Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Exp Parasitol ; 163: 8-15, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26821294

RESUMO

Cryptosporidium parvum is one of the most radioresistant organisms identified to date. In a previous study, we found that thioredoxin peroxidase (CpTPx) was significantly upregulated in this species following exposure to high dose (10 kGy) of γ-irradiation. To assess the potential of CpTPx to confer radioprotection in mammalian cells, it was expressed in COS-7 African green monkey kidney cells (CpTPx-COS7). For comparison, the thioredoxin peroxidase of Cryptosporidium muris (CmTPx) was also expressed in these cells (CmTPx-COS7 cells), which has been confirmed to have lesser antioxidant activity than CpTPx in the previous study. Notably, the survival rates of CpTPx-COS7 cells were significantly higher (12-22%) at 72 h after 8 Gy irradiation than CmTPx-COS7 or non-transfected COS-7 (ntCOS-7) counterparts. In addition, CpTPx revealed a 50% of ROS reduction in irradiated CpTPx-COS7 cells, while γ-H2AX DNA damage marker expression was not significantly changed. Furthermore, the amount of apoptosis only increased to about 120% after 2-8 Gy irradiation compared to 200-300% increase observed in ntCOS-7 cells. CmTPx was shown to have antioxidant and DNA damage protection activities; however, these activities were always lower than those of CpTPx. These results suggest that the potent antioxidant and protective activities of CpTPx are well conserved in this cell-based system and that CpTPx contributed to the radioprotection of mammalian cells through its exceptional antioxidant activity.


Assuntos
Antioxidantes/metabolismo , Células COS/enzimologia , Cryptosporidium parvum/enzimologia , Raios gama , Peroxirredoxinas/biossíntese , Animais , Células COS/parasitologia , Células COS/efeitos da radiação , Chlorocebus aethiops , Cryptosporidium parvum/efeitos da radiação , Regulação Enzimológica da Expressão Gênica , Microscopia Confocal , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção
2.
J Vet Pharmacol Ther ; 39(5): 478-87, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26970544

RESUMO

Research on drug metabolism and pharmacokinetics in large animal species including the horse is scarce because of the challenges in conducting in vivo studies. The metabolic reactions catalyzed by cytochrome P450s (CYPs) are central to drug pharmacokinetics. This study elucidated the characteristics of equine CYPs using diazepam (DZP) as a model compound as this drug is widely used as an anesthetic and sedative in horses, and is principally metabolized by CYPs. Diazepam metabolic activities were measured in vitro using horse and rat liver microsomes to clarify the species differences in enzyme kinetic parameters of each metabolite (temazepam [TMZ], nordiazepam [NDZ], p-hydroxydiazepam [p-OH-DZP], and oxazepam [OXZ]). In both species microsomes, TMZ was the major metabolite, but the formation rate of p-OH-DZP was significantly less in the horse. Inhibition assays with a CYP-specific inhibitors and antibody suggested that CYP3A was the main enzyme responsible for DZP metabolism in horse. Four recombinant equine CYP3A isoforms expressed in Cos-7 cells showed that CYP3A96, CYP3A94, and CYP3A89 were important for TMZ formation, whereas CYP3A97 exhibited more limited activity. Phylogenetic analysis suggested diversification of CYP3As in each mammalian order. Further study is needed to elucidate functional characteristics of each equine CYP3A isoform for effective use of diazepam in horses.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Diazepam/farmacocinética , Cavalos/metabolismo , Hipnóticos e Sedativos/farmacocinética , Animais , Células COS/enzimologia , Células COS/metabolismo , Chlorocebus aethiops , Citocromo P-450 CYP3A/genética , Diazepam/análogos & derivados , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Nordazepam/farmacocinética , Oxazepam/farmacocinética , Filogenia , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Temazepam/farmacocinética
3.
Nat Genet ; 21(4): 424-8, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10192396

RESUMO

High-density lipoprotein (HDL) cholesterol levels are inversely associated with risk of atherosclerotic cardiovascular disease. At least 50% of the variation in HDL cholesterol levels is genetically determined, but the genes responsible for variation in HDL levels have not been fully elucidated. Lipoprotein lipase (LPL) and hepatic lipase (HL), two members of the triacylglyerol (TG) lipase family, both influence HDL metabolism and the HL (LIPC) locus has been associated with variation in HDL cholesterol levels in humans. We describe here the cloning and in vivo functional analysis of a new member of the TG lipase family. In contrast to other family members, this new lipase is synthesized by endothelial cells in vitro and thus has been termed endothelial lipase (encoded by the LIPG gene). EL is expressed in vivo in organs including liver, lung, kidney and placenta, but not in skeletal muscle. In contrast to LPL and HL, EL has a lid of only 19 residues. EL has substantial phospholipase activity, but less triglyceride lipase activity. Overexpression of EL in mice reduced plasma concentrations of HDL cholesterol and its major protein apolipoprotein A-I. The endothelial expression, enzymatic profile and in vivo effects of EL suggest that it may have a role in lipoprotein metabolism and vascular biology.


Assuntos
Endotélio Vascular/enzimologia , Lipase/genética , Lipase/metabolismo , Lipoproteínas HDL/metabolismo , Sequência de Aminoácidos , Animais , Anticolesterolemiantes/farmacologia , Apolipoproteína A-I/genética , Northern Blotting , Células COS/enzimologia , Células Cultivadas , HDL-Colesterol/sangue , HDL-Colesterol/efeitos dos fármacos , Clonagem Molecular , Endotélio Vascular/citologia , Feminino , Humanos , Lipoproteínas HDL/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Placenta , Gravidez , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Homologia de Sequência de Aminoácidos , Transfecção
4.
Blood Cells Mol Dis ; 42(2): 159-66, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19167250

RESUMO

Gaucher disease is an autosomal recessive disorder. It is characterized by the accumulation of glucosylceramide in lysosomes of mononuclear phagocyte system, attributable to acid beta-glucosidase deficiency. The main consequences of this disease are hepatosplenomegaly, skeletal lesions and, sometimes, neurological manifestations. At sub-inhibitory concentrations, several competitive inhibitors act as chemical chaperones by inducing protein stabilization and increasing enzymatic activity. Here we tested two iminosugars (NB-DNJ and NN-DNJ) and four aminocyclitols with distinct degrees of lipophilicity as pharmacological chaperones for glucocerebrosidase (GBA). We report an increase in the activity of GBA using NN-DNJ, NB-DNJ and aminocyclitol 1 in stably transfected cell lines, and an increment with NN-DNJ and aminocyclitol 4 in patient fibroblasts. These results on specific mutations validate the use of chemical chaperones as a therapeutic approach for Gaucher disease. However, the development and analysis of new compounds is required in order to find more effective therapeutic agents that are active on a broader range of mutations.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Ciclitóis/farmacologia , Doença de Gaucher/enzimologia , Glucosilceramidase/genética , Dobramento de Proteína/efeitos dos fármacos , 1-Desoxinojirimicina/farmacologia , Animais , Células COS/efeitos dos fármacos , Células COS/enzimologia , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Doença de Gaucher/genética , Doença de Gaucher/patologia , Genótipo , Glucosilceramidase/antagonistas & inibidores , Glucosilceramidase/química , Glucosilceramidase/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imino Açúcares/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
5.
J Cell Biol ; 135(4): 939-51, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8922378

RESUMO

Peroxisome-to-mitochondrion mistargeting of the homodimeric enzyme alanine:glyoxylate aminotransferase 1 (AGT) in the autosomal recessive disease primary hyperoxaluria type 1 (PH1) is associated with the combined presence of a normally occurring Pro(11)Leu polymorphism and a PH1-specific Gly170Arg mutation. The former leads to the formation of a novel NH2-terminal mitochondrial targeting sequence (MTS), which although sufficient to direct the import of in vitro-translated AGT into isolated mitochondria, requires the additional presence of the Gly170Arg mutation to function efficiently in whole cells. The role of this mutation in the mistargeting phenomenon has remained elusive. It does not interfere with the peroxisomal targeting or import of AGT. In the present study, we have investigated the role of the Gly170Arg mutation in AGT mistargeting. In addition, our studies have led us to examine the relationship between the oligomeric status of AGT and the peroxisomal and mitochondrial import processes. The results obtained show that in vitro-translated AGT rapidly forms dimers that do not readily exchange subunits. Although the presence of the Pro(11)Leu or Gly170Arg substitutions alone had no effect on dimerization, their combined presence abolished homodimerization in vitro. However, AGT containing both substitutions was still able to form heterodimers in vitro with either normal AGT or AGT containing either substitution alone. Expression of various combinations of normal and mutant, as well as epitope-tagged and untagged forms of AGT in whole cells showed that normal AGT rapidly dimerizes in the cytosol and is imported into peroxisomes as a dimer. This dimerization prevents mitochondrial import, even when the AGT possesses an MTS generated by the Pro(11)Leu substitution. The additional presence of the Gly170Arg substitution impairs dimerization sufficiently to allow mitochondrial import. Pharmacological inhibition of mitochondrial import allows AGT containing both substitutions to be imported into peroxisomes efficiently, showing that AGT dimerization is not a prerequisite for peroxisomal import.


Assuntos
Alanina Transaminase/genética , Alanina Transaminase/metabolismo , Hiperoxalúria Primária/enzimologia , Microcorpos/enzimologia , Mitocôndrias/enzimologia , Transaminases , Alanina Transaminase/antagonistas & inibidores , Animais , Arginina/genética , Transporte Biológico/fisiologia , Células COS/enzimologia , Dimerização , Glicina/genética , Humanos , Leucina/genética , Fígado/enzimologia , Mutação Puntual/fisiologia , Polimorfismo Genético , Prolina/genética
6.
J Cell Biol ; 143(2): 511-21, 1998 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-9786959

RESUMO

Effects of fatty acids on translocation of the gamma- and epsilon-subspecies of protein kinase C (PKC) in living cells were investigated using their proteins fused with green fluorescent protein (GFP). gamma-PKC-GFP and epsilon-PKC-GFP predominated in the cytoplasm, but only a small amount of gamma-PKC-GFP was found in the nucleus. Except at a high concentration of linoleic acid, all the fatty acids examined induced the translocation of gamma-PKC-GFP from the cytoplasm to the plasma membrane within 30 s with a return to the cytoplasm in 3 min, but they had no effect on gamma-PKC-GFP in the nucleus. Arachidonic and linoleic acids induced slow translocation of epsilon-PKC-GFP from the cytoplasm to the perinuclear region, whereas the other fatty acids (except for palmitic acid) induced rapid translocation to the plasma membrane. The target site of the slower translocation of epsilon-PKC-GFP by arachidonic acid was identified as the Golgi network. The critical concentration of fatty acid that induced translocation varied among the 11 fatty acids tested. In general, a higher concentration was required to induce the translocation of epsilon-PKC-GFP than that of gamma-PKC-GFP, the exceptions being tridecanoic acid, linoleic acid, and arachidonic acid. Furthermore, arachidonic acid and the diacylglycerol analogue (DiC8) had synergistic effects on the translocation of gamma-PKC-GFP. Simultaneous application of arachidonic acid (25 MicroM) and DiC8 (10 microM) elicited a slow, irreversible translocation of gamma-PKC- GFP from the cytoplasm to the plasma membrane after rapid, reversible translocation, but a single application of arachidonic acid or DiC8 at the same concentration induced no translocation. These findings confirm the involvement of fatty acids in the translocation of gamma- and epsilon-PKC, and they also indicate that each subspecies has a specific targeting mechanism that depends on the extracellular signals and that a combination of intracellular activators alters the target site of PKCs.


Assuntos
Ácidos Graxos/farmacologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Animais , Ácido Araquidônico/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Células COS/efeitos dos fármacos , Células COS/enzimologia , Cálcio/metabolismo , Quelantes/farmacologia , Diglicerídeos/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde , Indicadores e Reagentes , Proteínas Luminescentes , Proteína Quinase C-épsilon , Proteínas Recombinantes de Fusão/metabolismo
7.
J Cell Biol ; 134(6): 1513-7, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8830778

RESUMO

RPTP mu is a receptor-like protein tyrosine phosphatase that mediates homophilic cell-cell interactions. Surface expression of RPTP mu is restricted to cell-cell contacts and is upregulated with increasing cell density, suggesting a role for RPTP mu in contact-mediated signaling. It was recently reported (Brady-Kalnay, S.M., D.L. Rimm, and N.K. Tonks. 1995. J. Cell Biol. 130:977-986) that RPTP mu binds directly to cadherin/catenin complexes, and thus may regulate the tyrosine phosphorylation of such complexes. Here we report that this concept needs revision. Through reciprocal precipitations using a variety of antibodies against RPTP mu, cadherins, and catenins, we show that RPTP mu does not interact with cadherin/catenin complexes, even when assayed under very mild lysis conditions. We find that the anti-RPTP mu antiserum used by others precipitates cadherins in a nonspecific manner independent of RPTP mu. We conclude that, contrary to previous claims, RPTP mu does not interact with cadherin complexes and thus is unlikely to directly regulate cadherin/catenin function.


Assuntos
Caderinas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Superfície Celular/metabolismo , Transativadores , Animais , Anticorpos Monoclonais , Células COS/química , Células COS/enzimologia , Caderinas/análise , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/metabolismo , Epitélio/química , Epitélio/enzimologia , Expressão Gênica/fisiologia , Pulmão/citologia , Proteínas de Membrana/genética , Vison , Testes de Precipitina , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/imunologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores , Receptores de Superfície Celular/genética , beta Catenina
8.
J Cell Biol ; 134(6): 1519-29, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8830779

RESUMO

Cadherins are calcium-dependent cell adhesion molecules that play fundamental roles in embryonic development, tissue morphogenesis, and cancer. A prerequisite for their function is association with the actin cytoskeleton via the catenins. Tyrosine phosphorylation of beta-catenin, which correlates with a reduction in cadherin-dependent cell adhesion, may provide cells with a mechanism to regulate cadherin activity. Here we report that beta-catenin immune precipitates from PC12 cells contain tyrosine phosphatase activity which dephosphorylates beta-catenin in vitro. In addition, we show that a member of the leukocyte antigen-related protein (LAR)-related transmembrane tyrosine phosphatase family (LAR-PTP) associates with the cadherin-catenin complex. This association required the amino-terminal domain of beta-catenin but does not require the armadillo repeats, which mediate association with cadherins. The interaction also is detected in PC9 cells, which lack alpha-catenin. Thus, the association is not mediated by alpha-catenin or by cadherins. Interestingly, LAR-PTPs are phosphorylated on tyrosine in a TrkA-dependent manner, and their association with the cadherin-catenin complex is reduced in cells treated with NGF. We propose that changes in tyrosine phosphorylation of beta-catenin mediated by TrkA and LAR-PTPs control cadherin adhesive function during processes such as neurite outgrowth.


Assuntos
Caderinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptor trkA , Transativadores , Sequência de Aminoácidos , Animais , Células COS/química , Células COS/enzimologia , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/química , Regulação para Baixo/efeitos dos fármacos , Fibronectinas/química , Imunoglobulinas/química , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Fatores de Crescimento Neural/farmacologia , Células PC12/química , Células PC12/enzimologia , Fosforilação , Testes de Precipitina , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases/química , Ratos , Tirosina/metabolismo , beta Catenina
9.
J Cell Biol ; 143(2): 501-10, 1998 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-9786958

RESUMO

Phosphatidylinositol 4,5-bisphosphate (PtdIns[4,5]P2) pools that bind pleckstrin homology (PH) domains were visualized by cellular expression of a phospholipase C (PLC)delta PH domain-green fluorescent protein fusion construct and analysis of confocal images in living cells. Plasma membrane localization of the fluorescent probe required the presence of three basic residues within the PLCdelta PH domain known to form critical contacts with PtdIns(4, 5)P2. Activation of endogenous PLCs by ionophores or by receptor stimulation produced rapid redistribution of the fluorescent signal from the membrane to cytosol, which was reversed after Ca2+ chelation. In both ionomycin- and agonist-stimulated cells, fluorescent probe distribution closely correlated with changes in absolute mass of PtdIns(4,5)P2. Inhibition of PtdIns(4,5)P2 synthesis by quercetin or phenylarsine oxide prevented the relocalization of the fluorescent probe to the membranes after Ca2+ chelation in ionomycin-treated cells or during agonist stimulation. In contrast, the synthesis of the PtdIns(4,5)P2 imaged by the PH domain was not sensitive to concentrations of wortmannin that had been found inhibitory of the synthesis of myo-[3H]inositol- labeled PtdIns(4,5)P2. Identification and dynamic imaging of phosphoinositides that interact with PH domains will further our understanding of the regulation of such proteins by inositol phospholipids.


Assuntos
Proteínas Sanguíneas/metabolismo , Cálcio/farmacologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfoproteínas , Células 3T3/química , Células 3T3/enzimologia , Animais , Proteínas Sanguíneas/química , Células COS/química , Células COS/enzimologia , Membrana Celular/química , Membrana Celular/enzimologia , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Proteínas de Fluorescência Verde , Indicadores e Reagentes , Inositol 1,4,5-Trifosfato/metabolismo , Inositol 1,4,5-Trifosfato/farmacologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Proteínas Luminescentes , Camundongos , Fosfatidilinositol 4,5-Difosfato/análise , Fosfatidilinositol 4,5-Difosfato/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Transfecção , Trítio , Fosfolipases Tipo C/química , Fosfolipases Tipo C/metabolismo
10.
J Cell Biol ; 155(6): 1065-79, 2001 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-11739414

RESUMO

SHIP-2 is a phosphoinositidylinositol 3,4,5 trisphosphate (PtdIns[3,4,5]P3) 5-phosphatase that contains an NH2-terminal SH2 domain, a central 5-phosphatase domain, and a COOH-terminal proline-rich domain. SHIP-2 negatively regulates insulin signaling. In unstimulated cells, SHIP-2 localized in a perinuclear cytosolic distribution and at the leading edge of the cell. Endogenous and recombinant SHIP-2 localized to membrane ruffles, which were mediated by the COOH-terminal proline-rich domain. To identify proteins that bind to the SHIP-2 proline-rich domain, yeast two-hybrid screening was performed, which isolated actin-binding protein filamin C. In addition, both filamin A and B specifically interacted with SHIP-2 in this assay. SHIP-2 coimmunoprecipitated with filamin from COS-7 cells, and association between these species did not change after epidermal growth factor stimulation. SHIP-2 colocalized with filamin at Z-lines and the sarcolemma in striated muscle sections and at membrane ruffles in COS-7 cells, although the membrane ruffling response was reduced in cells overexpressing SHIP-2. SHIP-2 membrane ruffle localization was dependent on filamin binding, as SHIP-2 was expressed exclusively in the cytosol of filamin-deficient cells. Recombinant SHIP-2 regulated PtdIns(3,4,5)P3 levels and submembraneous actin at membrane ruffles after growth factor stimulation, dependent on SHIP-2 catalytic activity. Collectively these studies demonstrate that filamin-dependent SHIP-2 localization critically regulates phosphatidylinositol 3 kinase signaling to the actin cytoskeleton.


Assuntos
Actinas/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Sequência de Aminoácidos , Animais , Células COS/efeitos dos fármacos , Células COS/enzimologia , Membrana Celular/enzimologia , Proteínas Contráteis/genética , Fator de Crescimento Epidérmico/farmacologia , Filaminas , Humanos , Inositol Polifosfato 5-Fosfatases , Melanoma , Camundongos , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/química , Músculo Esquelético/química , Músculo Esquelético/citologia , Miocárdio/química , Miocárdio/citologia , Sondas de Oligonucleotídeos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/análise , Monoéster Fosfórico Hidrolases/genética , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido , Leveduras
11.
Biochem J ; 386(Pt 2): 341-8, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15537392

RESUMO

We previously reported that acute agonist activation of G(i/o)-coupled receptors inhibits adenylate cyclase (AC) type VIII activity, whereas agonist withdrawal following chronic activation of these receptors induces AC-VIII superactivation. Three splice variants of AC-VIII have been identified, which are called AC-VIII-A, -B and -C (with AC-VIII-B missing the glycosylation domain and AC-VIII-C lacking most of the C1b area). We report here that AC-VIII-A and -B, but not -C, are inhibited by acute mu-opioid and dopaminergic type D2 receptor activation, indicating that the C1b area of AC-VIII has an important role in AC inhibition by G(i/o)-coupled receptor activation. On the other hand the glycosylation sites in AC-VIII did not play a role in AC-VIII regulation. Although AC-VIII-A and -C differed in their capacity to be inhibited by acute agonist exposure, agonist withdrawal after prolonged treatment led to a similar superactivation of all three splice variants, with no significant change in AC-VIII expression. AC-VIII superactivation was not affected by pre-incubation with a cell permeable cAMP analogue, indicating that the superactivation does not depend on the agonist-induced reduction in cAMP levels. The superactivated AC-VIII-A, -B and -C were similarly re-inhibited by re-application of agonist (morphine or quinpirole), returning the activity to control levels. These results demonstrate marked differences in the agonist inhibition of the AC-VIII splice variants before, but not after, superactivation.


Assuntos
Adenilil Ciclases/fisiologia , Processamento Alternativo/fisiologia , Variação Genética/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Adenilil Ciclases/metabolismo , Animais , Células COS/química , Células COS/efeitos dos fármacos , Células COS/enzimologia , Células COS/metabolismo , Chlorocebus aethiops , Colforsina/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Vetores Genéticos , Ionomicina/farmacologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Toxina Pertussis/farmacologia , Receptores de Dopamina D2 , Fatores de Tempo
12.
Biochem J ; 386(Pt 1): 153-60, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15458386

RESUMO

Intestinal alk-SMase (alkaline sphingomyelinase) is an ectoenzyme related to the NPP (nucleotide phosphodiesterase) family. It has five potential N-glycosylation sites and predicated transmembrane domains at both the N- and C-termini. The amino acid residues forming the two metal-binding sites in NPP are conserved, and those of the active core are modified. We examined the functional changes of the enzyme induced by deglycosylation and mutagenesis. Treating alk-SMase cDNA-transfected COS-7 cells with tunicamycin rendered the expressed enzyme completely inactive. Mutations of the five potential N-glycosylation sites individually and in combination showed that these sites were all glycosylated and deficient glycosylation decreased the enzyme activity. Immunogold labelling showed that the wild-type enzyme was mainly located in the plasma membrane, whereas the C-terminal domain-truncated enzyme was released into the medium. Deglycosylation blocked the release of the enzyme that accumulated in endosome-like structures. The enzyme activity was also decreased by mutations of the residues forming the putative metal-binding sites and the active core. Substitution of the active core sequence with that of NPP or mutation of T75 in the core abolished the enzyme activity against sphingomyelin but failed to render the enzyme NPP active. Our results indicate that alk-SMase activity is severely affected by defective N-glycosylation and structural alterations of the putative metal-binding sites and the predicted active core.


Assuntos
Cátions Bivalentes/metabolismo , Intestinos/enzimologia , Processamento de Proteína Pós-Traducional , Esfingomielina Fosfodiesterase/química , Animais , Sítios de Ligação , Células COS/enzimologia , Membrana Celular/enzimologia , Cricetinae , Cricetulus , Meios de Cultivo Condicionados , Endossomos/enzimologia , Glicosilação/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Proteínas de Membrana/análise , Mutagênese Sítio-Dirigida , Fosfodiesterase I/análise , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Esfingomielina Fosfodiesterase/análise , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo , Transfecção , Tunicamicina/farmacologia
13.
Cancer Res ; 60(17): 4725-8, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10987276

RESUMO

Irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11)] is metabolized by esterases to yield the potent topoisomerase I poison 7-ethyl-10-hydroxycamptothecin. One of the major side effects observed with CPT-11 is gastrointestinal toxicity, and we supposed that this might be due to local activation of CPT-11 within the gut. Carboxylesterase (CE) activity was detected in human gut biopsies, and extracts of these tissues converted CPT-11 to 7-ethyl-10-hydroxycamptothecin in vitro. Expression of a human intestinal CE cDNA in COS-7 cells produced extracts that demonstrated proficient CPT-11 activation and conferred sensitivity of cells to CPT-11. These results suggest that gut toxicity from CPT-11 may be due in part to direct drug conversion by CEs present within the small intestine.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Hidrolases de Éster Carboxílico/metabolismo , Intestinos/enzimologia , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/metabolismo , Biotransformação , Células COS/citologia , Células COS/efeitos dos fármacos , Células COS/enzimologia , Camptotecina/metabolismo , Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/genética , Divisão Celular/efeitos dos fármacos , DNA Complementar/genética , DNA Complementar/isolamento & purificação , DNA Complementar/metabolismo , Humanos , Irinotecano , Fígado/enzimologia , Dados de Sequência Molecular , Coelhos , Homologia de Sequência de Aminoácidos , Transfecção
14.
Cancer Res ; 60(21): 6061-7, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085528

RESUMO

Matrix metalloproteinases (MMPs) are overexpressed in a variety of tumor tissues and cell lines, and their expression is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed two mutated anthrax toxin protective antigen (PA) proteins in which the furin protease cleavage site is replaced by sequences selectively cleaved by MMPs. These MMP-targeted PA proteins were activated rapidly and selectively on the surface of MMP-overexpressing tumor cells. The activated PA proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A. The toxicity of the mutated PA proteins for MMP-overexpressing cells was blocked by hydroxamate inhibitors of MMPs, including BB94, and by a tissue inhibitor of matrix metalloproteinases (TIMP-2). The mutated PA proteins killed MMP-overexpressing tumor cells while sparing nontumorigenic normal cells when these were grown together in a coculture model, indicating that PA activation occurred on the tumor cell surface and not in the supernatant. This method of achieving cell-type specificity is conceptually distinct from, and potentially synergistic with, the more common strategy of retargeting a protein toxin by fusion to a growth factor, cytokine, or antibody.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas/farmacocinética , Toxinas Bacterianas/toxicidade , Metaloproteinases da Matriz/metabolismo , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/genética , Sítios de Ligação , Biotransformação , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Células COS/enzimologia , Chlorocebus aethiops , Técnicas de Cocultura , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/enzimologia , Humanos , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/biossíntese , Metaloproteinases da Matriz Associadas à Membrana , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Metaloendopeptidases/biossíntese , Metaloendopeptidases/metabolismo , Mutação , Células Tumorais Cultivadas , Células Vero/enzimologia
15.
Cancer Res ; 56(20): 4735-42, 1996 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8840992

RESUMO

The gene for the bacterial enzyme carboxypeptidase G2 (CPG2) was expressed internally in mammalian cells. Mammalian-expressed CPG2 had kinetic properties indistinguishable from bacterially expressed CPG2. Human tumor cell lines A2780, SK-OV-3 (ovarian adenocarcinomas), LS174T, and WiDr (colon carcinomas) were engineered to express constitutively either CPG2 or bacterial beta-galactosidase. These cell lines were subjected to a gene-directed enzyme prodrug therapy regime, using the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA). The lines which expressed CPG2 had enhanced sensitivity to CMDA. Comparing IC50S, WiDr-CPG2 and SK-OV-3-CPG2 were 11-16-fold more sensitive, whereas A2780-CPG2 and LS174T-CPG2 were approximately 95-fold more sensitive than the corresponding control lines. CPG2-expressing cells and control cells were mixed in differing proportions and then treated with prodrug. Total kill occurred when only approximately 12% of cells expressed CPG2 with the WiDr and SK-OV-3 lines and when only 4-5% of cells expressed CPG2 with the LS174T and A2780 lines, indicating a substantial bystander effect. These results establish this CPG2 enzyme/CMDA prodrug system as an effective combination for the gene-directed enzyme prodrug therapy approach.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Colo/enzimologia , Neoplasias do Colo/terapia , Terapia Genética/métodos , Glutamatos/uso terapêutico , Compostos de Mostarda Nitrogenada/uso terapêutico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/terapia , Pró-Fármacos/uso terapêutico , gama-Glutamil Hidrolase/genética , Animais , Antibacterianos/farmacologia , Células COS/enzimologia , Neoplasias do Colo/genética , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Vetores Genéticos/genética , Gentamicinas/farmacologia , Humanos , Mutagênese Sítio-Dirigida , Neoplasias Ovarianas/genética , Transfecção , Células Tumorais Cultivadas , gama-Glutamil Hidrolase/análise , gama-Glutamil Hidrolase/biossíntese
16.
Cancer Res ; 58(12): 2646-51, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9635592

RESUMO

We have isolated a cDNA encoding a rabbit carboxylesterase (CE; EC 3.1.1.1) that converts the camptothecin-derived prodrug irinotecan (CPT-11) to the potent topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin. NH2-terminal amino acid sequencing of a purified rabbit CE allowed the design of redundant oligonucleotides to perform PCR from rabbit liver cDNA. DNA sequencing of the PCR product confirmed the identity of the clone, and after both 5' and 3' rapid amplification of cDNA ends, oligonucleotide primers were designed to amplify the entire cDNA. The 1698-bp open reading frame encoded a 565-amino acid protein containing the characteristic CE B-1 and B-2 motifs, a hydrophobic NH2-terminal leader sequence, and the COOH-terminal residues HIEL that are thought to be responsible for protein localization in the endoplasmic reticulum. Transient expression of the cDNA in COS-7 cells resulted in CE activity in cell extracts and increased the sensitivity of cells to CPT-11. Additionally, stable expression of the rabbit liver CE cDNA in the human glioma U-373 MG cell line resulted in a 56-fold decrease in the IC50 value for CPT-11, whereas the expression of a human alveolar macrophage cDNA encoding a highly homologous CE produced no change in drug sensitivity.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/genética , DNA Complementar/isolamento & purificação , Pró-Fármacos/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Sequência de Bases , Células COS/efeitos dos fármacos , Células COS/enzimologia , Camptotecina/química , Camptotecina/metabolismo , Camptotecina/farmacologia , Carboxilesterase , Hidrolases de Éster Carboxílico/isolamento & purificação , Hidrolases de Éster Carboxílico/metabolismo , Sobrevivência Celular/efeitos dos fármacos , DNA Complementar/química , Glioma/enzimologia , Humanos , Irinotecano , Fígado/enzimologia , Dados de Sequência Molecular , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Coelhos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Oncogene ; 13(6): 1323-33, 1996 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-8808707

RESUMO

PEA3, a member of the Ets family of transcriptional regulatory proteins, binds to the PEA3 promoter element and stimulates transcription through this site. The activity of the PEA3 element is regulated by mitogens, activated receptor tyrosine kinases, and oncogenic members of the Ras signal transduction pathway. However, it is not clear whether PEA3 mediates transcriptional regulation by these agents because a number of different Ets proteins can functionally interact with the PEA3 element. To specifically learn whether the activity of PEA3 is regulated, we investigated the ability of constitutively-activated Ras (Ha-RasV12) and signaling proteins downstream of Ras to alter PEA3-dependent reporter gene expression in COS cells. Ha-RasV12 and activated proteins in both the extra-cellular regulated kinase (ERK) and the stress-activated protein kinase (SAPK) or Jun N-terminal kinase (JNK) cascades independently stimulated PEA3-mediated gene expression. Ha-RasV12 stimulation of PEA3 activity was reduced by dominant-negative mutants in each of these protein kinase cascades, suggesting that Ras activates PEA3 through both pathways. Furthermore, the ability of unique activators of each kinase cascade to stimulate PEA3-dependent gene expression was selectively reduced by dominant-negative mutants within the homologous but not the heterologous pathway. Hence two distinct mitogen-activated protein kinase (MAPK) cascades regulate PEA3 activity. PEA3 was phosphorylated in vivo at serine residues consistent with the possibility that it may be a direct target of MAPKs.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno , Fatores de Transcrição/fisiologia , Animais , Células COS/enzimologia , Células COS/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Ativação Enzimática , Expressão Gênica , Genes ras/fisiologia , MAP Quinase Quinase 1 , MAP Quinase Quinase 4 , Fosforilação , Biossíntese de Proteínas , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-raf , Serina/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Proteínas ras/fisiologia
18.
Oncogene ; 17(9): 1079-87, 1998 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-9764818

RESUMO

The RET proto-oncogene encodes two isoforms of a receptor tyrosine kinase which plays a role in neural crest and kidney development. Ret ligands have been recently identified as the neuron survival factor GDNF (Glial-Derived Neurotrophic Factor) and Neurturin. Somatic rearrangements of RET, designated RET/PTCs, have been frequently detected in papillary thyroid carcinomas. In addition, distinct germ-line mutations of RET gene have been associated with the inherited cancer syndromes MEN (Multiple Endocrine Neoplasia) 2A, 2B and FMTC (Familial Medullar Thyroid Carcinomas) as well as with the congenital megacolon or Hirschsprung's disease, thus enlightening a significant role of this receptor gene in diverse human pathologic conditions. In this study, by performing classical inhibition experiments using synthetic phosphopeptides and by site-directed mutagenesis of the putative docking site, we have determined that for Grb2 the latter is provided by the tyrosine 620 of Ret/ptc2 long isoform (corresponding to Tyr 1096 on proto-Ret). However, in intact cells, the interaction of Grb2 with the two short and long Ret isoforms expressed separately is of similar strength, thus suggesting that Ret short isoform interaction with Grb2 could be mediated not only by Shc but also by a molecule that binds preferentially to this isoform. This possibility is supported by the evidence that the mutant Ret/ptc2Y620F long isoform displays a weak coimmunoprecipitation with Grb2 and that this mutant, lacking the docking site for Grb2 but owing all the others phosphotyrosines, surprisingly displays a reduced transforming activity compared to that of the two WTs oncogenes. We thus conclude that in intact cells both Ret isoforms bind to Grb2, although with different modalities. In addition, the present results are in agreement with the possibility that different signal transduction pathways are associated with the two isoforms of Ret.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Proteínas de Drosophila , Isoenzimas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células 3T3/citologia , Células 3T3/enzimologia , Células 3T3/metabolismo , Substituição de Aminoácidos/genética , Animais , Sítios de Ligação , Células COS/citologia , Células COS/enzimologia , Células COS/metabolismo , Extratos Celulares/química , Clonagem Molecular , Proteína Adaptadora GRB2 , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Glutationa Transferase/genética , Peptídeos e Proteínas de Sinalização Intracelular , Isoenzimas/genética , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fenilalanina , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas/química , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Tirosina/genética , Domínios de Homologia de src
19.
Oncogene ; 19(37): 4221-9, 2000 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10980595

RESUMO

Ribosomal S6 kinases (RSKs) are serine/threonine kinases activated by mitogenic signals through the Mitogen-Activated Protein Kinases/Extracellular Signal-Regulated Kinases (MAPK/ERK). RSKs contain two heterologous complete protein kinase domains. Phosphorylation by ERK of the C-terminal kinase domain allows activation of the N-terminal kinase domain, which mediates substrate phosphorylation. In human, there are three isoforms of RSK (RSK1, RSK2, RSK3), whose functional specificity remains undefined. Importantly, we have shown that mutations in the RSK2 gene lead to the Coffin-Lowry syndrome (CLS). In this study, we characterize two monoclonal antibodies raised against phosphorylated forms of the N- and C-terminal domain of RSK2 (P-S227 and P-T577, respectively). Using these two antibodies, we show that stress signals, such as UV light, induce phosphorylation and activation of the three RSKs to an extent which is comparable to Epidermal Growth Factor (EGF)-mediated activation. The use of specific kinase inhibitors indicates that UV-induced phosphorylation and activation of RSK2 is mediated by the MAPK/ERK pathway, but that the Stress-Activated Protein Kinase 2 (SAPK2)/p38 pathway is also involved. These results modify the view of RSKs as kinases restricted to the mitogenic response and reveal a previously unappreciated role of MAPKs in stress induced signaling. Oncogene (2000) 19, 4221 - 4229


Assuntos
Isoenzimas/efeitos da radiação , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Processamento de Proteína Pós-Traducional/efeitos da radiação , Proteínas Quinases S6 Ribossômicas/efeitos da radiação , Estresse Fisiológico/fisiopatologia , Raios Ultravioleta , Células 3T3/enzimologia , Células 3T3/efeitos da radiação , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Células COS/enzimologia , Células COS/efeitos da radiação , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/enzimologia , Fibroblastos/efeitos da radiação , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Dados de Sequência Molecular , Fosforilação/efeitos da radiação , Estrutura Terciária de Proteína , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/imunologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
20.
Biochim Biophys Acta ; 1502(3): 433-46, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11068186

RESUMO

We have systematically investigated the molecular defects resulting in a primary lipoprotein lipase (LPL) deficiency in a Japanese male infant (proband SH) with fasting hyperchylomicronemia. Neither LPL activity nor immunoreactive LPL mass was detected in pre- or postheparin plasma from proband SH. DNA sequence analysis of the LPL gene of proband SH revealed homozygosity for a novel missense mutation of F270L (Phe(270)-->Leu/TTT(1065)-->TTG) in exon 6. The function of the mutant F270L LPL was determined by both biochemical and immunocytochemical studies. In vitro expression experiments on the mutant F270L LPL cDNA in COS-1 cells demonstrated that the mutant LPL protein was synthesized as a catalytically inactive form and its total amount was almost equal to that of the normal LPL. Moreover, the synthesized mutant LPL was non-releasable by heparin because the intracellular transport of the mutant LPL to the cell surface - by which normal LPL becomes heparin-releasable - was impaired due to the abnormal structure of the mutant LPL protein. These findings explain the failure to detect LPL activities and masses in pre- and postheparin plasma of the proband. The mutant F270L allele generated an XcmI restriction enzyme site in exon 6 of the LPL gene. The carrier status of F270L in the proband's family members was examined by digestion with XcmI. The proband was ascertained to be homozygous for the F270L mutation and his parents and sister were all heterozygous. The LPL activities and masses of the parents and the sister (carriers) were half or less than half of the control values. Regarding the phenotype of the carriers, the mother with a sign of hyperinsulinemia manifested hypertriglyceridemia (type IV hyperlipoproteinemia), whereas the healthy father and the sister were normolipidemic. Hyperinsulinemia may be a strong determinant of hypertriglyceridemia in subjects with heterozygous LPL deficiency.


Assuntos
Hiperlipoproteinemias/genética , Lipase Lipoproteica/deficiência , Mutação de Sentido Incorreto , Alelos , Animais , Células COS/enzimologia , Heterozigoto , Humanos , Hiperlipoproteinemias/sangue , Hiperlipoproteinemias/enzimologia , Imuno-Histoquímica , Recém-Nascido , Lipase/análise , Lipase/sangue , Lipase Lipoproteica/sangue , Lipase Lipoproteica/genética , Fígado/enzimologia , Masculino , Linhagem , Mapeamento por Restrição , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA