Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Brain ; 146(8): 3528-3541, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36732302

RESUMEN

Biallelic loss-of-function variants in SMPD4 cause a rare and severe neurodevelopmental disorder with progressive congenital microcephaly and early death. SMPD4 encodes a sphingomyelinase that hydrolyses sphingomyelin into ceramide at neutral pH and can thereby affect membrane lipid homeostasis. SMPD4 localizes to the membranes of the endoplasmic reticulum and nuclear envelope and interacts with nuclear pore complexes (NPC). We refine the clinical phenotype of loss-of-function SMPD4 variants by describing five individuals from three unrelated families with longitudinal data due to prolonged survival. All individuals surviving beyond infancy developed insulin-dependent diabetes, besides presenting with a severe neurodevelopmental disorder and microcephaly, making diabetes one of the most frequent age-dependent non-cerebral abnormalities. We studied the function of SMPD4 at the cellular and organ levels. Knock-down of SMPD4 in human neural stem cells causes reduced proliferation rates and prolonged mitosis. Moreover, SMPD4 depletion results in abnormal nuclear envelope breakdown and reassembly during mitosis and decreased post-mitotic NPC insertion. Fibroblasts from affected individuals show deficient SMPD4-specific neutral sphingomyelinase activity, without changing (sub)cellular lipidome fractions, which suggests a local function of SMPD4 on the nuclear envelope. In embryonic mouse brain, knockdown of Smpd4 impairs cortical progenitor proliferation and induces premature differentiation by altering the balance between neurogenic and proliferative progenitor cell divisions. We hypothesize that, in individuals with SMPD4-related disease, nuclear envelope bending, which is needed to insert NPCs in the nuclear envelope, is impaired in the absence of SMPD4 and interferes with cerebral corticogenesis and survival of pancreatic beta cells.


Asunto(s)
Diabetes Mellitus , Microcefalia , Humanos , Animales , Ratones , Membrana Nuclear/química , Membrana Nuclear/metabolismo , Microcefalia/genética , Microcefalia/metabolismo , Esfingomielina Fosfodiesterasa/análisis , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Poro Nuclear/metabolismo , Mitosis , Diabetes Mellitus/metabolismo
2.
Nat Immunol ; 10(7): 761-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19525969

RESUMEN

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.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Vesículas Secretoras/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Linfocitos T/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL5/metabolismo , Femenino , Granzimas/genética , Granzimas/metabolismo , Immunoblotting , Sinapsis Inmunológicas/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Perforina/genética , Perforina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vesículas Secretoras/enzimología , Esfingomielina Fosfodiesterasa/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
3.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25056906

RESUMEN

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.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Proteínas Portadoras/metabolismo , Disentería Bacilar/inmunología , Mitocondrias/inmunología , Proteínas Mitocondriales/metabolismo , Shigella/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Western Blotting , Caspasas/metabolismo , Proliferación Celular , Células Cultivadas , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Femenino , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/inmunología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Shigella/patogenicidad , Transducción de Señal , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
4.
Eur J Immunol ; 44(3): 728-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24272050

RESUMEN

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.


Asunto(s)
Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Deficiencia de Riboflavina/inmunología , Deficiencia de Riboflavina/metabolismo , Animales , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/inmunología , Flavina-Adenina Dinucleótido/biosíntesis , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , Fagocitos/inmunología , Fagocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Nature ; 460(7259): 1159-63, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19641494

RESUMEN

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.


Asunto(s)
NADPH Oxidasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular , Grupo Citocromo b/metabolismo , Activación Enzimática , Fibroblastos , Mononucleótido de Flavina/metabolismo , Flavina-Adenina Dinucleótido/biosíntesis , Flavina-Adenina Dinucleótido/metabolismo , Células HeLa , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Unión Proteica , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/química
6.
Hepatology ; 50(6): 1773-82, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19937696

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Hepatitis B/inmunología , Interferones/fisiología , Interleucina-6/fisiología , Células Cultivadas , Citocinas/biosíntesis , Hepatitis B/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Factor Nuclear 1-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/antagonistas & inhibidores , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Transcripción Genética , Replicación Viral
7.
Gastroenterology ; 134(1): 239-47, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166356

RESUMEN

BACKGROUND & AIMS: The final goal in hepatitis B therapy is eradication of the hepatitis B virus (HBV) replication template, the so-called covalently closed circular DNA (cccDNA). Current antiviral treatment of chronic hepatitis B depends on interferon alpha or nucleoside analogues inhibiting the viral reverse transcriptase. Despite treatment, cccDNA mostly persists in the host cell nucleus, continues to produce hepatitis B surface antigen (HBsAg), and causes relapsing disease. We therefore aimed at eliminating persistently infected hepatocytes carrying HBV cccDNA by redirecting cytolytic T cells toward HBsAg-producing cells. METHODS: We designed chimeric T-cell receptors directed against HBV surface proteins present on HBV-infected cells and used them to graft primary human T cells with antibody-like specificity. The receptors were composed of a single chain antibody fragment directed against HBV S or L protein fused to intracellular signalling domains of CD3xi and the costimulatory CD28 molecule. RESULTS: Our results show that these chimeric receptors, when retrovirally delivered and expressed on the cell surface, enable primary human T cells to recognize HBsAg-positive hepatocytes, release interferon gamma and interleukin 2, and, most importantly, lyse HBV replicating cells. When coincubated with HBV-infected primary human hepatocytes, these engineered, antigen-specific T cells selectively eliminated HBV-infected and thus cccDNA-positive target cells. CONCLUSIONS: Elimination of HBV cccDNA-positive hepatocytes following antiviral therapy is a major therapeutic goal in chronic hepatitis B, and adoptive transfer of grafted T cells provides a promising novel therapeutic approach. However, T-cell therapy may also cause liver damage and therefore needs further preclinical evaluation.


Asunto(s)
Complejo CD3/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Virus de la Hepatitis B/fisiología , Hepatitis B/inmunología , Hepatocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/fisiología , Técnicas de Cultivo de Célula , Quimera , Hepatitis B/patología , Humanos , Fragmentos de Inmunoglobulinas/fisiología , Especificidad del Receptor de Antígeno de Linfocitos T/fisiología
8.
PLoS One ; 14(8): e0220133, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31369589

RESUMEN

BACKGROUND: Hypobaric hypoxia has been reported to cause endothelial cell and platelet dysfunction implicated in the formation of microvascular lesions, and in its extremes may contribute to vascular leakage in high altitude pulmonary edema or blood brain barrier disruption leading to cerebral micro-hemorrhage (MH). Platelet function in the development of microvascular lesions remained ill defined, and is still incompletely understood. In this study platelet- and endothelial cell-derived extracellular vesicles (PEV and EEV, respectively) and cell adhesion molecules were characterized in plasma samples of members of a high altitude expedition to delineate the contribution of platelets and endothelial cells to hypobaric hypoxia-induced vascular dysfunction. METHODS AND FINDINGS: In this observational study, platelet and endothelial cell-derived extracellular vesicles were analysed by flow-cytometry in plasma samples from 39 mountaineers participating in a medical research climbing expedition to Himlung Himal, Nepal, 7,050m asl. Megakaryocyte/platelet-derived AnnexinVpos, PECAM-1 (CD31) and glycoprotein-1b (GP1b, CD42b) positive extracellular vesicles (PEV) constituted the predominant fraction of EV in plasma samples up to 6,050m asl. Exposure to an altitude of 7,050m led to a marked decline of CD31pos CD42neg EEV as well as of CD31pos CD42bpos PEV at the same time giving rise to a quantitatively prevailing CD31neg CD42blow/neg subpopulation of AnnexinVpos EV. An almost hundredfold increase in the numbers of this previously unrecognized population of CD31neg CD42blow/neg EV was observed in all participants reaching 7,050m asl. CONCLUSIONS: The emergence of CD31neg CD42blow/neg EV was observed in all participants and thus represents an early hypoxic marker at extreme altitude. Since CD31 and CD42b are required for platelet-endothelial cell interactions, these hypobaric hypoxia-dependent quantitative and phenotypic changes of AnnexinVpos EV subpopulations may serve as early and sensitive indicators of compromised vascular homeostasis.


Asunto(s)
Altitud , Anexina A5/sangre , Células Endoteliales/patología , Vesículas Extracelulares/patología , Hipoxia/fisiopatología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/sangre , Aclimatación , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Persona de Mediana Edad
9.
Sci Signal ; 12(568)2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755476

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular/química , Listeria monocytogenes/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , FN-kappa B/metabolismo , Transducción de Señal
10.
Cell Host Microbe ; 23(3): 324-337.e5, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29544096

RESUMEN

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.


Asunto(s)
Antígenos CD18/inmunología , Interacciones Huésped-Patógeno/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Antígeno de Macrófago-1/inmunología , Fagocitosis , Animales , Autofagia , Modelos Animales de Enfermedad , Listeria monocytogenes/patogenicidad , Lisosomas , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2/metabolismo , Fagosomas , Especies Reactivas de Oxígeno/metabolismo , Esfingomielina Fosfodiesterasa , Factores de Virulencia
11.
EMBO J ; 26(14): 3308-21, 2007 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-17599063

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Factores de Necrosis Tumoral/farmacología , Células 3T3 , Animales , Células COS , Chlorocebus aethiops , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Ratones , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP cdc42/metabolismo
12.
Blood ; 109(9): 3982-8, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17185461

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/metabolismo , Ácidos Borónicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Enfermedad de Hodgkin/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/biosíntesis , Pirazinas/farmacología , Células de Reed-Sternberg/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/biosíntesis , Linfocitos B/patología , Bortezomib , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos/genética , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Humanos , Células Jurkat , Proteínas de Neoplasias/genética , Células de Reed-Sternberg/patología , Proteína Inhibidora de la Apoptosis Ligada a X/genética
13.
J Biol Chem ; 281(19): 13784-13793, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16517606

RESUMEN

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.


Asunto(s)
Esfingomielina Fosfodiesterasa/metabolismo , Secuencia de Aminoácidos , Línea Celular , Regulación Enzimológica de la Expresión Génica , Humanos , Magnesio , Datos de Secuencia Molecular , Transporte de Proteínas , Saccharomyces cerevisiae/metabolismo , Esfingomielina Fosfodiesterasa/química , Esfingomielinas/metabolismo
14.
Vaccine ; 24(16): 3127-36, 2006 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-16497417

RESUMEN

Human immunodeficiency virus type 1-encoded Tat protein is a transactivating factor essentially required for viral replication. Tat binds specifically to the transactivation response RNA stem loop, which is formed at the 5' end of all viral transcripts. The TAR binding motif of Tat also contains a protein transduction domain, PTD that mediates not only nuclear localization of Tat but is also capable of transducing cargo across cellular membranes. In order to target a Tat antagonist directly to the TAR binding site in the nucleus, we engineered a chimeric protein consisting of the Tat-derived PTD fused to the anti-Tat single chain antibody scFvtat1 that binds intracellularly to Tat. Recombinant scFvtat1-PTD(TAT) fusion antibody retained both, anti-Tat specificity and PTD(TAT)-mediated transduction-competence leading to its nuclear accumulation within living cells. Incubation of Jurkat T cells with scFvtat1-PTD(TAT) suppressed Tat-dependent transcription of a HIV-1 reporter gene by >80%. Transfection of a scFvtat1-PTD(TAT) expression plasmid in HEK293 cells resulted in diffuse cytoplasmic and nuclear expression. ScFvtat1-PTD(TAT) did not inhibit HIV-1 Tat translocation to the nucleus, yet showed increased inhibition of 78%, indicating a nuclear site of scFvtat1-PTD(TAT) action. Strikingly, the PTD(TAT) alone showed 55% inhibition in the HIV-1 luciferase reporter assay, indicating competition with HIV-1 Tat binding to the TAR element. The results of this study suggest that Tat traffic can only marginally be affected by anti-Tat antibodies and that effective inhibition of Tat function requires both competition with HIV Tat for TAR binding mediated by PTD(TAT) and steric hindrance mediated by the scFvtat1 moiety.


Asunto(s)
Productos del Gen tat/antagonistas & inhibidores , Productos del Gen tat/inmunología , Anticuerpos Anti-VIH/metabolismo , VIH-1 , Secuencias de Aminoácidos , Fraccionamiento Celular , Núcleo Celular/química , Citoplasma/química , Genes Reporteros , Anticuerpos Anti-VIH/genética , Anticuerpos Anti-VIH/inmunología , Humanos , Immunoblotting , Células Jurkat , Luciferasas/análisis , Luciferasas/genética , Microscopía Confocal , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
15.
J Biol Chem ; 280(21): 20804-13, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15743760

RESUMEN

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.


Asunto(s)
Mitocondrias/ultraestructura , Proteínas Proto-Oncogénicas c-bcl-2/química , Esfingomielina Fosfodiesterasa/fisiología , Rayos Ultravioleta , Apoptosis , Caspasa 3 , Caspasa 8 , Caspasas/metabolismo , Catepsina D/metabolismo , Membrana Celular/química , Sistema Libre de Células , Ceramidas/farmacología , Citocromos c/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Enfermedades de Niemann-Pick/enzimología , Conformación Proteica/efectos de los fármacos , Conformación Proteica/efectos de la radiación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/efectos de la radiación , ARN Interferente Pequeño/farmacología , Transducción de Señal , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Proteína X Asociada a bcl-2
16.
EMBO J ; 21(18): 4785-95, 2002 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12234919

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/química , Proteínas del Tejido Nervioso/química , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Síndrome de Chediak-Higashi/genética , Síndrome de Chediak-Higashi/metabolismo , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transducción de Señal/fisiología
17.
J Immunol ; 169(9): 5161-70, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12391233

RESUMEN

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.


Asunto(s)
Antígenos CD/fisiología , Proteína Quinasa C/metabolismo , Proteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/inmunología , Esfingomielina Fosfodiesterasa/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antígenos CD/química , Antígenos CD/genética , Células COS , Línea Celular , Activación Enzimática/genética , Activación Enzimática/inmunología , Células HeLa , Humanos , Líquido Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Pruebas de Precipitina , Unión Proteica/genética , Unión Proteica/inmunología , Mapeo de Interacción de Proteínas/métodos , Proteína Quinasa C/química , Proteína Quinasa C/genética , Proteínas/química , Proteínas/genética , Receptores de Cinasa C Activada , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral , Secuencias Repetitivas de Aminoácido , Transducción de Señal/genética , Esfingomielina Fosfodiesterasa/química , Esfingomielina Fosfodiesterasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA