Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 12(568)2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755476

RESUMO

A major function of macrophages during infection is initiation of the proinflammatory response, leading to the secretion of cytokines that help to orchestrate the immune response. Here, we identify reactive oxygen species (ROS) as crucial mediators of proinflammatory signaling leading to cytokine secretion in Listeria monocytogenes-infected macrophages. ROS produced by NADPH oxidases (Noxes), such as Nox2, are key components of the macrophage response to invading pathogens; however, our data show that the ROS that mediated proinflammatory signaling were produced by mitochondria (mtROS). We identified the inhibitor of κB (IκB) kinase (IKK) complex regulatory subunit NEMO [nuclear factor κB (NF-κB) essential modulator] as a target for mtROS. Specifically, mtROS induced intermolecular covalent linkage of NEMO through disulfide bonds formed by Cys54 and Cys347, which was essential for activation of the IKK complex and subsequent signaling through the extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) and NF-κB pathways that eventually led to the secretion of proinflammatory cytokines. We thus identify mtROS-dependent disulfide linkage of NEMO as an essential regulatory step of the proinflammatory response of macrophages to bacterial infection.


Assuntos
Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Cisteína/química , Cisteína/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Interações Hospedeiro-Patógeno , Peptídeos e Proteínas de Sinalização Intracelular/química , Listeria monocytogenes/fisiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais
2.
Cell Host Microbe ; 23(3): 324-337.e5, 2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29544096

RESUMO

The intracellular pathogen Listeria monocytogenes (L.m.) is targeted by the autophagic machinery, but the molecular mechanisms involved and consequences for anti-listerial immunity remain enigmatic. Here, we demonstrate that L.m. infection of macrophages in vivo exclusively evokes LC3-associated phagocytosis (LAP), but not canonical autophagy, and that targeting of L.m. by LAP is required for anti-listerial immunity. The pathway leading to LAP induction in response to L.m. infection emanates from the ß2 integrin Mac-1 (CR3, integrin αMß2), a receptor recognizing diverse microbial ligands. Interaction of L.m. with Mac-1 induces acid sphingomyelinase-mediated changes in membrane lipid composition that facilitate assembly and activation of the phagocyte NAPDH oxidase Nox2. Nox2-derived reactive oxygen species then trigger LC3 recruitment to L.m.-containing phagosomes by LAP. By promoting fusion of L.m.-containing phagosomes with lysosomes, LAP increases exposure of L.m. to bactericidal acid hydrolases, thereby enhancing anti-listerial activity of macrophages and immunity of mice.


Assuntos
Antígenos CD18/imunologia , Interações Hospedeiro-Patógeno/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Antígeno de Macrófago 1/imunologia , Fagocitose , Animais , Autofagia , Modelos Animais de Doenças , Listeria monocytogenes/patogenicidade , Lisossomos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 2/metabolismo , Fagossomos , Espécies Reativas de Oxigênio/metabolismo , Esfingomielina Fosfodiesterase , Fatores de Virulência
3.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25056906

RESUMO

The X-linked inhibitor of apoptosis protein (XIAP) is a potent caspase inhibitor, best known for its anti-apoptotic function in cancer. During apoptosis, XIAP is antagonized by SMAC, which is released from the mitochondria upon caspase-mediated activation of BID. Recent studies suggest that XIAP is involved in immune signaling. Here, we explore XIAP as an important mediator of an immune response against the enteroinvasive bacterium Shigella flexneri, both in vitro and in vivo. Our data demonstrate for the first time that Shigella evades the XIAP-mediated immune response by inducing the BID-dependent release of SMAC from the mitochondria. Unlike apoptotic stimuli, Shigella activates the calpain-dependent cleavage of BID to trigger the release of SMAC, which antagonizes the inflammatory action of XIAP without inducing apoptosis. Our results demonstrate how the cellular death machinery can be subverted by an invasive pathogen to ensure bacterial colonization.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Proteínas de Transporte/metabolismo , Disenteria Bacilar/imunologia , Mitocôndrias/imunologia , Proteínas Mitocondriais/metabolismo , Shigella/imunologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/fisiologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Western Blotting , Caspases/metabolismo , Proliferação de Células , Células Cultivadas , Disenteria Bacilar/microbiologia , Disenteria Bacilar/patologia , Feminino , Hepatócitos/imunologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Técnicas Imunoenzimáticas , Integrases/metabolismo , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/imunologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Shigella/patogenicidade , Transdução de Sinais , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
4.
Eur J Immunol ; 44(3): 728-41, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24272050

RESUMO

Riboflavin, also known as vitamin B2 , is converted by riboflavin kinase (RFK) into flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), which are essential cofactors of dehydrogenases, reductases, and oxidases including the phagocytic NADPH oxidase 2 (Nox2). Riboflavin deficiency is common in young adults and elderly individuals, who are at the coincidental risk for listeriosis. To address the impact of acute riboflavin deficiency on host defense against Listeria monocytogenes (L.m.), we generated conditional RFK knockout (KO) strains of mice. Phagocyte-specific RFK KO impaired the capability of phagocytes to control intracellular L.m., which corresponded to a greater susceptibility of mice to in vivo challenge with L.m. The oxidative burst of RFK-deficient phagocytes in response to L.m. infection was significantly reduced. Mechanistically, TNF-induced priming of Nox2, which is needed for oxidative burst, was defective in RFK-deficient phagocytes. Lack of riboflavin in wild-type macrophages for only 6 h shut down TNF-induced, RFK-mediated de novo FMN/FAD generation, which was accompanied by diminished ROS production and impaired anti-listerial activity. Vice versa, ROS production by riboflavin-deprived macrophages was rapidly restored by riboflavin supplementation. Our results suggest that acute riboflavin deficiency immediately impairs priming of Nox2, which is of crucial relevance for an effective phagocytic immune response in vivo.


Assuntos
Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Deficiência de Riboflavina/imunologia , Deficiência de Riboflavina/metabolismo , Animais , Modelos Animais de Doenças , Resistência à Doença/imunologia , Flavina-Adenina Dinucleotídeo/biossíntese , Imunidade Inata , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , NADPH Oxidase 2 , Fagócitos/imunologia , Fagócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Hepatology ; 50(6): 1773-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19937696

RESUMO

UNLABELLED: With about 350 million virus carriers, hepatitis B virus (HBV) infection remains a major health problem. HBV is a noncytopathic virus causing persistent infection, but it is still unknown whether host recognition of HBV may activate an innate immune response. We describe that upon infection of primary human liver cells, HBV is recognized by nonparenchymal cells of the liver, mainly by liver macrophages (Kupffer cells), although they are not infected. Within 3 hours, this recognition leads to the activation of nuclear factor kappa B (NF-kappaB) and subsequently to the release of interleukin-6 (IL-6) and other proinflammatory cytokines (IL-8, TNF-alpha, IL-1beta), but does not induce an interferon response. The activation of proinflammatory cytokines, however, is transient, and even inhibits responsiveness toward a subsequent challenge. IL-6 released by Kupffer cells after activation of NF-kappaB controls HBV gene expression and replication in hepatocytes at the level of transcription shortly after infection. Upon binding to its receptor complex, IL-6 activates the mitogen-activated protein kinases exogenous signal-regulated kinase 1/2, and c-jun N-terminal kinase, which inhibit expression of hepatocyte nuclear factor (HNF) 1alpha and HNF 4alpha, two transcription factors essential for HBV gene expression and replication. CONCLUSION: Our results demonstrate recognition of HBV patterns by nonparenchymal liver cells, which results in IL-6-mediated control of HBV infection at the transcriptional level. Thus, IL-6 ensures early control of the virus, limiting activation of the adaptive immune response and preventing death of the HBV-infected hepatocyte. This pattern recognition may be essential for a virus, which infects a new host with only a few virions. Our data also indicate that therapeutic neutralization of IL-6 for treatment of certain diseases may represent a risk if the patient is HBV-infected.


Assuntos
Regulação da Expressão Gênica , Hepatite B/imunologia , Interferons/fisiologia , Interleucina-6/fisiologia , Células Cultivadas , Citocinas/biossíntese , Hepatite B/metabolismo , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/genética , Vírus da Hepatite B/fisiologia , Fator 1-alfa Nuclear de Hepatócito/antagonistas & inibidores , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/antagonistas & inibidores , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Transcrição Gênica , Replicação Viral
6.
Nature ; 460(7259): 1159-63, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19641494

RESUMO

Reactive oxygen species (ROS) produced by NADPH oxidase function as defence and signalling molecules related to innate immunity and various cellular responses. The activation of NADPH oxidase in response to plasma membrane receptor activation depends on the phosphorylation of cytoplasmic oxidase subunits, their translocation to membranes and the assembly of all NADPH oxidase components. Tumour necrosis factor (TNF) is a prominent stimulus of ROS production, but the molecular mechanisms by which TNF activates NADPH oxidase are poorly understood. Here we identify riboflavin kinase (RFK, formerly known as flavokinase) as a previously unrecognized TNF-receptor-1 (TNFR1)-binding protein that physically and functionally couples TNFR1 to NADPH oxidase. In mouse and human cells, RFK binds to both the TNFR1-death domain and to p22(phox), the common subunit of NADPH oxidase isoforms. RFK-mediated bridging of TNFR1 and p22(phox) is a prerequisite for TNF-induced but not for Toll-like-receptor-induced ROS production. Exogenous flavin mononucleotide or FAD was able to substitute fully for TNF stimulation of NADPH oxidase in RFK-deficient cells. RFK is rate-limiting in the synthesis of FAD, an essential prosthetic group of NADPH oxidase. The results suggest that TNF, through the activation of RFK, enhances the incorporation of FAD in NADPH oxidase enzymes, a critical step for the assembly and activation of NADPH oxidase.


Assuntos
NADPH Oxidases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Linhagem Celular , Grupo dos Citocromos b/metabolismo , Ativação Enzimática , Fibroblastos , Mononucleotídeo de Flavina/metabolismo , Flavina-Adenina Dinucleotídeo/biossíntese , Flavina-Adenina Dinucleotídeo/metabolismo , Células HeLa , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/química , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ligação Proteica , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/química
7.
Nat Immunol ; 10(7): 761-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525969

RESUMO

Granule-mediated cytotoxicity is the main effector mechanism of cytotoxic CD8+ T cells. We report that CD8+ T cells from acid sphingomyelinase (ASMase)-deficient (ASMase-KO) mice are defective in exocytosis of cytolytic effector molecules; this defect resulted in attenuated cytotoxic activity of ASMase-KO CD8+ T cells and delayed elimination of lymphocytic choriomeningitis virus from ASMase-KO mice. Cytolytic granules of ASMase-KO and wild-type CD8+ T cells were equally loaded with granzymes and perforin, and correctly directed to the immunological synapse. In wild-type CD8+ T cells, secretory granules underwent shrinkage by 82% after fusion with the plasma membrane. In ASMase-KO CD8+ T cells, the contraction of secretory granules was markedly impaired. Thus, ASMase is required for contraction of secretory granules and expulsion of cytotoxic effector molecules.


Assuntos
Citotoxicidade Imunológica/imunologia , Vesículas Secretórias/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Linfócitos T/imunologia , Animais , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Quimiocina CCL5/metabolismo , Feminino , Granzimas/genética , Granzimas/metabolismo , Immunoblotting , Sinapses Imunológicas/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Masculino , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Perforina/genética , Perforina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vesículas Secretórias/enzimologia , Esfingomielina Fosfodiesterase/genética , Linfócitos T/citologia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
8.
EMBO J ; 26(14): 3308-21, 2007 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-17599063

RESUMO

The WD-repeat protein factor associated with nSMase activity (FAN) is a member of the family of TNF receptor adaptor proteins that are coupled to specific signaling cascades. However, the precise functional involvement of FAN in specific cellular TNF responses remain unclear. Here, we report the involvement of FAN in TNF-induced actin reorganization and filopodia formation mediated by activation of Cdc42. The pleckstrin-homology (PH) domain of FAN specifically binds to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P), which targets FAN to the plasma membrane. Site-specific mutagenesis revealed that the ability of FAN to mediate filopodia formation was blunted either by the destruction of the PtdIns(4,5)P binding motif, or by the disruption of intramolecular interactions between the PH domain and the adjacent beige and Chediak-Higashi (BEACH) domain. Furthermore, FAN was shown to interact with the actin cytoskeleton in TNF-stimulated cells via direct filamentous actin (F-actin) binding. The results of this study suggest that PH-mediated plasma membrane targeting of FAN is critically involved in TNF-induced Cdc42 activation and cytoskeleton reorganization.


Assuntos
Actinas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fatores de Necrose Tumoral/farmacologia , Células 3T3 , Animais , Células COS , Chlorocebus aethiops , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Camundongos , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/metabolismo
9.
Blood ; 109(9): 3982-8, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17185461

RESUMO

The proteasome inhibitor bortezomib has been shown to possess promising antitumor activity and significant efficacy against a variety of malignancies. Different studies demonstrated that bortezomib breaks the chemoresistance in different tumor cells basically by altering nuclear factor-kappaB (NF-kappaB) activity. NF-kappaB has been shown to be constitutively active in most primary Hodgkin-Reed-Sternberg (H-RS) cells in lymph node sections and in Hodgkin lymphoma (HL) cell lines and was suggested to be a central molecular switch in apoptosis resistance in HL. Here we report a bimodal effect of bortezomib in HL cells. Whereas high-dose bortezomib induced direct cytotoxicity that correlated with decreased NF-kappaB activity, low-dose bortezomib sensitized HL cells against a variety of cytotoxic drugs without altering NF-kappaB action. Strikingly, bortezomib induced marked XIAP down-regulation at the posttranslational level that was independent of the NF-kappaB status. Similarly, RNA interference (RNAi)-mediated XIAP down-regulation generated susceptibility to cytostatic agents. The results identify XIAP as an NF-kappaB-independent target of bortezomib action that controls the chemoresistant phenotype of HL cells.


Assuntos
Antineoplásicos/farmacologia , Linfócitos B/metabolismo , Ácidos Borônicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Doença de Hodgkin/metabolismo , NF-kappa B/metabolismo , Proteínas de Neoplasias/biossíntese , Pirazinas/farmacologia , Células de Reed-Sternberg/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/biossíntese , Linfócitos B/patologia , Bortezomib , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistencia a Medicamentos Antineoplásicos/genética , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/genética , Doença de Hodgkin/patologia , Humanos , Células Jurkat , Proteínas de Neoplasias/genética , Células de Reed-Sternberg/patologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
10.
J Biol Chem ; 281(19): 13784-13793, 2006 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-16517606

RESUMO

Two genes encoding neutral sphingomyelinases-1 and -2 (sphingomyelin phosphodiesterases-2 and -3) have been recently identified that hydrolyze sphingomyelin to phosphorylcholine and ceramide. Data bank searches using a peptide sequence derived from a previously purified bovine neutral sphingomyelinase (nSMase) allowed us to identify a cDNA encoding a novel human sphingomyelinase, nSMase3, that shows only a little homology to nSMase1 and -2. nSMase3 was biochemically characterized by overexpression in a yeast strain, JK9-3ddeltaIsc1p, lacking endogenous SMase activity. Similar to nSMase2, nSMase3 is Mg2+-dependent and shows optimal activity at pH 7, which is enhanced in the presence of phosphatidylserine and inhibited by scyphostatin. nSMase3 is ubiquitously expressed as a 4.6-kb mRNA species. nSMase3 lacks an N-terminal signal peptide, yet contains a 23-amino-acid transmembrane domain close to the C terminus, which is indicative for the family of C-tail-anchored integral membrane proteins. Cellular localization studies with hemagglutinin-tagged nSMase3 demonstrated colocalization with markers of the endoplasmic reticulum as well as with Golgi markers. Tumor necrosis factor stimulates rapid activation of nSMase3 in MCF7 cells with peak activity at 1.5 min, which was impaired by expression of dominant negative FAN.


Assuntos
Esfingomielina Fosfodiesterase/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Regulação Enzimológica da Expressão Gênica , Humanos , Magnésio , Dados de Sequência Molecular , Transporte Proteico , Saccharomyces cerevisiae/metabolismo , Esfingomielina Fosfodiesterase/química , Esfingomielinas/metabolismo
11.
J Biol Chem ; 280(21): 20804-13, 2005 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15743760

RESUMO

Ultraviolet light-induced apoptosis can be caused by DNA damage but also involves immediate-early cell death cascades characteristic of death receptor signaling. Here we show that the UV light-induced apoptotic signaling pathway is unique, targeting Bax activation at the mitochondrial membrane independent of caspase-8 or cathepsin D activity. Cells deficient in acid sphingomyelinase (ASMase) do not show UV light-induced Bax activation, cytochrome c release, or apoptosis. In ASMase-deficient cells, the apoptotic UV light response is restored by stable or transient expression of human ASMase. Bax conformational change in ASMase(-/-) cells is also caused by synthetic C(16)-ceramide acting on intact cells or isolated mitochondria. The results suggest that UV light-triggered ASMase activation is essentially required for Bax conformational change leading to mitochondrial release of pro-apoptotic factors like cytochrome c and Smac.


Assuntos
Mitocôndrias/ultraestrutura , Proteínas Proto-Oncogênicas c-bcl-2/química , Esfingomielina Fosfodiesterase/fisiologia , Raios Ultravioleta , Apoptose , Caspase 3 , Caspase 8 , Caspases/metabolismo , Catepsina D/metabolismo , Membrana Celular/química , Sistema Livre de Células , Ceramidas/farmacologia , Citocromos c/metabolismo , Imunofluorescência , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Doenças de Niemann-Pick/enzimologia , Conformação Proteica/efeitos dos fármacos , Conformação Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/efeitos da radiação , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Proteína X Associada a bcl-2
12.
J Immunol ; 169(9): 5161-70, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391233

RESUMO

Factor associated with neutral sphingomyelinase activation (FAN) represents a p55 TNFR (TNF-R55)-associated protein essential for the activation of neutral sphingomyelinase. By means of the yeast interaction trap system, we have identified the scaffolding protein receptor for activated C-kinase (RACK)1 as an interaction partner of FAN. Mapping studies in yeast revealed that RACK1 is recruited to the C-terminal WD-repeat region of FAN and binds to FAN through a domain located within WD repeats V to VII of RACK1. Our data indicate that binding of both proteins is not mediated by linear motifs but requires folding into a secondary structure, such as the multibladed propeller characteristic of WD-repeat proteins. The interaction of FAN and RACK1 was verified in vitro by glutathione S-transferase-based coprecipitation assays as well as in eukaryotic cells by coimmunoprecipitation experiments. Colocalization studies in transfected cells suggest that TNF-R55 forms a complex with FAN and that this complex recruits RACK1 to the plasma membrane. Furthermore, activation of N-SMase by TNF was strongly enhanced when RACK1, FAN, and a noncytotoxic TNF-R55 mutant were expressed concurrently, suggesting RACK1 as a modulator of N-SMase activation. Together, these findings implicate RACK1 as a novel component of the signaling pathways of TNF-R55.


Assuntos
Antígenos CD/fisiologia , Proteína Quinase C/metabolismo , Proteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/imunologia , Esfingomielina Fosfodiesterase/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Antígenos CD/química , Antígenos CD/genética , Células COS , Linhagem Celular , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Células HeLa , Humanos , Líquido Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Células Jurkat , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Testes de Precipitina , Ligação Proteica/genética , Ligação Proteica/imunologia , Mapeamento de Interação de Proteínas/métodos , Proteína Quinase C/química , Proteína Quinase C/genética , Proteínas/química , Proteínas/genética , Receptores de Quinase C Ativada , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Sequências Repetitivas de Aminoácidos , Transdução de Sinais/genética , Esfingomielina Fosfodiesterase/química , Esfingomielina Fosfodiesterase/genética
13.
EMBO J ; 21(18): 4785-95, 2002 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-12234919

RESUMO

The BEACH domain is highly conserved in a large family of eukaryotic proteins, and is crucial for their functions in vesicle trafficking, membrane dynamics and receptor signaling. However, it does not share any sequence homology with other proteins. Here we report the crystal structure at 2.9 A resolution of the BEACH domain of human neurobeachin. It shows that the BEACH domain has a new and unusual polypeptide backbone fold, as the peptide segments in its core do not assume regular secondary structures. Unexpectedly, the structure also reveals that the BEACH domain is in extensive association with a novel, weakly conserved pleckstrin-homology (PH) domain. Consistent with the structural analysis, biochemical studies show that the PH and BEACH domains have strong interactions, suggesting they may function as a single unit. Functional studies in intact cells demonstrate the requirement of both the PH and the BEACH domains for activity. A prominent groove at the interface between the two domains may be used to recruit their binding partners.


Assuntos
Proteínas de Transporte/química , Proteínas do Tecido Nervoso/química , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Síndrome de Chediak-Higashi/genética , Síndrome de Chediak-Higashi/metabolismo , Cristalografia por Raios X , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA