Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Pept Res ; 63(1): 1-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14984567

ABSTRACT

A large body of data suggests that the linker histones family (H1) affects gene expression. Investigation of the linker histones role is then of a major interest in cell cycle studies with implications in gene therapy. Indeed, it has been shown that in most tissues a switch of histone subtypes occurs when the cells cease to divide. To investigate linker histone role in gene or transgene expression, an antibody against subtypes of H1 would be useful for immunoprecipitation experiments and further assays measuring H1subtypes-DNA interactions in living cells. In order to produce an antibody against the H1e subtype of linker histones, two synthetic peptides derived from two regions of the H1e mouse histone protein were examined for their potential, [as keyhole limpet hemocyanin (KLH) conjugates] to elicit polyclonal anti-H1e antibodies in New Zealand white rabbits. Selection of the peptide sequences was based on amino acid differences within the different classes of histones and between mice and rabbit histones as well. The evaluation of their potential immunogenic properties was based on examination of peptide hydropathy using predicting algorithms. Immunoglobulins (IgG) obtained from immunized and nonimmunized rabbits were tested using enzyme-linked immunosorbent assay (ELISA) procedures, Western immunoblot, and immunofluorescence experiments. Results showed that the selected synthetic peptides gave rise to a high-titer polyclonal antibody able to recognize the H1e histone under various conditions. This polyclonal antibody did not cross-react with other histones. To our knowledge, this is the first antibody produced against the mouse H1e linker histone.


Subject(s)
Antibodies/immunology , Histones/immunology , Peptides/immunology , Amino Acid Sequence , Animals , Antibodies/blood , Blotting, Western , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Histones/analysis , Histones/chemistry , Mice , Molecular Sequence Data , Peptides/chemical synthesis , Rabbits
2.
Science ; 294(5550): 2368-71, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11743206

ABSTRACT

Sickle cell disease (SCD) is caused by a single point mutation in the human betaA globin gene that results in the formation of an abnormal hemoglobin [HbS (alpha2betaS2)]. We designed a betaA globin gene variant that prevents HbS polymerization and introduced it into a lentiviral vector we optimized for transfer to hematopoietic stem cells and gene expression in the adult red blood cell lineage. Long-term expression (up to 10 months) was achieved, without preselection, in all transplanted mice with erythroid-specific accumulation of the antisickling protein in up to 52% of total hemoglobin and 99% of circulating red blood cells. In two mouse SCD models, Berkeley and SAD, inhibition of red blood cell dehydration and sickling was achieved with correction of hematological parameters, splenomegaly, and prevention of the characteristic urine concentration defect.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy , Genetic Vectors , Globins/genetics , HIV-1/genetics , Anemia, Sickle Cell/genetics , Animals , Disease Models, Animal , Erythrocytes/metabolism , Gene Expression , Globins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hemoglobin, Sickle/metabolism , Humans , Lentivirus/genetics , Locus Control Region , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxyhemoglobins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thalassemia/genetics , Thalassemia/therapy , Transduction, Genetic , Transgenes , beta-Globins
3.
Nature ; 413(6855): 488-94, 2001 Oct 04.
Article in English | MEDLINE | ID: mdl-11586351

ABSTRACT

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening systemic illness of abrupt onset and unknown cause. Proteolysis of the blood-clotting protein von Willebrand factor (VWF) observed in normal plasma is decreased in TTP patients. However, the identity of the responsible protease and its role in the pathophysiology of TTP remain unknown. We performed genome-wide linkage analysis in four pedigrees of humans with congenital TTP and mapped the responsible genetic locus to chromosome 9q34. A predicted gene in the identified interval corresponds to a segment of a much larger transcript, identifying a new member of the ADAMTS family of zinc metalloproteinase genes (ADAMTS13). Analysis of patients' genomic DNA identified 12 mutations in the ADAMTS13 gene, accounting for 14 of the 15 disease alleles studied. We show that deficiency of ADAMTS13 is the molecular mechanism responsible for TTP, and suggest that physiologic proteolysis of VWF and/or other ADAMTS13 substrates is required for normal vascular homeostasis.


Subject(s)
Metalloendopeptidases/genetics , Mutation , Purpura, Thrombotic Thrombocytopenic/genetics , von Willebrand Factor/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 9 , DNA Mutational Analysis , Female , Humans , Male , Metalloendopeptidases/blood , Metalloendopeptidases/physiology , Molecular Sequence Data , Multigene Family , Pedigree , Physical Chromosome Mapping , Purpura, Thrombotic Thrombocytopenic/congenital , Purpura, Thrombotic Thrombocytopenic/enzymology
4.
Blood ; 98(7): 2022-7, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567985

ABSTRACT

The mammalian beta-globin locus is a multigenic, developmentally regulated, tissue-specific locus from which gene expression is regulated by a distal regulatory region, the locus control region (LCR). The functional mechanism by which the beta-globin LCR stimulates transcription of the linked beta-like globin genes remains unknown. The LCR is composed of a series of 5 DNaseI hypersensitive sites (5'HSs) that form in the nucleus of erythroid precursors. These HSs are conserved among mammals, bind transcription factors that also bind to other parts of the locus, and compose the functional components of the LCR. To test the hypothesis that individual HSs have unique properties, homologous recombination was used to construct 5 lines of mice with individual deletions of each of the 5'HSs of the endogenous murine beta-globin LCR. Here it is reported that deletion of 5'HS1 reduces expression of the linked genes by up to 24%, while deletion of 5'HS4 leads to reductions of up to 27%. These deletions do not perturb the normal stage-specific expression of genes from this multigenic locus. In conjunction with previous studies of deletions of the other HSs and studies of deletion of the entire LCR, it is concluded that (1) none of the 5'HSs is essential for nearly normal expression; (2) none of the HSs is required for proper developmental expression; and (3) the HSs do not appear to synergize either structurally or functionally, but rather form independently and appear to contribute additively to the overall expression from the locus.


Subject(s)
Base Sequence , Deoxyribonuclease I/metabolism , Globins/genetics , Locus Control Region/genetics , Sequence Deletion , Age Factors , Animals , Binding Sites , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/metabolism , Globins/metabolism , Mice , Mice, Knockout/genetics , Recombination, Genetic , Tissue Distribution
5.
Mol Cell Biol ; 21(9): 2969-80, 2001 May.
Article in English | MEDLINE | ID: mdl-11287603

ABSTRACT

The major distal regulatory sequence for the beta-globin gene locus, the locus control region (LCR), is composed of multiple hypersensitive sites (HSs). Different models for LCR function postulate that the HSs act either independently or synergistically. To test these possibilities, we have constructed a series of expression cassettes in which the gene encoding the enhanced green fluorescent protein (EGFP) is under the control of DNA fragments containing single and multiple HSs of the LCR. LCR DNA fragments containing only the minimal region needed for position-independent expression (HS cores) or containing cores plus flanking sequences (HS units) were compared to ascertain whether conserved sequences between the HS cores contributed to enhancement. Expression of these constructs was measured after targeted integration into three defined loci in murine erythroleukemia cells using recombinase-mediated cassette exchange. At all three marked loci, synergistic enhancement of expression was observed in cassettes containing a combination of HS2, HS3, and HS4 units. In contrast, HS2, HS3, and HS4 cores (without flanking sequences) give an activity equivalent to the sum of the activities of the individual HS cores. These data suggest a model in which an HS core plus flanking regions, bound by specific proteins, forms a structure needed for interaction with other HS units to confer strong enhancement by the LCR. The three targeted integration sites differ substantially in their permissivity for expression, but even the largest LCR construct tested could not overcome these position effects to confer equal expression at all three sites.


Subject(s)
Enhancer Elements, Genetic , Globins/genetics , Locus Control Region , Binding Sites , Humans
6.
Mol Cell Biol ; 21(1): 298-309, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11113204

ABSTRACT

We have inserted two expression cassettes at tagged reference chromosomal sites by using recombinase-mediated cassette exchange in mammalian cells. The three sites of integration displayed either stable or silencing position effects that were dominant over the different enhancers present in the cassettes. These position effects were strongly dependent on the orientation of the construct within the locus, with one orientation being permissive for expression and the other being nonpermissive. Orientation-specific silencing, which was observed at two of the three site tested, was associated with hypermethylation but not with changes in chromatin structure, as judged by DNase I hypersensitivity assays. Using CRE recombinase, we were able to switch in vivo the orientation of the transgenes from the permissive to the nonpermissive orientation and vice versa. Switching from the permissive to the nonpermissive orientation led to silencing, but switching from the nonpermissive to the permissive orientation did not lead to reactivation of the transgene. Instead, transgene expression occurred dynamically by transcriptional oscillations, with 10 to 20% of the cells expressing at any given time. This result suggested that the cassette had been imprinted (epigenetically tagged) while it was in the nonpermissive orientation. Methylation analysis revealed that the methylation state of the inverted cassettes resembled that of silenced cassettes except that the enhancer had selectively lost some of its methylation. Sorting of the expressing and nonexpressing cell populations provided evidence that the transcriptional oscillations of the epigenetically tagged cassette are associated with changes in the methylation status of regulatory elements in the transgene. This suggests that transgene methylation is more dynamic than was previously assumed.


Subject(s)
Chromatin/genetics , Gene Expression Regulation/genetics , Mutagenesis, Insertional/genetics , Transgenes/genetics , Viral Proteins , Animals , Azacitidine/pharmacology , DNA Methylation/drug effects , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic/genetics , Flow Cytometry , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Genes, Reporter/genetics , Genomic Imprinting/genetics , Globins/genetics , Hydroxamic Acids/pharmacology , In Situ Hybridization, Fluorescence , Integrases/metabolism , Locus Control Region/genetics , Mice , Promoter Regions, Genetic/genetics , Transcriptional Activation/drug effects , Transfection , Tumor Cells, Cultured
7.
Mol Cell Biol ; 20(24): 9103-12, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11094062

ABSTRACT

We have developed a strategy to introduce in vitro-methylated DNA into defined chromosomal locations. Using this system, we examined the effects of methylation on transcription, chromatin structure, histone acetylation, and replication timing by targeting methylated and unmethylated constructs to marked genomic sites. At two sites, which support stable expression from an unmethylated enhancer-reporter construct, introduction of an in vitro-methylated but otherwise identical construct results in specific changes in transgene conformation and activity, including loss of the promoter DNase I-hypersensitive site, localized hypoacetylation of histones H3 and H4 within the reporter gene, and a block to transcriptional initiation. Insertion of methylated constructs does not alter the early replication timing of the loci and does not result in de novo methylation of flanking genomic sequences. Methylation at the promoter and gene is stable over time, as is the repression of transcription. Surprisingly, sequences within the enhancer are demethylated, the hypersensitive site forms, and the enhancer is hyperacetylated. Nevertheless, the enhancer is unable to activate the methylated and hypoacetylated reporter. Our findings suggest that CpG methylation represses transcription by interfering with RNA polymerase initiation via a mechanism that involves localized histone deacetylation. This repression is dominant over a remodeled enhancer but neither results in nor requires region-wide changes in DNA replication or chromatin structure.


Subject(s)
Chromatin/metabolism , DNA Methylation , DNA Replication/genetics , Gene Targeting/methods , Histones/metabolism , Transcription, Genetic/genetics , Viral Proteins , Acetylation , Animals , Artificial Gene Fusion , Blotting, Southern , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA/metabolism , DNA-Directed RNA Polymerases/metabolism , Enhancer Elements, Genetic/genetics , Flow Cytometry , Gene Expression Regulation , Genes, Reporter/genetics , Humans , Integrases/genetics , Integrases/metabolism , Plasmids/genetics , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic/genetics , Time Factors , Transgenes/genetics
8.
Blood Cells Mol Dis ; 26(4): 331-47, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11042035

ABSTRACT

When present in the homozygous form, hemoglobin C (HbC, CC disease) increases red cell density, a feature that is the major factor underlying the pathology in patients with SC disease (Fabry et al., JCI 70, 1315, 1982). The basis for the increased red cell density has not yet been fully defined. We have generated a HbC mouse in which the most successful founder expresses 56% human alpha and 34% human beta(C). We introduced knockouts (KO) of mouse alpha- and beta-globins in various combinations. In contrast to many KO mice, all partial KOs have normal MCH. Full KOs that express exclusively HbC and no mouse globins have minimally reduced MCH (13. 7 +/- 0.3 pg/cell vs 14.5 +/- 1.0 for C57BL/6) and a ratio of beta- to alpha-globin chains of 0.88 determined by chain synthesis; hence, these mice are not thalassemic. Mice with beta(C) > 30% have increased MCHC, dense reticulocytes, and increased K:Cl cotransport. Red cell morphology studied by SEM is strikingly similar to that of human CC cells with bizarre folded cells. We conclude that red cells of these mice have many properties that closely parallel the pathology of human disease in which HbC is the major determinant of pathogenesis. These studies also establish the existence of the interactions with other gene products that are necessary for pleiotropic effects (red cell dehydration, elevated K:Cl cotransport, morphological changes) that are also present in these transgenic mice, validating their usefulness in the analysis of pathophysiological events induced by HbC in red cells.


Subject(s)
Hemoglobin C/genetics , Alanine Transaminase/blood , Animals , Biological Transport , Bone Marrow/pathology , Breeding , Calcium/pharmacology , Cations/metabolism , Chromatography, High Pressure Liquid , Erythrocyte Indices , Erythrocytes/cytology , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Female , Founder Effect , Gene Expression , Genotype , Globins/genetics , Globins/metabolism , Hemoglobin C/metabolism , Humans , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Potassium/metabolism , Reticulocytes/cytology , Splenectomy
9.
J Biol Chem ; 275(33): 25831-9, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10827181

ABSTRACT

The human alpha-globin gene complex includes three functional globin genes (5'-zeta2-alpha2-alpha1-3') regulated by a common positive regulatory element named HS-40 displaying strong erythroid-specific enhancer activity. How this enhancer activity can be shared between different promoters present at different positions in the same complex is poorly understood. To address this question, we used homologous recombination to target the insertion of marker genes driven by cytomegalovirus or long terminal repeat promoters in both possible orientations either upstream or downstream from the HS-40 region into the single human alpha-globin gene locus present in hybrid mouse erythroleukemia cells. We also used CRE recombinase-mediated cassette exchange to target the insertion of a tagged alpha-globin gene at the same position downstream from HS-40. All these insertions led to a similar decrease in the HS-40-dependent transcription of downstream human alpha-globin genes in differentiated cells. Interestingly, this decrease is associated with the strong activation of the proximal newly inserted alpha-globin gene, whereas in marked contrast, the transcription of the non-erythroid marker genes remains insensitive to HS-40. Taken together, these results indicate that the enhancer activity of HS-40 can be trapped by non-erythroid promoters in both upstream and downstream directions without necessarily leading to their own activation.


Subject(s)
Enhancer Elements, Genetic , Globins/genetics , Globins/metabolism , Animals , Cell Nucleus/metabolism , Chromosomes, Human, Pair 16 , Cytomegalovirus/genetics , DNA Nucleotidyltransferases/metabolism , Humans , Mice , Models, Genetic , Mutagenesis, Insertional , Plasmids/metabolism , Promoter Regions, Genetic , Recombination, Genetic , Ribonucleases/metabolism , Terminal Repeat Sequences , Transcription, Genetic , Tumor Cells, Cultured
10.
Genomics ; 63(3): 417-24, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10704289

ABSTRACT

The mouse beta-globin gene cluster is regulated, at least in part, by a locus control region (LCR) composed of several developmentally stable DNase I hypersensitive sites located upstream of the genes. In this report, we examine the level of expression of the beta(min) and beta(maj) genes in adult mice in which HS2, HS3, or HS5,6 has been either deleted or replaced by a selectable marker via homologous recombination in ES cells. Primer extension analysis of RNA extracted from circulating reticulocytes and HPLC analysis of globin chains from peripheral red blood cells revealed that all mutations that reduce the overall output of the locus preferentially decrease beta(min) expression over beta(maj). The implications of these findings for the mechanism by which the LCR controls expression of the beta(maj) and beta(min) promoters are discussed.


Subject(s)
Gene Expression Regulation , Globins/genetics , Locus Control Region/genetics , Mice/genetics , Sequence Deletion , Animals , Base Sequence , Chromatin/ultrastructure , Chromatography, High Pressure Liquid , DNA Mutational Analysis , Erythroid Precursor Cells/metabolism , Female , Gene Targeting , Genotype , Globins/biosynthesis , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Genetic , Molecular Sequence Data , Recombination, Genetic
11.
Hum Mol Genet ; 9(4): 631-6, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10699186

ABSTRACT

Expression of a construct integrated at different genomic locations often varies because of position effects that have been subcategorized as stable (decreased level of expression) and variegating (decreased proportion of expressing cells). It is well established that locus control regions (LCRs) generally overcome position effects in transgenes. However, whether stable and variegated position effects are equally overcome by an intact LCR has not been determined. We report that single-copy yeast artificial chromosome transgenes containing an unmodified human beta -globin locus were not subject to detectable stable position effects but did undergo mild to severe variegating position effects at three of the four non-centromeric integration sites tested. We also find that, at a given integration site, the distance and the orientation of the LCR relative to the regulated gene contributes to the likelihood of variegating position effects, and can affect the magnitude of its transcriptional enhancement. DNase I hypersensitive site (HSS) formation varies with the proportion of expressing cells, not the level of gene expression, suggesting that silencing of the transgene is associated with a lack of HSS formation in the LCR region. We conclude that transcriptional enhancement and variegating position effects are caused by fundamentally different but inter-dependent mechanisms.


Subject(s)
Chromosomes, Artificial, Yeast/genetics , Gene Expression Regulation , Globins/genetics , Transgenes , Animals , Cells, Cultured , Centromere/genetics , Chromosome Inversion , DNA Transposable Elements , Deoxyribonuclease I/metabolism , Globins/biosynthesis , Humans , In Situ Hybridization, Fluorescence , Locus Control Region , Mice , Mice, Transgenic , Spleen/cytology
12.
J Mol Biol ; 292(4): 779-85, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10525404

ABSTRACT

Expression of experimental constructs in mammalian cells or transgenic animals is difficult to control because it is markedly influenced by position effects. This has limited both the analysis of cis -DNA regulatory elements for transcription and replication, and the physiological analysis of proteins expressed from transgenes. We report here two new methods based on the concept of recombinase-mediated cassette exchange (RMCE) to perform site-specific chromosomal integration. The first method permits the exchange of a negative selectable marker pre-localized on the chromosome with a transgene via a CRE-mediated double recombination between inverted Lox sites. Integration efficiency is close to 100 % of negatively selected mouse erythroleukemia cells and ranges from 10 to 50 % in embryonic stem cells. The second method allows RMCE with no selection at all except for cells that have taken up plasmid transiently. While less efficient, this technique permits novel experimental approaches. We find that integration of a transgene at a given genomic site leads to reproducible expression. RMCE should be useful to develop artificial genetic loci that impart specific and reproducible regulation of transgenes in higher eukaryotes. This should facilitate the analysis of cis -regulatory DNA elements governing expression and position effects, improve our control over the physiological effects of transgenes, and accelerate the development of animal models for complex human diseases.


Subject(s)
Chromosomes/genetics , Genetic Markers/genetics , Integrases/metabolism , Mutagenesis, Insertional/methods , Mutagenesis, Site-Directed/genetics , Viral Proteins , Animals , Attachment Sites, Microbiological/genetics , Clone Cells/metabolism , Cloning, Molecular , Gene Expression , Genes, Reporter/genetics , Genetic Vectors/genetics , Genome , Mice , Mutagenesis, Insertional/genetics , Recombination, Genetic/genetics , Stem Cells/metabolism , Transfection , Transgenes/genetics , Tumor Cells, Cultured
13.
Blood Cells Mol Dis ; 25(2): 110-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10389593

ABSTRACT

Our current strategy for gene therapy of sickle cell anemia involves retroviral vectors capable of transducing "designer" globin genes that code for novel anti-sickling globins (while resisting digestion by a ribozyme), coupled with the expression of a hammerhead ribozyme that can selectively cleave the human beta s mRNA. In this report, we have tested in vivo an anti-beta s hammerhead ribozyme embedded within a cDNA coding for the luciferase reporter gene driven by the human beta-globin promoter and hyper-sensitive sites 3 and 4 of the locus control region. We have created mice transgenic for this luciferase-ribozyme construct and bred the ribozyme transgene into mice that were already transgenic for the human beta s gene. We then measured expression of the beta s transgene at the protein and RNA levels by HPLC and primer extension. The presence of the ribozyme was associated with a statistically significant reduction in the level of beta s mRNA in spleen stress reticulocytes (from 60.5 +/- 4.1% to 52.9 +/- 4.2%) and in the percentage of beta s globin chains in very young mice (from 44.5 +/- 0.6% to 40.8 +/- 0.7%). These results demonstrate that it is possible to decrease the concentration of beta s chains and mRNA with the help of a hammerhead ribozyme. While the enormous amount of globin mRNA in reticulocytes is a challenge for ribozyme technology, the exquisite dependence of the delay time for formation of Hb S nuclei on the concentration of Hb S in red blood cells suggests that even a modest reduction in Hb S concentration would have therapeutic value.


Subject(s)
Hemoglobin, Sickle/genetics , RNA, Antisense/genetics , RNA, Catalytic/genetics , RNA, Messenger/genetics , Amino Acid Sequence , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Animals , Base Sequence , Chromatography, High Pressure Liquid , DNA Primers , Female , Genetic Engineering , Genetic Therapy , Globins/genetics , Globins/metabolism , Hemoglobin, Sickle/analysis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , RNA/genetics , RNA/metabolism , RNA, Antisense/physiology , RNA, Catalytic/physiology , RNA, Messenger/metabolism , Time Factors , Transgenes
14.
Mol Cell Biol ; 19(7): 4907-17, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373540

ABSTRACT

By using recombinase-mediated cassette exchange, a method that allows integration of single copies of different constructs at the same predetermined chromosomal location, several expression cassettes have been integrated at a randomly chosen locus in the genome of mouse erythroleukemia cells. The cassettes studied contain the human beta-globin promoter fused to lacZ coding sequences either alone or linked to DNase I-hypersensitive site HS2, HS3, or HS234 (a large locus control region fragment containing HS2, HS3, and HS4) of the human beta-globin locus control region. Analysis of expression of these cassettes revealed mosaic expression patterns reminiscent of, but clearly different from, position effect variegation. Further investigations demonstrated that these mosaic expression patterns are caused by dynamic activation and inactivation of the transcription unit, resulting in oscillations of expression. These oscillations occur once in every few cell cycles at a rate specific for the enhancer present at the locus. DNase I sensitivity studies revealed that the chromatin is accessible and that DNase-hypersensitive sites were present whether or not the transcription unit is active, suggesting that the oscillations occur between transcriptionally competent and transcriptionally active chromatin conformations, rather than between open and closed chromatin conformations. Treatment of oscillating cells with trichostatin A eliminates the oscillations only after the cells have passed through late G1 or early S, suggesting that these oscillations might be caused by changes in histone acetylation patterns.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Globins/genetics , Transcription, Genetic , Animals , Deoxyribonuclease I , G1 Phase , Humans , Hydroxamic Acids/pharmacology , Leukemia, Erythroblastic, Acute , Mice , Mosaicism , S Phase , Transcriptional Activation , Tumor Cells, Cultured
15.
Mol Cell Biol ; 19(5): 3714-26, 1999 May.
Article in English | MEDLINE | ID: mdl-10207095

ABSTRACT

A constitutive DNase I-hypersensitive site 5' of the chicken beta-globin locus, termed 5'HS4 or cHS4, has been shown to insulate a promoter from the effect of an upstream enhancer and to reduce position effects on mini-white expression in Drosophila cells; on the basis of these findings, it has been designated a chromatin insulator. We have examined the effect of the cHS4 insulator in a system that assays both the level of gene expression and the rate of transcriptional silencing. Because transgenes flanked by insulator elements are shielded from position effects in Drosophila cells, we tested the ability of cHS4 to protect transgenes from position effects in mammalian cells. Flanking of an expression vector with the cHS4 insulator in a colony assay did not increase the number of G418-resistant colonies. Using lox/cre-based recombinase-mediated cassette exchange to control integration position, we studied the effect of cHS4 on the silencing of an integrated beta-geo reporter at three genomic sites in K562 erythroleukemia cells. In this assay, enhancers act to suppress silencing but do not increase expression levels. While cHS4 blocked enhancement at each integration site, the strength of the effect varied from site to site. Furthermore, at some sites, cHS4 inhibited the enhancer effect either when placed between the enhancer and the promoter or when placed upstream of the enhancer. These results suggest that the activity of cHS4 is not dominant in all contexts and is unlikely to prevent silencing at all genomic integration sites.


Subject(s)
Enhancer Elements, Genetic/genetics , Globins/genetics , Integrases , Suppression, Genetic , Animals , Chickens , DNA Nucleotidyltransferases/genetics , Drug Resistance/genetics , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Gentamicins/pharmacology , Humans , Mammals , Recombinases , Transgenes/genetics , Tumor Cells, Cultured
16.
Acta Haematol ; 102(4): 172-9, 1999.
Article in English | MEDLINE | ID: mdl-10725758

ABSTRACT

We have observed that a severe decrease in the number of erythroid progenitor cells follows the long-term culture (LTC) of human primitive stem cells on an established mouse fibroblast feeder layer. This suggests that one, or several, factors necessary to support erythroid differentiation might be missing in these culture conditions. To improve the erythroid differentiation and stem cell output of LTCs, we have examined the hypothesis that the factors that regulate pluripotent stem cell proliferation and erythroid commitment can be found in media conditioned by embryonic stem (ES) cells. We have found that media conditioned by undifferentiated and differentiating ES cells can affect differentiation patterns in both short-term culture and LTC assays. Medium conditioned by undifferentiated ES cells increases the numbers of estimated LTC-initiating cells (est.LTC-IC) and potentiates granulocytic differentiation. In contrast, medium conditioned by ES cells undergoing differentiation increases the number of est.LTC-IC and is a powerful promoter of erythroid differentiation. In the presence of this medium, the number of erythroid progenitors generated after 5 weeks of LTC was increased up to 100-fold as compared with controls, and the number of est.LTC-IC was amplified up to 140-fold. These results offer a new approach for the identification of factors implicated in stem cell proliferation, self-renewal and commitment. In addition, the improved LTC conditions for the expansion of stem cells without reduction of their in vitro differentiation potential should be useful for a wide range of applications.


Subject(s)
Hematinics/metabolism , Hematopoiesis , Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Hematinics/isolation & purification , Hematinics/pharmacology , Hematopoiesis/drug effects , Humans , Mice , Stromal Cells
17.
Blood ; 92(11): 4375-82, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834244

ABSTRACT

Hemoglobin (Hb) S-Oman has two mutations in the beta-chains. In addition to the classic betaS mutation (beta6 Glu --> Val), it contains a second mutation in the same chain (beta121 Glu --> Lys) identical to that of HbOARAB. We have studied a pedigree of heterozygous carriers of HbS-Oman that segregates into two types of patients: those expressing about 20% HbS-Oman and concomitant -/ thalassemia and those with about 14% of HbS-Oman and concomitant -/- thalassemia. The higher expressors of S-Oman have a sickle cell anemia (SS) clinical syndrome of moderate intensity, while the lower expressors have no clinical syndrome, and are comparable to the solitary case first described in Oman. In addition, the higher expressors exhibit a unique form of irreversibly sickled cell reminiscent of a "yarn and knitting needle" shape, in addition to folded and target cells. The CSAT of S-Oman is identical to that of S-Antilles, another supersickling hemoglobin, whose carriers express the abnormal hemoglobin at 40% to 50%, with a very similar clinical picture to HbS-Oman. Because the level of expression is so different and the clinical picture so similar, and based on the hemolysates CSAT's, we conclude that HbS-Oman produces pathology beyond its sickling tendencies. A clue for this additional pathogenesis is found in the fact that homozygous HbOARAB, which has the same second substitution as S-Oman, has a moderately severe hemolytic anemia; when HbOARAB is combined with HbS, it makes the phenotype of this double heterozygote as severe as SS. Properties of HbS-Oman red blood cells (RBCs) include reticulocytes that are much denser than normal (similar to those of SC and CC disease), a decrease in the Km for Ca2+ needed to activate the Gardos' channel (making this transporter more sensitive to Ca2+), increased association of HbS-Oman with the RBC membrane, the presence of dense cells by isopycnic gradient, the presence of folded cells, and abundant nidus of polymerization under the membrane. Other properties include a clear increase in volume and N-ethylmaleimide-stimulated K:Cl cotransport in RBCs expressing more than 20% HbS-Oman. We conclude that the pathology of heterozygous S-Oman is the product of the sickling properties of the beta6 Val mutation which are enhanced by the second mutation at beta121. In addition, the syndrome is further enhanced by a hemolytic anemia induced by the mutation at beta121. We speculate that this pathology results from the abnormal association of the highly positively charged HbS-Oman (3 charges different from normal hemoglobin) with the RBC membrane.


Subject(s)
Hemoglobin, Sickle/genetics , Hemoglobins, Abnormal/genetics , Mutation , Syndrome , Adolescent , Adult , Anemia, Sickle Cell/genetics , Child , Female , Heterozygote , Humans , Male , Pedigree , alpha-Thalassemia/genetics
18.
Blood ; 90(9): 3332-44, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9345015

ABSTRACT

Efficient integration of transgenes at preselected chromosomal locations was achieved in mammalian cells by recombinase-mediated-cassette-exchange (RMCE), a novel procedure that makes use of the CRE recombinase together with Lox sites bearing different spacer regions. We have applied RMCE to the study of the human beta-globin gene Locus Control Region by integrating at the same genetic locus in MEL cells, a LacZ gene driven by the human beta-globin promoter linked to HS2 and HS3 alone or in combination with HS4. Expression studies at the cell population level and in individual cells before and after induction of differentiation with hemin or DMSO show that the presence of these enhancers is associated with variegated patterns of expression. We were able to show that the LCR fragments tested act by controlling both the probability of expression and the rate of transcription of the linked beta-globin promoter. Both of these factors were also dependent on the state of differentiation of the MELc and on the presence of a second transcription unit located in cis. The ability to manipulate by RMCE constructs integrated into chromosomes should help in the creation of complex, rationally designed, artificial genetic loci.


Subject(s)
DNA Nucleotidyltransferases/genetics , Enhancer Elements, Genetic , Gene Targeting/methods , Globins/genetics , Integrases , Transcription, Genetic , Animals , Chromosomes/genetics , Gene Expression Regulation , Humans , Mice , Promoter Regions, Genetic/genetics , Recombinases
19.
Am J Hematol ; 54(1): 30-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8980258

ABSTRACT

HS-26, the mouse homologue of HS-40, is the major regulatory element of the mouse alpha-globin gene locus. Like HS-40, HS-26 is located within an intron of a house-keeping gene; comparison of the nucleotide sequences of HS-26 and HS-40 reveals conservation of the sequences and positions of several DNA binding motifs in the 5' regions of both elements (3 GATA, 2 NFE-2, and 1 CACCC sites) and the absence in HS-26 of three CACCC sites and one GATA site that are present in the 3' region of HS-40, suggesting that the two elements might not be identical. We report here that when HS-26 is linked to a 1.5 kb Pstl human alpha-globin gene fragment, it has a weak enhancer activity in induced MEL cells and in transgenic embryos, and it does not have any detectable activity in adult transgenic mice. This suggests that HS-26 does not have Locus Control Region (LCR) activity but can act as an enhancer during the embryonic life when integrated at a permissive locus. To further test the importance of HS-26 at its natural locus, we have generated embryonic stem cells and chimeric animals in which 350 bp containing HS-26 have been replaced by a neomycin resistance gene by homologous recombination. The sizes of the chimeras' red cells were then estimated by measuring forward scattering on a FacsScan apparatus in hypotonic conditions. This revealed that a fraction of the chimeric animals' red cells were smaller than normal mouse red cells and were very similar to cells from mice heterozygous for alpha-thalassemia. Density gradient analysis also suggested the presence of thalassemic cells. These results indicated that despite its lack of LCR activity, HS-26 is important for the regulation of the mouse alpha-globin gene locus.


Subject(s)
Enhancer Elements, Genetic , Globins/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Mice, Transgenic , Transcription, Genetic
20.
Blood ; 79(7): 1846-54, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1558976

ABSTRACT

Erythrocyte (RBC) protein 4.2 (P4.2)-deficiency observed in Japanese individuals results in a hemolytic anemia associated with abnormally shaped (spherocytic, ovalocytic, and elliptocytic), osmotically fragile RBCs, the clinical presentation of which resembles hereditary spherocytosis (HS). By sodium dodecyl sulfate-polyacrylamide gel electrophoresis, P4.2-deficient individuals contain less than 1% of the normal membrane content of P4.2 and immunologic analysis shows that the P4.2 present exists as an equimolar doublet of 74-Kd and 72-Kd bands, in contrast to normal RBC membranes where a discrete 74-Kd band is not observed. RBC membranes from both of the biologic parents of a P4.2-deficient individual contained both the 74-Kd and the 72-Kd bands, demonstrating their heterozygosity for the P4.2 defect. The molecular basis of Japanese P4.2-deficiency was investigated by reverse transcription of total reticulocyte RNA, followed by polymerase chain reaction (PCR) amplification, subcloning, and sequencing. The complete cDNA sequence of a P4.2-deficient patient showed a single point mutation that changes codon 142 from GCT (alanine) to ACT (threonine) (Protein 4.2NIPPON). The mutation also eliminated an HgaI restriction site, therefore allowing rapid screening for the presence of the mutation. Screening of PCR-amplified genomic DNA showed that the mutation was present in the homozygous state in four (eight chromosomes) unrelated Japanese P4.2-deficient individuals and absent in 35 (70 chromosomes) P4.2-normal controls (including 15 Japanese [30 chromosomes]). The presence of the mutation was confirmed by allele-specific hybridization. The mutation occurred in an alternatively spliced exon that is present in two of four P4.2 mRNA splicing isoforms. These results demonstrate that Japanese P4.2-deficiency is closely associated with the P4.2 gene and does not arise secondarily to a defect in another membrane protein, and further suggest that the P4.2-deficiency is related to the pathogenesis of the hemolytic anemia in this variant form of recessively inherited spherocytosis.


Subject(s)
Alanine/genetics , Anemia, Hemolytic/genetics , Blood Proteins/genetics , DNA/chemistry , Threonine/genetics , Amino Acid Sequence , Anemia, Hemolytic/blood , Base Sequence , Blood Proteins/chemistry , Blood Proteins/deficiency , Blotting, Western , Codon , Cytoskeletal Proteins , Erythrocyte Membrane/chemistry , Humans , Japan , Membrane Proteins , Molecular Sequence Data , Mutation , Nucleic Acid Hybridization , Peptide Fragments/chemistry , Polymerase Chain Reaction , RNA Splicing , RNA, Messenger/genetics , Reticulocytes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL