Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Biol Chem ; 296: 100631, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33823153

RESUMEN

TREM2 is a pattern recognition receptor, expressed on microglia and myeloid cells, detecting lipids and Aß and inducing an innate immune response. Missense mutations (e.g., R47H) of TREM2 increase risk of Alzheimer's disease (AD). The soluble ectodomain of wild-type TREM2 (sTREM2) has been shown to protect against AD in vivo, but the underlying mechanisms are unclear. We show that Aß oligomers bind to cellular TREM2, inducing shedding of the sTREM2 domain. Wild-type sTREM2 bound to Aß oligomers (measured by single-molecule imaging, dot blots, and Bio-Layer Interferometry) inhibited Aß oligomerization and disaggregated preformed Aß oligomers and protofibrils (measured by transmission electron microscopy, dot blots, and size-exclusion chromatography). Wild-type sTREM2 also inhibited Aß fibrillization (measured by imaging and thioflavin T fluorescence) and blocked Aß-induced neurotoxicity (measured by permeabilization of artificial membranes and by loss of neurons in primary neuronal-glial cocultures). In contrast, the R47H AD-risk variant of sTREM2 is less able to bind and disaggregate oligomeric Aß but rather promotes Aß protofibril formation and neurotoxicity. Thus, in addition to inducing an immune response, wild-type TREM2 may protect against amyloid pathology by the Aß-induced release of sTREM2, which blocks Aß aggregation and neurotoxicity. In contrast, R47H sTREM2 promotes Aß aggregation into protofibril that may be toxic to neurons. These findings may explain how wild-type sTREM2 apparently protects against AD in vivo and why a single copy of the R47H variant gene is associated with increased AD risk.


Asunto(s)
Péptidos beta-Amiloides/química , Amiloide/química , Glicoproteínas de Membrana/fisiología , Proteínas Mutantes/metabolismo , Mutación , Neuronas/patología , Síndromes de Neurotoxicidad/patología , Receptores Inmunológicos/fisiología , Enfermedad de Alzheimer , Amiloide/metabolismo , Animales , Ratones , Ratones Noqueados , Proteínas Mutantes/genética , Neuronas/metabolismo , Síndromes de Neurotoxicidad/etiología
2.
J Cell Sci ; 126(Pt 9): 2124-33, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23447676

RESUMEN

Parkin and DJ-1 are two multi-functional proteins linked to autosomal recessive early-onset Parkinson's disease (PD) that have been shown to functionally interact by as-yet-unknown mechanisms. We have delineated the mechanisms by which parkin controls DJ-1. Parkin modulates DJ-1 transcription and protein levels via a signaling cascade involving p53 and the endoplasmic reticulum (ER)-stress-induced active X-box-binding protein-1S (XBP-1S). Parkin triggers the transcriptional repression of p53 while p53 downregulates DJ-1 protein and mRNA expressions. We show that parkin-mediated control of DJ-1 is fully p53-dependent. Furthermore, we establish that p53 lowers the protein and mRNA levels of XBP-1S. Accordingly, we show that parkin ultimately upregulates XBP-1 levels. Subsequently, XBP-1S physically interacts with the DJ-1 promoter, thereby enhancing its promoter trans-activation, mRNA levels and protein expression. This data was corroborated by the examination of DJ-1 in both parkin- and p53-null mice brains. This transcriptional cascade is abolished by pathogenic parkin mutations and is independent of its ubiquitin-ligase activity. Our data establish a parkin-dependent ER-stress-associated modulation of DJ-1 and identifies p53 and XBP-1 as two major actors acting downstream of parkin in this signaling cascade in cells and in vivo. This work provides a mechanistic explanation for the increase in the unfolded protein response observed in PD pathology, i.e. that it is due to a defect in parkin-associated control of DJ-1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Mutantes/genética , Proteínas Oncogénicas/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Peroxirredoxinas , Proteína Desglicasa DJ-1 , Factores de Transcripción del Factor Regulador X , Transducción de Señal/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Proteína 1 de Unión a la X-Box
3.
J Neurosci ; 32(34): 11820-34, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22915123

RESUMEN

In Alzheimer disease (AD), the perturbation of the endoplasmic reticulum (ER) calcium (Ca²âº) homeostasis has been linked to presenilins, the catalytic core in γ-secretase complexes cleaving the amyloid precursor protein (APP), thereby generating amyloid-ß (Aß) peptides. Here we investigate whether APP contributes to ER Ca²âº homeostasis and whether ER Ca²âº could in turn influence Aß production. We show that overexpression of wild-type human APP (APP(695)), or APP harboring the Swedish double mutation (APP(swe)) triggers increased ryanodine receptor (RyR) expression and enhances RyR-mediated ER Ca²âº release in SH-SY5Y neuroblastoma cells and in APP(swe)-expressing (Tg2576) mice. Interestingly, dantrolene-induced lowering of RyR-mediated Ca²âº release leads to the reduction of both intracellular and extracellular Aß load in neuroblastoma cells as well as in primary cultured neurons derived from Tg2576 mice. This Aß reduction can be accounted for by decreased Thr-668-dependent APP phosphorylation and ß- and γ-secretases activities. Importantly, dantrolene diminishes Aß load, reduces Aß-related histological lesions, and slows down learning and memory deficits in Tg2576 mice. Overall, our data document a key role of RyR in Aß production and learning and memory performances, and delineate RyR-mediated control of Ca²âº homeostasis as a physiological paradigm that could be targeted for innovative therapeutic approaches.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Enfermedad de Alzheimer/genética , Aminofenoles/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Encéfalo/citología , Cafeína/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/uso terapéutico , Células Cultivadas , Citosol/efectos de los fármacos , Citosol/metabolismo , Dantroleno/farmacología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Inhibidores Enzimáticos/uso terapéutico , Conducta Exploratoria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Maleimidas/uso terapéutico , Aprendizaje por Laberinto/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Ratones Transgénicos , Relajantes Musculares Centrales/farmacología , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/patología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Purinas/uso terapéutico , ARN Mensajero/metabolismo
4.
Bioorg Med Chem ; 21(4): 1018-29, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23294826

RESUMEN

The 3-alkoxy-7-amino-4-chloro-isocoumarins JLK-6 and JLK-2 have been shown to markedly reduce the production of Amyloid ß-peptide (Aß) by Amyloid-ß Precursor Protein (APP) expressing HEK293 cells by affecting the γ-secretase cleavage of APP, with no effect on the cleavage of the Notch receptor. This suggested that these compounds do not directly inhibit the presenilin-dependent γ-secretase complex but more likely interfere with an upstream target involved in γ-secretase-associated pathway. The mechanism of action of these compounds is unknown and there are high fundamental and therapeutical interests to unravel their target. Isocoumarin compounds were previously shown to behave as potent mechanism-based irreversible inhibitors of serine proteases, suggesting that the JLK-directed target could belong to such enzyme family. To get further insight into structure-activity relationships and to develop more potent isocoumarin derivatives, we have synthesized and evaluated a series of isocoumarin analogues with modifications at positions 3, 4 and 7. In particular, the 7-amino group was substituted with either acyl, urethane, alkyl or aryl groups, which could represent additional interaction sites. Altogether, the results highlighted the essential integrity of the 3-alkoxy-7-amino-4-chloro-isocoumarin scaffold for Aß-lowering activity and supported the involvement of a serine protease, or may be more generally, a serine hydrolase. The newly reported 7-N-alkyl series produced the most active compounds with an IC(50) between 10 and 30µM. Finally, we also explored peptide boronates, a series of reversible serine protease inhibitors, previously shown to also lower cellular Aß production. The presented data suggested they could act on the same target or interfere with the same pathway as isocoumarins derivatives.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Isocumarinas/química , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Ácidos Borónicos/química , Células HEK293 , Humanos , Isocumarinas/síntesis química , Isocumarinas/metabolismo , Serina Proteasas/metabolismo , Transfección
5.
J Cell Sci ; 122(Pt 21): 4003-8, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19889971

RESUMEN

The senile plaques found in the brains of patients with Alzheimer's disease are mainly due to the accumulation of amyloid beta-peptides (A beta) that are liberated by gamma-secretase, a high molecular weight complex including presenilins, PEN-2, APH-1 and nicastrin. The depletion of each of these proteins disrupts the complex assembly into a functional protease. Here, we describe another level of regulation of this multimeric protease. The depletion of both presenilins drastically reduces Pen2 mRNA levels and its promoter transactivation. Furthermore, overexpression of presenilin-1 lowers Pen2 promoter transactivation, a phenotype abolished by a double mutation known to prevent presenilin-dependent gamma-secretase activity. PEN-2 expression is decreased by depletion of beta-amyloid precursor protein (APP) and increased by the APP intracellular domain (AICD). We show that AICD and APP complement for Pen2 mRNA levels in APP/APLP1-2 knockout fibroblasts. Interestingly, overexpression of presenilin-2 greatly increases Pen2 promoter transactivation. The opposite effect triggered by both presenilins was reminiscent of our previous study, which showed that these two proteins elicit antagonistic effects on p53. Therefore, we examined the contribution of p53 on Pen2 transcription. Pen2 promoter transactivation, and Pen2 mRNA and protein levels were drastically reduced in p53(-/-) fibroblasts. Furthermore, PEN-2 expression could be rescued by p53 complementation in p53- and APP-deficient cells. Interestingly, PEN-2 expression was also reduced in p53-deficient mouse brain. Overall, our study describes a p53-dependent regulation of PEN-2 expression by other members of the gamma-secretase complex, namely presenilins.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Proteínas de la Membrana/genética , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Presenilina-1/genética , Presenilina-2/genética , Proteína p53 Supresora de Tumor/genética
6.
J Neurochem ; 109(1): 248-56, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19187443

RESUMEN

Several lines of data previously indicated that N-terminally truncated forms of amyloid-beta (Abeta) peptides are likely the earliest and more abundant species immunohistochemically detectable in Alzheimer's disease-affected brains. It is noteworthy that the free N-terminal residue of full-length Abeta (fl-Abeta) is an aspartyl residue, suggesting that Abeta could be susceptible to exopeptidasic attack by aminopeptidase A (APA)-like proteases. In this context, we have examined whether APA could target Abeta peptides in both cell-free and cellular models. We first show that the general aminopeptidase inhibitor amastatin as well as two distinct aminopeptidase A inhibitors EC33 and pl302 both significantly increase the recovery of genuine fl-Abeta peptides generated by cells over-expressing Swedish-mutated beta amyloid precursor protein (APP) while the aminopeptidase N blocker pl250 did not modify fl-Abeta recovery. In agreement with this observation, we establish that over-expressed APA drastically reduces, in a calcium dependent manner, fl-Abeta but not APP IntraCellular Domain in a cell-free model of Abeta production. In agreement with the above data, we show that recombinant APA degrades fl-Abeta in a pl302-sensitive manner. Interestingly, we also show that EC33 and pl302 lower staurosporine-stimulated activation of caspase-3 in wild-type fibroblasts but not in betaAPP/beta-amyloid precursor protein-like protein 2 (APLP2) double knockout fibroblasts, suggesting that protecting endogenous fl-Abeta physiological production triggers neuroprotective phenotype. By contrast, EC33 does not modify staurosporine-induced caspase-3 activation in wild-type and Swedish-mutated betaAPP-HEK293 expressing cells that display exacerbated production of Abeta. Overall, our data establish that APA contributes to the N-terminal truncation of Abeta and suggest that this cleavage is likely abrogating a protective function associated with physiological but not supraphysiological levels of genuine fl-Abeta peptides.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Glutamil Aminopeptidasa/fisiología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Animales , Línea Celular , Glutamil Aminopeptidasa/antagonistas & inhibidores , Glutamil Aminopeptidasa/genética , Humanos , Ratones , Ratones Noqueados , Péptidos/fisiología , Estructura Terciaria de Proteína
7.
J Neurochem ; 110(1): 275-83, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19457123

RESUMEN

Amyloid-beta (Abeta) peptides production is thought to be a key event in the neurodegenerative process ultimately leading to Alzheimer's disease (AD) pathology. A bulk of studies concur to propose that the C-terminal moiety of Abeta is released from its precursor beta-amyloid precursor protein by a high molecular weight enzymatic complex referred to as gamma-secretase, that is composed of at least, nicastrin (NCT), Aph-1, Pen-2, and presenilins (PS) 1 or 2. They are thought to harbor the gamma-secretase catalytic activity. However, several lines of evidence suggest that additional gamma-secretase-like activities could potentially contribute to Abeta production. By means of a quenched fluorimetric substrate (JMV2660) mimicking the beta-amyloid precursor protein sequence targeted by gamma-secretase, we first show that as expected, this probe allows monitoring of an activity detectable in several cell systems including the neuronal cell line telencephalon specific murine neurons (TSM1). This activity is reduced by DFK167, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), and LY68458, three inhibitors known to functionally interact with PS. Interestingly, JMV2660 but not the unrelated peptide JMV2692, inhibits Abeta production in an in vitrogamma-secretase assay as expected from a putative substrate competitor. This activity is enhanced by PS1 and PS2 mutations known to be responsible for familial forms of AD and reduced by aspartyl mutations inactivating PS or in cells devoid of PS or NCT. However, we clearly establish that residual JMV2660-hydrolysing activity could be recovered in PS- and NCT-deficient fibroblasts and that this activity remained inhibited by DFK167. Overall, our study describes the presence of a proteolytic activity displaying gamma-secretase-like properties but independent of PS and still blocked by DFK167, suggesting that the PS-dependent complex could not be the unique gamma-secretase activity responsible for Abeta production and delineates PS-independent gamma-secretase activity as a potential additional therapeutic target to fight AD pathology.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/biosíntesis , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Presenilinas/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Línea Celular , Células Cultivadas , Dipéptidos/farmacología , Fibroblastos , Humanos , Ratones , Fragmentos de Péptidos/farmacología , Placa Amiloide/efectos de los fármacos , Placa Amiloide/metabolismo , Presenilinas/metabolismo
8.
Biochem Biophys Res Commun ; 371(1): 69-74, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18405662

RESUMEN

The presenilin (PS)-dependent gamma-secretase activity refers to a high molecular mass-complex including, besides PS1 or PS2, three other proteins recently identified, namely nicastrin, Aph-1, and Pen-2. This proteolytic complex has been shown to contribute to both gamma- and epsilon-cleavages of the beta-amyloid precursor protein (betaAPP), thereby generating beta-amyloid peptides (Abeta) and the APP intracellular domain (AICD), respectively. TMP21, a member of the p24 cargo protein family, was recently shown to interact with PS complexes. Interestingly, TMP21 modulates gamma-secretase-mediated Abeta production but does not regulate epsilon-secretase-derived AICD formation [F. Chen, H. Hasegawa, G. Schmitt-ulms, T. Kawarai, C. Bohm, T. Katayama, Y. Gu, N. Sanjo, M. Glista, E. Rogaeva, Y. Wakutami, R. Pardossi-Piquard, X. Ruan, A. Tandon, F. Checler, P. Marambaud, K. Hansen, D. Westaway, P. St. George-Hyslop, P. Fraser, TMP21 is a presenilin complex component that modulates gamma- but not epsilon-secretase activities, Nature 440 (2006) 1208-1212]. Here we investigate the functional incidence of the over-expression or depletion of TMP21 on both intracellular and secreted Abeta recoveries and AICD-associated phenotypes. First we confirm that TMP21 depletion yields increased levels of secreted Abeta40. However, we demonstrate that both staurosporine-stimulated caspase-3 activation, p53 and neprilysin expression and activity were not affected by TMP21 over-expression or depletion. Overall, our functional data further reinforce the view that TMP21 behaves as a regulator of gamma- but not epsilon-cleavages generated by PS-dependent gamma-secretase complex.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Neprilisina/genética , Neprilisina/metabolismo , Proteínas de Transporte Nucleocitoplasmático , Fragmentos de Péptidos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
J Neurosci ; 26(23): 6377-85, 2006 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-16763046

RESUMEN

Presenilins (PSs) are part of the gamma-secretase complex that produces the amyloid beta-peptide (Abeta) from its precursor [beta-amyloid precursor protein (betaAPP)]. Mutations in PS that cause familial Alzheimer's disease (FAD) increase Abeta production and trigger p53-dependent cell death. We demonstrate that PS deficiency, catalytically inactive PS mutants, gamma-secretase inhibitors, and betaAPP or amyloid precursor protein-like protein 2 (APLP2) depletion all reduce the expression and activity of p53 and lower the transactivation of its promoter and mRNA expression. p53 expression also is diminished in the brains of PS- or betaAPP-deficient mice. The gamma- and epsilon-secretase-derived amyloid intracellular C-terminal domain (AICD) fragments (AICDC59 and AICDC50, respectively) of betaAPP trigger p53-dependent cell death and increase p53 activity and mRNA. Finally, PS1 mutations enhance p53 activity in human embryonic kidney 293 cells and p53 expression in FAD-affected brains. Thus our study shows that AICDs control p53 at a transcriptional level, in vitro and in vivo, and that FAD mutations increase p53 expression and activity in cells and human brains.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Anciano , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas , Encéfalo/metabolismo , Estudios de Casos y Controles , Caspasa 3 , Caspasas/metabolismo , Muerte Celular , Línea Celular , Activación Enzimática , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Mutación , Proteínas del Tejido Nervioso/deficiencia , Fragmentos de Péptidos/metabolismo , Presenilina-1 , Presenilina-2 , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Transcripción Genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
10.
Curr Alzheimer Res ; 4(4): 423-6, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17908046

RESUMEN

Amyloid beta-peptide (Abeta), which plays a central role in Alzheimer Disease, is generated by presenilin-dependent and presenilin-independent gamma-secretase cleavages of beta-amyloid precursor protein (betaAPP). We report that the presenilins (PS1 and PS2) also regulate p53-associated cell death. Thus, we established that PS deficiency, catalytically inactive PS mutants, gamma-secretase inhibitors and betaAPP or APLP2 depletion reduced the expression and activity of p53, and lowered the transactivation of its promoter and mRNA levels. p53 expression was also reduced in the brains or betaAPP-deficient mice or in brains where both PS had been invalidated by double conditional knock out. AICDC59 and AICDC50, the gamma- and epsilon-secretase-derived C-terminal fragments of betaAPP, respectively, trigger the activation of caspase-3, p53-dependent cell death, and increase p53 activity and mRNA. Finally, HEK293 cells expressing PS1 harboring familial AD (FAD) mutations or FAD-affected brains, all display enhanced p53 activity and p53 expression. Our studies demonstrate that AICDs control p53 at a transcriptional level, in vitro and in vivo and unravel a still unknown function for presenilins.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Péptidos beta-Amiloides/química , Animales , Humanos , Modelos Biológicos , Estructura Terciaria de Proteína
11.
Sci Rep ; 7: 40313, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28098160

RESUMEN

The prion protein (PrP) evolved from the subbranch of ZIP metal ion transporters comprising ZIPs 5, 6 and 10, raising the prospect that the study of these ZIPs may reveal insights relevant for understanding the function of PrP. Building on data which suggested PrP and ZIP6 are critical during epithelial-to-mesenchymal transition (EMT), we investigated ZIP6 in an EMT paradigm using ZIP6 knockout cells, mass spectrometry and bioinformatic methods. Reminiscent of PrP, ZIP6 levels are five-fold upregulated during EMT and the protein forms a complex with NCAM1. ZIP6 also interacts with ZIP10 and the two ZIP transporters exhibit interdependency during their expression. ZIP6 contributes to the integration of NCAM1 in focal adhesion complexes but, unlike cells lacking PrP, ZIP6 deficiency does not abolish polysialylation of NCAM1. Instead, ZIP6 mediates phosphorylation of NCAM1 on a cluster of cytosolic acceptor sites. Substrate consensus motif features and in vitro phosphorylation data point toward GSK3 as the kinase responsible, and interface mapping experiments identified histidine-rich cytoplasmic loops within the ZIP6/ZIP10 heteromer as a novel scaffold for GSK3 binding. Our data suggests that PrP and ZIP6 inherited the ability to interact with NCAM1 from their common ZIP ancestors but have since diverged to control distinct posttranslational modifications of NCAM1.


Asunto(s)
Antígeno CD56/metabolismo , Proteínas de Transporte de Catión/metabolismo , Transición Epitelial-Mesenquimal , Adhesiones Focales/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Catión/química , Citoesqueleto/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Histidina/metabolismo , Humanos , Integrinas/metabolismo , Ratones , Modelos Biológicos , Ácido N-Acetilneuramínico/metabolismo , Fosforilación , Proteínas Priónicas/metabolismo , Unión Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína
12.
EMBO Mol Med ; 9(10): 1366-1378, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28855301

RESUMEN

We have characterised the proteolytic cleavage events responsible for the shedding of triggering receptor expressed on myeloid cells 2 (TREM2) from primary cultures of human macrophages, murine microglia and TREM2-expressing human embryonic kidney (HEK293) cells. In all cell types, a soluble 17 kDa N-terminal cleavage fragment was shed into the conditioned media in a constitutive process that is inhibited by G1254023X and metalloprotease inhibitors and siRNA targeting ADAM10. Inhibitors of serine proteases and matrix metalloproteinases 2/9, and ADAM17 siRNA did not block TREM2 shedding. Peptidomimetic protease inhibitors highlighted a possible cleavage site, and mass spectrometry confirmed that shedding occurred predominantly at the H157-S158 peptide bond for both wild-type and H157Y human TREM2 and for the wild-type murine orthologue. Crucially, we also show that the Alzheimer's disease-associated H157Y TREM2 variant was shed more rapidly than wild type from HEK293 cells, possibly by a novel, batimastat- and ADAM10-siRNA-independent, sheddase activity. These insights offer new therapeutic targets for modulating the innate immune response in Alzheimer's and other neurological diseases.


Asunto(s)
Enfermedad de Alzheimer/genética , Glicoproteínas de Membrana/metabolismo , Proteolisis , Receptores Inmunológicos/metabolismo , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Animales Recién Nacidos , Medios de Cultivo Condicionados , Células HEK293 , Humanos , Cetocolesteroles/farmacología , Macrófagos/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Inmunológicos/genética
13.
J Mol Cell Biol ; 5(2): 132-42, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23359614

RESUMEN

We previously established that besides its canonical function as E3-ubiquitin ligase, parkin also behaves as a transcriptional repressor of p53. Here we show that parkin differently modulates presenilin-1 and presenilin-2 expression and functions at transcriptional level. Thus, parkin enhances/reduces the protein expression, promoter activity and mRNA levels of presenilin-1 and presenilin-2, respectively, in cells and in vivo. This parkin-associated function is independent of its ubiquitin-ligase activity and remains unrelated to its capacity to repress p53. Accordingly, physical interaction of endogenous or overexpressed parkin with presenilins promoters is demonstrated by chromatin immunoprecipitation assays (ChIP). Furthermore, we identify a consensus sequence, the deletion of which abolishes parkin-dependent modulation of presenilins-1/2 and p53 promoter activities. Interestingly, electrophoretic mobility shift assays (EMSA) revealed a physical interaction between this consensus sequence and wild-type but not mutated parkin. Finally, we demonstrate that the RING1-IBR-RING2 domain of parkin harbors parkin's potential to modulate presenilins promoters. This transcriptional control impacts on presenilins-associated phenotypes, since parkin increases presenilin-1-associated γ-secretase activity and reduces presenilin-2-linked caspase-3 activation. Overall, our data delineate a promoter responsive element targeted by parkin that drives differential regulation of presenilin-1 and presenilin-2 transcription with functional consequences for γ-secretase activity and cell death.


Asunto(s)
Presenilina-1/metabolismo , Presenilina-2/metabolismo , Regiones Promotoras Genéticas/fisiología , Transcripción Genética/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Muerte Celular/fisiología , Activación Enzimática/fisiología , Ratones , Ratones Noqueados , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Presenilina-1/genética , Presenilina-2/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
14.
J Alzheimers Dis ; 30(1): 145-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22406447

RESUMEN

One of the major pathological hallmarks of brains affected with Alzheimer's disease (AD) is the senile plaque, an extracellular deposit mainly composed of a set of highly insoluble peptides of various lengths (39-43 amino acids) referred to as amyloid-ß (Aß) peptides. Aß peptides are derived from combined proteolytic cleavages undergone on the amyloid-ß protein precursor (AßPP) by a set of enzymes called secretases. Several lines of anatomical and biological evidence suggest that Aß peptides would not account for all pathological stigmata and molecular dysfunctions taking place in AD. In amyloidogenic and non-amyloidogenic pathways, AßPP first undergoes ß- or α-secretases-mediated cleavages yielding C99 and C83, respectively. These two membrane-embedded C-terminal fragments are both potential targets of subsequent γ-secretase-mediated proteolysis. The latter cleavage not only generates either p3 or Aß peptides but similarly gives rise to an AßPP IntraCellular Domain (AICD fragment) that could modulate the transcription of several genes linked to AD pathology. It is therefore striking that AICD theoretically derives from both amyloidogenic and non-amyloidogenic AßPP processing pathways. Here we show that AICD predominantly derives from C99 by means of recombinant substrates and transiently transfected cells expressing C99. Our data suggest a preferred pathogenic pathway for AICD production and suggests that this fragment, in addition to C99 and Aß peptides, could contribute to AD pathology.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Líquido Intracelular/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Línea Celular Transformada , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Humanos , Inmunoprecipitación , Líquido Intracelular/efectos de los fármacos , Neuroblastoma/patología , Fragmentos de Péptidos/farmacología , Estructura Terciaria de Proteína , Proteolisis/efectos de los fármacos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato/efectos de los fármacos , Factores de Tiempo , Transfección
15.
J Biol Chem ; 284(24): 16298-16307, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19369254

RESUMEN

Complexes involved in the gamma/epsilon-secretase-regulated intramembranous proteolysis of substrates such as the amyloid-beta precursor protein are composed primarily of presenilin (PS1 or PS2), nicastrin, anterior pharynx defective-1 (APH1), and PEN2. The presenilin aspartyl residues form the catalytic site, and similar potentially functional polar transmembrane residues in APH1 have been identified. Substitution of charged (E84A, R87A) or polar (Q83A) residues in TM3 had no effect on complex assembly or activity. In contrast, changes to either of two highly conserved histidines (H171A, H197A) located in TM5 and TM6 negatively affected PS1 cleavage and altered binding to other secretase components, resulting in decreased amyloid generating activity. Charge replacement with His-to-Lys substitutions rescued nicastrin maturation and PS1 endoproteolysis leading to assembly of the formation of structurally normal but proteolytically inactive gamma-secretase complexes. Substitution with a negatively charged side chain (His-to-Asp) or altering the structural location of the histidines also disrupted gamma-secretase binding and abolished functionality of APH1. These results suggest that the conserved transmembrane histidine residues contribute to APH1 function and can affect presenilin catalytic activity.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Presenilinas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico/genética , Catálisis , Células Cultivadas , Secuencia Conservada , Endopeptidasas , Fibroblastos/citología , Fibroblastos/fisiología , Histidina/genética , Humanos , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Péptido Hidrolasas/química , Estructura Terciaria de Proteína , Transfección
16.
J Biol Chem ; 283(15): 10037-47, 2008 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-18263584

RESUMEN

Beta-amyloid (Abeta) peptides that accumulate in Alzheimer disease are generated from the beta-amyloid precursor protein (betaAPP) by cleavages by beta-secretase BACE1 and by presenilin-dependent gamma-secretase activities. Very few data document a putative cross-talk between these proteases and the regulatory mechanisms underlying such interaction. We show that presenilin deficiency lowers BACE1 maturation and affects both BACE1 activity and promoter transactivation. The specific gamma-secretase inhibitor DFK167 triggers the decrease of BACE1 activity in wild-type but not in presenilin-deficient fibroblasts. This decrease is also elicited by catalytically inactive gamma-secretase. The overexpression of APP intracellular domain (AICD), the gamma/epsilon-secretase-derived C-terminal product of beta-amyloid precursor protein, does not modulate BACE1 activity or promoter transactivation in fibroblasts and does not alter BACE1 expression in AICD transgenic brains of mice. A DFK167-sensitive increase of BACE1 activity is observed in cells overexpressing APPepsilon (the N-terminal product of betaAPP generated by epsilon-secretase cleavage harboring the Abeta domain but lacking the AICD sequence), suggesting that the production of Abeta could account for the modulation of BACE1. Accordingly, we show that HEK293 cells overexpressing wild-type betaAPP exhibit a DFK167-sensitive increase in BACE1 promoter transactivation that is increased by the Abeta-potentiating Swedish mutation. This effect was mimicked by exogenous application of Abeta42 but not Abeta40 or by transient transfection of cDNA encoding Abeta42 sequence. The IkappaB kinase inhibitor BMS345541 prevents Abeta-induced BACE1 promoter transactivation suggesting that NFkappaB could mediate this Abeta-associated phenotype. Accordingly, the overexpression of wild-type or Swedish mutated betaAPP does not modify the transactivation of BACE1 promoter constructs lacking NFkappaB-responsive element. Furthermore, APP/beta-amyloid precursor protein-like protein deficiency does not affect BACE1 activity and expression. Overall, these data suggest that physiological levels of endogenous Abeta are not sufficient per se to modulate BACE1 promoter transactivation but that exacerbated Abeta production linked to wild-type or Swedish mutated betaAPP overexpression modulates BACE1 promoter transactivation and activity via an NFkappaB-dependent pathway.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , FN-kappa B/metabolismo , Elementos de Respuesta , Activación Transcripcional , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Fibroblastos/enzimología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/genética , Presenilinas/genética , Presenilinas/metabolismo , Inhibidores de Proteasas/farmacología , Estructura Terciaria de Proteína/genética , Ratas , Elementos de Respuesta/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
17.
J Biol Chem ; 282(14): 10516-25, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-17276981

RESUMEN

The presenilin-dependent gamma-secretase activity, which is responsible for the generation of amyloid beta-peptide, is a high molecular weight complex composed of at least four components, namely, presenilin-1 (or presenilin-2), nicastrin, Aph-1, and Pen-2. Previous data indicated that presenilins, which are thought to harbor the catalytic core of the complex, also control p53-dependent cell death. Whether the other components of the gamma-secretase complex could also modulate the cell death process in mammalian neurons remained to be established. Here, we examined the putative contribution of Aph-1 and Pen-2 in the control of apoptosis in TSM1 cells from a neuronal origin. We show by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and DNA fragmentation analyses that the overexpression of Aph-1a, Aph-1b, or Pen-2 drastically lowered staurosporine-induced cellular toxicity. In support of an apoptosis rather than necrosis process, Aph-1 and Pen-2 also lower staurosporine- and etoposide-induced caspase-3 expression and diminished caspase-3 activity and poly(ADP-ribose) polymerase inactivation. The Aph-1 and Pen-2 anti-apoptotic phenotype was associated with a drastic reduction of p53 expression and activity and lowered p53 mRNA transcription. Furthermore, the Aph-1- and Pen-2-associated reduction of staurosporine-induced caspase-3 activation was fully abolished by p53 deficiency. Conversely, Aph-1a, Aph-1b, and Pen-2 gene inactivation increases both caspase-3 activity and p53 mRNA levels. Finally, we show that Aph-1 and Pen-2 did not trigger an anti-apoptotic response in cells devoid of presenilins or nicastrin, whereas the protective response was still observed in fibroblasts devoid of beta-amyloid precursor protein and amyloid precursor protein like-protein 2. Furthermore, Aph-1- and Pen-2-associated protection against staurosporine-induced caspase-3 activation was not affected by the gamma-secretase inhibitors N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester and difluoromethylketone. Altogether, our study indicates that Aph-1 and Pen-2 trigger an anti-apoptotic response by lowering p53-dependent control of caspase-3. Our work also demonstrates that this phenotype is strictly dependent on the molecular integrity of the gamma-secretase complex but remains independent of the gamma-secretase catalytic activity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/biosíntesis , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Caspasa 3/metabolismo , Línea Celular , Endopeptidasas , Inhibidores Enzimáticos/toxicidad , Humanos , Glicoproteínas de Membrana/metabolismo , Péptido Hidrolasas , Fenotipo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Presenilinas/metabolismo , Estaurosporina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA