Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 360
Filtrar
1.
Verh K Acad Geneeskd Belg ; 71(1-2): 43-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19739397

RESUMO

In 1963 I started the Laboratory of Molecular Biology (LMB) at the University of Ghent. Molecular Biology was then a new scientific discipline. Nucleic acids (NA) could be sequenced, manipulated, and recombined to form genetic information which never before had existed. Cloning of DNA-segments allowed multiplying a single molecule a billion-fold. By 1975 recombinant DNA-technology had sufficiently progressed that one could start the pursuit of a medically important goal. Our choice was to go for Interferon, a mysterious substance which could protect against viral infection, and might possibly be used as an anti-cancer agent if available in unlimited quantities. Fortunately, Piet De Somer, then Rector of the KUL, was one of the pioneers in interferon research. He encouraged his young colleague Erik De Clercq to collaborate with us. Erik brought extensive interferon expertise and reagents to the collaboration. But molecular biologists work with NA and interferon is a protein. There was a missing link which was provided by Jean Content of the Brussels Pasteur Institute. Jean had developed a system to convert interferon mRNA into protein, which was send to Leuven for quantification. This close collaboration between 3 laboratories led in January 1980 to the cloning of the human fibroblast, now interferon-beta, gene, and to the primary structure of the protein. However, 2 months earlier, Tadatsugu Taniguchi had succeeded already to obtain such a clone. But we were the first to express the clone in E. coli, and this was the definite proof that the cloned gene coded for human interferon-beta.


Assuntos
Clonagem Molecular , Expressão Gênica , Interferon beta/genética , DNA Recombinante , Humanos , Biologia Molecular
2.
Protein Expr Purif ; 23(2): 226-32, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11676596

RESUMO

The p55 tumor necrosis factor receptor (TNF-RI) is the main receptor by which TNF exerts its effects. The signaling capacity largely depends on the presence of an intact C-terminal protein-protein interaction domain, a so-called death domain (DD). Here we report the expression and purification of the human TNF-RI DD as a fusion with the Escherichia coli thioredoxin A (TRX) protein. When expressed under control of the bacteriophage T7 promoter, TRX-DD accumulates as a soluble protein in the cytoplasm of E. coli. The TRX-DD protein was released from the cells into the periplasmic fraction after osmotic shock. Due to self-association of the DD, a large part of the material appeared as multimers; it could be removed by selective precipitation and a combination of ion-exchange and size-exclusion chromatography. This purification protocol yielded 30 mg of purified, monomeric protein from 1 liter of shake-flask culture. The purified TRX-DD was found to be functional as it still bound to the TNF-RI-associated DD protein and the intracellular part of TNF-RI. We conclude that TRX-DD is correctly folded and can be used for further structure/function analysis.


Assuntos
Antígenos CD/genética , Receptores do Fator de Necrose Tumoral/genética , Antígenos CD/química , Antígenos CD/isolamento & purificação , Clonagem Molecular , Escherichia coli , Humanos , Estrutura Terciária de Proteína , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/isolamento & purificação , Receptores Tipo I de Fatores de Necrose Tumoral , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Tiorredoxinas/genética
3.
Cell Death Differ ; 8(8): 829-40, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11526436

RESUMO

In L929sAhFas cells, tumor necrosis factor (TNF) leads to necrotic cell death, whereas agonistic anti-Fas antibodies elicit apoptotic cell death. Apoptosis, but not necrosis, is correlated with a rapid externalization of phosphatidylserine and the appearance of a hypoploid population. During necrosis no cytosolic and organelle-associated active caspase-3 and -7 fragments are detectable. The necrotic process does not involve proteolytic generation of truncated Bid; moreover, no mitochondrial release of cytochrome c is observed. Bcl-2 overexpression slows down the onset of necrotic cell death. In the case of apoptosis, active caspases are released to the culture supernatant, coinciding with the release of lactate dehydrogenase. Following necrosis, mainly unprocessed forms of caspases are released. Both TNF-induced necrosis and necrosis induced by anti-Fas in the presence of the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone are prevented by the serine protease inhibitor N-tosyl-L-phenylalanine chloromethylketone and the oxygen radical scavenger butylated hydroxyanisole, while Fas-induced apoptosis is not affected.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Mitocôndrias/metabolismo , Necrose , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspases/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Humanos , Cinética , Camundongos , Mitocôndrias/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/efeitos dos fármacos , Receptor fas/metabolismo
4.
Verh K Acad Geneeskd Belg ; 63(2): 153-7; discussion 158-60, 2001.
Artigo em Holandês, Inglês | MEDLINE | ID: mdl-11441855
5.
J Biol Chem ; 276(40): 37426-30, 2001 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-11448951

RESUMO

The bioactivity of tumor necrosis factor (TNF) is mediated by two TNF receptors (TNF-Rs), more particularly TNF-RI and TNF-RII. Although human TNF (hTNF) and murine TNF (mTNF) are very homologous, hTNF binds only to mTNF-RI. By measuring the binding of a panel of mTNF/hTNF chimeras to both mTNF-R, we pinpointed the TNF region that mediates the interaction with mTNF-RII. Using site-specific mutagenesis, we identified amino acids 71-73 and 89 as the main interacting residues. Mutein hTNF-S71D/T72Y/H73 Delta/T89E interacts with both types of mTNF-R and is active in CT6 cell proliferation assays mediated by mTNF-RII. Mutein mTNF-D71S/Y72T/Delta 73H/E89T binds to mTNF-RI only and is no longer active on CT6 cells. However, the L929s cytotoxicity of this mutein (an effect mediated by mTNF-RI triggering) was also 100-fold lower than that of wild-type mTNF due to enhanced dissociation during incubation at subnanomolar concentrations. The additional mutation of amino acid 102, resulting in the mutein mTNF-D71S/Y72T/Delta 73H/E89T/P102Q, restored the trimer stability, which led to an enhanced specific activity on L929s cells. Hence the specific activity of a TNF species is governed not only by its receptor binding characteristics but also by its trimer stability after incubation at subnanomolar concentrations. In conclusion, the mutation of TNF amino acids 71-73, 89, and 102 is sufficient to obtain a mTNF mutein selective for mTNF-RI and a hTNF mutein that, unlike wild-type hTNF, also acts on mTNF-RII.


Assuntos
Antígenos CD/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Cromatografia em Gel , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/genética
6.
J Biol Chem ; 276(29): 27098-103, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11371574

RESUMO

Incubation of murine tumor necrosis factor (mTNF) at subnanomolar concentrations results in partial dissociation of the trimers, coinciding with a decrease in bioactivity. Using size-exclusion chromatography, we observed that the conversion of labeled mTNF to monomers is not only prevented by coincubation with an excess of unlabeled mTNF but also with unlabeled human TNF (hTNF). Moreover, after coincubation of mTNF and hTNF four different TNF complexes were revealed by native polyacrylamide gel electrophoresis, viz. homotrimeric mTNF and hTNF, as well as two complexes with an intermediate migration pattern. Analytical gel filtration in combination with native polyacrylamide gel electrophoresis and Western blot immunodetection indicated that these new complexes consisted of heterotrimeric TNF molecules. We conclude that an exchange of monomers takes place during coincubation of two different species of TNF, which results in homotrimeric and heterotrimeric TNF. To assess receptor interaction in vitro, TNF heterotrimeric molecules were used as obtained after incubation of mTNF with labeled hTNF (which only binds to mTNF receptor I) or with labeled mutein mTNF75 (specific for mTNF receptor II). These heterotrimers were retained by both mTNF receptors, which means that the mTNF subunits incorporated in heterotrimeric complexes still can bind to both types of TNF receptor. In addition, the gradual decrease in mTNF bioactivity during preincubation at subnanomolar concentrations was prevented by the presence of mutein mTNF75, which is inactive in an L929 cytotoxicity assay, indicating that heterotrimerization can influence the overall bioactivity.


Assuntos
Fator de Necrose Tumoral alfa/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Biopolímeros , Cromatografia em Gel , Eletroforese em Gel de Poliacrilamida , Humanos , Camundongos , Especificidade da Espécie , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/isolamento & purificação
7.
J Immunol ; 165(12): 7050-7, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11120833

RESUMO

Due to their multispecificity and versatility, bispecific Abs (BsAbs) are promising therapeutic tools in tomorrow's medicine. Especially intermediate-sized BsAbs that combine body retention with tissue penetration are valuable for therapy but necessitate expression systems that favor heterodimerization of the binding sites for large-scale application. To identify heterodimerization domains to which single-chain variable fragments (scFv) can be fused, we compared the efficiency of heterodimerization of CL and CH1 constant domains with complete L and Fd chains in mammalian cells. We found that the isolated CL:CH1 domain interaction was inefficient for secretion of heterodimers. However, when the complete L and Fd chains were used, secretion of L:Fd heterodimers was highly successful. Because these Fab chains contribute a binding moiety, C-terminal fusion of a scFv molecule to the L and/or Fd chains generated BsAbs or trispecific Abs (TsAbs) of intermediate size (75-100 kDa). These disulfide-stabilized bispecific Fab-scFv ("bibody") and trispecific Fab-(scFv)(2) ("tribody") heterodimers represent up to 90% of all secreted Ab fragments in the mammalian expression system and possess fully functional binding moieties. Furthermore, both molecules recruit and activate T cells in a tumor cell-dependent way, whereby the trispecific derivative can exert this activity to two different tumor cells. Thus we propose the use of the disulfide-stabilized L:Fd heterodimer as an efficient platform for production of intermediate-sized BsAbs and TsAbs in mammalian expression systems.


Assuntos
Anticorpos Biespecíficos/biossíntese , Fragmentos Fab das Imunoglobulinas/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Adjuvantes Imunológicos/síntese química , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/farmacologia , Sítios de Ligação de Anticorpos/genética , Linhagem Celular , Citotoxicidade Imunológica/genética , Dimerização , Estabilidade de Medicamentos , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/metabolismo , Fragmentos Fab das Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/imunologia , Células Tumorais Cultivadas
8.
Immunity ; 13(2): 223-31, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10981965

RESUMO

Hypotension and shock observed in sepsis, SIRS, and tumor necrosis factor (TNF) or cytokine-based cancer treatment are the consequence of excessive nitric oxide (NO) production and subsequent soluble guanylate cyclase (sGC)-mediated vascular smooth muscle relaxation. We demonstrate here that, while NO synthase (NOS) inhibitors exacerbated toxicity, inhibitors of sGC activation protected against TNF-induced lethality, bradycardia, and hypotension. Importantly, sGC inhibition did not interfere with the antitumor activity of TNF. Using NOS inhibitors or iNOS-deficient animals, we furthermore observed that no protection against TNF toxicity could be obtained in the absence of NO. These data imply that iNOS- (and not eNOS-) derived NO is an endogenous protective molecule indispensable to survive a TNF challenge and exerting this beneficial effect via sGC-independent mechanisms.


Assuntos
Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Choque/metabolismo , Choque/prevenção & controle , Fator de Necrose Tumoral alfa/toxicidade , Animais , Inibidores Enzimáticos/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II
9.
J Biol Chem ; 275(48): 37596-603, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10988295

RESUMO

Tumor necrosis factor (TNF) induces a typical apoptotic cell death program in various cell lines by interacting with the p55 tumor necrosis factor receptor (TNF-R55). In contrast, triggering of the fibrosarcoma cell line L929sA gives rise to characteristic cellular changes resulting in necrosis. The intracellular domain of TNF-R55 can be subdivided into two parts: a membrane-proximal domain (amino acids 202-325) and a C-terminal death domain (DD) (amino acids 326-413), which has been shown to be necessary and sufficient for apoptosis. Structure/function analysis of TNF-R55-mediated necrosis in L929sA cells demonstrated that initiation of necrotic cell death, as defined by swelling of the cells, rapid membrane permeabilization, absence of nuclear condensation, absence of DNA hypoploidy, and generation of mitochondrial reactive oxygen intermediates, is also confined to the DD. The striking synergistic effect of the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone on TNF-induced necrosis was also observed with receptors solely containing the DD. TNF-R55-mediated necrosis is not affected by the dominant negative deletion mutant of the Fas-associated death domain (FADD-(80-205)) that lacks the N-terminal death effector domain. Moreover, overexpression of FADD-(80-205) in L929sA is cytotoxic and insensitive to CrmA, while the cytotoxicity due to overexpression of the deletion mutant FADD-(1-111) lacking the DD is prevented by CrmA. These results demonstrate that the death domain of FADD can elicit an active necrotic cell death pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Antígenos CD/química , Antígenos CD/metabolismo , Proteínas de Transporte/metabolismo , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Virais , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Proteína de Domínio de Morte Associada a Fas , Citometria de Fluxo , Humanos , Camundongos , Necrose , Receptores Tipo I de Fatores de Necrose Tumoral , Serpinas/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
10.
J Biol Chem ; 275(43): 33876-82, 2000 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-10913145

RESUMO

Tumor necrosis factor (TNF)-induced cell death in the fibrosarcoma cell line L929 occurs independently of caspase activation and cytochrome c release. However, it is dependent on mitochondria and is characterized by increased production of reactive oxygen intermediates that are essential to the death process. To identify signaling molecules involved in this TNF-induced, reactive oxygen intermediate-dependent cell death pathway, we performed a comparative study by two-dimensional gel electrophoresis of phosphoproteins from a mitochondria-enriched fraction derived from TNF-treated and control cells. TNF induced rapid and persistent phosphorylation of the phosphorylation-responsive regulator of the microtubule (MT) dynamics, oncoprotein 18 (Op18). By using induced overexpression of wild type Op18 and phosphorylation site-deficient mutants S25A/S38A and S16A/S63A in L929 cells, we show that TNF-induced phosphorylation on each of the four Ser residues of Op18 promotes cell death and that Ser(16) and Ser(63) are the primary sites. This hyperphosphorylation of Op18 is known to completely turn off its MT-destabilizing activity. As a result, TNF treatment of L929 cells induced elongated and extremely tangled microtubules. These TNF-induced changes to the MT network were also observed in cells overexpressing wild type Op18 and, to a lesser extent, in cells overexpressing the S25A/S38A mutant. No changes in the MT network were observed upon TNF treatment of cells overexpressing the S16A/S63A mutant, and these cells were desensitized to TNF-induced cell death. These findings indicate that TNF-induced MT stabilization is mediated by hyperphosphorylation of Op18 and that this promotes cell death. The data suggest that Op18 and the MT network play a functional role in transduction of the cell death signal to the mitochondria.


Assuntos
Proteínas dos Microtúbulos , Microtúbulos/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Imidazóis/farmacologia , Camundongos , Microtúbulos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosforilação , Piridinas/farmacologia , Estatmina , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
11.
FEBS Lett ; 465(1): 47-52, 2000 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-10620704

RESUMO

Time kinetics of phosphatidyl serine (PS) exposure were compared to other apoptotic parameters following different apoptotic stimuli. Our data indicate that anti-Fas treatment of L929sAhFas cells results in rapid exposure of PS, which precedes decrease in mitochondrial transmembrane potential (DeltaPsi(m)) and release of cytochrome c, indicating that PS exposure occurs independently of these mitochondrial events. Also during TNF-, etoposide- or staurosporine-mediated apoptosis in PC60 RI/RII cells, PS-positive cells were observed before they had a decreased DeltaPsi(m). However, during growth factor depletion-induced death of 32D cells, both phenomena seemed to occur at the same time.


Assuntos
Apoptose , Grupo dos Citocromos c/metabolismo , Mitocôndrias/metabolismo , Fosfatidilserinas/metabolismo , Animais , Linhagem Celular , Etoposídeo , Substâncias de Crescimento/deficiência , Humanos , Potenciais da Membrana , Camundongos , Estaurosporina , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa , Receptor fas/genética , Receptor fas/farmacologia
12.
Biofactors ; 10(2-3): 145-56, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10609876

RESUMO

TNF is produced during inflammation and induces, among other activities, cell death in sensitive tumour cells. We previously reported an increased generation of ROS in TNF-treated L929 fibrosarcoma cells prior to cell death. These ROS are of mitochondrial origin and participate in the cell death process. Presently, we focus on the identification of parameters that control ROS production and subsequent cytotoxicity. From the cytotoxic properties and susceptibility to scavenging of TNF-induced ROS as compared to pro-oxidant-induced ROS we conclude that TNF-mediated ROS generation and their lethal action are confined to the inner mitochondrial membrane. Oxidative substrates, electron-transport inhibitors, glutathione and thiol-reactive agents but also caspase inhibitors modulate TNF-induced ROS production and imply the existence of a negative regulator of ROS production. Inactivation of this regulator by a TNF-induced reduction of NAD(P)H levels and/or formation of intraprotein disulfides would be responsible for ROS generation.


Assuntos
Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Fibrossarcoma , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , NAD/metabolismo , NADP/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/fisiologia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
13.
Cytokine ; 11(8): 617-25, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10433810

RESUMO

Injection of tumour necrosis factor (TNF) in animals causes severe liver cell toxicity, especially when D-(+)-galactosamine (GalN) is co-administered. After challenge with TNF/GalN, serum complement activity (CH50 and APCH50) decreased dramatically, suggesting strong activation of both the classical and the alternative pathways. TNF or GalN alone had no such effect. A cleavage product of complement protein C3 [C3(b)] was deposited on the surface of hepatocytes of TNF/GalN-treated mice. Intravenous administration of cobra venom factor (CVF), which depletes complement, inhibited the development of hepatitis. However, CVF pretreatment also protected C3-deficient mice. Pretreatment of mice with a C1q-depleting antibody did not prevent TNF/GalN lethality, although the anti-C1q antibody had depleted plasma C1q. Factor B-deficient and C3-deficient mice, generated by gene targeting, proved to be as sensitive to TNF/GalN as control mice. Furthermore, induction of lethal shock by platelet-activating factor, an important mediator in TNF-induced hepatic failure, was not reduced in C3-deficient mice. These data indicate that complement, although activated, plays no major role in the generation of acute lethal hepatic failure in this model and that CVF-induced protection is independent of complement depletion.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/imunologia , Ativação do Complemento/imunologia , Fígado/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Ativação do Complemento/efeitos dos fármacos , Complemento C1q/análise , Complemento C3b/análise , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Venenos Elapídicos/farmacologia , Feminino , Galactosamina/toxicidade , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA
14.
Oncogene ; 18(29): 4182-90, 1999 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-10435631

RESUMO

A20 is a Cys2/Cys2 zinc finger protein which is induced by a variety of inflammatory stimuli and which has been characterized as an inhibitor of cell death by a yet unknown mechanism. In order to clarify its molecular mechanism of action, we used the yeast two-hybrid system to screen for proteins that interact with A20. A cDNA fragment was isolated which encoded a portion of a novel protein (TXBP151), which was recently found to be a human T-cell leukemia virus type-I (HTLV-I) Tax-binding protein. The full-length 2386 bp TXBP151 mRNA encodes a protein of 86 kDa. Like A20, overexpression of TXBP151 could inhibit apoptosis induced by tumour necrosis factor (TNF) in NIH3T3 cells. Moreover, transfection of antisense TXBP151 partially abolished the anti-apoptotic effect of A20. Furthermore, apoptosis induced by TNF or CD95 (Fas/APO-1) was associated with proteolysis of TXBP151. This degradation could be inhibited by the broad-spectrum caspase inhibitor zVAD-fmk or by expression of the cowpox virus-derived inhibitor CrmA, suggesting that TXBP151 is a novel substrate for caspase family members. TXBP151 was indeed found to be specifically cleaved in vitro by members of the caspase-3-like subfamily, viz. caspase-3, caspase-6 and caspase-7. Thus TXBP151 appears to be a novel A20-binding protein which might mediate the anti-apoptotic activity of A20, and which can be processed by specific caspases.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Neoplasias , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Proteínas Virais , Dedos de Zinco , Células 3T3 , Clorometilcetonas de Aminoácidos/farmacologia , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Proteínas de Transporte/isolamento & purificação , Linhagem Celular , Clonagem Molecular , Cisteína Endopeptidases , Inibidores de Cisteína Proteinase/farmacologia , DNA Complementar/genética , Proteínas de Ligação a DNA , Dactinomicina/farmacologia , Genes , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares , Inibidores da Síntese de Ácido Nucleico/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Ligação Proteica , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Serpinas/fisiologia , Especificidade por Substrato , Transfecção , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/fisiologia
15.
J Cell Biol ; 145(7): 1471-82, 1999 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-10385526

RESUMO

The zinc finger protein A20 is a tumor necrosis factor (TNF)- and interleukin 1 (IL-1)-inducible protein that negatively regulates nuclear factor-kappa B (NF-kappaB)-dependent gene expression. However, the molecular mechanism by which A20 exerts this effect is still unclear. We show that A20 does not inhibit TNF- induced nuclear translocation and DNA binding of NF-kappaB, although it completely prevents the TNF- induced activation of an NF-kappaB-dependent reporter gene, as well as TNF-induced IL-6 and granulocyte macrophage-colony stimulating factor gene expression. Moreover, NF-kappaB activation induced by overexpression of the TNF receptor-associated proteins TNF receptor-associated death domain protein (TRADD), receptor interacting protein (RIP), and TNF recep- tor-associated factor 2 (TRAF2) was also inhibited by expression of A20, whereas NF-kappaB activation induced by overexpression of NF-kappaB-inducing kinase (NIK) or the human T cell leukemia virus type 1 (HTLV-1) Tax was unaffected. These results demonstrate that A20 inhibits NF-kappaB-dependent gene expression by interfering with a novel TNF-induced and RIP- or TRAF2-mediated pathway that is different from the NIK-IkappaB kinase pathway and that is specifically involved in the transactivation of NF-kappaB. Via yeast two-hybrid screening, we found that A20 binds to a novel protein, ABIN, which mimics the NF-kappaB inhibiting effects of A20 upon overexpression, suggesting that the effect of A20 is mediated by its interaction with this NF-kappaB inhibiting protein, ABIN.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Quinases Ativadas por Mitógeno , NF-kappa B/metabolismo , Proteínas/metabolismo , Ativação Transcricional , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cisteína Endopeptidases , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA , Ativação Enzimática/efeitos dos fármacos , Produtos do Gene tax/genética , Produtos do Gene tax/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-6/genética , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Dados de Sequência Molecular , NF-kappa B/antagonistas & inibidores , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/química , Proteínas/genética , Proteína Serina-Treonina Quinases de Interação com Receptores , Transdução de Sinais/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Receptor de TNF , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF , Transativadores/genética , Transativadores/fisiologia , Ativação Transcricional/efeitos dos fármacos , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/farmacologia , Leveduras/genética , Dedos de Zinco , Proteínas Quinases p38 Ativadas por Mitógeno , Quinase Induzida por NF-kappaB
16.
Eur J Biochem ; 260(1): 166-75, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10091596

RESUMO

The A/Victoria/3/75 (H3N2-subtype) hemagglutinin (HA) gene was engineered for expression in Pichia pastoris as a soluble secreted molecule. The HA cDNA lacking the C-terminal transmembrane anchor-coding sequence was fused to the Saccharomyces cerevisiae alpha-mating factor secretion signal and placed under control of the methanol-inducible P. pastoris alcohol oxidase 1 (AOX1) promoter. Growth of transformants on methanol-containing medium resulted in the secretion of recombinant non-cleaved soluble hemagglutinin (HA0s). Remarkably, the pH of the induction medium had an important effect on the expression level, the highest level being obtained at pH 8.0. The gel filtration profile and the reactivity against a panel of different HA-conformation specific monoclonal antibodies indicated that HA0s was monomeric. Analysis of the N-linked glycans revealed a typical P. pastoris type of glycosylation, consisting of glycans with 10-12 glycosyl residues. Mice immunized with purified soluble hemagglutinin (HA0s) showed complete protection against a challenge with 10 LD50 of mouse-adapted homologous virus (X47), whereas all control mice succumbed. Heterologous challenge with X31 virus [A/Aichi/2/68 (H3N2-subtype)], resulted in significantly higher survival rates in the immunized group compared with the control group. These results, together with the safety, reliability and economic potential of P. pastoris, as well as the flexibility and fast adaptation of the expression system may allow development of an effective recombinant influenza vaccine.


Assuntos
Hemaglutininas Virais/imunologia , Imunização , Vacinas contra Influenza/imunologia , Orthomyxoviridae/imunologia , Oxirredutases do Álcool/genética , Amidoidrolases/metabolismo , Animais , Vetores Genéticos/genética , Glicosilação , Hemaglutininas Virais/genética , Vacinas contra Influenza/biossíntese , Fator de Acasalamento , Camundongos , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase , Peptídeos/genética , Pichia/genética , Pichia/metabolismo , Polissacarídeos/química , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Saccharomyces cerevisiae/genética , Proteínas Virais/genética , Proteínas Virais/imunologia
17.
Acta Crystallogr D Biol Crystallogr ; 55(Pt 4): 772-8, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10089307

RESUMO

The 1.4 A resolution structure of recombinant mouse tumour-necrosis factor alpha (mTNF) at 100 K has been determined. The crystals are triclinic, space group P1, with unit-cell parameters a = 48.06, b = 48.18, c = 51.01 A, alpha = 114.8, beta = 103.6, gamma = 91.1 degrees. The structure was refined to a final crystallographic R value of 19.7% (Rfree = 23.3%), including 3477 protein atoms, one 2-propanol molecule, one Tris molecule and 240 water molecules. Throughout the crystal lattice, the trimers are differently packed compared with human TNF, which was crystallized in the tetragonal space group P41212 and refined to 2.6 A resolution. The structures of mTNF and human TNF are very similar, diverging mainly in regions that are either flexible and/or involved in crystal packing. Some loops in mTNF which contain residues important for receptor binding are better resolved than in human TNF, such as the surface-exposed loops 30-34 and 144-147, which are also important for receptor specificity. Compared with human TNFs, the channel formed by the three monomers in mTNF is narrower. One 2-propanol molecule trapped in the trimeric channel could be a lead compound for the design of TNF inhibitors.


Assuntos
Fator de Necrose Tumoral alfa/química , Sequência de Aminoácidos , Animais , Antígenos CD/metabolismo , Cristalografia por Raios X , Humanos , Camundongos , Dados de Sequência Molecular , Conformação Proteica , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Especificidade por Substrato
18.
Genomics ; 55(3): 284-9, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10049582

RESUMO

Administration of recombinant murine tumor necrosis factor (TNF) to mice results in lethal shock, characterized by hypotension, hypothermia, and dramatic induction of cytokines released in the circulation, such as interleukin-6 (IL-6). The sensitivity of mice to the effects of murine TNF varies from strain to strain. DBA/2 mice were found to be considerably more resistant to TNF than C57BL/6 mice. The resistance proved to be dominant since (C57BL/6 x DBA/2)F1 mice were also resistant. Using BXD recombinant inbred mice and a dose of TNF lethal for C57BL/6 but not for DBA/2 mice, we found that the resistance to TNF links to loci coding for corticosteroid-binding globulin (Cbg), alpha1-protease inhibitor (Spi1), contrapsin (Spi2) and the contrapsin-regulating gene Spi2r that form a gene cluster on chromosome 12. Quantitative trait-loci analysis of TNF-induced induction of IL-6 and of hypothermia also points to the importance of this locus (P < 0.0002 and P = 0.017, respectively), more particularly the Cbg and Spi2 loci, in the resistance to TNF. We propose to name the locus "TNF protection locus." The data suggest that endogenous protease inhibitors and/or glucocorticoids play a significant role in the attenuation of TNF-induced lethal shock. This study also demonstrates that loci affecting important biological responses can be identified with very high resolution using recombinant inbred mice.


Assuntos
Fator de Necrose Tumoral alfa/toxicidade , Animais , Relação Dose-Resposta a Droga , Genótipo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Característica Quantitativa Herdável , Proteínas Recombinantes/farmacologia , Síndrome de Resposta Inflamatória Sistêmica/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
19.
J Immunol ; 162(2): 837-45, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9916706

RESUMO

TNF is known to regulate macrophage (Mphi) migration, but the signaling pathways mediating this response have not been established. Here we report that stimulation of the 55-kDa TNF receptor (TNFR-1) induced an overall decrease in filamentous actin (F-actin), inhibited CSF-1- and Cdc42-dependent filopodium formation, and stimulated macropinocytosis. Using a panel of TNFR-1 mutants, the regions of the receptor required for each of these responses were mapped. The decrease in F-actin required both the death domain and the membrane proximal part of the receptor, whereas inhibition of filopodium formation and increased pinocytosis were only dependent upon a functional death domain. When the TNF-induced decrease in F-actin was inhibited using either receptor mutants or the compound D609, TNF-stimulated actin reorganization at the cell cortex became apparent. This activity was dependent upon the FAN-binding region of TNFR-1. We conclude that different domains of TNFR-1 mediate distinct changes in the Mphi cytoskeleton, and that the ability of TNF to inhibit Mphi chemotaxis may be due to decreased filopodium formation downstream of Cdc42.


Assuntos
Actinas/fisiologia , Proteínas de Ciclo Celular/fisiologia , Inibição de Migração Celular , Proteínas de Ligação ao GTP/fisiologia , Macrófagos/fisiologia , Proteínas Quinases Ativadas por Mitógeno , Pseudópodes/fisiologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Animais , Antígenos CD/química , Antígenos CD/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Membrana Celular/fisiologia , Células Cultivadas , Proteínas de Ligação ao GTP/antagonistas & inibidores , Leucemia P388 , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Fragmentos de Peptídeos/fisiologia , Pinocitose/efeitos dos fármacos , Pseudópodes/enzimologia , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral , Acetato de Tetradecanoilforbol/farmacologia , Proteína cdc42 de Ligação ao GTP , Proteínas Quinases p38 Ativadas por Mitógeno
20.
Oncogene ; 18(54): 7719-30, 1999 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-10618712

RESUMO

Cell death is an essential phenomenon in normal development and homeostasis, but also plays a crucial role in various pathologies. Our understanding of the molecular mechanisms involved has increased exponentially, although it is still far from complete. The morphological features of a cell dying either by apoptosis or by necrosis are remarkably conserved for quite different cell types derived from lower or higher organisms. At the molecular level, several gene products play a similar, crucial role in a major cell death pathway in a worm and in man. However, one should not oversimplify. It is now evident that there are multiple pathways leading to cell death, and some cells may have the required components for one pathway, but not for another, or contain endogenous inhibitors which preclude a particular pathway. Furthermore, different pathways can co-exist in the same cell and are switched on by specific stimuli. Apoptotic cell death, reported to be non-inflammatory, and necrotic cell death, which may be inflammatory, are two extremes, while the real situation is usually more complex. We here review the distinguishing features of the various cell death pathways: caspases (cysteine proteases cleaving after particular aspartate residues), mitochondria and/or reactive oxygen species are often, but not always, key components. As these various caspase-dependent and caspase-independent cell death pathways are becoming better characterized, we may learn to differentiate them, fill in the many gaps in our understanding, and perhaps exploit the knowledge acquired for clinical benefit.


Assuntos
Apoptose , Morte Celular , Espécies Reativas de Oxigênio/fisiologia , Animais , Caspases/metabolismo , Humanos , Inflamação , Mitocôndrias/fisiologia , Necrose , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA