Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Vavilovskii Zhurnal Genet Selektsii ; 25(2): 234-245, 2021 Mar.
Artículo en Ruso | MEDLINE | ID: mdl-35083398

RESUMEN

72 clinical strains of Klebsiella spp. isolated from samples obtained from humans in Novosibirsk, Russia, were analyzed. Species identification of strains was performed using 16S rRNA and rpoB gene sequences. It was revealed that Klebsiella pneumoniae strains were dominant in the population (57 strains), while the remaining 15 strains were K. grimontii, K. aerogenes, K. oxytoca and K. quasipneumoniae. By molecular serotyping using the wzi gene sequence, K. pneumoniae strains were assigned to twenty-one K-serotypes with a high proportion of virulent K1- and K2-serotypes. It was found that K. pneumoniae strains isolated from the hospitalized patients had a higher resistance to antibiotics compared to the other Klebsiella species. Real-time PCR revealed that the population contained genes of the blaSHV, blaTEM, blaCTX families and the blaOXA-48 gene, which are the genetic determinants of beta-lactam resistance. It has been shown that the presence of the blaCTX sequence correlated with the production of extended-spectrum beta-lactamases, and phenotypic resistance to carbapenems is due to the presence of the blaOXA-48 gene. At the same time, the carbapenemase genes vim, ndm, kpc, imp were not detected. Among the aminoglycoside resistance genes studied, the aph(6)-Id and aadA genes were found, but their presence did not always coincide with phenotypic resistance. Resistance to fluoroquinolones in the vast majority of strains was accompanied by the presence of the aac(6')-IB-cr, oqxA, oqxB, qnrB, and qnrS genes in various combinations, while the presence of the oqxA and/or oqxB genes alone did not correlate with resistance to fluoroquinolones. Thus, the detection of blaCTX and blaOXA-48 can be used to quickly predict the production of extended-spectrum beta-lactamases and to determine the resistance of Klebsiella to carbapenems. The detection of the aac(6')-Ib-cr and/or qnrB/qnrS genes can be used to quickly determine resistance to fluoroquinolones.

2.
J Med Genet ; 46(10): 711-5, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19584064

RESUMEN

AIM: To evaluate the nature and frequency of ATP-binding cassette subfamily B member 4 (ABCB4) gene variants in a series of French patients with intrahepatic cholestasis of pregnancy (ICP). METHODS: In this prospective study, the entire ABCB4 gene coding sequence was analysed by DNA sequencing in 50 unrelated women with ICP defined by pruritus and raised serum alanine aminotransferase activity or bile acid concentration, with recovery after delivery. Genomic variants detected in patients with ICP were sought in 107 control pregnant women. Patients with ICP and controls were of Caucasian origin. RESULTS: Eight genomic variants were observed. One nonsense mutation (p.Arg144Stop) and two missense mutations (p.Ser320Phe and p.Thr775Met) were revealed each in one heterozygous patient. A third missense mutation (p.Arg590Gln) was detected in three heterozygous patients and in two homozygous patients also homozygous for a particular haplotype of three single-nucleotide polymorphisms (c.175C>T, c.504T>C, c.711A>T). The chromosomal frequency of the p.Arg590Gln variant was significantly different between the ICP and control group (7.0% vs 0.5%; p = 0.0017; OR 16.03, 95% CI 1.94 to 132.16). An association was also found between allele T of the c.504T>C silent nucleotide polymorphism and ICP (68.0% vs 53.7%; p = 0.017; OR 1.83, 95% CI 1.08 to 3.11). The chromosomal frequency of the p.Arg652Gly variant did not differ between the ICP and control group (p = 0.40). CONCLUSIONS: This study shows that 16% of Caucasian patients with ICP bear ABCB4 gene mutations, and confirms the significant involvement of this gene in the pathogenesis of this complex disorder.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Colestasis Intrahepática/genética , Mutación , Polimorfismo de Nucleótido Simple , Complicaciones del Embarazo/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Adulto , Secuencia de Aminoácidos , Colestasis Intrahepática/complicaciones , Femenino , Frecuencia de los Genes , Humanos , Datos de Secuencia Molecular , Embarazo , Estudios Prospectivos , Alineación de Secuencia
3.
Mol Cell Biol ; 18(10): 5852-60, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9742102

RESUMEN

Two distinct genes encode the closely related signal transducer and activator of transcription proteins STAT5A and STAT5B. The molecular mechanisms of gene regulation by STAT5 and, particularly, the requirement for both STAT5 isoforms are still undetermined. Only a few STAT5 target genes, among them the CIS (cytokine-inducible SH2-containing protein) gene, have been identified. We cloned the human CIS gene and studied the human CIS gene promoter. This promoter contains four STAT binding elements organized in two pairs. By electrophoretic mobility shift assay studies using nuclear extracts of UT7 cells stimulated with erythropoietin, we showed that these four sequences bound to STAT5-containing complexes that exhibited different patterns and affinities: the three upstream STAT binding sequences bound to two distinct STAT5-containing complexes (C0 and C1) and the downstream STAT box bound only to the slower-migrating C1 band. Using nuclear extracts from COS-7 cells transfected with expression vectors for the prolactin receptor, STAT5A, and/or STAT5B, we showed that the C1 complex was composed of a STAT5 tetramer and was dependent on the presence of STAT5A. STAT5B lacked this property and bound with a stronger affinity than did STAT5A to the four STAT sequences as a homodimer (C0 complex). This distinct biochemical difference between STAT5A and STAT5B was confirmed with purified activated STAT5 recombinant proteins. Moreover, we showed that the presence on the same side of the DNA helix of a second STAT sequence increased STAT5 binding and that only half of the palindromic STAT binding sequence was sufficient for the formation of a STAT5 tetramer. Again, STAT5A was essential for this cooperative tetrameric association. This property distinguishes STAT5A from STAT5B and could be essential to explain the transcriptional regulation diversity of STAT5.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas de la Leche , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células COS , Núcleo Celular/metabolismo , Citocinas/metabolismo , ADN Complementario , Dimerización , Humanos , Ratones , Datos de Secuencia Molecular , Factor de Transcripción STAT5 , Proteínas Supresoras de la Señalización de Citocinas , Transcripción Genética , Activación Transcripcional , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
4.
Oncogene ; 15(16): 1995-9, 1997 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-9365246

RESUMEN

Abnormal production of erythropoietin (Epo) has been described in several human and murine erythroleukemia. The murine IW32 cell line is derived from an F-MuLV-induced erythroleukemia. An autocrine Epo production due to the rearrangement of one Epo allele has been previously described (Beru et al., 1989). However, the exact mechanism leading to the transcriptional activation of the abnormal Epo gene was unknown. In this study, we show that this deregulated expression results from a deletion within chromosome 5. The Epo gene in the abnormal allele is under the control of the G-protein beta2 subunit gene promoter and the expressed mRNA results from the fusion of the non coding exon 1 of the G-protein beta2 subunit gene to a truncated Epo exon 1 gene. This resulting abnormal cDNA allows the expression of a normal Epo protein.


Asunto(s)
Eritropoyetina/genética , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica/genética , Reordenamiento Génico , Leucemia Eritroblástica Aguda/genética , Animales , Secuencia de Bases , ADN Complementario , Humanos , Leucemia Eritroblástica Aguda/patología , Ratones , Datos de Secuencia Molecular , Células Tumorales Cultivadas
5.
Oncogene ; 20(18): 2197-204, 2001 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-11402314

RESUMEN

In this study, we show that upon thrombopoietin (Tpo) stimulation the two adapter proteins Gab1 and Gab2 are strongly tyrosine phosphorylated and associated with Shc, SHP2, PI 3-kinase and Grb2 in mpl-expressing UT7 cells. Although Gab1 and Gab2 seem to mediate overlapping biological signals in many cells, only Gab1 is expressed and phosphorylated in response to Tpo in primary human megakaryocytic progenitors; furthermore, it associates with the same proteins. Although a low level of tyrosine phosphorylated IRS-2 protein is also detected in PI 3-kinase immunoprecipitates, Gab proteins are the essential proteins associated with PI 3-kinase after Tpo stimulation. We demonstrate that, albeit no association is detected between the Tpo receptor mpl and Gab proteins, Y112 located in the C-terminal cytoplasmic domain of mpl is required for Gab1/2 tyrosine phosphorylation. Gab proteins are not tyrosine phosphorylated after Tpo stimulation of UT-7 and Ba/F3 cells expressing a mpl mutant lacking Y112. Moreover, no activation of the PI 3-kinase/Akt pathway is observed in cells expressing this mpl mutant. Finally, we show that this mutant does not allow cell proliferation, thereby confirming that PI 3-kinase activation is required for Tpo-induced cell proliferation.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas , Trombopoyetina/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , Activación Enzimática/efectos de los fármacos , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Conejos , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trombopoyetina/genética , Tirosina/metabolismo
6.
J Clin Pharmacol ; 21(2): 65-71, 1981.
Artículo en Inglés | MEDLINE | ID: mdl-7229119

RESUMEN

Lofexidine, a new centrally acting antihypertensive agent, was compared in a double-blind study with clonidine in the treatment of mild (standing diastolic blood pressure 95-104 mm Hg) or moderate (105-129 mm Hg) essential hypertension. In dialy dosages of 0.2 or 0.4 mg, monotherapy with lofexidine produced significant decreases in blood pressure and heart rate that were not different from those with clonidine. Blood pressure and heart rate were not different from those with clonidine. Blood pressure control (supine and standing diastolic pressure less than 90 mm Hg) occurred in seven of eight mild hypertensives treated with lofexidine and in seven of ten treated with clonidine. In moderate hypertension, three of 11 and seven of ten, ten of the 14 responders to clonidine required dosages of 0.4 mg daily or less. The maximum dosage tested was 1.6 mg daily. Concomitant hydrochlorothiazide therapy was given to eight of the lofexidine responders and 12 of the clonidine responders. For both drugs, drowsiness and dry mouth were the chief complaints. Neither agent changed standard clinical biochemistries except for decreased potassium and increased bicarbonate concentrations due to concurrent diuretic therapy. Lofexidine to have clinical characteristics similar to those of clonidine. Each of these agents is best used in lower doses, which are frequently effective and less likely to produce symptomatic complaints.


Asunto(s)
Antihipertensivos , Clonidina/análogos & derivados , Clonidina/uso terapéutico , Hipertensión/tratamiento farmacológico , Clonidina/efectos adversos , Método Doble Ciego , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad
7.
Eur J Biochem ; 233(3): 793-9, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8521844

RESUMEN

We have previously shown that the +79 to +135 fragment of the human erythropoietin receptor (Epo-R) acts negatively on the transcriptional activity and confers erythroid specificity to the gene [Maouche, L., Cartron, J.-P. & Chrétien, S. (1994) Nucleic Acids Res. 22, 338-346]. In this work, we demonstrate that this effect is mediated by a CCACC motif that binds weakly to the simian virus 40 protein 1 (Sp1) factor and that the increase of the affinity for Sp1 augments transcription inhibition. The repression is not restricted to the human Epo-R promoter, although it seems more efficient on heterologous promoters of erythroid genes. In chloramphenicol acetyl transferase constructs containing the mouse Epo-R promoter, rearranged by retroviral long terminal repeat (LTR) insertion of murine erythroleukemia cell lines, we found that positioning the CCACC motif 3' to the LTR represses the transcriptional activity mediated by the LTR in non-erythroid cells. These results demonstrate that Epo-R gene expression is negatively regulated by a CCACC or a GC box-binding factor, which is most likely identical to the Sp1 transcription protein. Further data suggest that Sp1-mediated negative regulation is not the result of a direct competition between Sp1 and another DNA-binding protein.


Asunto(s)
Receptores de Eritropoyetina/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Sitios de Unión , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Receptores de Eritropoyetina/genética , Proteínas Represoras/genética , Análisis de Secuencia
8.
Nucleic Acids Res ; 22(3): 338-46, 1994 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-8127671

RESUMEN

To analyse the 5'-flanking sequences required for the tissue specific transcription of the human erythropoietin receptor (hEpo-R) gene, a DNA region spanning nucleotides -1050 to +135 relative to the transcription initiation site (+1) was explored. Our studies indicate that a minimum promoter (-76/+33) containing GATA and SP1 binding sites at positions -45 and -20 is not sufficient to confer erythroid specific expression to a reporter gene. Erythroid specificity of the promoter was observed either with the (-1050/+33 construct) which contains a cluster of Alu repetitive elements or with the addition of the 135 bp down to the transcription initiation site (-76/+135 construct) which exert a negative control on the promoter activity with a major effect in non erythroid tissues. The latter region can be subdivided on two distinct domains: the +1/+78 region that exerts a positive effect and the +79/+135 region that has a negative effect on the Epo-R promoter activity measured by CAT assays. The first region contains three CANNTG motifs, whereas the second contains an SP1/CACCC motif at position +85. These findings reveal a complex regulation of the hEpo-R gene and provide a working model useful to explain how the minimal promoter, containing GATA/SP1, can be positively and negatively regulated during erythroid differentiation.


Asunto(s)
Regiones Promotoras Genéticas , Receptores de Eritropoyetina/genética , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Cartilla de ADN/química , Proteínas de Unión al ADN/metabolismo , Células Precursoras Eritroides/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Transcripción Genética
9.
J Biol Chem ; 266(34): 23380-5, 1991 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-1660472

RESUMEN

The structure of the murine erythropoietin receptor was studied using antibodies against the intracellular part of the cloned erythropoietin receptor chain. These antibodies precipitated erythropoietin-receptor complexes from Triton X-100-solubilized cells. When the complexes were cross-linked by disuccinimidyl suberate, the 85- and 100-kDa erythropoietin-cross-linked proteins previously described were immunoprecipitated. However, these proteins were not precipitated when the complexes were denatured and reduced before immunoprecipitation. Using 1-ethyl 3-(3-dimethylaminopropyl)carbodiimide, we observed erythropoietin cross-linking with a protein of 66 kDa in addition to the 100- and 85-kDa proteins. Only the 66-kDa erythropoietin-cross-linked protein was immunoprecipitated by anti-receptor antibodies after denaturation and reduction of the complex. Thus, our results suggest that the 85- and 100-kDa proteins previously evidenced by cross-linking are associated with the cloned chain of the receptor to form a multimeric complex but these proteins seem immunologically unrelated to the cloned chain. We observed that reducing the length of molecules able to cross-link amino groups decreased the efficiency of cross-linking with the 100-kDa protein and only the 85-kDa protein was cross-linked with erythropoietin using 1,5-difluoro-2,4-dinitrobenzene. These results suggest that the 85- and 100-kDa proteins occupate slightly different positions relative to the erythropoietin molecule bound to the receptor.


Asunto(s)
Eritropoyetina/metabolismo , Receptores de Superficie Celular/química , Animales , Anticuerpos , Células Cultivadas , Clonación Molecular , Reactivos de Enlaces Cruzados , Ratones , Pruebas de Precipitina , Desnaturalización Proteica , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Receptores de Eritropoyetina
10.
J Biol Chem ; 275(24): 18375-81, 2000 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-10849444

RESUMEN

The binding of erythropoietin (Epo) to its receptor leads to the transient phosphorylation of the Epo receptor (EpoR) and the activation of intracellular signaling pathways. Inactivation mechanisms are simultaneously turned on, and Epo-induced signaling pathways return to nearly basal levels after 30-60 min of stimulation. We show that proteasomes control these inactivation mechanisms. In cells treated with the proteasome inhibitors N-Ac-Leu-Leu-norleucinal (LLnL) or lactacystin, EpoR tyrosine phosphorylation and activation of intracellular signaling pathways (Jak2, STAT5, phosphatidylinositol 3-kinase) were sustained for at least 2 h. We show that this effect was due to the continuous replenishment of the cell surface pool of EpoRs in cells treated with proteasome inhibitors. Proteasome inhibitors did not modify the internalization and degradation of Epo.EpoR complexes, but they allowed the continuous replacement of the internalized receptors by newly synthesized receptors. Proteasome inhibitors did not modify the synthesis of EpoRs, but they allowed their transport to the cell surface. N-Ac-Leu-Leu-norleucinal, but not lactacystin, also inhibited the degradation of internalized Epo.EpoR complexes, most probably through cathepsin inhibition. The internalized EpoRs were not tyrosine-phosphorylated, and they did not activate intracellular signaling pathways. Our results show that the proteasome controls the down-regulation of EpoRs in Epo-stimulated cells by inhibiting the cell surface replacement of internalized EpoRs.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Regulación hacia Abajo , Complejos Multienzimáticos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Eritropoyetina/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Eritropoyetina/fisiología , Humanos , Fosforilación , Complejo de la Endopetidasa Proteasomal , Proteínas Recombinantes/metabolismo , Transducción de Señal , Tirosina/metabolismo
11.
Ann Intern Med ; 121(4): 252-8, 1994 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7741833

RESUMEN

OBJECTIVE: To evaluate the safety and efficacy of controlled-release niacin in patients with hyperlipoproteinemia. DESIGN: A retrospective cohort study. SETTING: A Department of Veterans Affairs Medical Center. PATIENTS: A consecutive sample of 969 predominantly elderly male veterans treated for dyslipoproteinemia with controlled-release niacin between October 1988 and October 1991. MAIN OUTCOME MEASURES: Primary outcomes were lipid levels and lipoprotein cholesterol response, alternations in levels of hepatic enzymes and blood chemistry test results, and characterization of niacin-induced hepatotoxicity abstracted from the patient's medical, laboratory, and pharmacy records. RESULTS: 93% (896 of 969) of the cohort was evaluable. Patients (age, 61.7 years [9.4 years], mean [SD]) were treated for 1 to 36 months (13.0 months [9.7 months]) with an average maintenance dose of 1.67 g/d (0.8 g/d). Niacin was discontinued in 48.5% (435 of 896) of the patients primarily because of adverse effects. Poor glycemic control led to discontinuation in 40.6% (43 of 106) of the patients with diabetes mellitus. The lipoprotein response was dose-related and favorable (levels of total cholesterol, -19.1%; low-density lipoprotein cholesterol, -24.0%; high-density lipoprotein cholesterol, +5.7%; and triglycerides, -32.5%). Statistically but not clinically meaningful dose-related increases were seen in levels of liver enzymes and serum glucose (aspartate aminotransferase, +29%; alanine aminotransferase, +23%; alkaline phosphatase, +25%; and glucose, +7%; P = 0.0001). Twenty of 896 (2.2%) and 42 of 896 (4.7%) patients met biochemical criteria for probable and for possible or probable niacin-induced hepatotoxicity, respectively. Predisposing factors included high dose, alcohol use, preexisting liver disease, and concurrent oral sulfonylurea therapy. CONCLUSIONS: Controlled-release niacin is effective in treating dyslipoproteinemia in selected middle-aged and elderly veterans, but approximately one half of patients discontinued the drug because of adverse effects or other causes including noncompliance. Niacin should be avoided in patients with hepatic dysfunction or a history of liver disease, patients with diabetes mellitus, and patients who abuse alcohol. Because controlled-release niacin seems to be more potent than crystalline niacin, product substitution without dose adjustment should be avoided.


Asunto(s)
Hiperlipoproteinemias/tratamiento farmacológico , Niacina/uso terapéutico , Anciano , Preparaciones de Acción Retardada , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hiperlipoproteinemias/sangre , Hiperlipoproteinemias/enzimología , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/enzimología , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Niacina/efectos adversos , Estudios Retrospectivos , Resultado del Tratamiento
12.
J Biol Chem ; 273(51): 33985-90, 1998 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-9852052

RESUMEN

Erythropoietin (Epo) regulates the proliferation and differentiation of erythroid cells through interaction with a cell surface receptor (EpoR) that belongs to the cytokine receptor family. The Jak2 tyrosine kinase was previously shown to bind to the EpoR, to be activated upon Epo stimulation, and to play a critical role in Epo-induced proliferation. However, little is known about the role of other tyrosine kinases in Epo signaling. In this paper, we examined whether Syk was involved in EpoR activation. Coimmunoprecipitation experiments showed that the phosphorylated EpoR was associated with the Syk kinase in activated UT7 cells. The interaction of Epo with its receptor led to an increased kinase activity. The use of recombinant Syk Src homology 2 (SH2) domains expressed in tandem or individually revealed that both N- and C-SH2 domains of Syk participated in EpoR binding with a major contribution of the C-terminal SH2 domain. Far Western blotting further indicated that Syk directly binds to the EpoR and that the interaction of Syk with EpoR only occurred after Epo activation. These data suggest that phosphorylation of EpoR on tyrosine residues may mediate Syk binding to the receptor through interaction between the two SH2 domains of Syk and tyrosines of the receptor. We propose that in addition to Jak2, Syk protein kinase may be a component of EpoR signaling.


Asunto(s)
Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Eritropoyetina/farmacología , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Eritropoyetina/metabolismo , Glutatión Transferasa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Reacción en Cadena de la Polimerasa , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes , Transducción de Señal , Quinasa Syk , Transfección , Dominios Homologos src
13.
J Biol Chem ; 272(42): 26173-8, 1997 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-9334184

RESUMEN

In this report, we demonstrate that insulin receptor substrate-2 (IRS-2) is phosphorylated on tyrosine following treatment of UT-7 cells with erythropoietin. We have investigated the expression of IRS-1 and IRS-2 in several cell lines with erythroid and/or megakaryocytic features, and we observed that IRS-2 was expressed in all cell lines tested. In contrast, we did not detect the expression of IRS-1 in these cells. In response to erythropoietin, IRS-2 was immediately phosphorylated on tyrosine, with maximal phosphorylation between 1 and 5 min. Tyrosine-phosphorylated IRS-2 was associated with phosphatidylinositol 3-kinase and with a 140-kDa protein that comigrated with the phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase, SHIP. Moreover, IRS-2 was constitutively associated with the erythropoietin receptor. We did not observe the association of IRS-2 with JAK2, Grb2, or PTP1D. Using BaF3 cells transfected with mutated erythropoietin receptors, we demonstrate that neither the tyrosine residues of the intracellular domain nor the last 109 amino acids of the erythropoietin receptor are required for erythropoietin-induced IRS-2 tyrosine phosphorylation. Altogether, our results indicate that erythropoietin-induced IRS-2 tyrosine phosphorylation could account for the previously reported activation of phosphatidylinositol 3-kinase mediated by erythropoietin receptors mutated in the phosphatidylinositol 3-kinase-binding site (Damen, J., Cutler, R. L., Jiao, H., Yi, T., and Krystal, G. (1995) J. Biol. Chem. 270, 23402-23406; Gobert, S., Porteu, F., Pallu, S., Muller, O., Sabbah, M., Dusanter-Fourt, I., Courtois, G., Lacombe, C., Gisselbrecht, S., and Mayeux, P. (1995) Blood 86, 598-606).


Asunto(s)
Eritropoyetina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Tirosina/metabolismo , Activación Enzimática , Humanos , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Fosforilación , Proteínas Recombinantes , Células Tumorales Cultivadas
14.
J Biol Chem ; 274(48): 33966-72, 1999 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-10567359

RESUMEN

We showed that erythropoietin induced rapid glycosylphosphatidylinositol (GPI) hydrolysis and tyrosine phosphorylation of phospholipase C (PLC)-gamma(2) in FDC-P1 cells transfected with the wild-type erythropoietin-receptor. Erythropoietin-induced tyrosine phosphorylation of PLC-gamma(2) was time- and dose-dependent. By using FDC-P1 cells transfected with an erythropoietin receptor devoid of tyrosine residues, we showed that both effects required the tyrosine residues of intracellular domain on the erythropoietin receptor. Erythropoietin-activated PLC-gamma(2) hydrolyzed purified [(3)H]GPI indicating that GPI hydrolysis and PLC-gamma(2) activation under erythropoietin stimulation were correlated. Results obtained on FDC-P1 cells transfected with erythropoietin receptor mutated on tyrosine residues suggest that tyrosines 343, 401, 464, and/or 479 are involved in erythropoietin-induced GPI hydrolysis and tyrosine phosphorylation of PLC-gamma(2), whereas tyrosines 429 and/or 431 seem to be involved in an inhibition of both effects. Thus, our results suggest that erythropoietin regulates GPI hydrolysis via tyrosine phosphorylation of its receptor and PLC-gamma(2) activation.


Asunto(s)
Eritropoyetina/farmacología , Glicosilfosfatidilinositoles/metabolismo , Animales , Línea Celular , Hidrólisis/efectos de los fármacos , Isoenzimas/metabolismo , Mutación , Fosfolipasa C gamma , Fosforilación/efectos de los fármacos , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Transfección , Fosfolipasas de Tipo C/metabolismo , Tirosina/metabolismo
15.
Drug Intell Clin Pharm ; 19(7-8): 575-80, 1985.
Artículo en Inglés | MEDLINE | ID: mdl-3928310

RESUMEN

A cost-benefit evaluation of a clinical pharmacist-managed anticoagulation clinic (AC) was performed. Outpatient and hospital records were examined for 26 patients in the treatment group with an AC clinic and 26 patients in the control group. Therapeutic prothrombin times were maintained within the treatment group to a significantly greater extent than within the control group (p less than 0.001). The AC was successful in preventing hospitalizations resulting from hemorrhage or thromboembolism (p less than 0.01). The abnormal prothrombin times on admission in the control group correlated with hemorrhagic and thromboembolic admissions (p less than 0.05, p less than 0.005, respectively). Patients were hospitalized 3.22 days and .048 days per patient-treatment-year in the control and treatment groups, respectively. The net savings in reduced hospitalization costs per year in the treatment group was $211776. The benefit:cost ratio (B:C) was 6.55, suggesting the program is socially valuable. This clinical pharmacist-managed AC was effective in maintaining therapeutic prothrombin times, and reducing the incidence of hospitalization resulting from anticoagulation complications, and can be cost-justified based on a cost-benefit analysis.


Asunto(s)
Análisis Costo-Beneficio , Servicio Ambulatorio en Hospital/economía , Servicio de Farmacia en Hospital/economía , Warfarina/uso terapéutico , California , Femenino , Hemorragia/prevención & control , Hospitales con más de 500 Camas , Humanos , Masculino , Persona de Mediana Edad , Farmacéuticos , Tiempo de Protrombina , Distribución Aleatoria , Recurrencia , Estudios Retrospectivos , Tromboembolia/prevención & control , Factores de Tiempo , Warfarina/efectos adversos
16.
Blood ; 78(10): 2557-63, 1991 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-1668607

RESUMEN

The genomic and complementary DNAs of the human erythropoietin receptor (hEpo-R) have been isolated and characterized from a genomic placental library and from two cDNA libraries prepared from bone marrow and fetal liver. The five different partial cDNAs isolated were aberrant in the predicted reading frames as compared with the Epo-R protein sequence, because all retained insert sequences that may represent splicing intermediates (three clones), cloning artifact (one clone), or a new sequence at a splice junction (one clone) of the gene. The cDNAs were used to isolate several genomic clones encompassing the complete hEpo-R gene. This gene, which encodes a 508-amino acid polypeptide chain of predicted M(r) 55,000, is organized into eight exons spread over 6 kb of DNA and exhibited a high degree of sequence homology (81.6% in the coding region) and structural organization with its murine counterpart. Primer extension analysis indicated that the transcription initiation site is located 141 bp upstream of the initiation codon. Sequence homology 320 bp upstream of the cap site was significantly lower (60%) and diverged completely further upstream as compared with the murine gene. Similarly, the human and murine sequences were largely divergent downstream of the stop codon, indicating that a strong conservation during evolution was restricted to the coding sequence of the Epo-R protein. The 320-bp region upstream of the cap site does not contain the typical TATA or CAAT boxes present in many tissue-specific genes, but does include potential binding sites for the ubiquitous Sp1 and the erythroid-specific GATA-1 trans-activating factors. These boxes are well conserved in sequence and position relative to the cap site within the promoter region of the human and murine genes, but the CACCC boxes present in the murine gene are absent in the human gene.


Asunto(s)
Eritroblastos/fisiología , Receptores de Superficie Celular/genética , Animales , Secuencia de Bases , Médula Ósea/fisiología , Clonación Molecular/métodos , ADN/genética , ADN/aislamiento & purificación , Eritropoyetina/metabolismo , Exones , Femenino , Feto , Biblioteca de Genes , Humanos , Intrones , Hígado/fisiología , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Placenta/fisiología , Reacción en Cadena de la Polimerasa , Embarazo , Regiones Promotoras Genéticas , Receptores de Eritropoyetina , Homología de Secuencia de Ácido Nucleico , Bazo/fisiología , TATA Box
17.
Eur J Biochem ; 162(1): 105-10, 1987 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-3816774

RESUMEN

Porphobilinogen deaminase (hydroxymethylbilane synthase; EC 4.3.1.8), the third enzyme of the heme biosynthetic pathway, catalyzes the stepwise condensation of four porphobilinogen units to yield hydroxymethylbilane, which is in turn converted to uroporphyrinogen III by cosynthetase. We compared the apparent molecular mass of porphobilinogen deaminase from erythropoietic and from non-erythropoietic cells by sodium dodecyl sulfate/polyacrylamide gel electrophoresis and immune-blotting. The results indicate that two isoforms of porphobilinogen deaminase can be distinguished and differ by 2000 Da. Analysis of cell-free translation products directed by mRNAs from human erythropoietic spleen and from human liver demonstrates that the two isoforms of porphobilinogen deaminase are encoded by distinct messenger RNAs. We cloned and sequenced cDNAs complementary to the non-erythropoietic form of porphobilinogen deaminase encoding RNA. Comparison of these sequences to that of human erythropoietic mRNA [Raich et al. (1986) Nucleic Acids Res. 14, 5955-5968] revealed that the two mRNA species differ by their 5' extremity. From the mRNA sequences we could deduce that an additional peptide of 17 amino acid residues at the NH2 terminus of the non-erythropoietic isoform of porphobilinogen deaminase accounts for its higher molecular mass. RNase mapping experiments demonstrate that the two porphobilinogen deaminase mRNAs are distributed according to a strict tissue-specificity, the erythropoietic form being restricted to erythropoietic cells. We propose that a single porphobilinogen deaminase gene is transcribed from two different promoters, yielding the two forms of porphobilinogen deaminase mRNAs. Our present finding may have some relevance for further understanding the porphobilinogen deaminase deficiency in certain cases of acute intermittent porphyria with an enzymatic defect restricted in non-erythropoietic cells.


Asunto(s)
Amoníaco-Liasas/genética , Hidroximetilbilano Sintasa/genética , Isoenzimas/genética , Hígado/enzimología , Bazo/enzimología , Secuencia de Bases , Sistema Libre de Células , ADN/análisis , Eritropoyesis , Regulación de la Expresión Génica , Humanos , Hidroximetilbilano Sintasa/análisis , Isoenzimas/análisis , Linfocitos/enzimología , Peso Molecular , Biosíntesis de Proteínas , Transcripción Genética
18.
J Biol Chem ; 276(3): 2007-14, 2001 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-11053408

RESUMEN

To identify new proteins involved in erythropoietin (Epo) signal transduction, we purified the entire set of proteins reactive with anti-phosphotyrosine antibodies from Epo-stimulated UT7 cells. Antisera generated against these proteins were used to screen a lambdaEXlox expression library. One of the isolated cDNAs encodes Gbeta2, the beta2 subunit of heterotrimeric GTP-binding proteins. Gbeta and Galpha(i) coprecipitated with the Epo receptor (EpoR) in extracts from human and murine cell lines and from normal human erythroid progenitor cells. In addition, in vitro Gbeta associated with a fusion protein containing the intracellular domain of the EpoR. Using EpoR mutants, we found that the distal part of the EpoR (between amino acids 459-479) was required for Gi binding. Epo activation of these cells induced the release of the Gi protein from the EpoR. Moreover in isolated cell membranes, Epo treatment inhibited ADP-ribosylation of Gi and increased the binding of GTP. Our results show that heterotrimeric Gi proteins associate with the C-terminal end of the EpoR. Receptor activation leads to the activation and dissociation of Gi from the receptor, suggesting a functional role of Gi protein in Epo signal transduction.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Receptores de Eritropoyetina/metabolismo , Animales , Citoplasma/metabolismo , Eritropoyetina/farmacología , Humanos , Ratones , Unión Proteica , Receptores de Eritropoyetina/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
19.
J Biol Chem ; 266(11): 6952-6, 1991 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-1849897

RESUMEN

We have characterized the structure of the erythropoietin receptor gene promoter in normal murine erythroid tissues and in Friend-induced tumor cells. Using primer extension analysis, we identified two distinct transcriptional start sites, which were located 2 base pairs apart in anemic spleens, fetal liver, Friend-induced tumoral spleens, and mouse erythro-leukemia cells. In contrast, transcription was initiated 37 base pairs upstream of the normal cap sites in T3Cl-2, a Friend virus-induced murine erythroleukemia cell line. Also, the erythropoietin receptor mRNA in T3Cl-2 was overexpressed when compared with other erythroleukemia cell lines. We found that abnormal transcription occurring in T3Cl-2 cells resulted from an erythropoietin receptor gene alteration. Indeed, one erythropoietin receptor allele was rearranged by insertion of a spleen focus-forming virus long terminal repeat within the noncoding region of the first exon, 45 bases upstream of the ATG initiation codon and in the same 5'----3' orientation. The transcription of the rearranged allele was shown to be directed from the long terminal repeat promoter, leading to a long terminal repeat-erythropoietin receptor fusion transcript, whereas the normal erythropoietin receptor allele was weakly transcribed. Such altered receptor gene activation may provide a positive pressure in the development of tumorigenic erythroleukemia.


Asunto(s)
Elementos Transponibles de ADN , Eritropoyetina/metabolismo , Regulación Viral de la Expresión Génica , Genes , Receptores de Superficie Celular/genética , Secuencias Repetitivas de Ácidos Nucleicos , Virus Formadores de Foco en el Bazo/genética , Animales , Secuencia de Bases , Línea Celular , ADN/genética , ADN/aislamiento & purificación , Leucemia Experimental , Ratones , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Sondas ARN , ARN Mensajero/genética , Receptores de Eritropoyetina , Mapeo Restrictivo , Transcripción Genética , Activación Transcripcional
20.
EMBO J ; 10(7): 1809-16, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2050118

RESUMEN

The human GATA1, hGATA1 (previously called NF-E1, GF-1 or Eryf-1), a major sequence-specific DNA-binding protein of the erythrocytic lineage, is a member of a zinc-finger family of DNA-binding proteins. We report here the cloning of a human cDNA for a new member of this family. This member, called hGATA3, has 85% amino acid homology with hGATA1 in the DNA-binding domain and no homology elsewhere in the protein. Unlike hGATA1, hGATA3 is not localized on the X chromosome and we map it to the 10p15 band of the human genome. Northern blot analysis indicates that this factor is a T-cell specific transcription factor, present before activation and up-regulated during T-cell activation. The encoded hGATA3 protein, made in an in vitro transcription-translation assay, binds the WGATAR motif present in the human T-cell receptor (TCR) delta gene enhancer and, by transfection in HeLa cells, we show that hGATA3 can transactivate this TCR delta gene enhancer. Interestingly this enhancer binds and is also transactivated by hGATA1. Conversely, the promoter of the human glycophorin B (GPB), which is erythroid-specific and contains two WGATAR motifs, binds and is transactivated by hGATA1 and, to a lesser extent, by hGATA3. These results indicate that the activation of specific genes by hGATA1 or hGATA3 is partly governed by the lineage expression of these two factors during haematopoiesis and that, in the T-cell lineage, hGATA3 binds the human TCR delta gene enhancer and is involved in its expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Receptores de Antígenos de Linfocitos T/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 10 , Clonación Molecular , Proteínas de Unión al ADN/biosíntesis , Elementos de Facilitación Genéticos , Factores de Unión al ADN Específico de las Células Eritroides , Femenino , Factor de Transcripción GATA1 , Genes Sintéticos , Glicoforinas/genética , Células HeLa/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T/metabolismo , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/biosíntesis , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA