Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638593

RESUMEN

Salt stress tolerance of crop plants is a trait with increasing value for future food production. In an attempt to identify proteins that participate in the salt stress response of barley, we have used a cDNA library from salt-stressed seedling roots of the relatively salt-stress-tolerant cv. Morex for the transfection of a salt-stress-sensitive yeast strain (Saccharomyces cerevisiae YSH818 Δhog1 mutant). From the retrieved cDNA sequences conferring salt tolerance to the yeast mutant, eleven contained the coding sequence of a jacalin-related lectin (JRL) that shows homology to the previously identified JRL horcolin from barley coleoptiles that we therefore named the gene HvHorcH. The detection of HvHorcH protein in root extracellular fluid suggests a secretion under stress conditions. Furthermore, HvHorcH exhibited specificity towards mannose. Protein abundance of HvHorcH in roots of salt-sensitive or salt-tolerant barley cultivars were not trait-specific to salinity treatment, but protein levels increased in response to the treatment, particularly in the root tip. Expression of HvHorcH in Arabidopsis thaliana root tips increased salt tolerance. Hence, we conclude that this protein is involved in the adaptation of plants to salinity.


Asunto(s)
Hordeum/genética , Lectinas/genética , Lectinas de Plantas/genética , Proteínas de Plantas/genética , Raíces de Plantas/genética , Estrés Salino/genética , Adaptación Fisiológica/genética , Regulación de la Expresión Génica de las Plantas/genética , Fenotipo , Salinidad , Tolerancia a la Sal/genética , Plantones/genética , Estrés Fisiológico/genética
2.
EMBO J ; 30(2): 379-94, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21157428

RESUMEN

We previously demonstrated that tumour necrosis factor (TNF)-induced ceramide production by endosomal acid sphingomyelinase (A-SMase) couples to apoptosis signalling via activation of cathepsin D and cleavage of Bid, resulting in caspase-9 and caspase-3 activation. The mechanism of TNF-mediated A-SMase activation within the endolysosomal compartment is poorly defined. Here, we show that TNF-induced A-SMase activation depends on functional caspase-8 and caspase-7 expression. The active forms of all three enzymes, caspase-8, caspase-7 and A-SMase, but not caspase-3, colocalize in internalized TNF receptosomes. While caspase-8 and caspase-3 are unable to induce activation of purified pro-A-SMase, we found that caspase-7 mediates A-SMase activation by direct interaction resulting in proteolytic cleavage of the 72-kDa pro-A-SMase zymogen at the non-canonical cleavage site after aspartate 253, generating an active 57 kDa A-SMase molecule. Caspase-7 down modulation revealed the functional link between caspase-7 and A-SMase, confirming proteolytic cleavage as one further mode of A-SMase activation. Our data suggest a signalling cascade within TNF receptosomes involving sequential activation of caspase-8 and caspase-7 for induction of A-SMase activation by proteolytic cleavage of pro-A-SMase.


Asunto(s)
Caspasa 7/metabolismo , Caspasa 8/metabolismo , Endosomas/metabolismo , Activación Enzimática/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Western Blotting , Línea Celular , Ceramidas/metabolismo , Cromatografía en Capa Delgada , Clonación Molecular , Activación Enzimática/genética , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Células Jurkat , Ratones , Microscopía Confocal
3.
Gastroenterology ; 146(1): 278-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24120475

RESUMEN

BACKGROUND & AIMS: Tumor necrosis factor-related apoptosis inducing ligand (TRAIL-R1) (TNFRSF10A) and TRAIL-R2 (TNFRSF10B) on the plasma membrane bind ligands that activate apoptotic and other signaling pathways. Cancer cells also might have TRAIL-R2 in the cytoplasm or nucleus, although little is known about its activities in these locations. We investigated the functions of nuclear TRAIL-R2 in cancer cell lines. METHODS: Proteins that interact with TRAIL-R2 initially were identified in pancreatic cancer cells by immunoprecipitation, mass spectrometry, and immunofluorescence analyses. Findings were validated in colon, renal, lung, and breast cancer cells. Functions of TRAIL-R2 were determined from small interfering RNA knockdown, real-time polymerase chain reaction, Drosha-activity, microRNA array, proliferation, differentiation, and immunoblot experiments. We assessed the effects of TRAIL-R2 overexpression or knockdown in human pancreatic ductal adenocarcinoma (PDAC) cells and their ability to form tumors in mice. We also analyzed levels of TRAIL-R2 in sections of PDACs and non-neoplastic peritumoral ducts from patients. RESULTS: TRAIL-R2 was found to interact with the core microprocessor components Drosha and DGCR8 and the associated regulatory proteins p68, hnRNPA1, NF45, and NF90 in nuclei of PDAC and other tumor cells. Knockdown of TRAIL-R2 increased Drosha-mediated processing of the let-7 microRNA precursor primary let-7 (resulting in increased levels of mature let-7), reduced levels of the let-7 targets (LIN28B and HMGA2), and inhibited cell proliferation. PDAC tissues from patients had higher levels of nuclear TRAIL-R2 than non-neoplastic pancreatic tissue, which correlated with increased nuclear levels of HMGA2 and poor outcomes. Knockdown of TRAIL-R2 in PDAC cells slowed their growth as orthotopic tumors in mice. Reduced nuclear levels of TRAIL-R2 in cultured pancreatic epithelial cells promoted their differentiation. CONCLUSIONS: Nuclear TRAIL-R2 inhibits maturation of the microRNA let-7 in pancreatic cancer cell lines and increases their proliferation. Pancreatic tumor samples have increased levels of nuclear TRAIL-R2, which correlate with poor outcome of patients. These findings indicate that in the nucleus, death receptors can function as tumor promoters and might be therapeutic targets.


Asunto(s)
Apoptosis/fisiología , Carcinoma Ductal Pancreático/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/metabolismo , Humanos , Neoplasias Renales/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
4.
Acta Neuropathol ; 125(6): 795-813, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23604588

RESUMEN

In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and prion diseases, deposits of aggregated disease-specific proteins are found. Oligomeric aggregates are presumed to be the key neurotoxic agent. Here we describe the novel oligomer modulator anle138b [3-(1,3-benzodioxol-5-yl)-5-(3-bromophenyl)-1H-pyrazole], an aggregation inhibitor we developed based on a systematic high-throughput screening campaign combined with medicinal chemistry optimization. In vitro, anle138b blocked the formation of pathological aggregates of prion protein (PrP(Sc)) and of α-synuclein (α-syn), which is deposited in PD and other synucleinopathies such as dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Notably, anle138b strongly inhibited all prion strains tested including BSE-derived and human prions. Anle138b showed structure-dependent binding to pathological aggregates and strongly inhibited formation of pathological oligomers in vitro and in vivo both for prion protein and α-synuclein. Both in mouse models of prion disease and in three different PD mouse models, anle138b strongly inhibited oligomer accumulation, neuronal degeneration, and disease progression in vivo. Anle138b had no detectable toxicity at therapeutic doses and an excellent oral bioavailability and blood-brain-barrier penetration. Our findings indicate that oligomer modulators provide a new approach for disease-modifying therapy in these diseases, for which only symptomatic treatment is available so far. Moreover, our findings suggest that pathological oligomers in neurodegenerative diseases share structural features, although the main protein component is disease-specific, indicating that compounds such as anle138b that modulate oligomer formation by targeting structure-dependent epitopes can have a broad spectrum of activity in the treatment of different protein aggregation diseases.


Asunto(s)
Encéfalo/efectos de los fármacos , Enfermedad de Parkinson/terapia , Enfermedades por Prión/terapia , Priones/efectos de los fármacos , Pirazoles/agonistas , Pirimidinas/agonistas , Animales , Encéfalo/metabolismo , Encéfalo/patología , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedades por Prión/etiología , Enfermedades por Prión/metabolismo , Priones/metabolismo , Rotenona/farmacología , alfa-Sinucleína/farmacología
5.
J Clin Invest ; 116(12): 3204-10, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17143329

RESUMEN

Prion diseases are fatal neurodegenerative diseases characterized by the accumulation of PrP(Sc), the infectious and protease-resistant form of the cellular prion protein (PrP(C)). We generated lentivectors expressing PrP(C)-specific short hairpin RNAs (shRNAs) that efficiently silenced expression of the prion protein gene (Prnp) in primary neuronal cells. Treatment of scrapie-infected neuronal cells with these lentivectors resulted in an efficient and stable suppression of PrP(Sc) accumulation. After intracranial injection, lentiviral shRNA reduced PrP(C) expression in transgenic mice carrying multiple copies of Prnp. To test the therapeutic potential of lentiviral shRNA, we used what we believe to be a novel approach in which the clinical situation was mimicked. We generated chimeric mice derived from lentivector-transduced embryonic stem cells. Depending on the degree of chimerism, these animals carried the lentiviral shRNAs in a certain percentage of brain cells and expressed reduced levels of PrP(C). Importantly, in highly chimeric mice, survival after scrapie infection was significantly extended. Taken together, these data suggest that lentivector-mediated RNA interference could be an approach for the treatment of prion disease.


Asunto(s)
Lentivirus/genética , Priones/metabolismo , Interferencia de ARN , Scrapie/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/terapia , Priones/genética , Priones/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Scrapie/genética , Scrapie/terapia , Ovinos , Análisis de Supervivencia
6.
J Neurochem ; 106(3): 1459-70, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18498440

RESUMEN

The physiological function of the cellular prion protein (PrP(c)) is unclear. PrP(c) associates with lipid rafts, highly glycolipid-rich membrane domains containing a large variety of signaling molecules, e.g., sphingolipids (SL). In this study, we investigated possible connections between PrP(c) and sphingolipid-associated signaling pathways. Using PrP(c)-wt and PrP(c)-k.o. hippocampal cell lines and mouse brains we showed higher activity of neutral and acid sphingomyelinase (SMase) in PrP(c)-k.o.-groups, while ceramide and sphingomyelin-levels were unchanged. Furthermore, despite lower basal expression levels of sphingosine kinase (SphK) in PrP(c)-k.o.-groups, the levels of its metabolite sphingosine-1-phosphate were increased, whereas S1P(3)-receptor expression was higher in PrP(c)-wt-groups again. In addition, we detected enhanced activity of phospholipase D1, an enzyme that seems to be suitable to act as a connector between the S1P(3) receptor and continuative signaling. Finally, evidence for an impact on downstream signaling cascades, especially activation of the PI3K/Akt pathway, was found. In summary, our data suggest that PrP(c) is involved in sphingolipid-associated signaling, modulating pathways that exert anti-apoptotic functions, hence indicating that PrP(c) plays a role in neuroprotection.


Asunto(s)
Proteínas PrPC/fisiología , Transducción de Señal/fisiología , Esfingolípidos/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/metabolismo , Proteínas PrPC/genética
7.
J Neurochem ; 103(6): 2291-300, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17919292

RESUMEN

The polymorphisms at amino acid residues 136, 154, and 171 in ovine prion protein (PrP) have been associated with different susceptibility to scrapie: animals expressing PrP(ARQ) [PrP(Ala136/Arg154/Gln171)] show vulnerability, whereas those that express PrP(ARR) [PrP(Ala136/Arg154/Arg171)] are resistant to scrapie. The aim of this study was to evaluate the in vitro toxic effects of PrP(ARR) and PrP(ARQ) variants in relation with their structural characteristics. We show that both peptides cause cell death inducing apoptosis but, unexpectedly, the scrapie resistant PrP(ARR) form was more toxic than the scrapie susceptible PrP(ARQ) variant. Moreover, the alpha-helical conformation of PrP(ARR) was less stable than that of PrP(ARQ) and the structural determinants responsible of these different conformational stabilities were characterized by spectroscopic analysis. We observed that PrP toxicity was inversely related to protein structural stability, being the unfolded conformation more toxic than the native one. However, the PrP(ARQ) variant displays a higher propensity to form large aggregates than PrP(ARR). Interestingly, in the presence of small amounts of PrP(ARR), PrP(ARQ) aggregability was reduced to levels similar to that of PrP(ARR). Thus, in contrast to PrP(ARR) toxicity, scrapie transmissibility seems to reside in the more stable conformation of PrP(ARQ) that allows the formation of large amyloid fibrils.


Asunto(s)
Péptidos/química , Péptidos/toxicidad , Priones/química , Priones/toxicidad , Scrapie/metabolismo , Amiloide/química , Amiloide/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/toxicidad , Estructura Secundaria de Proteína/fisiología , Scrapie/fisiopatología , Oveja Doméstica , Análisis Espectral
8.
Vet Microbiol ; 123(4): 358-66, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17482774

RESUMEN

The cellular prion protein (PrP(C)) is a copper binding protein. The molecular features of the Cu(2+) binding sites have been investigated and characterized by spectroscopic experiments on PrP(C)-derived peptides and the correctly folded human full-length PrP(C) (hPrP-[23-231]). These experiments allowed us to distinguish two different configurations of copper binding. The different copper complexes depend on sequence context, buffer conditions and stoichiometry of copper. The combined information of spectroscopic data from our EXAFS, EPR and ENDOR experiments was used to create models for these two copper complexes. A large number of conformations of these models were calculated using molecular mechanics computations, and the simulated spectra of these structures were compared with our experimental data. Common features and differences of the copper binding motifs are discussed in this paper and it remains for future investigations to study whether different configurations are associated with different functional states of PrP(C).


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Enfermedades por Prión/metabolismo , Priones/química , Priones/metabolismo , Animales , Cobre/metabolismo , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Conformación Proteica , Espectrometría por Rayos X/métodos
9.
Oncotarget ; 8(12): 20067-20085, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28223543

RESUMEN

Acid sphingomyelinase (A-SMase) plays an important role in the initiation of CD95 signaling by forming ceramide-enriched membrane domains that enable clustering and activation of the death receptors. In TNF-R1 and TRAIL-R1/R2 signaling, A-SMase also contributes to the lysosomal apoptosis pathway triggered by receptor internalization. Here, we investigated the molecular mechanism of CD95-mediated A-SMase activation, demonstrating that A-SMase is located in internalized CD95-receptosomes and is activated by the CD95/CD95L complex in a biphasic manner.Since several caspases have been described to be involved in the activation of A-SMase, we evaluated expression levels of caspase-8, caspase-7 and caspase-3 in CD95-receptosomes. The occurrence of cleaved caspase-8 correlated with the first peak of A-SMase activity and translocation of the A-SMase to the cell surface which could be blocked by the caspase-8 inhibitor IETD.Inhibition of CD95-internalization selectively reduced the second phase of A-SMase activity, suggesting a fusion between internalized CD95-receptosomes and an intracellular vesicular pool of A-SMase. Further analysis demonstrated that caspase-7 activity correlates with the second phase of the A-SMase activity, whereas active caspase-3 is present at early and late internalization time points. Blocking caspases-7/ -3 by DEVD reduced the second phase of A-SMase activation in CD95-receptosomes suggesting the potential role of caspase-7 or -3 for late A-SMase activation.In summary, we describe a biphasic A-SMase activation in CD95-receptosomes indicating (I.) a caspase-8 dependent translocation of A-SMase to plasma membrane and (II.) a caspase-7 and/or -3 dependent fusion of internalized CD95-receptosomes with intracellular A-SMase-containing vesicles.


Asunto(s)
Linfocitos B/patología , Caspasas/metabolismo , Proteína Ligando Fas/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Receptor fas/metabolismo , Apoptosis , Linfocitos B/enzimología , Inhibidores de Caspasas/farmacología , Caspasas/química , Membrana Celular/metabolismo , Proliferación Celular , Activación Enzimática , Humanos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
10.
PLoS One ; 12(12): e0188989, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29220360

RESUMEN

Prion diseases have been linked to impaired copper homeostasis and copper induced-oxidative damage to the brain. Divalent metal ions, such as Cu2+ and Zn2+, bind to cellular prion protein (PrPC) at octapeptide repeat (OR) and non-OR sites within the N-terminal half of the protein but information on the impact of such binding on conversion to the misfolded isoform often derives from studies using either OR and non-OR peptides or bacterially-expressed recombinant PrP. Here we created new transgenic mouse lines expressing PrP with disrupted copper binding sites within all four histidine-containing OR's (sites 1-4, H60G, H68G, H76G, H84G, "TetraH>G" allele) or at site 5 (composed of residues His-95 and His-110; "H95G" allele) and monitored the formation of misfolded PrP in vivo. Novel transgenic mice expressing PrP(TetraH>G) at levels comparable to wild-type (wt) controls were susceptible to mouse-adapted scrapie strain RML but showed significantly prolonged incubation times. In contrast, amino acid replacement at residue 95 accelerated disease progression in corresponding PrP(H95G) mice. Neuropathological lesions in terminally ill transgenic mice were similar to scrapie-infected wt controls, but less severe. The pattern of PrPSc deposition, however, was not synaptic as seen in wt animals, but instead dense globular plaque-like accumulations of PrPSc in TgPrP(TetraH>G) mice and diffuse PrPSc deposition in (TgPrP(H95G) mice), were observed throughout all brain sections. We conclude that OR and site 5 histidine substitutions have divergent phenotypic impacts and that cis interactions between the OR region and the site 5 region modulate pathogenic outcomes by affecting the PrP globular domain.


Asunto(s)
Histidina/química , Proteínas Priónicas/química , Scrapie/patología , Animales , Ratones , Ratones Transgénicos
11.
Neuroreport ; 17(5): 527-30, 2006 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-16543819

RESUMEN

In normal brains and cultured cells, cellular prion protein (PrP) is partially found as N-terminally truncated fragments, designated C1 and C2. The cleavage of recombinant PrP to a fragment corresponding to C1 can be mediated by the protease plasmin (Pln) in vitro, suggesting that plasmin might be responsible for the generation of the C1 fragment in vivo as well. The cleavage pattern of PrP found in both brain lysates and other tissues of plasminogen knock-out mice, however, is unaltered. The presence of C1 fragment in homogenates from plasminogen-deficient mice in a comparable ratio with full-length PrP as can be found in wild-type animals indicates that other proteases in addition to plasmin are responsible for PrP cleavage in vivo.


Asunto(s)
Plasminógeno/fisiología , Priones/metabolismo , Animales , Western Blotting , Fibrinolisina/metabolismo , Genotipo , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasminógeno/deficiencia , Plasminógeno/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Biochem Biophys Methods ; 63(3): 213-21, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15967508

RESUMEN

Prions, which mainly consist of the scrapie isoform of the prion protein (PrP(Sc)), induce the misfolding of the physiological prion protein (PrP(C)). The Protein Misfolding Cyclic Amplification (PMCA), a process consisting of sonication and incubation, is one of the few methods thought to model autocatalytic prion replication and generation of proteinase K (PK)-resistant PrP (PrPres) in vitro. Here we show for the first time that the amplification may be achieved through direct as well as indirect sonication (water bath sonication using sealed sample containers), allowing the PMCA method to be automated. The automated method may serve as a valuable tool in high throughput screening for the diagnosis or compound identification for treatment of prion disease. The in vitro amplification process is weakly facilitated by divalent cations such as Mn, Zn and Ni, but not Cu, however, the presence of metal ions decreases the stability of PrPres against proteinase K digestion.


Asunto(s)
Biofisica/métodos , Priones/química , Animales , Automatización , Cationes , Cobre/química , Cricetinae , Ácido Edético/farmacología , Endopeptidasa K/química , Iones , Magnesio/química , Mesocricetus , Níquel/química , Pliegue de Proteína , Sonicación , Temperatura , Factores de Tiempo , Agua/química , Zinc/química
14.
Biochem J ; 382(Pt 1): 353-62, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15130091

RESUMEN

Dewaste et al. [Dewaste, Moreau, De Smedt, Bex, De Smedt, Wuytaack, Missiaen and Erneux (2003) Biochem. J. 374, 41-49] showed that over-expressed EGFP (enhanced green fluorescent protein) fused to Ins(1,4,5)P3 3-kinase B (IP3K-B) co-localizes with the cytoskeleton, as well as with the endoplasmic reticulum and the plasma membrane. The domains responsible for these subcellular localizations are not yet identified. For the endogenous enzyme, we confirmed both actin and endoplasmic reticulum localization by employing a high affinity antibody against IP3K-B. F-actin targeting is exclusively dependent on the non-catalytic N-terminal region of IP3K-B. By expressing fragments of this N-terminal domain as EGFP-fusion proteins and inspecting transfected cells by confocal microscopy, we characterized a distinct 63-amino-acid domain comprising amino acids 108-170 of the enzyme which is responsible for F-actin targeting. A truncation of this fragment from both sides revealed that the full size of this segment is essential for this function. Deletion of this segment in a full-length over-expressed IP3K-B-EGFP-fusion protein completely abolished F-actin interaction. Direct interaction of this actin-binding segment with only F-actin, but not with G-actin, was observed in vitro using a bacterially expressed, affinity-purified GST (glutathione S-transferase)-Rattus norvegicus IP3K (aa 108-170) fusion protein. Helix-breaking mutations within this isolated segment abolished the F-actin binding properties both in vitro and when over-expressed in cells, indicating that an intact secondary structure is essential for actin targeting. The segment shows sequence similarities to the actin-binding region in IP3K-A, but no similarity to other actin-binding domains.


Asunto(s)
Actinas/metabolismo , Péptidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Citoesqueleto de Actina/química , Animales , Western Blotting/métodos , Línea Celular , Clonación Molecular , Citoesqueleto/química , Citoesqueleto/enzimología , Retículo Endoplásmico/química , Retículo Endoplásmico/enzimología , Humanos , Isoenzimas/biosíntesis , Túbulos Renales Proximales/química , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/metabolismo , Microscopía Fluorescente/métodos , Células PC12/química , Células PC12/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Estructura Terciaria de Proteína/fisiología , Ratas
15.
Eur J Cell Biol ; 90(6-7): 467-75, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21144616

RESUMEN

Receptors that belong to the family of death-receptors including TNF receptor-1 (TNF-R1), CD95 (Fas, APO-1) and TRAIL receptors (TRAIL-R1, TRAIL R2/DR4/DR5) transduce signals resulting in entirely different biological outcomes: They promote cell death via apoptosis but are also capable of inducing anti-apoptotic signals through the transcription factor nuclear factor NF-κB or activation of the proliferative MAPK/ERK protein kinase cascade resulting in cell protection and tissue regeneration. Recent findings revealed a regulatory role of receptor internalization and its intracellular trafficking in selectively transmitting signals that lead either to apoptosis or to the survival of the cell, providing a clue to the understanding of these contradictory biological phenomena. In this chapter we review our data obtained during the Collaborative Research Center 415 (CRC 415) focusing on the compartmentalization of TNF-R1 and CD95 pro and anti-apoptotic signaling. We will address the role of internalization in determining the fate of the receptors. We suggest that fusion of internalized TNF-receptosomes with trans-Golgi vesicles is a novel mechanism to transduce death signals along the endosomal trafficking route. The roles of acid sphingomyelinase, the lipid second messenger ceramide, and the aspartate-protease cathepsin D as novel players in the cell death scenario is also highlighted. We report on the regulation of NF-κB signaling by recruitment of the endosomal E3-ubiquitin ligases CARP-2 and CARP-1 during TNF-receptosome trafficking. The biological significance of TNF receptor-1 compartmentalization is demonstrated by the strategy of adenoviruses to impede TNF-R1 internalization and by this preventing host cell apoptosis.


Asunto(s)
Receptores de Muerte Celular/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo , Animales , Apoptosis/fisiología , Humanos , Transducción de Señal , Fracciones Subcelulares/metabolismo
16.
ChemMedChem ; 6(10): 1928-37, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21755599

RESUMEN

Transmissible spongiform encephalopathies (TSE) or prion diseases belong to a category of fatal and so far untreatable neurodegenerative conditions. All prion diseases are characterized by both degeneration in the central nervous system (CNS) in humans and animals and the deposition and accumulation of Proteinase K-resistant prion protein (PrP(res)). Until now, no pharmaceutical product has been available to cure these diseases or to alleviate their associated symptoms. Here, a cell-culture screening system is described that allows for the large-scale analysis of the PrP(res) inhibitory potential of a library of compounds and the identification of structural motifs leading potent compounds able to cause PrP(res) clearance at the cellular level. Based on different scrapie-infected cell lines, 10,000 substances were tested, out of which 530 potential inhibitors were identified. After re-screening and validation using a series of dilutions, 14 compounds were identified as the most effective. These 14 compounds were then used for therapeutic studies in a mouse bioassay to test and verify their in vivo potency. Two compounds exhibited therapeutic potential in the mouse model by significantly extending the survival time of intracerebrally infected mice, when treated 90 days after infection with scrapie.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Proteínas PrPSc/antagonistas & inhibidores , Proteínas PrPSc/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Ratones , Proteínas PrPSc/metabolismo , Scrapie/tratamiento farmacológico
17.
J Mol Med (Berl) ; 88(7): 729-40, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20354842

RESUMEN

Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) and agonistic anti-DR4/TRAIL-R1 and anti-DR5/TRAIL-R2 antibodies are currently under clinical investigation for treatment of different malignancies. TRAIL activates DR4 and DR5 and thereby triggers apoptotic and non-apoptotic signaling pathways, but possible different roles of DR4 or DR5 in these responses has poorly been addressed so far. In the present work, we analyzed cell viability, DISC formation as well as IL-8 and NF-kappaB activation side by side in responses to TRAIL and agonistic antibodies against DR4 (mapatumumab) and against DR5 (lexatumumab) in pancreatic ductal adenocarcinoma cells. We found that all three reagents are able to activate cell death and pro-inflammatory signaling. Death-inducing signaling complex (DISC) analysis revealed that mapatumumab and lexatumumab induce formation of homocomplexes of either DR4 or DR5, whereas TRAIL additionally stimulated the formation of heterocomplexes of both receptors. Notably, blocking of receptors using DR4- and DR5-specific Fab fragments indicated that TRAIL exerted its function predominantly via DR4. Interestingly, inhibition of PKC by Goe6983 enabled DR5 to trigger apoptotic signaling in response to TRAIL and also strongly enhanced lexatumumab-mediated cell death. Our results suggest the existence of mechanisms that silence DR5 for TRAIL- but not for agonistic-antibody treatment.


Asunto(s)
Neoplasias Pancreáticas/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Interleucina-8/metabolismo , Células Jurkat , FN-kappa B/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Factor de Necrosis Tumoral/genética
18.
Biopolymers ; 88(6): 840-7, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17922496

RESUMEN

The prion protein is usually pictured as globular structured C-terminal domain that is linked to an extended flexible N-terminal tail. However, in its physiological form, it is a glycoprotein tethered to the cell surface via a C-terminal GPI anchor. The low solubility of PrP even without GPI anchor and its strong tendency for aggregation has forced most structural investigations to be performed at low pH and mostly with N-terminally truncated variants. In the present study, we have used a synthetic peptide related to the PrP tetra-octarepeat region, i.e., the sequence (Pro-His-Gly-Gly-Gly-Trp-Gly-Gln)(4), for NMR structural analysis of its preferred conformation in DPC micelles as membrane mimic. Well-defined and identical loops are observed between the four octarepeats that are linked by flexible Gly-Gly-Gly sequences. Interaction with the micelles is mainly through the tryptophan residues that appear to act as anchors. Copper binding to the peptide in the presence of DPC micelles revealed marked conformational rearrangements although binding to the micelles is preserved. Interestingly, titration experiments point to cooperative effects for the four binding sites. A destabilization of the DPC micelles by the peptide parallels the destabilizing effect of the prion protein on membranes so that the octarepeat region appears to be very membrane-active. How the physico-chemical properties reported here are linked to the function and significance of the prion protein remains a puzzle as long as the functional mechanism of the prion protein is not precisely elucidated. Nevertheless, our results emphasize the strong influence of the (membrane) environment on the PrP properties.


Asunto(s)
Cobre/química , Membranas Artificiales , Oligopéptidos/química , Fosforilcolina/análogos & derivados , Priones/química , Sitios de Unión , Dicroismo Circular/métodos , Humanos , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética/métodos , Micelas , Microscopía Fluorescente/métodos , Modelos Moleculares , Imitación Molecular , Oligopéptidos/síntesis química , Fosforilcolina/química , Conformación Proteica , Soluciones/química , Termodinámica
19.
Eur Biophys J ; 36(3): 239-52, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17225136

RESUMEN

The cellular prion protein (PrP(C)) is a Cu(2+) binding protein connected to the outer cell membrane. The molecular features of the Cu(2+) binding sites have been investigated and characterized by spectroscopic experiments on PrP(C)-derived peptides and the recombinant human full-length PrP(C )(hPrP-[23-231]). The hPrP-[23-231] was loaded with (63)Cu under slightly acidic (pH 6.0) or neutral conditions. The PrP(C)/Cu(2+)-complexes were investigated by extended X-ray absorption fine structure (EXAFS), electron paramagnetic resonance (EPR), and electron nuclear double resonance (ENDOR). For comparison, peptides from the copper-binding octarepeat domain were investigated in different environments. Molecular mechanics computations were used to select sterically possible peptide/Cu(2+) structures. The simulated EPR, ENDOR, and EXAFS spectra of these structures were compared with our experimental data. For a stoichiometry of two octarepeats per copper the resulting model has a square planar four nitrogen Cu(2+) coordination. Two nitrogens belong to imidazole rings of histidine residues. Further ligands are two deprotonated backbone amide nitrogens of the adjacent glycine residues and an axial oxygen of a water molecule. Our complex model differs significantly from those previously obtained for shorter peptides. Sequence context, buffer conditions and stoichiometry of copper show marked influence on the configuration of copper binding to PrP(C).


Asunto(s)
Cobre/química , Modelos Químicos , Modelos Moleculares , Proteínas PrPC/química , Proteínas PrPC/ultraestructura , Sitios de Unión , Simulación por Computador , Humanos , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína
20.
J Biol Chem ; 281(14): 9373-84, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16455657

RESUMEN

The misfolded infectious isoform of the prion protein (PrP(Sc)) is thought to replicate in an autocatalytic manner by converting the cellular form (PrP(C)) into its pathogenic folding variant. The similarity in the amino acid sequence of PrP(C) and PrP(Sc) influences the conversion efficiency and is considered as the major determinant for the species barrier. We performed in vitro conversion reactions on wild-type and mutated PrP(C) to determine the role of the primary sequence for the high susceptibility of bank voles to scrapie. Different conversion efficiencies obtained with bank vole and mouse PrP(C) in reactions with several prion strains were due to differences at amino acid residues 155 and 170. However, the conversion efficiencies obtained with mouse and vole PrP(C) in reactions with sheep scrapie did not correlate with the susceptibility of the respective species to this prion strain. This discrepancy between in vitro and in vivo data may indicate that at least in the case of scrapie transmission to bank voles additional host factors can strongly modulate the species barrier. Furthermore, in vitro conversion reactions with different prion strains revealed that the degree of alteration of the conversion efficiency induced by amino acid exchanges was varying according to the prion strain. These results support the assumption that the repertoire of conformations adopted by a certain PrP(C) primary sequence is decisive for its convertibility to the strain-specific PrP(Sc) conformation.


Asunto(s)
Arvicolinae/fisiología , Proteínas PrPC/química , Proteínas PrPSc/química , Conformación Proteica , Scrapie/transmisión , Secuencia de Aminoácidos , Animales , Susceptibilidad a Enfermedades , Ratones , Datos de Secuencia Molecular , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA