Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Brain Behav Immun ; 93: 156-171, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33444731

RESUMO

BACKGROUND: Myocardial infarction (MI) coinciding with depression worsens prognosis. Although Tumor Necrosis Factor alpha (TNF) is recognized to play a role in both conditions, the therapeutic potential of TNF inhibition is disappointing. TNF activates two receptors, TNFR1 and TNFR2, associated with opposite effects. Therefore, anti-inflammatory treatment with specific TNF receptor interference was compared to non-specific TNF inhibition regarding effects on heart, (neuro)inflammation, brain and behavior in mice with MI. METHODS: Male C57BL/6 mice were subjected to MI or sham surgery. One hour later, MI mice were randomized to either non-specific TNF inhibition by Enbrel, specific TNFR1 antagonist-, or specific TNFR2 agonist treatment until the end of the protocol. Control sham and MI mice received saline. Behavioral evaluation was obtained day 10-14 after surgery. Eighteen days post-surgery, cardiac function was measured and mice were sacrificed. Blood and tissue samples were collected for analyses of (neuro)inflammation. RESULTS: MI mice displayed left ventricular dysfunction, without heart failure, (neuro) inflammation or depressive-like behavior. Both receptor-specific interventions, but not Enbrel, doubled early post-MI mortality. TNFR2 agonist treatment improved left ventricular function and caused hyper-ramification of microglia, with no effect on depressive-like behavior. In contrast, TNFR1 antagonist treatment was associated with enhanced (neuro)inflammation: more plasma eosinophils and monocytes; increased plasma Lcn2 and hippocampal microglia and astrocyte activation. Moreover, increased baseline heart rate, with reduced beta-adrenergic responsiveness indicated sympathetic activation, and coincided with reduced exploratory behavior in the open field. Enbrel did not affect neuroinflammation nor behavior. CONCLUSION: Early receptor interventions, but not non-specific TNF inhibition, increased mortality. Apart from this undesired effect, the general beneficial profile after TNFR2 stimulation, rather than the unfavourable effects of TNFR1 inhibition, would render TNFR2 stimulation preferable over non-specific TNF inhibition in MI with comorbid depression. However, follow-up studies regarding optimal timing and dosing are needed.


Assuntos
Infarto do Miocárdio , Receptores Tipo I de Fatores de Necrose Tumoral , Animais , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/complicações , Receptores Tipo II do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa
2.
Cell Death Differ ; 14(12): 2021-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17703232

RESUMO

Variants of human TRAIL (hTRAIL) and human CD95L (hCD95L), encompassing the TNF homology domain (THD), interact with the corresponding receptors and stimulate CD95 and TRAILR2 signaling after cross-linking. The murine counterparts (mTRAIL, mCD95L) showed no or only low receptor binding and were inactive/poorly active after cross-linking. The stalk region preceding the THD of mCD95L conferred secondary aggregation and restored CD95 activation in the absence of cross-linking. A corresponding variant of mTRAIL, however, was still not able to activate TRAIL death receptors, but gained good activity after cross-linking. Notably, disulfide-bonded fusion proteins of the THD of mTRAIL and mCD95L with a subdomain of the tenascin-C (TNC) oligomerization domain, which still assembled into trimers, efficiently interacted with their cognate cellular receptors and robustly stimulated CD95 and TRAILR2 signaling after secondary cross-linking. Introduction of the TNC domain also further enhanced the activity of THD encompassing variants of hTRAIL and hCD95L. Thus, spatial fixation of the N-terminus of the THD appears necessary in some TNF ligands to ensure proper receptor binding. This points to yet unanticipated functions of the stalk and/or transmembrane region of TNF ligands for the functionality of these molecules and offers a broadly applicable option to generate recombinant soluble ligands of the TNF family with superior activity.


Assuntos
Proteína Ligante Fas/química , Proteína Ligante Fas/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/química , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Reagentes de Ligações Cruzadas/farmacologia , Humanos , Células Jurkat , Camundongos , Proteínas Mutantes/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade/efeitos dos fármacos , Relação Estrutura-Atividade , Tenascina/metabolismo
3.
Cell Death Differ ; 14(4): 765-74, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17053806

RESUMO

To achieve tumor cell-restricted activation of CD95, we developed a CD95L fusion protein format, in which CD95L activity is only unmasked upon antibody-mediated binding to tumor cells and subsequent processing by tumor-associated proteases, such as matrix metalloproteases (MMPs) and urokinase plasminogen activator (uPA). On target-negative, but MMP- and uPA-expressing HT1080 tumor cells, the CD95L prodrugs were virtually inactive. On target antigen-expressing HT1080 cells, however, the CD95L prodrugs showed an apoptotic activity comparable to soluble CD95L artificially activated by crosslinking. CD95 activation by the CD95L prodrugs was preceded by prodrug processing. Apoptosis was blocked by inhibitors of MMPs or uPA and by neutralizing antibodies recognizing the targeted cell surface antigen or the CD95L moiety of the prodrugs. In a xenotransplantation tumor model, local application of the prodrug reduced the growth of target antigen-expressing, but not antigen-negative tumor cells, verifying targeted CD95L prodrug activation in vivo.


Assuntos
Antineoplásicos/metabolismo , Apoptose/fisiologia , Biomarcadores Tumorais/metabolismo , Proteína Ligante Fas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Pró-Fármacos/metabolismo , Receptor fas/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Apoptose/efeitos dos fármacos , Membrana Celular/enzimologia , Proteína Ligante Fas/química , Proteína Ligante Fas/farmacologia , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/metabolismo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Proteínas Recombinantes , Testes de Toxicidade Crônica , Células Tumorais Cultivadas , Receptor fas/efeitos dos fármacos
4.
Oncogene ; 25(56): 7434-9, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16751802

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted considerable attention for its potential use in tumor therapy, as some recombinant variants of this ligand induce apoptosis in tumor cells without harming most normal cells. Here, we show that TRAIL strongly induces the expression of the proinflammatory cytokines interleukin-8 and monocyte chemoattractant protein 1 and enhances the invasion of apoptosis-resistant pancreatic ductal adenocarcinoma cells in vitro by upregulation of the urokinase-type plasminogen activator expression. Most importantly, we also demonstrate for the first time that TRAIL treatment results in strongly increased distant metastasis of pancreatic tumors in vivo. We orthotopically transplanted human pancreatic ductal adenocarcinoma cells to the pancreata of severe combined immunodeficiency mice and observed a dramatic increase in metastatic spread including a sixfold increase in the volume and fourfold increase in the number of liver metastases upon TRAIL treatment. Our results point to the necessity to carefully evaluate in vivo side effects of TRAIL and to select therapy conditions that not only enhance apoptosis induction but in addition prevent proinvasive and proinflammatory non-apoptotic TRAIL signaling.


Assuntos
Carcinoma Ductal Pancreático/patologia , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Animais , Humanos , Camundongos , Camundongos SCID
5.
Cell Death Differ ; 13(2): 273-84, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16052236

RESUMO

Tumor necrosis factor (TNF) prodrugs are fusion proteins comprised of an N-terminal single-chain antibody variable fragment (scFv) targeting a TNF effector and a C-terminal TNF receptor (TNFR)1-derived inhibitor module. Introduction of matrix metalloproteinase (MMP)-2 recognition motifs between TNF and the TNFR1 fragment allowed activation by recombinant MMP-2 and MMP-expressing HT1080 cells. Processing by endogeneous MMPs required specific membrane binding of the TNF prodrug via the targeting scFv, ensuring strictly antigen-dependent activation. Interestingly, TNF bioactivity of the processed prodrug was approximately 1000-fold higher upon scFv-mediated targeting, and signaled juxtatropic cell death also to antigen-negative cells. Microscopical analyses of TNFR2 clustering and TNF receptor-associated factor 2 recruitment at contact sites to adjacent cells revealed the formation of stable TNFR complexes by target-bound, processed prodrug, resembling the increased signal capacity of natural, membrane-expressed TNF. MMP-2-sensitive TNF prodrugs represent novel cytokine-based reagents for targeted cancer therapy, which should be exploitable for MMP-overexpressing tumors.


Assuntos
Morte Celular , Membrana Celular/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Pró-Fármacos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Biomarcadores Tumorais/análise , Western Blotting , Linhagem Celular Tumoral , Membrana Celular/química , Citometria de Fluxo , Humanos , Metaloproteinase 2 da Matriz/genética , Peptídeo Hidrolases/metabolismo , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Ligação Proteica , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Curr Biol ; 8(2): 113-6, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9427646

RESUMO

Fas/Apo1 and other cytotoxic receptors of the tumor necrosis factor receptor (TNFR) family contain a cytoplasmic death domain (DD) [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] that activates the apoptotic process by interacting with the DD-containing adaptor proteins TNFR-associated DD protein (TRADD) [12] [13] and Fas-associated DD protein (FADD/MORT1) [14] [15], leading to the activation of cysteine proteases of the caspase family [16]. Stimulation of Fas/Apo1 leads to the formation of a receptor-bound death-inducing signaling complex (DISC), consisting of FADD and two different forms of caspase-8 [17] [18] [19]. Transient expression of a dominant-negative mutant of FADD impairs TNFR60-mediated and Fas/Apo1-mediated apoptosis [13] [20], but has no effect on TNF-related apoptosis-inducing ligand (TRAIL/Apo2L)-induced cell death [7] [8] [9] [10] [21]. To study the function of FADD in DD-receptor signaling in more detail, we established HeLa cells that stably expressed a green fluorescent protein (GFP)-tagged dominant-negative mutant of FADD, GFP-DeltaFADD. Interestingly, expression of this mutant inhibited cell death induced by TNFR60, Fas/Apo1 and TRAIL-R/Apo2. In addition, GFP-DeltaFADD did not interfere with TNF-mediated gene induction or with activation of NF-kappaB or Jun N-terminal kinase (JNK), demonstrating that FADD is part of the TNFR60-initiated apoptotic pathway but does not play a role in TNFR60-mediated gene induction. Fas/Apo1-mediated activation of JNK was unaffected by the expression of GFP-DeltaFADD, suggesting that in Fas/Apo1 signaling the apoptotic pathway and the activation of JNK diverge at a level proximal to the receptor, upstream of or parallel to FADD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose , Proteínas de Transporte/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor fas/metabolismo , Proteínas de Transporte/genética , Proteína de Domínio de Morte Associada a Fas , Regulação da Expressão Gênica , Células HeLa , Humanos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ativação Transcricional
7.
Mol Cell Biol ; 21(12): 3964-73, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11359904

RESUMO

The caspase 8 homologue FLICE-inhibitory protein (cFLIP) is a potent negative regulator of death receptor-induced apoptosis. We found that cFLIP can be upregulated in some cell lines under critical involvement of the NF-kappaB pathway, but NF-kappaB activation was clearly not sufficient for cFLIP induction in all cell lines. Treatment of SV80 cells with the proteasome inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132) or geldanamycin, a drug interfering with tumor necrosis factor (TNF)-induced NF-kappaB activation, inhibited TNF-induced upregulation of cFLIP. Overexpression of a nondegradable IkappaBalpha mutant (IkappaBalpha-SR) or lack of IkappaB kinase gamma expression completely prevented phorbol myristate acetate-induced upregulation of cFLIP mRNA in Jurkat cells. These data point to an important role for NF-kappaB in the regulation of the cFLIP gene. SV80 cells normally show resistance to TNF-related apoptosis-inducing ligand (TRAIL) and TNF, as apoptosis can be induced only in the presence of low concentrations of cycloheximide (CHX). However, overexpression of IkappaBalpha-SR rendered SV80 cells sensitive to TRAIL-induced apoptosis in the absence of CHX, and cFLIP expression was able to reverse the proapoptotic effect of NF-kappaB inhibition. Western blot analysis further revealed that cFLIP, but not TRAF1, A20, and cIAP2, expression levels rapidly decrease upon CHX treatment. In conclusion, these data suggest a key role for cFLIP in the antiapoptotic response of NF-kappaB activation.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas I-kappa B , Peptídeos e Proteínas de Sinalização Intracelular , NF-kappa B/metabolismo , Anticorpos Monoclonais/farmacologia , Apoptose , Benzoquinonas , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Antígenos CD40/imunologia , Proteínas de Transporte/genética , Linhagem Celular , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Interleucina-1/farmacologia , Células Jurkat , Lactamas Macrocíclicas , Leupeptinas/farmacologia , Mutação , Inibidor de NF-kappaB alfa , Quinonas/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
9.
Cytokine Growth Factor Rev ; 10(1): 15-26, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10379909

RESUMO

Just four years ago the first two members of a new family of molecules involved in signal transduction by members of the TNF receptor superfamily were described and designated TNF Receptor Associated Factors (TRAFs). In the meantime six human and murine TRAFs as well as a TRAF protein from C. elegans have been molecularly cloned. From our current point of view, TRAF proteins appear to represent multifunctional signal adaptors, tightly embedded in a network of signals culminating in the activation of kinase cascades that finally lead to the activation of c-Jun N-terminal kinase. p38 mitogen activated protein kinase, and the transcription factor NF-kappaB, thereby also affecting the balance between survival and cell death. Some of the activities of the individual TRAF family members may be redundant although transgenic knockout animal models have already shown that crucial signaling pathways for single TRAF molecules in vivo can be defined.


Assuntos
Citocinas/fisiologia , Família Multigênica , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Humanos , Dados de Sequência Molecular , Receptores do Fator de Necrose Tumoral/genética , Homologia de Sequência de Aminoácidos , Proteínas da Matriz Viral
10.
Oncogene ; 20(5): 571-80, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11313989

RESUMO

One of the main functions of the tumor suppressor p53 is the induction of programmed cell death. Here we investigated in detail the molecular mechanisms that underlay p53 transactivation-dependent apoptosis in the human colon cancer cell line DLD-1. Although p53 upregulated the death receptors Fas, TRAIL-R1 and TRAIL-R2 in this cell line, p53-induced cell death occurred without detectable caspase-8 activation whereas, activation of caspase-9 and caspase-3 was readily observed. In addition to the upregulation of death receptors, p53 induced the pro-apoptotic Bcl-2 family members Bik and Bak and downregulated the anti-apoptotic Bcl-xL protein. Moreover, in RNase protection assay analyses as well as in reporter gene analyses we found a p53-dependent upregulation of the death receptor-inhibitory protein cFLIP. Together, these data argue for a p53-mediated activation of the mitochondrial pathway of apoptosis. In contrast to recently published data obtained in different cellular systems, there was no evidence for an essential role of NF-kappaB in p53-induced cell death. Moreover, induction of p53 interfered with TNF-induced NF-kappaB activation independently from apoptosis-induction.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/genética , Caspases/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , NF-kappa B/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Sequência de Bases , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/biossíntese , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , NF-kappa B/genética , Ativação Transcricional/fisiologia , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Regulação para Cima
11.
Oncogene ; 20(30): 4101-6, 2001 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11494138

RESUMO

TNF-related apoptosis-inducing ligand (TRAIL) is a typical member of the tumor necrosis factor (TNF) ligand family that is expressed as a type II membrane protein (memTRAIL) and signals apoptosis via the death domain-containing receptors TRAIL-R1 and -2. Soluble recombinant derivatives of TRAIL (sTRAIL) are considered as novel tumors therapeutics because of their selective apoptosis inducing activity in a variety of human tumors but not in normal cells. Using antagonistic antigen-binding fragment (Fab) preparations of TRAIL-R1- and TRAIL-R2-specific antibodies, we demonstrate in this study that TRAIL-R1 becomes activated by both the soluble and the membrane-bound form of the ligand, whereas TRAIL-R2 becomes only activated by memTRAIL or soluble TRAIL secondarily cross-linked by antibodies. Furthermore, we show that the restricted signal capacity of sTRAIL can be readily converted into a fully signal competent memTRAIL-like molecule, i.e. a TRAIL-R2 stimulating ligand, by genetic fusion to an antibody derivative that allows antigen-dependent 'immobilization' of the fusion protein to cell surfaces. We conclude that antibody targeting-dependent activation can be used to design selective therapeutics derived of those ligands of the TNF family that are biologically inactive in their soluble form.


Assuntos
Antígenos de Superfície/imunologia , Glicoproteínas de Membrana/farmacologia , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Especificidade de Anticorpos , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Células COS , Chlorocebus aethiops , Desenho de Fármacos , Células HeLa/efeitos dos fármacos , Humanos , Fragmentos Fab das Imunoglobulinas , Células Jurkat/efeitos dos fármacos , Células KB/efeitos dos fármacos , Ligantes , Glicoproteínas de Membrana/química , Proteínas de Membrana/química , Proteínas de Membrana/farmacologia , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/fisiologia , Estrutura Terciária de Proteína , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/fisiologia , Solubilidade , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/química
12.
Oncogene ; 19(40): 4563-73, 2000 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11030145

RESUMO

Caspase-8 plays an essential role in apoptosis triggered by death receptors. Through the cleavage of Bid, a proapoptotic Bcl-2 member, it further activates the mitochondrial cytochrome c/Apaf-1 pathway. Because caspase-8 can be processed also by anticancer drugs independently of death receptors, we investigated its exact role and order in the caspase cascade. We show that in Jurkat cells either deficient for caspase-8 or overexpressing its inhibitor c-FLIP apoptosis mediated by CD95, but not by anticancer drugs was inhibited. In the absence of active caspase-8, anticancer drugs still induced the processing of caspase-9, -3 and Bid, indicating that Bid cleavage does not require caspase-8. Overexpression of Bcl-x(L) prevented the processing of caspase-8 as well as caspase-9, -6 and Bid in response to drugs, but was less effective in CD95-induced apoptosis. Similar responses were observed by overexpression of a dominant-negative caspase-9 mutant. To further determine the order of caspase-8 activation, we employed MCF7 cells lacking caspase-3. In contrast to caspase-9 that was cleaved in these cells, anticancer drugs induced caspase-8 activation only in caspase-3 transfected MCF7 cells. Thus, our data indicate that, unlike its proximal role in receptor signaling, in the mitochondrial pathway caspase-8 rather functions as an amplifying executioner caspase.


Assuntos
Antineoplásicos/farmacologia , Apoptose/fisiologia , Caspases/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Mitocôndrias/fisiologia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Clorometilcetonas de Aminoácidos/farmacologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/biossíntese , Caspases/deficiência , Caspases/genética , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Etoposídeo/farmacologia , Humanos , Células Jurkat/efeitos dos fármacos , Células Jurkat/enzimologia , Mitomicina/farmacologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia , Proteína bcl-X , Receptor fas/fisiologia
13.
Cell Death Differ ; 10(1): 45-65, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12655295

RESUMO

A single mouse click on the topic tumor necrosis factor (TNF) in PubMed reveals about 50,000 articles providing one or the other information about this pleiotropic cytokine or its relatives. This demonstrates the enormous scientific and clinical interest in elucidating the biology of a molecule (or rather a large family of molecules), which began now almost 30 years ago with the description of a cytokine able to exert antitumoral effects in mouse models. Although our understanding of the multiple functions of TNF in vivo and of the respective underlying mechanisms at a cellular and molecular level has made enormous progress since then, new aspects are steadily uncovered and it appears that still much needs to be learned before we can conclude that we have a full comprehension of TNF biology. This review shortly covers some general aspects of this fascinating molecule and then concentrates on the molecular mechanisms of TNF signal transduction. In particular, the multiple facets of crosstalk between the various signalling pathways engaged by TNF will be addressed.


Assuntos
Apoptose/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas/metabolismo , Fator 1 Associado a Receptor de TNF , Fator 2 Associado a Receptor de TNF
14.
Mech Dev ; 100(1): 109-13, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11118894

RESUMO

TNF-receptor associated factors (TRAFs) comprise a family of adaptor proteins that act as downstream signal transducers of the TNF receptor superfamily and the Toll/interleukin-1 receptor family. The mammalian TRAFs 2, 5 and 6 are known to activate JNK- and NF-kappaB signaling pathways, whereas the function of the other three mammalian family members, TRAF 1, 3 and 4 is less well characterized. Vertebrate TRAFs have a very similar structure with the exception of TRAF1: aside the characteristic C-terminal TRAF domain, they share a N-terminal RING finger followed by five or, in the case of TRAF4, seven regularly spaced zinc fingers. Two TRAF homologues are present in the genome of Drosophila melanogaster, DTRAF1 and DTRAF2 (also known as DTRAF6) and both have been implicated in the Toll-receptor pathways leading to the activation of NF-kappa B and JNK. DTRAF1 is most closely related to mammalian TRAF4 which is predominantly expressed during nervous system development and in ephitelial progenitor cells. In order to gain insight into possible roles of DTRAF1 during development, we have performed a detailed transcriptional analysis of the gene at various embryonic and larval stages.


Assuntos
Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero/metabolismo , Larva/metabolismo , Biossíntese de Proteínas , Proteínas/genética , Animais , Blastoderma/metabolismo , DNA Complementar/metabolismo , Mesoderma/metabolismo , Modelos Genéticos , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator 1 Associado a Receptor de TNF , Fatores de Tempo , Transcrição Gênica
15.
Plant Physiol ; 109(4): 1231-1238, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12228664

RESUMO

Using high-performance liquid chromatography and nuclear magnetic resonance we identified vicianin as the cyanogenic compound of Phlebodium aureum. The (R)-hydroxynitrile lyase involved during cyanogenesis in the catabolism of the aglycon ([R]-mandelonitrile) was purified to apparent homogeneity. The purified holoenzyme is a homomultimer with subunits of Mr = 20,000. At least three isoforms of the enzyme exist. In contrast to other hydroxynitrile lyases, mandelonitrile lyase (MDL) from P. aureum was not inhibited by sulfhydryl- or hydroxyl-modifying reagents, suggesting a different catalytic mechanism. The enzyme is active over a broad temperature range, with maximum activity between 35 and 50[deg]C, and a pH optimum at 6.5. In contrast to (R)-MDLs isolated from several species of the Rosaceae family, (R)-MDL from P. aureum is not a flavoprotein. The substrate specificity was investigated using immobilized enzyme and diisopropyl ether as solvent. The addition of cyanide to aromatic and heterocyclic carbonyls is catalyzed by this (R)-MDL, whereas aliphatic carbonyls are poorly converted.

16.
Cell Signal ; 13(6): 389-400, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11384837

RESUMO

The TNF-receptor-associated factor (TRAF) family is a phylogenetically conserved group of scaffold proteins that link receptors of the IL-1R/Toll and TNF receptor family to signalling cascades, leading to the activation of NF-kappaB and mitogen-activated protein kinases. Furthermore, TRAF proteins serve as a docking platform for a variety of regulators of these signalling pathways and are themselves often regulated at the transcriptional and posttranslational level. In this review, we address the structural and molecular basis of TRAF protein functions and highlight their role in cytokine signalling.


Assuntos
Família Multigênica , Proteínas Serina-Treonina Quinases , Receptores de Citocinas/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais , Animais , Proteínas de Transporte/metabolismo , Ativação Enzimática , Humanos , Cinética , Sistema de Sinalização das MAP Quinases , Glicoproteínas de Membrana/metabolismo , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral/genética , Fator 1 Associado a Receptor de TNF , Transcrição Gênica , Fator de Necrose Tumoral alfa/metabolismo
17.
Curr Opin Biotechnol ; 11(6): 532-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11102786

RESUMO

(R)- as well as (S)-cyanohydrins are now easily available as a result of the excellent accessibility, the relatively high stability and the easy handling of hydroxynitrile lyases (HNLs). The optimization of reaction conditions (solvent, temperature, and using site-directed mutagenesis, etc.) has enabled HNL-catalyzed preparations of optically active cyanohydrins on a technical scale. The enantioselectivity of chiral metal-complex-catalyzed additions of trimethylsilyl cyanide to aldehydes has been improved, but is, by far, not yet competitive with the HNL-catalyzed reactions.


Assuntos
Aldeído Liases/metabolismo , Catálise , Nitrilas/química , Nitrilas/metabolismo , Plantas/enzimologia , Estereoisomerismo
18.
Cell Death Differ ; 22(11): 1727-41, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292758

RESUMO

From the beginning of research on receptors of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), agonistic antibodies have been used to stimulate TNFRSF receptors in vitro and in vivo. Indeed, CD95, one of the first cloned TNFRSF receptors, was solely identified as the target of cell death-inducing antibodies. Early on, it became evident from in vitro studies that valency and Fcγ receptor (FcγR) binding of antibodies targeting TNFRSF receptors can be of crucial relevance for agonistic activity. TNFRSF receptor-specific antibodies of the IgM subclass and secondary cross-linked or aggregation prone dimeric antibodies typically display superior agonistic activity compared with dimeric antibodies. Likewise, anchoring of antibodies to cell surface-expressed FcγRs potentiate their ability to trigger TNFRSF receptor signaling. However, only recently has the relevance of oligomerization and FcγR binding for the in vivo activity of antibody-induced TNFRSF receptor activation been straightforwardly demonstrated in vivo. This review discusses the crucial role of oligomerization and/or FcγR binding for antibody-mediated TNFRSF receptor stimulation in light of current models of TNFRSF receptor activation and especially the overwhelming relevance of these issues for the rational development of therapeutic TNFRSF receptor-targeting antibodies.


Assuntos
Anticorpos/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Membrana Celular/metabolismo , Humanos , Multimerização Proteica
19.
Protein Sci ; 10(5): 1015-22, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316882

RESUMO

The structure and function of hydroxynitrile lyase from Manihot esculenta (MeHNL) have been analyzed by X-ray crystallography and site-directed mutagenesis. The crystal structure of the MeHNL-S80A mutant enzyme has been refined to an R-factor of 18.0% against diffraction data to 2.1-A resolution. The three-dimensional structure of the MeHNL-S80A-acetone cyanohydrin complex was determined at 2.2-A resolution and refined to an R-factor of 18.7%. Thr11 and Cys81 involved in substrate binding have been substituted by Ala in site-directed mutagenesis. The kinetic measurements of these mutant enzymes are presented. Combined with structural data, the results support a mechanism for cyanogenesis in which His236 as a general base abstracts a proton from Ser80, thereby allowing proton transfer from the hydroxyl group of acetone cyanohydrin to Ser80. The His236 imidazolium cation then facilitates the leaving of the nitrile group by proton donating.


Assuntos
Aldeído Liases/química , Aldeído Liases/metabolismo , Substituição de Aminoácidos/genética , Manihot/enzimologia , Nitrilas/química , Nitrilas/metabolismo , Aldeído Liases/genética , Sítios de Ligação/genética , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , Substâncias Perigosas/metabolismo , Cinética , Manihot/genética , Modelos Moleculares , Mutação/genética , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
20.
Prog Mol Subcell Biol ; 34: 47-72, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14979664

RESUMO

A central event in innate immunity is the activation of the NF-kappaB signaling pathway and up-regulation of NF-kappaB-dependent defense genes. Attack of mammals as well as of insects by microorganisms leads, among other things, to the activation of receptors of the Toll-like receptor group. Various adaptor proteins involving members of the TNF receptor-associated factor (TRAF) family channel these receptor-generated signals to conserved intracellular kinase cascades that finally lead to the activation of NF-kappaB and JNK. In vertebrates, TRAF proteins link these pathways also to IL-1R-related molecules and members of the TNF receptor superfamily, which orchestrate a variety of immunoregulatory processes of the innate but also of the adaptive immune system. In this review, we will focus on the similarities but also the differences in TRAF-dependent signaling pathways of mammals and insects.


Assuntos
Drosophila/imunologia , Mamíferos/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Animais , Caenorhabditis elegans/imunologia , Drosophila/genética , Proteínas de Drosophila/imunologia , Imunidade Inata , Glicoproteínas de Membrana/imunologia , Modelos Biológicos , Mutação , NF-kappa B/imunologia , Receptores de Superfície Celular/imunologia , Transdução de Sinais , Receptor 5 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA