Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Mol Biochem Parasitol ; 110(2): 259-72, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11071281

ABSTRACT

The malaria parasite Plasmodium falciparum undergoes distinct morphologic changes during its 48-h life cycle inside human red blood cells. Parasite proteinases appear to play important roles at all stages of the erythrocytic cycle of human malaria. Proteases involved in erythrocyte rupture and invasion are possibly required to breakdown erythrocyte membrane skeleton. To identify such proteases, soluble cytosolic extract of isolated trophozoites/schizonts was incubated with erythrocyte membrane ghosts or spectrin-actin depleted inside-out vesicles, which were then analyzed by SDS-PAGE. In both cases, a new protein band of 155 kDa was detected. The N-terminal peptide sequencing established that the 155 kDa band represents truncated ankyrin. Immunoblot analysis using defined monoclonal antibodies confirmed that ankyrin was cleaved at the C-terminus. While the enzyme preferentially cleaved ankyrin, degradation of protein 4.1 was also observed at high concentrations of the enzyme. The optimal activity of the purified enzyme, using ankyrin as substrate, was observed at pH 7.0-7.5, and the activity was strongly inhibited by standard inhibitors of cysteine proteinases (cystatin, NEM, leupeptin, E-64 and MDL 28 170), but not by inhibitors of aspartic (pepstatin) or serine (PMSF, DFP) proteinases. Furthermore, we demonstrate that protease digestion of ankyrin substantially reduces its interaction with ankyrin-depleted membrane vesicles. Ektacytometric measurements showed a dramatic increase in the rate of fragmentation of ghosts after treatment with the protease. Although the role of ankyrin cleavage in vivo remains to be determined, based on our findings we postulate that the parasite-derived cysteine protease activity cleaves host ankyrin thus weakening the ankyrin-band 3 binding interactions and destabilizing the erythrocyte membrane skeleton, which, in turn, facilitates parasite release. Further characterization of the enzyme may lead to the development of novel antimalarial drugs.


Subject(s)
Ankyrins/metabolism , Cysteine Endopeptidases/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/enzymology , Animals , Ankyrins/chemistry , Cysteine Endopeptidases/isolation & purification , Erythrocyte Membrane/chemistry , Erythrocytes/chemistry , Humans
4.
Biochim Biophys Acta ; 1493(1-2): 242-5, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10978531

ABSTRACT

The cDNA for a novel Plasmodium cysteine protease (falcipain-2) has been isolated from a Plasmodium falciparum cDNA library. A 602 bp fragment was amplified from P. falciparum by PCR using degenerate oligonucleotide primers. The primers were designed based upon the amino acids flanking the active site cysteine and asparagine residues that are conserved in the eukaryotic cysteine proteases. This fragment was used to screen a P. falciparum cDNA library and isolated a 2.1 kb clone that encoded a novel cysteine protease. The sequence of the 2.1 kb clone predicted a 56 kDa protein containing a typical signal sequence, a prosequence and a 24.7 kDa mature protease with 37% identity to falcipain-1, a hemoglobin-degrading cysteine protease of P. falciparum. Northern blot analysis detected a 2.1 kb message in trophozoites. Taken together, we have isolated a novel cysteine protease of P. falciparum, which may play an important role at the late stages of the erythrocytic cycle of the parasite.


Subject(s)
Cysteine Endopeptidases/genetics , Genes, Protozoan , Plasmodium falciparum/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , Cysteine Endopeptidases/chemistry , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Gene Library , Molecular Sequence Data , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Alignment
6.
Blood ; 92(8): 2940-50, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9763581

ABSTRACT

We have previously identified a novel protein that mediates the attachment of erythroblasts to macrophages in vitro. This attachment promotes terminal maturation and enucleation of erythroblasts (Hanspal and Hanspal, Blood 84:3494, 1994). This protein is referred to here as Emp for erythroblast macrophage protein. Two immunologically related isoforms of Emp with apparent molecular weights of 33 kD and 36 kD were detected in macrophage membranes. The complete amino acid sequence of the larger isoform of Emp was deduced from the nucleotide sequence of a full-length 2.0-kb cDNA that was isolated from a human macrophage cDNA library using affinity-purified anti-Emp antibodies. Of the 2,005 bp, 1,185 bp encode for 395 amino acids representing 43 kD (the sodium dodecyl sulfate-polyacrylamide gel electrophoresis [SDS-PAGE] molecular mass is 36 kD). Northern blot analysis of human macrophage poly(A) RNA detected a message for Emp of 2.1 kb. The deduced amino acid sequence contains a putative transmembrane domain near the N-terminus. To investigate the structure/function relationships of Emp, recombinant fusion proteins of full-length and truncated Emp were produced in bacteria, COS-7, and HeLa cells. Cell binding assays showed that the N-terminus is exposed on the cell surface. The recombinant Emp functions as a cell attachment molecule when expressed in heterologous cells. Furthermore, we showed that the demise of erythroblasts in the absence of Emp-mediated erythroblast-macrophage association is accompanied by apoptosis. We postulate that Emp-mediated contact between erythroblasts and macrophages promotes terminal maturation of erythroid cells by suppressing apoptosis.


Subject(s)
Blood Proteins/isolation & purification , Erythroblasts/cytology , Macrophages/cytology , Amino Acid Sequence , Animals , Apoptosis/physiology , Base Sequence , Blood Proteins/physiology , COS Cells , Cell Adhesion , Cell Adhesion Molecules , Cloning, Molecular , Cytoskeletal Proteins , DNA, Complementary/genetics , Erythroblasts/metabolism , Erythropoiesis , HeLa Cells , Humans , Macrophages/metabolism , Molecular Sequence Data , Protein Isoforms/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
7.
Blood ; 92(1): 329-38, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9639533

ABSTRACT

Band 3, the anion transport protein of the erythrocyte membrane, exists in the membrane as a mixture of dimers (B3D) and tetramers (B3T). The dimers are not linked to the skeleton and constitute the free mobile band 3 fraction. The tetramers are linked to the skeleton by their interaction with ankyrin. In this report we have examined the temporal synthesis and assembly of band 3 oligomers into the plasma membrane during red cell maturation. The oligomeric state of newly synthesized band 3 in early and late erythroblasts was analyzed by size-exclusion high-pressure liquid chromatography of band 3 extracts derived by mild extraction of plasma membranes with the nonionic detergent C12E8 (octaethylene glycol n-dodecyl monoether). This analysis revealed that at the early erythroblast stage, the newly synthesized band 3 is present predominantly as tetramers, whereas at the late stages of erythroid maturation, it is present exclusively as dimers. To examine whether the dimers and tetramers exist in the membrane as preformed stable species or whether they are interconvertible, the fate of band 3 species synthesized during erythroblast maturation was examined by pulse-chase analysis. We showed that the newly synthesized band 3 dimers and tetramers are stable and that there is no interconversion between these species in erythroblast membranes. Pulse-chase analysis followed by cellular fractionation showed that, in early erythroblasts, the newly synthesized band 3 tetramers are initially present in the microsomal fraction and later incorporated stably into the plasma membrane fraction. In contrast, in late erythroblasts the newly synthesized band 3 dimers move rapidly to the plasma membrane fraction but then recycle between the plasma membrane and microsomal fractions. Fluorescence photobleaching recovery studies showed that significant fractions of B3T and B3D are laterally mobile in early and late erythroblast plasma membranes, respectively, suggesting that many B3T-ankyrin complexes are unattached to the membrane skeleton in early erythroblasts and that the membrane skeleton has yet to become tightly organized in late erythroblasts. We postulate that in early erythroblasts, band 3 tetramers are transported through microsomes and stably incorporated into the plasma membrane. However, when ankyrin synthesis is downregulated in late erythroblasts, it appears that B3D are rapidly transported to the plasma membrane but then recycled between the plasma membrane and microsomal compartments. These observations may suggest novel roles for membrane skeletal proteins in stabilizing integral membrane protein oligomers at the plasma membrane and in regulating the endocytosis of such proteins.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/biosynthesis , Erythroblasts/metabolism , Erythrocyte Membrane/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/chemistry , Cell Differentiation , Dimerization , Erythroblasts/cytology , Mice
8.
Blood ; 91(6): 2146-51, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9490702

ABSTRACT

Glycophorin A is the major transmembrane sialoglycoprotein of red blood cells. It has been shown to contribute to the expression of the MN and Wright blood group antigens, to act as a receptor for the malaria parasite Plasmodium falciparum and Sendai virus, and along with the anion transporter, band 3, may contribute to the mechanical properties of the red blood cell membrane. Several lines of evidence suggest a close interaction between glycophorin A and band 3 during their biosynthesis. Recently, we have generated mice where the band 3 expression was completely eliminated by selective inactivation of the AE1 anion exchanger gene, thus allowing us to study the effect of band 3 on the expression of red blood cell membrane proteins. In this report, we show that the band 3 -/- red blood cells contain protein 4.1, adducin, dematin, p55, and glycophorin C. In contrast, the band 3 -/- red blood cells are completely devoid of glycophorin A (GPA), as assessed by Western blot and immunocytochemistry techniques, whereas the polymerase chain reaction (PCR) confirmed the presence of GPA mRNA. Pulse-label and pulse-chase experiments show that GPA is not incorporated in the membrane and is rapidly degraded in the cytoplasm. Based on these findings and other published evidence, we propose that band 3 plays a chaperone-like role, which is necessary for the recruitment of GPA to the red blood cell plasma membrane.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/genetics , Erythrocyte Membrane/metabolism , Glycophorins/deficiency , Animals , Anion Exchange Protein 1, Erythrocyte/deficiency , Anion Exchange Protein 1, Erythrocyte/physiology , Biological Transport , Blood Proteins/analysis , Glycophorins/genetics , Glycophorins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA, Messenger/analysis
9.
Curr Opin Hematol ; 4(2): 142-7, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9107532

ABSTRACT

In the bone marrow, erythropoiesis occurs in distinct anatomic units called erythroblastic islands that consist of a central macrophage surrounded by a ring of developing erythroblasts. The formation of erythroblastic islands involves adhesive interactions between the central macrophage and the ring of developing erythroblasts, between adjacent erythroblasts in the ring, and between macrophages/erythroblasts and the components of extracellular matrix. These adhesive interactions are mediated by specific pairs of cell surface receptors and counterreceptors (ligands) including integrins, the immunoglobulin superfamily, and cadherins. Several lines of evidence have suggested that cell-cell interactions among various cell types in the bone marrow play an important role in supporting and regulating hematopoietic differentiation. Cell surface adhesion molecules expressed during erythroid development and the likely counterreceptors for these molecules are discussed in this review.


Subject(s)
Cell Communication/physiology , Erythropoiesis/physiology , Animals , Humans
10.
Blood ; 88(12): 4500-9, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8977242

ABSTRACT

The AE1 gene is expressed in erythrocytes and the A-type intercalated cells of the kidney distal collecting duct. Although the 5' end of the principal transcript expressed in murine erythroid cells has previously been mapped to a cluster of transcription start sites located immediately upstream of exon 1, the 5' end of the mouse kidney transcript has not been identified. Using the anchored polymerase chain reaction technique to analyze mouse kidney AE1 mRNA, we identified an internal transcription start site located within erythroid intron 3. This site defines an exon of 37 nucleotides that forms the 5' end of the mouse kidney AE1 transcript. AE1 transcripts beginning at this internal start site could not be detected in RNA isolated from purified erythroid progenitor cells or from erythroid cells undergoing erythropoietin-dependent terminal maturation, although transcripts derived from the upstream site were abundant, indicating that only the upstream promoter is active during erythropoiesis. Transient expression of reporter constructs in erythroid and nonerythroid cell lines identified a proximal upstream region of approximately 135 nucleotides that was active as a basal promoter. However, an additional approximately 200 nucleotides of upstream sequence was required for induced levels of activity in erythroid cells. Although our functional approach does not yet indicate the precise sequences required for erythroid induction, the AE1 gene upstream region contains potential GATA sites at -154, -141, and -60; an E-box at -163; CACCC or GGTGG motifs at -188, -121, and -88; and an AP-1/NF-E2-like site at -42.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/genetics , Animals , Erythroid Precursor Cells/cytology , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/cytology , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Transcription, Genetic , Tumor Cells, Cultured
11.
J Clin Invest ; 98(10): 2300-7, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8941647

ABSTRACT

We studied a patient with a severe spherocytic hemolytic anemia without family history of spherocytosis. Analysis of patient's erythrocyte membrane proteins revealed spectrin deficiency and a truncated alpha spectrin protein. We determined that the patient is a compound heterozygote with two mutations in alpha spectrin gene. Mutation in the paternal allele, designated alpha spectrin(PRAGUE), is a transition A to G in the penultimate position of intron 36 that leads to skipping of exon 37, frameshift, and production of the truncated alpha spectrin protein. The maternal allele, designated alpha spectrin(LEPRA), contains transition C-->T in position -99 of intron 30. This mutation enhances an alternative acceptor splice site 70 nucleotides upstream from the regular site. The alternative splicing causes a frameshift and premature termination of translation leading to a significant decrease in alpha spectrin production. The alpha(LEPRA) mutation is linked to a spectrin alphaIIa marker that was found to be associated with recessive or nondominant spectrin-deficient hereditary spherocytosis in approximately 50% of studied families. We conclude that the alpha(LEPRA) mutation combined in trans with the alpha(PRAGUE) mutation underlie the severe hemolytic anemia in the proband. We suggest that allele alpha spectrin(LEPRA) may be frequently involved in pathogenesis of recessive or nondominant spectrin-deficient hereditary spherocytosis.


Subject(s)
Mutation , Spectrin/genetics , Spherocytosis, Hereditary/genetics , Adult , Alleles , Alternative Splicing , Amino Acid Sequence , Base Sequence , Blotting, Western , Child , DNA, Complementary/analysis , Deoxyribonuclease EcoRI/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Electrophoresis, Polyacrylamide Gel , Erythrocytes/metabolism , Exons/genetics , Female , Genome, Human , Humans , Introns/genetics , Male , Membrane Proteins/analysis , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , Spectrin/biosynthesis
12.
Cell ; 86(6): 917-27, 1996 Sep 20.
Article in English | MEDLINE | ID: mdl-8808627

ABSTRACT

The red blood cell (RBC) membrane protein AE1 provides high affinity binding sites for the membrane skeleton, a structure critical to RBC integrity. AE1 biosynthesis is postulated to be required for terminal erythropoiesis and membrane skeleton assembly. We used targeted mutagenesis to assess AE1 function in vivo. RBCs lacking AE1 spontaneously shed membrane vesicles and tubules, leading to severe spherocytosis and hemolysis, but the levels of the major skeleton components, the synthesis of spectrin in mutant erythroblasts, and skeletal architecture are normal or nearly normal. The results indicate that AE1 does not regulate RBC membrane skeleton assembly in vivo but is essential for membrane stability. We postulate that stabilization is achieved through AE1-lipid interactions and that loss of these interactions is a key pathogenic event in hereditary spherocytosis.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Erythrocyte Membrane/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Base Sequence , Binding Sites , Cytoskeleton/metabolism , DNA Primers/genetics , Erythrocyte Membrane/ultrastructure , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Female , Gene Targeting , Hemolysis/genetics , Hemolysis/physiology , In Vitro Techniques , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Pregnancy , Spectrin/biosynthesis , Spherocytosis, Hereditary/blood , Spherocytosis, Hereditary/genetics
13.
Blood ; 87(6): 2538-45, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8630421

ABSTRACT

Red cell membrane protein analysis by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and direct quantitation by radioimmunoassay or cytofluorometry defines four distinct subsets of patients with hereditary spherocytosis: Patients with isolated spectrin deficiency, combined spectrin and ankyrin deficiency, band 3 deficiency, and protein 4.2 deficiency. In regard to the first group, only one mutation of beta spectrin has been reported in the literature. We describe a spectrin variant characterized by a truncated beta chain, and associated with hereditary spherocytosis and isolated spectrin deficiency. The clinical phenotype consists of a moderate hemolytic anemia with spherocytosis and frequent spiculation of the red cells. We present the biochemical characteristics of this mutant protein and show that it constitutes only 12% of the total spectrin on the membrane. We show that the truncation of the protein is the result of a single point mutation at position +1 (G-->A) of the donor consensus splice site of intron 17 leading to an aberrant beta spectrin transcriptional message lacking exons 16 and 17. To elucidate the basis for the decreased amount of the truncated protein on the membrane and the overall spectrin deficiency, we provide evidence that the mutated gene is transcribed but its mRNA is less abundant than its normal counterpart in reticulocytes; we also show that the mutant protein is synthesized in decreased amounts in the cytoplasm of erythroid progenitor cells, and appears to be susceptible to proteolytic degradation. This mutant spectrin underscores the importance of the regulatory role played by the beta spectrin molecule in the assembly of alphabeta spectrin heterodimers on the membrane.


Subject(s)
Point Mutation , Spectrin/deficiency , Spherocytosis, Hereditary/genetics , Base Sequence , Child, Preschool , Cytoplasm/metabolism , DNA Mutational Analysis , Electrophoresis, Polyacrylamide Gel , Endopeptidases , Erythrocyte Membrane/chemistry , Erythroid Precursor Cells/metabolism , Exons/genetics , Humans , Introns/genetics , Male , Molecular Sequence Data , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reticulocytes/metabolism , Spectrin/genetics
14.
J Clin Invest ; 96(6): 2623-9, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675627

ABSTRACT

We describe a spectrin variant characterized by a truncated beta chain and associated with hereditary spherocytosis. The clinical phenotype consists of a moderate hemolytic anemia with striking spherocytosis and mild spiculation of the red cells. We describe the biochemical characteristics of this truncated protein which constitutes only 10% of the total beta spectrin present on the membrane, resulting in spectrin deficiency. Analysis of reticulocyte cDNA revealed the deletion of exons 22 and 23. We show, using Southern blot analysis, that this truncation results from a 4.6-kb genomic deletion. To elucidate the basis for the decreased amount of the truncated protein on the membrane and the overall spectrin deficiency, we show that (a) the mutated gene is efficiently transcribed and its mRNA abundant in reticulocytes, (b) the mutant protein is normally synthesized in erythroid progenitor cells, (c) the stability of the mutant protein in the cytoplasm of erythroblasts parallels that of the normal beta spectrin, and (d) the abnormal protein is inefficiently incorporated into the membrane of erythroblasts. We conclude that the truncation within the beta spectrin leads to inefficient incorporation of the mutant protein into the skeleton despite its normal synthesis and stability. We postulate that this misincorporation results from conformational changes of the beta spectrin subunit affecting the binding of the abnormal heterodimer to ankyrin, and we provide evidence based on binding assays of recombinant synthetic peptides to inside-out-vesicles to support this model.


Subject(s)
Ankyrins/metabolism , Genetic Variation , Spectrin/deficiency , Spectrin/genetics , Spherocytosis, Hereditary/genetics , Base Sequence , Binding Sites , Blotting, Southern , Child, Preschool , Cloning, Molecular , Cytoplasm/metabolism , DNA Primers , Erythroblasts/metabolism , Erythrocyte Membrane/metabolism , Female , Humans , Macromolecular Substances , Male , Membrane Proteins/biosynthesis , Membrane Proteins/blood , Membrane Proteins/isolation & purification , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Polymerase Chain Reaction , Protein Conformation , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Reticulocytes/metabolism , Spectrin/chemistry , Spherocytosis, Hereditary/blood
15.
Br J Haematol ; 91(2): 502-10, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8547102

ABSTRACT

Spherocytic elliptocytosis is a phenotypic hybrid between hereditary spherocytosis (HS) and hereditary elliptocytosis (HE) characterized by the presence of spheroovalocytes and spherocytes which exhibit increased osmotic fragility, indicating a deficiency of surface area. Both the spherocytic red cell morphology and the increased osmotic fragility distinguish this clinical entity from common HE. In contrast to common HE, the molecular basis of spherocytic elliptocytosis is unknown. Here we describe two members of a family who both have the characteristic features of spherocytic HE. We show that the underlying defect involves a G to C transversion at the -1 position of the acceptor splice site upstream of exon X of beta spectrin leading to skipping of exon X from the mutant beta spectrin mRNA allele. The mutant mRNA is present in reticulocytes in similar amounts as the normal mRNA. Pulse-labelling of erythroblasts prepared from peripheral blood in a two-phase liquid-culture system reveals a decreased synthesis of the truncated beta spectrin, a finding which is likely to underlie the moderately severe spectrin deficiency in the two patients. In addition, this mutant spectrin, similar to the previously reported spectrins, is defective in spectrin heterodimer self-association. The spectrin deficiency, which represents a common finding in the majority of patients with HS, together with weakened spectrin heterodimer self-association, as found in the majority of patients with common HE, provides a molecular explanation for the phenotype of spherocytic elliptocytosis in this kindred and, most likely, in other patients carrying similar beta spectrin mutations.


Subject(s)
Elliptocytosis, Hereditary/genetics , Spectrin/deficiency , Spherocytosis, Hereditary/genetics , Base Sequence , DNA/analysis , Erythrocyte Membrane , Female , Humans , Molecular Sequence Data , Mutation , Spectrin/genetics
16.
Blood ; 84(10): 3494-504, 1994 Nov 15.
Article in English | MEDLINE | ID: mdl-7949103

ABSTRACT

Although the association of erythroblasts with macrophages has been well documented in the human bone marrow, the function and identification of the intimate contacts occurring between the membranes of these two cell types in the physiology of erythropoiesis is not known. Using in vitro cultures of human peripheral blood derived erythroid progenitors, we have shown the presence of erythroblastic islands consisting of a central macrophage surrounded by a ring of erythroblasts that undergo terminal maturation leading to enucleation. However, when cultures were carried in the absence of intact macrophages, erythroid cells matured to the late erythroblast stage but failed to enucleate. Furthermore, the number of erythroid cells was markedly reduced in macrophage-depleted cultures, suggesting that the erythroblast-macrophage contact promotes proliferation and terminal maturation of erythroid cells leading to their enucleation. To examine the molecule(s) involved in the interaction between erythroblasts and macrophages, we have used a cell attachment assay involving incubation of solubilized surface-labeled erythroblasts with macrophage membrane proteins separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to a nitrocellulose membrane. Erythroblast surface proteins specifically attached to a 30-kD protein from macrophage membranes, whereas no adhesion was seen to the protein standards. An apparently similar protein of 30 kD was also detected on erythroblasts and was shown to mediate erythroblast-erythroblast contact in addition to the erythroblast-macrophage contact. The extraction of plasma membranes with Triton X-100 showed that the 30-kD protein is linked to the membrane skeleton via an integral membrane protein both in erythroblasts and macrophages. Furthermore, our results show that the cell:cell interactions mediated by the 30-kD protein are calcium-independent and could be specifically inhibited by heparin. We conclude that the association of erythroblasts with macrophages promotes erythroid proliferation and maturation leading to erythroblast enucleation and that a 30-kD heparin-binding protein present on the surface of macrophages and erythroblasts is involved in this contact. This protein is capable of binding homotypic and heterotypic cells.


Subject(s)
Carrier Proteins/metabolism , Cell Adhesion/physiology , Erythroblasts/physiology , Heparin/pharmacology , High Mobility Group Proteins/metabolism , Macrophages/physiology , Carrier Proteins/isolation & purification , Cell Adhesion/drug effects , Cell Communication , Cell Division , Cells, Cultured , Culture Media, Conditioned , Electrophoresis, Polyacrylamide Gel , Erythroblasts/cytology , Erythroblasts/drug effects , Fibronectins/metabolism , HMGB1 Protein , HeLa Cells , High Mobility Group Proteins/isolation & purification , Humans , Macrophages/cytology , Macrophages/drug effects , Molecular Weight
17.
Blood ; 82(5): 1652-60, 1993 Sep 01.
Article in English | MEDLINE | ID: mdl-8364214

ABSTRACT

Hereditary pyropoikilocytosis (HPP) is a recessively inherited hemolytic anemia characterized by severe poikilocytosis and red blood cell fragmentation. HPP red blood cells are partially deficient in spectrin and contain a mutant alpha or beta-spectrin that is defective in terms of spectrin self-association. Although the nature of the latter defect has been studied in considerable detail and many mutations of alpha-spectrin and beta spectrin have been identified, the molecular basis of spectrin deficiency is unknown. Here we report two mechanisms underlying spectrin deficiency in HPP. The first mechanism involves a thalassemia-like defect characterized by a reduced synthesis of alpha-spectrin as shown by studies involving synthesis of spectrin in two unrelated HPP probands and their parents: One parent carries the elliptocytogenic spectrin mutation, whereas the other parent is fully asymptomatic. Peripheral blood mononuclear cells as a source of erythroid burst-forming unit (BFUe) were cultured in a two-phase liquid culture system that gives rise to terminally differentiated erythroblasts. Pulse-labeling studies of an equal number of erythroblasts or morphologically identical maturity showed that the synthesis of alpha-spectrin as well as the mRNA levels as measured by the competitive polymerase chain reaction (PCR) method are markedly reduced in the presumed asymptomatic carriers and the HPP probands. In contrast, the synthesis and mRNA levels of beta-spectrin were normal. These results constitute a direct demonstration of an alpha-spectrin synthetic defect in a subset of asymptomatic carriers of HPP and HPP probands. The second mechanism underlying spectrin deficiency involves increased degradation of mutant spectrin before its assembly on the membrane. This is evidenced by pulse labeling studies of erythroblasts from a patient with HPP associated with a homozygous state for spectrin alpha I/46 mutation (leu-pro mutation at AA 207 of alpha-spectrin). These studies showed that although spectrin is synthesized in the cytosol in normal amounts, the rate of turnover of alpha-spectrin is faster resulting in about 40% to 50% reduced assembly of alpha-spectrin and beta-spectrin on the membrane. Thus, spectrin deficiency in this case is at least in part caused by increased susceptibility of the mutant spectrin to degradation before its assembly on the membrane. We conclude that at least two separate mechanisms underlie the molecular basis of spectrin deficiency in HPP.


Subject(s)
Anemia, Hemolytic, Congenital/blood , Spectrin/deficiency , Spectrin/genetics , Adolescent , Adult , Anemia, Hemolytic, Congenital/genetics , Base Sequence , Cells, Cultured , Erythroblasts/metabolism , Erythrocytes/metabolism , Erythrocytes, Abnormal/metabolism , Female , Humans , Membrane Proteins/analysis , Molecular Sequence Data , Mutation , RNA, Messenger/analysis , Spectrin/biosynthesis
18.
Stem Cells ; 11 Suppl 1: 8-12, 1993 May.
Article in English | MEDLINE | ID: mdl-8318923

ABSTRACT

To study the biogenesis of red cell membrane skeleton at various stages of erythroid differentiation, we have chosen the following model systems: a) Rauscher erythroleukemia cell line representing the early stages of differentiation, b) Friend erythroleukemia cells, and c) in vitro cultured human erythroblasts. The latter two systems represent terminally differentiated erythroblasts. Using these model systems, we have shown asynchronous synthesis of membrane proteins during erythroid differentiation. At the early stages of erythroid development, the synthesis of spectrin, ankyrin and band 4.1 proteins is initiated before that of the band 3 protein. Following erythroid induction with erythropoietin and dimethylsulfoxide (DMSO), there is a dramatic increase in the synthesis of the band 3 protein without noticeable changes in the synthesis of other membrane proteins. This increase in band 3 synthesis is accompanied by increased stability and recruitment of the skeletal proteins into the membrane skeleton, leading to increased steady state levels. The progressive increase in band 3 synthesis continues during terminal maturation of erythroblasts. This is accompanied by increased stability and assembly of spectrin and ankyrin on the membrane, despite their reduced synthesis. These results point to a key role for the band 3 protein in anchoring and stabilizing these proteins into the permanent skeletal network. Finally, to detect defects of skeletal biosynthesis, we have extended these studies to a patient with severe hereditary spherocytosis characterized by a combined deficiency of spectrin and ankyrin. We have shown that this combined deficiency is a consequence of reduced ankyrin synthesis and mRNA content representing a thalassemia-like membrane protein mutation.


Subject(s)
Erythrocyte Membrane/metabolism , Animals , Ankyrins/biosynthesis , Cell Differentiation , Cells, Cultured , Erythrocyte Membrane/ultrastructure , Humans , Leukemia, Erythroblastic, Acute , Mice , Microscopy, Electron , Spectrin/biosynthesis , Tumor Cells, Cultured
20.
Eur J Cell Biol ; 58(2): 313-8, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1425768

ABSTRACT

While the temporal sequences of the synthesis and assembly of membrane skeletal proteins has been studied during erythroid maturation, relatively little is known about the events which initiate the assembly of membrane skeleton at the early stages of mammalian erythroid commitment. To investigate the early events that initiate the assembly of the membrane skeleton in mammalian erythroid cells, we have studied the synthesis and assembly of membrane skeletal proteins in murine Rauscher erythroleukemia virus-transformed cells. These cells are blocked in differentiation at around the early progenitor (burst forming unit-erythroid, BFUe) cell stage but can be induced to differentiate in vitro. Pulse-labeling studies reveal that Rauscher cells actively synthesize alpha spectrin, beta spectrin, ankyrin and band 4.1 proteins. However, the synthesis of the band 3 protein and its mRNA are barely detectable in these cells. The peripheral membrane skeletal components assemble only transiently in the membrane skeleton and turn over rapidly, resulting in about 20-fold lower steady state levels than are found in mature erythrocytes. Upon induction with erythropoietin and dimethyl sulfoxide, the mRNA level and synthesis of band 3 are increased about 50-fold. In contrast, the synthesis of spectrin, ankyrin and band 4.1 is increased only about 1.5 to 2.0-fold. However, after induction, the fraction of these proteins assembled on the membrane is increased, their half-lives on the membrane are nearly doubled with a concomitant 4 to 5-fold increase in their steady-state levels. These results suggest that the synthesis of peripheral membrane proteins is detected at the earliest stages of erythroid commitment and increases only slightly during further differentiation.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anion Exchange Protein 1, Erythrocyte/biosynthesis , Cytoskeletal Proteins , Erythrocyte Membrane/metabolism , Neuropeptides , Animals , Ankyrins/biosynthesis , Cytoskeleton/metabolism , Dimethyl Sulfoxide/pharmacology , Erythropoiesis , Membrane Proteins/biosynthesis , Mice , RNA, Messenger/analysis , Spectrin/biosynthesis , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...