Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 34(2): 567-82, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15821117

RESUMO

Retinoid X receptors (RXR) are members of the nuclear receptor superfamily of ligand-activated transcription factors that have been characterized in a wide variety of metazoan phyla. They act as heterodimer partners of other nuclear receptors, and in vertebrates also activate transcription as homodimers in the presence of a ligand, 9-cis retinoic acid. In order to test the hypothesis that retinoic acid signaling pathways involving RXRs are present in the Lophotrochozoa, we have sought to isolate conserved members of this family from the platyhelminth parasite Schistosoma mansoni and its intermediate host, the mollusk Biomphalaria glabrata. Here we report that an RXR ortholog from B. glabrata (BgRXR) is better conserved, compared with mouse RXRalpha, both in the DNA-binding domain (89% identity) and in the ligand-binding domain (LBD) (81% identity), than are arthropod homologs. In EMSA, BgRXR binds to the direct repeat response element DR1 as a homodimer or as a heterodimer with mammalian RARalpha, LXR, FXR or PPARalpha. When transfected alone into mammalian cell lines, BgRXR transactivated transcription of a reporter gene from the Apo-A1 promoter in the presence of 9-cis retinoic acid or DHA. Constructs with the Gal4 DNA binding domain fused to the hinge and LBDs of BgRXR were used to show that ligand-dependent activation of transcription by BgRXR required its intact AF-2 activation domain, and that the LBD can form homodimers. Finally, the binding of 9-cis retinoic acid preferentially protected the LBD of BgRXR from degradation by trypsin in a proteolysis protection assay. Our results show that BgRXR binds and is activated by retinoids and suggest that retinoid signaling pathways are conserved in the Lophotrochozoa. The nucleotide sequence reported in this paper has been submitted to the GenBank/EBI Data Bank with accession no. AY048663.


Assuntos
Biomphalaria/metabolismo , Receptores X de Retinoides/metabolismo , Retinoides/metabolismo , Transcrição Gênica , Ativação Transcricional , Sequência de Aminoácidos , Animais , Biomphalaria/genética , Dimerização , Genes Reporter , Camundongos , Dados de Sequência Molecular , Filogenia , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Receptores X de Retinoides/química , Receptores X de Retinoides/classificação , Receptores X de Retinoides/genética , Alinhamento de Sequência , Transdução de Sinais/fisiologia , Técnicas do Sistema de Duplo-Híbrido
2.
Arterioscler Thromb Vasc Biol ; 22(5): 717-26, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12006382

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors activated by fatty acids and derivatives. Although PPARalpha mediates the hypolipidemic action of fibrates, PPARgamma is the receptor for the antidiabetic glitazones. PPARalpha is highly expressed in tissues such as liver, muscle, kidney, and heart, where it stimulates the beta-oxidative degradation of fatty acids. PPARgamma is predominantly expressed in adipose tissues, where it promotes adipocyte differentiation and lipid storage. PPARbeta/delta is expressed in a wide range of tissues, and recent findings indicate a role for this receptor in the control of adipogenesis. Pharmacological and gene-targeting studies have demonstrated a physiological role for PPARs in lipid and lipoprotein metabolism. PPARalpha controls plasma lipid transport by acting on triglyceride and fatty acid metabolism and by modulating bile acid synthesis and catabolism in the liver. All 3 PPARs regulate macrophage cholesterol homeostasis. By enhancing cholesterol efflux, they stimulate the critical steps of the reverse cholesterol transport pathway. As such, PPARs control plasma levels of cholesterol and triglycerides, which constitute major risk factors for coronary heart disease. Furthermore, PPARalpha and PPARgamma regulate the expression of key proteins involved in all stages of atherogenesis, such as monocyte and lymphocyte recruitment to the arterial wall, foam cell formation, vascular inflammation, and thrombosis. Thus, by regulating gene transcription, PPARs modulate the onset and evolution of metabolic disorders predisposing to atherosclerosis and exert direct antiatherogenic actions at the level of the vascular wall.


Assuntos
Arteriosclerose/genética , Arteriosclerose/metabolismo , Metabolismo dos Lipídeos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Humanos , Proteínas Nucleares/metabolismo , Fenótipo
3.
J Clin Invest ; 107(11): 1423-32, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11390424

RESUMO

Statins are inhibitors of the rate-limiting enzyme in cholesterol synthesis, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. In addition to reducing LDL cholesterol, statin treatment increases the levels of the antiatherogenic HDL and its major apolipoprotein apoA-I. Here, we investigated the molecular mechanisms of apoA-I regulation by statins. Treatment with statins increased apoA-I mRNA levels in human HepG2 hepatoma cells, and this effect was reversed by the addition of mevalonate, implicating HMG-CoA reductase as the relevant target of these drugs. Pretreatment with Actinomycin D abolished the increase of apoA-I mRNA, indicating that statins act at the transcriptional level. Indeed, statins increased the human apoA-I promoter activity in transfected cells, and we have identified a statin response element that coincides with a PPARalpha response element known to confer fibrate responsiveness to this gene. The statin effect could be abolished not only by mevalonate, but also by geranylgeranyl pyrophosphate, whereas inhibition of geranylgeranyl transferase activity or treatment with an inhibitor of the Rho GTP-binding protein family increased PPARalpha activity. Using dominant negative forms of these proteins, we found that Rho A itself mediates this response. Because cotreatment with statins and fibrates activated PPARalpha in a synergistic manner, these observations provide a molecular basis for combination treatment with statins and fibrates in coronary heart disease.


Assuntos
Apolipoproteína A-I/biossíntese , Proteínas de Ligação a DNA/metabolismo , Fenofibrato/análogos & derivados , Piridinas/farmacologia , Quinolinas/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Linhagem Celular , Meios de Cultura Livres de Soro , Óxidos N-Cíclicos , Inibidores Enzimáticos/farmacologia , Fenofibrato/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lipoproteínas HDL/biossíntese , Lipoproteínas HDL/metabolismo , Mercaptoetanol/análogos & derivados , Fosforilação , Regiões Promotoras Genéticas/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia
4.
Biochem J ; 352 Pt 2: 583-91, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085954

RESUMO

The DNA-binding activity of the transcription nuclear factor kappaB (NF-kappaB) is regulated by a redox-control mechanism involving the reduction of a disulphide bond from a specific cysteine residue conserved in all members of the NF-kappaB family. Thioredoxin is involved in this redox control. DNA binding and transactivating capacity of NF-kappaB are up-regulated by inducible phosphorylation. Here we demonstrate that the conserved redox cysteine in the c-Rel protein is involved in the phosphorylation regulation of the protein. When this cysteine residue is mutated to an aspartic acid residue, the mutant protein loses its capacity to be phosphorylated and its DNA-binding activity. In addition, our results suggest that, when the conserved redox cysteine is chemically modified by N-ethylmaleimide and 2-chloro-1,3-dinitrobenzene, the protein c-Rel cannot be phosphorylated. In contrast, the protein in which the cysteine residue was replaced by a serine residue, creating a potential phosphorylation site, is highly phosphorylated and binds kappaB sequences. The protein could loose the redox regulation of the phosphorylation when the residue replacing the cysteine can be itself phosphorylated. We also show that specific inhibitors of thioredoxin reductases impair the phosphorylation of the c-Rel protein, suggesting that the redox regulation of the protein controls its phosphorylation.


Assuntos
Cisteína/metabolismo , DNA/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Embrião de Galinha , Primers do DNA , Inibidores Enzimáticos/farmacologia , Mutagênese , Oxirredução , Fosforilação , Proteínas Proto-Oncogênicas c-rel/química , Compostos de Sulfidrila/química , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores
5.
Infect Immun ; 62(10): 4176-85, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7927673

RESUMO

Cholera toxin (CTX) is composed of two subunits, subunit A, which possesses ADP-ribosyltransferase activity, and subunit B, which is responsible for receptor binding. It has previously been shown that agents that increase cyclic AMP (cAMP) levels in cells induce differentiation of PC12 cells into neurite-like cells. In this report, we show that as little as 100 pg of CTX per ml induces such changes. CTX was found to ADP-ribosylate at least four membrane proteins of PC12 cells in vitro and in vivo and to increase intracellular cAMP levels. We have developed an inducible ctx gene expression system in Vibrio cholerae by using the tac promoter. The culture medium of the CTX-producing bacteria was able to induce the morphological changes and the ADP-ribosylation of the PC12 cell membrane proteins. We have constructed two CTX-cross-reactive mutant proteins (CTX-CRM) by site-directed mutagenesis. The choice of glutamic acid 29 as the target amino acid was based on sequence similarities with other bacterial toxins. CTX-CRM-E29 delta, in which the Glu-29 of the A subunit was deleted, showed strongly reduced ADP-ribosyltransferase activity and did not induce significant morphological changes of PC12 cells. In contrast, CTX-CRM-E29D, in which the Glu-29 was replaced by an aspartic acid, was as active as the wild-type protein. We conclude that the ADP-ribosylation activity of CTX is important for the toxin-induced differentiation of PC12 cells. Pertussis toxin, which had no visible effect on PC12 cell morphology, was also able to ADP-ribosylate a membrane-bound protein(s) in vitro and in vivo. Pertussis toxin alone did not significantly increase cAMP levels in PC12 cells, but it acted synergistically with CTX.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Toxina da Cólera/toxicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Toxina da Cólera/biossíntese , Toxina da Cólera/genética , Colforsina/farmacologia , Cricetinae , AMP Cíclico/análise , Vetores Genéticos , Dados de Sequência Molecular , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Coelhos , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/toxicidade
6.
Oncogene ; 8(11): 3103-10, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8414512

RESUMO

Triiodothyronine (T3) positively regulates both the expression of the MyoD gene, a key myogenic regulator, and C2 muscle cell differentiation. To directly examine the role of its nuclear receptors in the control of myogenesis, we introduced a c-erbA expression vector into C2 muscle cells by transient or stable transfection. Our results show that c-erbA can play a potent role in the triggering of muscle terminal differentiation since its overexpression leads to: (1) a complete abrogation of the activity of the myogenesis inhibitor AP-1 (fos/jun) transcription factor; (2) an enhanced induction of MyoD expression upon T3 treatment; (3) the acquisition by T3 of the ability to trigger both growth arrest and terminal differentiation in the presence of large amounts of serum mitogens, a property that is otherwise specific to retinoic acid (RA). Thus, c-erbA is one of the two protooncogenes (with c-ski) that acts as positive regulator of muscle differentiation. Furthermore, the fact that c-erbA overexpression allows T3 to largely mimic the RA effects indicates that their biological differences in the modulation of myogenic program primarily rely on the differential expression of their receptors in C2 muscle cells rather than on an intrinsic specificity of their target genes.


Assuntos
Regulação da Expressão Gênica , Músculos/citologia , Proto-Oncogenes , Receptores dos Hormônios Tireóideos/genética , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Camundongos , Músculos/metabolismo , Proteína MyoD/análise , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores do Ácido Retinoico/genética , Transfecção , Tri-Iodotironina/farmacologia
7.
Genes Dev ; 5(11): 2033-47, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1682217

RESUMO

The v-erbA oncoprotein represents a retrovirus-transduced oncogenic version of the thyroid hormone (T3/T4) receptor c-erbA (type alpha). It contributes to virus-induced erythroleukemia by efficiently arresting differentiation of red cell progenitors and by suppressing transcription of erythrocyte-specific genes. Here, we show that v-erbA and c-erbA bind directly to sequences within the promoter of the erythrocyte-specific carbonic anhydrase II (CAII), a gene whose transcription is efficiently suppressed by v-erbA. This erbA-binding site confers thyroid hormone responsiveness to a heterologous promoter in transient expression experiments and is a target for efficient down-regulation of CAII transcription by the v-erbA oncoprotein. In stably transformed erythroblasts coexpressing the v-erbA oncoprotein and the c-erbA/T3 receptor at an approximately equimolar ratio, c-erbA activity is dominant over v-erbA. T3 efficiently induced erythroid differentiation in these cells, thus overcoming the v-erbA-mediated differentiation arrest. Likewise, T3 activated CAII transcription as well as transient expression of a T3-responsive reporter gene containing the CAII-specific erbA-binding site. The c-erbA-dependent activation of this CAII reporter construct could only be suppressed by very high amounts of v-erbA. Our results suggest that overexpression of v-erbA is required for its function as an oncoprotein.


Assuntos
Anidrases Carbônicas/genética , Diferenciação Celular/genética , Eritroblastos/citologia , Regulação da Expressão Gênica/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Oncogênicas de Retroviridae/genética , Sequência de Bases , Sítios de Ligação/genética , Northern Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Resistência a Medicamentos/genética , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Genes gag/genética , Células HeLa , Humanos , Luciferases/genética , Dados de Sequência Molecular , Neomicina/farmacologia , Proteínas Oncogênicas v-erbA , Plasmídeos/genética , Testes de Precipitina , Receptores dos Hormônios Tireóideos/genética , Tri-Iodotironina/farmacologia , Vaccinia virus/genética
8.
Genes Dev ; 4(10): 1663-76, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1979040

RESUMO

The v-erbA oncogene of avian erythroblastosis virus (AEV) encodes a ligand-independent mutated version of the chicken c-erbA alpha-encoded thyroid hormone receptor. The v-erbA gene product, a 75-kD gag/v-erbA fusion protein, is phosphorylated on Ser-16/17 of its v-erbA-encoded domain, and phosphorylation at this site is increased in vivo after activation of either the PKA or PKC signal transduction pathways. To test the hypothesis that phosphorylation of Ser-16/17 regulates gag/v-erbA protein function, mutant proteins in which Ser-16/17 had been changed to alanine or threonine residues were analyzed for their ability to inhibit erythroid differentiation of ts v-erbB or ts v-sea-transformed erythroblasts at nonpermissive temperature. Conversion of Ser-16/17 into alanine, although not affecting nuclear localization or DNA binding of the gag/erbA protein, prevented phosphorylation of the v-erbA-encoded domain of the protein both in unstimulated cells or after stimulation by PKA and PKC activators. The nonphosphorylatable AA-gag/v-erbA protein proved unable to inhibit temperature-induced differentiation of ts v-erbB and ts v-sea-transformed erythroblasts and to block expression of the erythrocyte-specific genes band 3 and carbonic anhydrase II. Back mutation of these alanine residues to serine resulted in the recovery of both normal phosphorylation levels and wild-type biological activity. In contrast, substitution of Ser-16/17 for threonine, which preserved phosphorylation in unstimulated cells but not PKA- and PKC-enhanced phosphorylation, resulted in a partially active gag/v-erbA protein. These results, together with the fact that the protein kinase inhibitor H7 resulted in both a dose-dependent inhibition of gag/v-erbA protein phosphorylation and the induction of terminal differentiation of AEV-transformed erythroblasts show that phosphorylation of gag/v-erbA protein is required for full biological activity. These results support the hypothesis that phosphorylation of the gag/v-erbA protein is important for transcriptional repression of at least some of its target genes in erythroid cells.


Assuntos
Alpharetrovirus/genética , Regulação Viral da Expressão Gênica/fisiologia , Oncogenes/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina , Sequência de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Embrião de Galinha , Eritroblastos/efeitos dos fármacos , Eritrócitos/fisiologia , Produtos do Gene gag/genética , Isoquinolinas/farmacologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/genética , Proteínas Oncogênicas v-erbA , Fenótipo , Fosforilação , Piperazinas/farmacologia , Inibidores de Proteínas Quinases , Proteínas Oncogênicas de Retroviridae/genética
9.
Oncogene ; 4(10): 1247-54, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2552374

RESUMO

The c-erbA alpha progenitor of the v-erbA oncogene of avian erythroblastosis virus (AEV) encodes a nuclear receptor for the thyroid hormone triiodothyronine (T3) which acts as a ligand-dependent transcription factor. As previously reported (Goldberg et al., EMBO J., 7, 2425-2433), the 46 kd chicken c-erbA alpha-encoded T3 receptor (ck-ErbA alpha) is phosphorylated at two major sites. Only one of these sites (Ser28/Ser29) is retained in the v-erbA-encoded P75gag-v-erbA protein. We report here the identification of the second phosphorylation site of ck-ErbA alpha as a single serine residue localized at position 12. We propose that casein kinase II, a protein kinase distributed in the cytosolic and nuclear compartments of a number of different tissues, is responsible for serine 12 phosphorylation on the following grounds. First, serine 12 is part of a sequence containing multiple acidic amino-acids, a feature common to all sites phosphorylated by casein kinase II in physiological substrates. Second, ck-ErbA alpha was found to be phosphorylated by purified casein kinase II in vitro at the same site, as defined by two-dimensional mapping experiments, as that observed in vivo. Third, conversion of serine 12 into an unphosphorylatable alanine residue by site directed mutagenesis abolishes the phosphorylation of ck-ErbA alpha by casein kinase II in vitro. Phosphorylation of serine 12 is likely to play a role in the modulation of ErbA alpha function since both serine 12 and the casein kinase II phosphorylation sequence motif are phylogenetically conserved in all known members of the c-erbA alpha gene family encoding T3 binding proteins. The codon specifying serine 12 in ck-ErbA alpha being precisely the point where recombination between gag and ck-c-erbA alpha occurred to generate v-erbA, our results furthermore suggest that deletion of serine 12 could contribute to the oncogenic activation of v-erbA.


Assuntos
Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proto-Oncogenes , Receptores dos Hormônios Tireóideos/metabolismo , Sequência de Aminoácidos , Caseína Quinases , Regulação Neoplásica da Expressão Gênica , Fosforilação , Proteínas Proto-Oncogênicas/genética , Transfecção
10.
Biochimie ; 71(2): 279-91, 1989 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2539867

RESUMO

In this review, we discuss the biological action and biochemical function of the v-erbA oncogene product, and the role of c-erbA proto-oncogene products as thyroid hormone receptors, as related to the molecular structure and function of the nuclear hormone receptors at large.


Assuntos
Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/fisiologia , Hormônios Tireóideos/metabolismo , Animais , Sequência de Bases , Galinhas , DNA/metabolismo , Humanos , Dados de Sequência Molecular , Proto-Oncogene Mas , Ratos , Hormônios Tireóideos/genética
11.
EMBO J ; 7(8): 2425-33, 1988 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2903825

RESUMO

The c-erbA proto-oncogene encodes a nuclear receptor for thyroid hormone (T3), which is believed to stimulate transcription from specific target promoters upon binding to cis-acting DNA sequence elements. The v-erbA oncogene of avian erythroblastosis virus (AEV) encodes a ligand-independent version of this nuclear receptor. The v-erbA product inhibits terminal differentiation of avian erythroblasts, presumably by affecting the transcription of specific genes. We show here that the c-erbA-encoded nuclear receptor (p46c-erbA) is phosphorylated on serine residues on two distinct sites. One of these sites, defined by the limit tryptic phosphopeptide 28SSQCLVK, is retained on the v-erbA-encoded P75gag-v-erbA protein. This site is located in the amino-terminal domain of these molecules, 21 amino acids upstream of the DNA-binding region. Phosphorylation of this site in both p46c-erbA and P75gag-v-erbA is enhanced 10-fold following treatment of cells with activators of either protein kinase C or cAMP-dependent protein kinase. Since cAMP-dependent protein kinase phosphorylates both p46c-erbA and P75gag-v-erbA in vitro at the same site as that observed in vivo, at least part of the cAMP-dependent phosphorylation of erbA molecules in cells could result from direct phosphorylation by this enzyme. The possible role phosphorylation may play in the function of the erbA-encoded transcriptional factors is discussed.


Assuntos
Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores dos Hormônios Tireóideos/metabolismo , Proteínas dos Retroviridae/metabolismo , Alpharetrovirus/genética , Animais , Sequência de Bases , Linhagem Celular Transformada , Embrião de Galinha , Ativação Enzimática , Regulação da Expressão Gênica , Proteínas Oncogênicas v-erbA , Fragmentos de Peptídeos/metabolismo , Mapeamento de Peptídeos , Fosforilação , Testes de Precipitina , Proto-Oncogenes , Receptores dos Hormônios Tireóideos/genética , Proteínas dos Retroviridae/genética
13.
J Immunol Methods ; 53(3): 313-9, 1982 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-7142712

RESUMO

Monoclonal antibodies can be purified directly from ascitic fluids by chromatography on a DEAE Affi-gel blue column. Optimal conditions were determined for the recovery of immunoglobulins free of contaminating protease and nuclease activities.


Assuntos
Antracenos , Anticorpos Monoclonais/isolamento & purificação , Líquido Ascítico/análise , Triazinas , Animais , Cromatografia em Gel , Cromatografia por Troca Iônica , Concentração de Íons de Hidrogênio , Métodos , Camundongos
15.
Biosci Rep ; 1(11): 885-92, 1981 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7030431

RESUMO

Hybridomas secreting antibodies against human urokinase have been produced by the cell-fusion technique (Köhler & Milstein, 1976). They belong to the IgG1 and IgG2 classes. Fixation and inhibition of the binding of 125 I-labelled urokinase, in radioimmunoassay, show that two of the monoclonal antibodies exhibit a high titer in ascitic fluids, a good sensitivity, and no cross reaction with other enzymes showing partial sequence homology with urokinase. Moreover, one of the monoclonal antibodies is able to inhibit the enzymatic activity of urokinase using a chromogenic substrate.


Assuntos
Anticorpos Monoclonais , Endopeptidases/imunologia , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Animais , Complexo Antígeno-Anticorpo , Fusão Celular , Células Clonais , Humanos , Hibridomas/enzimologia , Camundongos , Camundongos Endogâmicos/imunologia , Radioimunoensaio/métodos , Baço/imunologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...