/cytology , Mice , Mice, Knockout , Promoter Regions, Genetic
13.
Blood ; 108(4): 1183-8, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16638928

ABSTRACT

Previous studies have demonstrated that sickle cell disease (SCD) can be corrected in mouse models by transduction of hematopoietic stem cells with lentiviral vectors containing antisickling globin genes followed by transplantation of these cells into syngeneic recipients. Although self-inactivating (SIN) lentiviral vectors with or without insulator elements should provide a safe and effective treatment in humans, some concerns about insertional mutagenesis persist. An ideal correction would involve replacement of the sickle globin gene (beta(S)) with a normal copy of the gene (beta(A)). We recently derived embryonic stem (ES) cells from a novel knock-in mouse model of SCD and tested a protocol for correcting the sickle mutation by homologous recombination. In this paper, we demonstrate the replacement of the human beta(S)-globin gene with a human beta(A)-globin gene and the derivation of mice from these cells. The animals produce high levels of normal human hemoglobin (HbA) and the pathology associated with SCD is corrected. Hematologic values are restored to normal levels and organ pathology is ameliorated. These experiments provide a foundation for similar studies in human ES cells derived from sickle cell patients. Although efficient methods for production of human ES cells by somatic nuclear transfer must be developed, the data in this paper demonstrate that sickle cell disease can be corrected without the risk of insertional mutagenesis.


Subject(s)
Anemia, Sickle Cell/therapy , Embryo, Mammalian/metabolism , Genetic Therapy , Globins/genetics , Recombination, Genetic , Stem Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Animals , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian/cytology , Embryo, Mammalian/pathology , Genetic Vectors , Globins/biosynthesis , Humans , Insulator Elements/genetics , Lentivirus , Mice , Mice, Knockout , Mutagenesis, Insertional , Stem Cells/cytology , Stem Cells/pathology
14.
Nucleic Acids Res ; 32(16): e128, 2004 Sep 08.
Article in English | MEDLINE | ID: mdl-15356288

ABSTRACT

The construction of knockin vectors designed to modify endogenous genes in embryonic stem (ES) cells and the generation of mice from these modified cells is time consuming. The timeline of an experiment from the conception of an idea to the availability of mature mice is at least 9 months. We describe a method in which this timeline is typically reduced to 3 months. Knockin vectors are rapidly constructed from bacterial artificial chromosome clones by recombineering followed by gap-repair (GR) rescue, and mice are rapidly derived by injecting genetically modified ES cells into tetraploid blastocysts. We also describe a tandem affinity purification (TAP)/floxed marker gene plasmid and a GR rescue plasmid that can be used to TAP tag any murine gene. The combination of recombineering and tetraploid blastocyst complementation provides a means for large-scale TAP tagging of mammalian genes.


Subject(s)
Embryo, Mammalian/cytology , Gene Targeting/methods , Mice/genetics , Stem Cells , Animals , Blastocyst/ultrastructure , Cell Line , Chromosomes, Artificial, Bacterial , Cloning, Organism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Genetic Complementation Test , Injections , Kruppel-Like Transcription Factors , Polyploidy , Recombination, Genetic , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Biol Chem ; 279(26): 27518-24, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15084588

ABSTRACT

A new recombinant, human anti-sickling beta-globin polypeptide designated beta(AS3) (betaGly(16) --> Asp/betaGlu(22) --> Ala/betaThr(87) --> Gln) was designed to increase affinity for alpha-globin. The amino acid substitutions at beta22 and beta87 are located at axial and lateral contacts of the sickle hemoglobin (HbS) polymers and strongly inhibit deoxy-HbS polymerization. The beta16 substitution confers the recombinant beta-globin subunit (beta(AS3)) with a competitive advantage over beta(S) for interaction with the alpha-globin polypeptide. Transgenic mouse lines that synthesize high levels of HbAS3 (alpha(2)beta(AS3)(2)) were established, and recombinant HbAS3 was purified from hemolysates and then characterized. HbAS3 binds oxygen cooperatively and has an oxygen affinity that is comparable with fetal hemoglobin. Delay time experiments demonstrate that HbAS3 is a potent inhibitor of HbS polymerization. Subunit competition studies confirm that beta(AS3) has a distinct advantage over beta(S) for dimerization with alpha-globin. When equal amounts of beta(S)- and beta(AS3)-globin monomers compete for limiting alpha-globin chains up to 82% of the tetramers formed is HbAS3. Knock-out transgenic mice that express exclusively human HbAS3 were produced. When these mice were bred with knock-out transgenic sickle mice the beta(AS3) polypeptides corrected all hematological parameters and organ pathology associated with the disease. Expression of beta(AS3)-globin should effectively lower the concentration of HbS in erythrocytes of patients with sickle cell disease, especially in the 30% percent of these individuals who coinherit alpha-thalassemia. Therefore, constructs expressing the beta(AS3)-globin gene may be suitable for future clinical trials for sickle cell disease.


Subject(s)
Antisickling Agents/pharmacology , Fetal Hemoglobin/pharmacology , Globins/genetics , Hemoglobins/pharmacology , Adult , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Antisickling Agents/chemistry , Disease Models, Animal , Erythrocytes, Abnormal/cytology , Erythrocytes, Abnormal/metabolism , Erythrocytes, Abnormal/ultrastructure , Hematologic Tests , Hemoglobins/chemistry , Hemoglobins/genetics , Humans , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Liver/metabolism , Liver/pathology , Liver/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Spleen/metabolism , Spleen/pathology , Spleen/ultrastructure
16.
Blood ; 102(13): 4312-9, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12933581

ABSTRACT

Although sickle cell anemia was the first hereditary disease to be understood at the molecular level, there is still no adequate long-term treatment. Allogeneic bone marrow transplantation is the only available cure, but this procedure is limited to a minority of patients with an available, histocompatible donor. Autologous transplantation of bone marrow stem cells that are transduced with a stably expressed, antisickling globin gene would benefit a majority of patients with sickle cell disease. Therefore, the development of a gene therapy protocol that corrects the disease in an animal model and is directly translatable to human patients is critical. A method is described in which unmobilized, highly purified bone marrow stem cells are transduced with a minimum amount of self-inactivating (SIN) lentiviral vector containing a potent antisickling beta-globin gene. These cells, which were transduced in the absence of cytokine stimulation, fully reconstitute irradiated recipients and correct the hemolytic anemia and organ pathology that characterize the disease in humans. The mean increase of hemoglobin concentration was 46 g/L (4.6 g/dL) and the average lentiviral copy number was 2.2; therefore, a 21-g/L /vector copy increase (2.1-g/dL) was achieved. This transduction protocol may be directly translatable to patients with sickle cell disease who cannot tolerate current bone marrow mobilization procedures and may not safely be exposed to large viral loads.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Vectors/therapeutic use , Globins/genetics , Hematopoietic Stem Cell Transplantation , Lentivirus/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Animals, Congenic , Cell Separation , Defective Viruses/genetics , Disease Models, Animal , Female , Globins/deficiency , Kidney/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Radiation Chimera , Spleen/pathology , Transduction, Genetic , beta-Thalassemia/genetics
17.
Blood ; 101(3): 903-6, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12393544

ABSTRACT

We previously described a mouse line that contains green myelomonocytic cells due to the knock-in of enhanced green fluorescence protein (EGFP) into the lysozyme M gene.(1) We have now created a transgenic line with fluorescent erythroid cells using a beta-globin locus control region driving the enhanced cyan fluorescence protein (ECFP) gene. These mice exhibit cyan fluorescent cells specifically in the erythroid compartment and in megakaryocyte-erythroid progenitors. Crossing the animals with lysozyme EGFP mice yielded a line in which live erythroid and myeloid cells can readily be distinguished by fluorescence microscopy and by fluorescence-activated cell-sorter scanner. This cross allowed unambiguous identification of unstained mixed erythroid-myeloid colonies for the first time. The new mouse lines should become useful tools to dissect the branching between erythroid and myelomonocytic cells during in vitro differentiation of definitive multipotent progenitors.


Subject(s)
Erythrocytes/cytology , Luminescent Proteins/genetics , Myeloid Cells/cytology , Animals , Cell Line , Cytological Techniques , Erythrocytes/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Flow Cytometry , Globins/genetics , Green Fluorescent Proteins , Mice , Mice, Transgenic , Microscopy, Fluorescence , Muramidase/genetics , Myeloid Cells/metabolism , Transgenes/genetics
18.
J Biol Chem ; 278(6): 4194-204, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12401783

ABSTRACT

The intermittent vascular occlusion occurring in sickle cell disease (SCD) leads to ischemia-reperfusion injury and activation of inflammatory processes including enhanced production of reactive oxygen species and increased expression of inducible nitric-oxide synthase (NOS2). Appreciating that impaired nitric oxide-dependent vascular function and the concomitant formation of oxidizing and nitrating species occur in concert with increased rates of tissue reactive oxygen species production, liver and kidney NOS2 expression, tissue 3-nitrotyrosine (NO(2)Tyr) formation and apoptosis were evaluated in human SCD tissues and a murine model of SCD. Liver and kidney NOS2 expression and NO(2)Tyr immunoreactivity were significantly increased in SCD mice and humans, but not in nondiseased tissues. TdT-mediated nick end-label (TUNEL) staining showed apoptotic cells in regions expressing elevated levels of NOS2 and NO(2)Tyr in all SCD tissues. Gas chromatography mass spectrometry analysis revealed increased plasma protein NO(2)Tyr content and increased levels of hepatic and renal protein NO(2)Tyr derivatives in SCD (21.4 +/- 2.6 and 37.5 +/- 7.8 ng/mg) versus wild type mice (8.2 +/- 2.2 and 10 +/- 1.2 ng/mg), respectively. Western blot analysis and immunoprecipitation of SCD mouse liver and kidney proteins revealed one principal NO(2)Tyr-containing protein of 42 kDa, compared with controls. Enzymatic in-gel digestion and MALDI-TOF mass spectrometry identified this nitrated protein as actin. Electrospray ionization and fragment analysis by tandem mass spectrometry revealed that 3 of 15 actin tyrosine residues are nitrated (Tyr(91), Tyr(198), and Tyr(240)) at positions that significantly modify actin assembly. Confocal microscopy of SCD human and mouse tissues revealed that nitration led to morphologically distinct disorganization of filamentous actin. In aggregate, we have observed that the hemoglobin point mutation of sickle cell disease that mediates hemoglobin polymerization defects is translated, via inflammatory oxidant reactions, into defective cytoskeletal polymerization.


Subject(s)
Actins/metabolism , Anemia, Sickle Cell/metabolism , Cytoskeleton/metabolism , Nitric Oxide/physiology , Reactive Oxygen Species/metabolism , Tyrosine/metabolism , Actins/chemistry , Amino Acid Sequence , Anemia, Sickle Cell/enzymology , Animals , Humans , In Situ Nick-End Labeling , Kidney/enzymology , Kidney/metabolism , Liver/enzymology , Liver/metabolism , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...