Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes Immun ; 11(2): 113-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19865104

RESUMO

Resistance to blood-stage malaria in AcB55 and AcB61 is caused by a loss of function mutation in pyruvate kinase (Pklr(I90N)). Likewise, pyruvate kinase (PK) deficiency in humans is protective against Plasmodium replication in vitro. We identified a third AcB strain, AcB62 that also carries the Pklr(I90N) mutation. However, AcB62 mice were susceptible to P.chabaudi infection and showed high levels of parasite replication (54-62% peak parasitemia). AcB62 mice showed the hallmarks of PK deficiency-associated anemia similar to AcB55/61 with reticulocytosis, splenic red pulp expansion, tissue iron overload, and increased expression of iron metabolism proteins. This suggests that malaria susceptibility in AcB62 is not because of absence of PK deficiency-associated pathophysiology. To map novel genetic factors affecting malaria susceptibility in AcB62, we generated an informative F2 population using AcB62 (Pklr(I90N)) and CBA-Pk(slc) (Pklr(G338D)) as progenitors and identified a novel locus on chromosome 9 (Char10; LOD=7.24) that controls peak parasitemia. A weaker linkage to the Pklr region of chromosome 3 (LOD=3.7) was also detected, a finding that may reflect the segregation of the two defective Pklr alleles. AcB62 alleles at both loci are associated with higher peak parasitemia. These results identify Char10 as a novel locus modulating severity of malaria in the context of PK deficiency.


Assuntos
Cromossomos Humanos Par 9/genética , Suscetibilidade a Doenças/imunologia , Malária/genética , Piruvato Quinase/deficiência , Alelos , Anemia Hemolítica Congênita/genética , Anemia Hemolítica Congênita/imunologia , Animais , Estruturas Cromossômicas/genética , Humanos , Malária/imunologia , Camundongos , Camundongos Endogâmicos CBA , Mutação , Parasitemia/genética , Piruvato Quinase/genética
2.
Transfus Clin Biol ; 12(2): 123-30, 2005 Jun.
Artigo em Francês | MEDLINE | ID: mdl-15927501

RESUMO

Most of the iron required for erythropoiesis is provided by heme iron recycling following degradation of senescent erythrocytes by tissue macrophages. Accumulation of biochemical modifications at the red blood cell membrane during ageing (externalisation of phosphatidyl-serine, peroxydation of membrane-bound lipoproteins, loss of sialic acid residues and formation of senescence neoantigens) constitute a series of signals that will allow the macrophage to identify the red blood cells to be eliminated, through interaction with specific receptors. After this initial recognition step, the red blood cell is internalised by phagocytosis, and phagosome maturation, which can comprise recruitment of the endoplasmic reticulum, will favour degradation of red blood cell constituents. Heme is catabolised by heme oxygenase 1, anchored in the endoplasmic reticulum membrane. A fraction of the released iron will be recycled back to the plasma through ferroportin, a membrane-bound Fe (II) export molecule, and a fraction will retained within the ferritin molecules, to be released at later stages. Multiple evidence coming from human diseases (type 4 hemochromatosis) and animal models indicate that ferroportin is essential for heme iron recycling by macrophages. Furthermore, ferroportin seems to be the molecular target of hepcidin, this circulating peptide synthesized by the liver and acting as a negative regulator of intestinal iron absorption and iron recycling by macrophages. Perturbations in erythrophagocytosis play a physiopathological role in several diseases, including hemochromatosis, anemia of chronic disorders and thalassemia.


Assuntos
Peptídeos Catiônicos Antimicrobianos/fisiologia , Eritrócitos/química , Heme/metabolismo , Ferro/sangue , Macrófagos/fisiologia , Fagocitose , Anemia/sangue , Proteínas de Transporte de Cátions/sangue , Envelhecimento Eritrocítico , Eritropoese , Ferritinas/sangue , Hemocromatose/sangue , Hepatócitos/metabolismo , Hepcidinas , Humanos , Inflamação/sangue , Absorção Intestinal , Ferro/farmacocinética , Modelos Biológicos , Talassemia/sangue
3.
J Leukoc Biol ; 71(5): 890-904, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11994515

RESUMO

The natural resistance-associated macrophage protein 1 (Nramp1) is a proton-dependent transporter of divalent metals. We studied NRAMP1 expression during HL-60 differentiation induced by VD and VD agonists. NRAMP1 and CD14 gene expression differed in kinetics of induction, mRNA levels and stability, and response to VD combined with PMA, whereas a combination of VD and IFN-gamma induced similar up-regulation. NRAMP1 protein expression paralleled the accumulation of mRNA and was localized in the phagosomal membrane after phagocytosis. A promoter construct extending 647 bp upstream of NRAMP1 ATG showed myeloid-specific transcription in transient transfection assays, which was up-regulated by VD in HL-60. In HL-60 clones stably transfected with this construct, transcription was apparently induced through indirect VD genomic effects, and there was accordance between the levels of reporter transcription and endogenous NRAMP1 mRNA in response to VD but not to IFN-gamma. Thus, VD genomic effects stimulate NRAMP1 transcription and protein expression in maturing phagocytes.


Assuntos
Calcitriol/farmacologia , Proteínas de Transporte de Cátions/genética , Fagócitos/metabolismo , Ativação Transcricional , Região 5'-Flanqueadora , Calcitriol/agonistas , Proteínas de Transporte de Cátions/biossíntese , Diferenciação Celular , Sinergismo Farmacológico , Células HL-60 , Humanos , Interferon gama/farmacologia , Cinética , Células Mieloides/metabolismo , Fagócitos/citologia , Fagócitos/efeitos dos fármacos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Receptores de Calcitriol/fisiologia , Elementos de Resposta , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas
4.
Blood ; 98(13): 3823-30, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11739192

RESUMO

Divalent metal transporter 1 (DMT1) is the major transferrin-independent iron uptake system at the apical pole of intestinal cells, but it may also transport iron across the membrane of acidified endosomes in peripheral tissues. Iron transport and expression of the 2 isoforms of DMT1 was studied in erythroid cells that consume large quantities of iron for biosynthesis of hemoglobin. In mk/mk mice that express a loss-of-function mutant variant of DMT1, reticulocytes have a decreased cellular iron uptake and iron incorporation into heme. Interestingly, iron release from transferrin inside the endosome is normal in mk/mk reticulocytes, suggesting a subsequent defect in Fe(++) transport across the endosomal membrane. Studies by immunoblotting using membrane fractions from peripheral blood or spleen from normal mice where reticulocytosis was induced by erythropoietin (EPO) or phenylhydrazine (PHZ) treatment suggest that DMT1 is coexpressed with transferrin receptor (TfR) in erythroid cells. Coexpression of DMT1 and TfR in reticulocytes was also detected by double immunofluorescence and confocal microscopy. Experiments with isoform-specific anti-DMT1 antiserum strongly suggest that it is the non-iron-response element containing isoform II of DMT1 that is predominantly expressed by the erythroid cells. As opposed to wild-type reticulocytes, mk/mk reticulocytes express little if any DMT1, despite robust expression of TfR, suggesting a possible effect of the mutation on stability and targeting of DMT1 isoform II in these cells. Together, these results provide further evidence that DMT1 plays a central role in iron acquisition via the transferrin cycle in erythroid cells.


Assuntos
Anemia/sangue , Proteínas de Transporte de Cátions/sangue , Eritrócitos/química , Proteínas de Ligação ao Ferro , Animais , Transporte Biológico/genética , Células CHO , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/fisiologia , Cricetinae , Endossomos/química , Endossomos/metabolismo , Membrana Eritrocítica/metabolismo , Eritrócitos/ultraestrutura , Células Precursoras Eritroides/química , Células Precursoras Eritroides/ultraestrutura , Eritropoetina/farmacologia , Imunofluorescência , Heme/metabolismo , Hemoglobinas/biossíntese , Homozigoto , Ferro/sangue , Leucemia Eritroblástica Aguda , Camundongos , Camundongos Mutantes , Microscopia Confocal , Mutação , Fenil-Hidrazinas/farmacologia , Isoformas de Proteínas/genética , Contagem de Reticulócitos , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo , Transferrina/metabolismo , Células Tumorais Cultivadas
5.
Genomics ; 72(3): 331-7, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11401449

RESUMO

Loop-tail (Lp) is a semidominant mutation that affects neurulation in mice. Heterozygous animals are characterized by a looped-tail appearance (pig tail) and wobbly head movements while homozygous embryos exhibit a neural tube closure defect that extends from the caudal midbrain to the tip of the tail. The Lp gene has been finely mapped to the distal part of chromosome 1, and a positional cloning strategy has been initiated to isolate the defective gene. This study represents the characterization of a new Lp allele (Lp(m1Jus)) induced by N-ethyl-N-nitrosurea mutagenesis. Lp(m1Jus)/+ mice have a looped-tail appearance, and both Lp(m1Jus)/Lp(m1Jus) homozygotes and Lp/Lp(m1Jus) compound heterozygotes fail to initiate neural tube closure along most of the embryonic axis. These data indicate that the Lp(m1Jus) allele causes a neural tube defect and overall phenotype similar to that of the original Lp allele. Segregation analysis of 90 (Lp(m1Jus)/+ x C57BL/6J)F(1) x C57BL/6J looped-tail mice with seven markers that define the Lp genetic map (D1Mit455/D1Mit146/D1Mit148/D1Mit270-1 cM-D1Mit113-0.4 cM-Lp-0.2 cM-D1Mit149-0.8 cM-D1Mit115) showed significant linkage between Lp(m1Jus) and all loci analyzed (P < 0.0001). Eight crossovers were detected with the proximal cluster of D1Mit455, D1Mit146, D1Mit148, and D1Mit270, indicating a recombination rate higher than expected in this region, and a single recombinant was encountered with the distal markers D1Mit149 and D1Mit115. Based on these phenotypic and genetic data, Lp(m1Jus) is most likely allelic to Lp, thereby representing a valuable additional tool for the positional cloning of the Lp gene and its subsequent molecular characterization.


Assuntos
Alelos , Defeitos do Tubo Neural/genética , Alquilantes/toxicidade , Animais , Mapeamento Cromossômico , Cruzamentos Genéticos , Etilnitrosoureia/toxicidade , Feminino , Teste de Complementação Genética , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/embriologia , Fenótipo , Gravidez
6.
Blood ; 97(4): 1138-40, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11159549

RESUMO

Iron overload is highly prevalent, but its molecular pathogenesis is poorly understood. Recently, DMT1 was shown to be a major apical iron transporter in absorptive cells of the duodenum. In vivo, it is the only transporter known to be important for the uptake of dietary non-heme iron from the gut lumen. The expression and subcellular localization of DMT1 protein in 3 mouse models of iron overload were examined: hypotransferrinemic (Trf(hpx)) mice, Hfe knockout mice, and B2m knockout mice. Interestingly, in Trf(hpx) homozygotes, DMT1 expression was strongly induced in the villus brush border when compared to control animals. This suggests that DMT1 expression is increased in response to iron deficiency in the erythron, even in the setting of systemic iron overload. In contrast, no increase was seen in DMT1 expression in animals with iron overload resembling human hemochromatosis. Therefore, it does not appear that changes in DMT1 levels are primarily responsible for iron loading in hemochromatosis.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas de Transporte de Cátions , Regulação da Expressão Gênica , Sobrecarga de Ferro/genética , Proteínas de Ligação ao Ferro , Proteínas de Membrana/biossíntese , Animais , Western Blotting , Células CHO , Proteínas de Transporte/genética , Cricetinae , Cricetulus , Modelos Animais de Doenças , Duodeno/metabolismo , Genótipo , Antígenos HLA/genética , Hemocromatose/genética , Hemocromatose/metabolismo , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Absorção Intestinal , Ferro/farmacocinética , Sobrecarga de Ferro/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transferrina/deficiência , Transferrina/genética , Microglobulina beta-2/deficiência , Microglobulina beta-2/genética
7.
Blood ; 96(12): 3964-70, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11090085

RESUMO

Microcytic anemia (mk) mice and Belgrade (b) rats are severely iron deficient because of impaired intestinal iron absorption and defective iron metabolism in peripheral tissues. Both animals carry a glycine to arginine substitution at position 185 in the iron transporter known as Nramp2/DMT1 (divalent metal transporter 1). DMT1 messenger RNA (mRNA) and protein expression has been examined in the gastrointestinal tract of mk mice. Northern blot analysis indicates that, by comparison to mk/+ heterozygotes, mk/mk homozygotes show a dramatic increase in the level of DMT1 mRNA in the duodenum. This increase in RNA expression is paralleled by a concomitant increase of the 100-kd DMT1 isoform I protein expression in the duodenum. Immunohistochemical analyses show that, as for normal mice on a low-iron diet, DMT1 expression in enterocytes of mk/mk mice is restricted to the duodenum. However, and in contrast to normal enterocytes, little if any expression of DMT1 is seen at the apical membrane in mk/mk mice. These results suggest that the G185R mutation, which was shown to impair the transport properties of DMT1, also affects the membrane targeting of the protein in mk/mk enterocytes. This loss of function of DMT1 is paralleled by a dramatic increase in expression of the defective protein in mk/mk mice. This is consistent with a feedback regulation of DMT1 expression by iron stores. (Blood. 2000;96:3964-3970)


Assuntos
Anemia/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions , Duodeno/química , Proteínas de Ligação ao Ferro , Proteínas de Membrana/genética , Alelos , Anemia/etiologia , Animais , Transporte Biológico , Proteínas de Transporte/imunologia , Duodeno/citologia , Duodeno/ultraestrutura , Immunoblotting , Imuno-Histoquímica , Ferro da Dieta/farmacologia , Camundongos , Camundongos Mutantes , Microvilosidades , RNA Mensageiro/metabolismo , Distribuição Tecidual
8.
Proc Assoc Am Physicians ; 111(4): 283-9, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10417735

RESUMO

Susceptibility to infectious diseases is under genetic control in humans. Animal models provide an ideal tool to study the genetic component of susceptibility and to identify candidate genes that can then be tested for association or linkage studies in human populations from endemic areas of disease. The Nramp1 gene was isolated by positional cloning the host resistance locus Bcg/Ity/Lsh, and mutations at this locus impair the resistance of mice to infections with intracellular parasites, such as Salmonella, Leishmania, and Mycobacterium. Allelic variants at the human Nramp1 homologue have recently been found to be associated with susceptibility to tuberculosis and leprosy in humans. The Nramp1 protein is an integral membrane protein expressed exclusively in the lysosomal compartment of monocytes and macrophages. After phagocytosis, Nramp1 is targeted to the membrane of the microbe-containing phagosome, where it may modify the intraphagosomal milieu to affect microbial replication. Although the biochemical mechanism of action of Nramp1 at that site remains unknown, Nramp homologues have been identified in many other animal species and actually define a protein family conserved from bacteria to humans. Some of these homologues have been shown to be divalent cation transporters. Recently, a second member of the mammalian Nramp family, Nramp2, was discovered and shown to be mutated in animal models of iron deficiency. The Nramp2 protein was subsequently shown to be the major transferrin-independent iron uptake system of the intestine. Together, these results suggest that Nramp1 may control intracellular microbial replication by actively removing iron or other divalent cations from the phagosomal space.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Transporte de Cátions , Predisposição Genética para Doença/genética , Imunidade Inata/genética , Proteínas de Ligação ao Ferro , Ativação de Macrófagos/efeitos dos fármacos , Proteínas de Membrana/fisiologia , Animais , Proteínas de Transporte/genética , Cátions/metabolismo , Frequência do Gene , Humanos , Infecções/genética , Infecções/imunologia , Transporte de Íons/genética , Ferro/metabolismo , Lisossomos/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , Modelos Moleculares , Família Multigênica , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/imunologia , Fagocitose/genética , Conformação Proteica
9.
Blood ; 93(12): 4406-17, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10361139

RESUMO

Genetic studies in animal models of microcytic anemia and biochemical studies of transport have implicated the Nramp2 gene in iron transport. Nramp2 generates two alternatively spliced mRNAs that differ at their 3' untranslated region by the presence or absence of an iron-response element (IRE) and that encode two proteins with distinct carboxy termini. Antisera raised against Nramp2 fusion proteins containing either the carboxy or amino termini of Nramp2 and that can help distinguish between the two Nramp2 protein isoforms (IRE: isoform I; non-IRE: isoform II) were generated. These antibodies were used to identify the cellular and subcellular localization of Nramp2 in normal tissues and to study possible regulation by dietary iron deprivation. Immunoblotting experiments with membrane fractions from intact organs show that Nramp2 is expressed at low levels throughout the small intestine and to a higher extent in kidney. Dietary iron starvation results in a dramatic upregulation of the Nramp2 isoform I in the proximal portion of the duodenum only, whereas expression in the rest of the small intestine and in kidney remains largely unchanged in response to the lack of dietary iron. In proximal duodenum, immunostaining studies of tissue sections show that Nramp2 protein expression is abundant under iron deplete condition and limited to the villi and is absent in the crypts. In the villi, staining is limited to the columnar absorptive epithelium of the mucosa (enterocytes), with no expression in mucus-secreting goblet cells or in the lamina propria. Nramp2 expression is strongest in the apical two thirds of the villi and is very intense at the brush border of the apical pole of the enterocytes, whereas the basolateral membrane of these cells is negative for Nramp2. These results strongly suggest that Nramp2 is indeed responsible for transferrin-independent iron uptake in the duodenum. These findings are discussed in the context of overall mechanisms of iron acquisition by the body.


Assuntos
Proteínas de Transporte/análise , Proteínas de Transporte de Cátions , Intestinos/ultraestrutura , Ferro da Dieta/farmacologia , Proteínas de Ligação ao Ferro , Ferro/metabolismo , Proteínas de Membrana/análise , Microvilosidades/química , Processamento Alternativo , Animais , Especificidade de Anticorpos , Células CHO , Proteínas de Transporte/genética , Cricetinae , Duodeno/química , Duodeno/ultraestrutura , Soros Imunes , Immunoblotting , Imuno-Histoquímica , Intestinos/química , Intestinos/efeitos dos fármacos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/análise , Distribuição Tecidual , Transfecção
10.
Infect Immun ; 67(5): 2225-32, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10225878

RESUMO

Mutations at the Nramp1 locus in vivo cause susceptibility to infection by unrelated intracellular microbes. Nramp1 encodes an integral membrane protein abundantly expressed in the endosomal-lysosomal compartment of macrophages and is recruited to the phagosomal membrane following phagocytosis. The mechanism by which Nramp1 affects the biochemical properties of the phagosome to control microbial replication is unknown. To devise an in vitro assay for Nramp1 function, we introduced a wild-type Nramp1(G169) cDNA into RAW 264.7 macrophages (which bear a homozygous mutant Nramp1(D169) allele and thus are permissive to replication of specific intracellular parasites). Recombinant Nramp1 was expressed in a membranous compartment in RAW264.7 cells and was recruited to the membrane of Salmonella typhimurium and Yersinia enterocolitica containing phagosomes. Evaluation of the antibacterial activity of RAW264.7 transfectants showed that expression of the recombinant Nramp1 protein abrogated intracellular replication of S. typhimurium. Studies with a replication-defective S. typhimurium mutant suggest that this occurs through an enhanced bacteriostatic activity. The effect of Nramp1 expression was specific, since (i) it was not seen in RAW264.7 transfectants overexpressing the closely related Nramp2 protein, and (ii) control RAW264.7 cells, Nramp1, and Nramp2 transfectants could all efficiently kill a temperature-sensitive, replication-defective mutant of S. typhimurium. Finally, increased antibacterial activity of the Nramp1 RAW264.7 transfectants was linked to increased phagosomal acidification, a distinguishing feature of primary macrophages expressing a wild-type Nramp1 allele. Together, these results indicate that transfection of Nramp1 cDNAs in the RAW264.7 macrophage cell line can be used as a direct assay to study both Nramp1 function and mechanism of action as well as to identify structure-function relationships in this protein.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte de Cátions , Macrófagos/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Alelos , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , DNA Complementar/genética , Expressão Gênica , Genes myc , Concentração de Íons de Hidrogênio , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Mycobacterium bovis/imunologia , Mycobacterium bovis/patogenicidade , Fagossomos/imunologia , Fagossomos/metabolismo , Fagossomos/microbiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Transfecção , Yersinia enterocolitica/genética , Yersinia enterocolitica/imunologia , Yersinia enterocolitica/patogenicidade
11.
J Exp Med ; 189(5): 831-41, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10049947

RESUMO

The natural resistance associated macrophage protein (Nramp) gene family is composed of two members in mammals, Nramp1 and Nramp2. Nramp1 is expressed primarily in macrophages and mutations at this locus cause susceptibility to infectious diseases. Nramp2 has a much broader range of tissue expression and mutations at Nramp2 result in iron deficiency, indicating a role for Nramp2 in iron metabolism. To get further insight into the function and mechanism of action of Nramp proteins, we have generated isoform specific anti-Nramp1 and anti-Nramp2 antisera. Immunoblotting experiments indicate that Nramp2 is present in a number of cell types, including hemopoietic precursors, and is coexpressed with Nramp1 in primary macrophages and macrophage cell lines. Nramp2 is expressed as a 90-100-kD integral membrane protein extensively modified by glycosylation (>40% of molecular mass). Subcellular localization studies by immunofluorescence and confocal microscopy indicate distinct and nonoverlapping localization for Nramp1 and Nramp2. Nramp1 is expressed in the lysosomal compartment, whereas Nramp2 is not detectable in the lysosomes but is expressed primarily in recycling endosomes and also, to a lower extent, at the plasma membrane, colocalizing with transferrin. These findings suggest that Nramp2 plays a key role in the metabolism of transferrin-bound iron by transporting free Fe2+ across the endosomal membrane and into the cytoplasm.


Assuntos
Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte de Cátions , Endocitose , Endossomos/química , Proteínas de Ligação ao Ferro , Ferro/metabolismo , Proteínas de Membrana/isolamento & purificação , Transferrina/isolamento & purificação , Animais , Especificidade de Anticorpos , Transporte Biológico , Compartimento Celular , Linhagem Celular , Imunofluorescência , Macrófagos/citologia , Glicoproteínas de Membrana , Camundongos , Modelos Biológicos , Monócitos/citologia
12.
Nucleic Acids Res ; 25(6): 1177-84, 1997 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9092627

RESUMO

We have recently elucidated the nature and function of transcription factors present in Jurkat, glial and neuronal cells that interact with modulatory region B, the nuclear receptor responsive element, in the long terminal repeat of human immunodeficiency virus type 1 (HIV-1). Considering the key role that the combination of host cell proteins plays in HIV-1 gene transcription, it appears essential to characterize proteins interacting with the adjacent region A. In vitro experiments revealed that the 5'-TGATTGGC-3'motif of region A is the target for at least three distinct proteins, one belonging to the nuclear factor I family, while two others are related to the cAMP response element binding (CREB) protein family. One of these proteins, present in DNA-protein complex C2, is formed by distinct polypeptides of relative molecular mass 43 000 and 50 000. We have purified the 43 kDa protein, which is distinct from CREB-43, and have shown that renatured p43 is able to specifically interact with site A. Transient expression experiments with vectors containing wild-type or mutant motif A revealed that basal HIV-1 gene transcription in Jurkat cells is regulated by antagonistic effects of the site A binding proteins.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Repetição Terminal Longa de HIV , Proteínas Nucleares/metabolismo , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/metabolismo , Sequências Reguladoras de Ácido Nucleico , Sequência de Bases , Sítios de Ligação , Cloranfenicol O-Acetiltransferase/biossíntese , Metilação de DNA , Células HeLa , Humanos , Células Jurkat , Proteínas Recombinantes/biossíntese , Transfecção , Raios Ultravioleta
13.
J Virol ; 69(11): 6634-42, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474072

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection of the neuronal and astroglial cells of the central nervous system has been proposed to contribute to HIV-1-associated dementia. Recently it was shown that differences in the nucleotide sequence of the long terminal repeat (LTR) of different HIV-1 strains govern the tissue-specific pattern of viral expression. The LTR from central nervous system-derived HIV-1 strains JR-FL and JR-CSF directs expression in the neurons of transgenic mice, in contrast with the lymphotropic LAI strain. By in vitro footprinting, gel retardation, and methylation interference experiments, we have studied the interactions of host cell proteins from human neuronal, glial, HeLa, and Jurkat T cells with the LTRs from the neurotropic JR-FL and JR-CSF strains, compared with the LAI strain. Proteins belonging to the nuclear receptor family bind with different affinities to variant -352 to -324 sites. Gel supershift assays with Jun and Fos antibodies showed that the AP-1 transcription factor present in the various cell types was unable to recognize the -352 to -324 and -306 to -285 AP-1 putative binding sites. Interestingly, Jun and Fos components of AP-1 interact with the variant TGGCTCA sequence located in the -247 to -222 region of both neurotropic strains. These interactions were cell type specific, since they were detected only with extracts from glial and HeLa cells and not from neuronal or Jurkat cells. Cotransfection experiments further revealed that the -247 to -222 sequence is able to mediate AP-1-induced transcriptional activation in glial and not neuronal cells.


Assuntos
DNA Viral/metabolismo , Repetição Terminal Longa de HIV , HIV-1/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Astrocitoma , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Variação Genética , HIV-1/classificação , HIV-1/metabolismo , Células HeLa , Humanos , Metilação , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neuroblastoma , Neuroglia/virologia , Neurônios/virologia , Proteínas Nucleares/isolamento & purificação , Oligodesoxirribonucleotídeos , Especificidade por Substrato , Fator de Transcrição AP-1/isolamento & purificação , Células Tumorais Cultivadas
14.
Neurochem Int ; 25(6): 545-53, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7894331

RESUMO

We have examined the expression of tropomyosin (TM) messenger RNAs (mRNAs) and protein isoforms in primary cultures of rat astroblasts during morphological changes. Three messenger RNA bands of 2.5, 1.8 and 1.2 kilobase pairs (kb) were detected by Northern blot. Using an antibody cross-reacting with all tropomyosin isoforms, we found that rat cerebellar neonatal astroblasts expressed three tropomyosin protein isoforms termed TM-As1, TM-As2 and TM-As3 (As for Astroblast) with respective molecular masses of 38,000, 33,000 and 31,000. Treatment of cells with agents which promote or mimick the action of cyclic AMP, or with growth factors, is known to induce astroblast morphological alteration from flat, polygonal epitheloid cells into star-shaped, process-bearing cells. In the presence of dibutyryl cAMP (dBcAMP), forskolin or basic fibroblast growth factor (bFGF), these morphological changes were found to be associated with dramatic decreases of the three mRNA transcripts and also of the three protein isoforms. This decrease was reversed upon removal of the drugs. The pattern of the tropomyosin protein isoforms in cultured astroblasts showed that TM-Asl, the most immunoreactive isoform recovered in the cytoskeletal insoluble cell fraction, had a developmental profile similar to that of F-actin. Therefore this isoform, which belongs to the high-molecular-mass family of proteins known to interact strongly with F-actin, could specifically be involved in the regulation/control of F-actin stability and thus be associated with the plasticity of astroblasts.


Assuntos
Astrócitos/metabolismo , AMP Cíclico/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Tropomiosina/biossíntese , Animais , Células Cultivadas , Colforsina/farmacologia , Expressão Gênica/fisiologia , RNA Mensageiro/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...