Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37446396

RESUMEN

The pan Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor fasudil acts as a vasodilator and has been used as a medication for post-cerebral stroke for the past 29 years in Japan and China. More recently, based on the involvement of ROCK inhibition in synaptic function, neuronal survival, and processes associated with neuroinflammation, it has been suggested that the drug may be repurposed for neurodegenerative diseases. Indeed, fasudil has demonstrated preclinical efficacy in many neurodegenerative disease models. To facilitate an understanding of the wider biological processes at play due to ROCK inhibition in the context of neurodegeneration, we performed a global gene expression analysis on the brains of Alzheimer's disease model mice treated with fasudil via peripheral IP injection. We then performed a comparative analysis of the fasudil-driven transcriptional profile with profiles generated from a meta-analysis of multiple neurodegenerative diseases. Our results show that fasudil tends to drive gene expression in a reverse sense to that seen in brains with post-mortem neurodegenerative disease. The results are most striking in terms of pathway enrichment analysis, where pathways perturbed in Alzheimer's and Parkinson's diseases are overwhelmingly driven in the opposite direction by fasudil treatment. Thus, our results bolster the repurposing potential of fasudil by demonstrating an anti-neurodegenerative phenotype in a disease context and highlight the potential of in vivo transcriptional profiling of drug activity.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Animales , Ratones , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Ratones Transgénicos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Quinasas Asociadas a rho/metabolismo
2.
Sci Rep ; 12(1): 18639, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36329114

RESUMEN

Clusterin (CLU) is one of the most significant genetic risk factors for late onset Alzheimer's disease (AD). However, the mechanisms by which CLU contributes to AD development and pathogenesis remain unclear. Studies have demonstrated that the trafficking and localisation of glycosylated CLU proteins is altered by CLU-AD mutations and amyloid-ß (Aß), which may contribute to AD pathogenesis. However, the roles of non-glycosylated and glycosylated CLU proteins in mediating Aß toxicity have not been studied in human neurons. iPSCs with altered CLU trafficking were generated following the removal of CLU exon 2 by CRISPR/Cas9 gene editing. Neurons were generated from control (CTR) and exon 2 -/- edited iPSCs and were incubated with aggregated Aß peptides. Aß induced changes in cell death and neurite length were quantified to determine if altered CLU protein trafficking influenced neuronal sensitivity to Aß. Finally, RNA-Seq analysis was performed to identify key transcriptomic differences between CLU exon 2 -/- and CTR neurons. The removal of CLU exon 2, and the endoplasmic reticulum (ER)-signal peptide located within, abolished the presence of glycosylated CLU and increased the abundance of intracellular, non-glycosylated CLU. While non-glycosylated CLU levels were unaltered by Aß25-35 treatment, the trafficking of glycosylated CLU was altered in control but not exon 2 -/- neurons. The latter also displayed partial protection against Aß-induced cell death and neurite retraction. Transcriptome analysis identified downregulation of multiple extracellular matrix (ECM) related genes in exon 2 -/- neurons, potentially contributing to their reduced sensitivity to Aß toxicity. This study identifies a crucial role of glycosylated CLU in facilitating Aß toxicity in human neurons. The loss of these proteins reduced both, cell death and neurite damage, two key consequences of Aß toxicity identified in the AD brain. Strikingly, transcriptomic differences between exon 2 -/- and control neurons were small, but a significant and consistent downregulation of ECM genes and pathways was identified in exon 2 -/- neurons. This may contribute to the reduced sensitivity of these neurons to Aß, providing new mechanistic insights into Aß pathologies and therapeutic targets for AD.


Asunto(s)
Enfermedad de Alzheimer , Clusterina , Humanos , Clusterina/genética , Clusterina/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo
3.
Brain Commun ; 4(6): fcac267, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36349119

RESUMEN

Establishing preclinical models of Alzheimer's disease that predict clinical outcomes remains a critically important, yet to date not fully realized, goal. Models derived from human cells offer considerable advantages over non-human models, including the potential to reflect some of the inter-individual differences that are apparent in patients. Here we report an approach using induced pluripotent stem cell-derived cortical neurons from people with early symptomatic Alzheimer's disease where we sought a match between individual disease characteristics in the cells with analogous characteristics in the people from whom they were derived. We show that the response to amyloid-ß burden in life, as measured by cognitive decline and brain activity levels, varies between individuals and this vulnerability rating correlates with the individual cellular vulnerability to extrinsic amyloid-ß in vitro as measured by synapse loss and function. Our findings indicate that patient-induced pluripotent stem cell-derived cortical neurons not only present key aspects of Alzheimer's disease pathology but also reflect key aspects of the clinical phenotypes of the same patients. Cellular models that reflect an individual's in-life clinical vulnerability thus represent a tractable method of Alzheimer's disease modelling using clinical data in combination with cellular phenotypes.

4.
Alzheimers Res Ther ; 13(1): 73, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33795014

RESUMEN

BACKGROUND: Major depression (MD) is the most prevalent psychiatric disease in the population and is considered a prodromal stage of the Alzheimer's disease (AD). Despite both diseases having a robust genetic component, the common transcriptomic signature remains unknown. METHODS: We investigated the cognitive and emotional behavioural responses in 3- and 6-month-old APP/PSEN1-Tg mice, before ß-amyloid plaques were detected. We studied the genetic and pathway deregulation in the prefrontal cortex, striatum, hippocampus and amygdala of mice at both ages, using transcriptomic and functional data analysis. RESULTS: We found that depressive-like and anxiety-like behaviours, as well as memory impairments, are already present at 3-month-old APP/PSEN1-Tg mutant mice together with the deregulation of several genes, such as Ciart, Grin3b, Nr1d1 and Mc4r, and other genes including components of the circadian rhythms, electron transport chain and neurotransmission in all brain areas. Extending these results to human data performing GSEA analysis using DisGeNET database, it provides translational support for common deregulated gene sets related to MD and AD. CONCLUSIONS: The present study sheds light on the shared genetic bases between MD and AD, based on a comprehensive characterization from the behavioural to transcriptomic level. These findings suggest that late MD could be an early manifestation of AD.


Asunto(s)
Enfermedad de Alzheimer , Trastorno Depresivo Mayor , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Animales , Comorbilidad , Depresión , Trastorno Depresivo Mayor/epidemiología , Trastorno Depresivo Mayor/genética , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Transcriptoma
5.
J Alzheimers Dis ; 77(3): 1353-1368, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32831200

RESUMEN

BACKGROUND: Previous studies suggest that Dickkopf-1 (DKK1), an inhibitor of Wnt signaling, plays a role in amyloid-induced toxicity and hence Alzheimer's disease (AD). However, the effect of DKK1 expression on protein expression, and whether such proteins are altered in disease, is unknown. OBJECTIVE: We aim to test whether DKK1 induced protein signature obtained in vitro were associated with markers of AD pathology as used in the amyloid/tau/neurodegeneration (ATN) framework as well as with clinical outcomes. METHODS: We first overexpressed DKK1 in HEK293A cells and quantified 1,128 proteins in cell lysates using aptamer capture arrays (SomaScan) to obtain a protein signature induced by DKK1. We then used the same assay to measure the DKK1-signature proteins in human plasma in two large cohorts, EMIF (n = 785) and ANM (n = 677). RESULTS: We identified a 100-protein signature induced by DKK1 in vitro. Subsets of proteins, along with age and apolipoprotein E ɛ4 genotype distinguished amyloid pathology (A + T-N-, A+T+N-, A+T-N+, and A+T+N+) from no AD pathology (A-T-N-) with an area under the curve of 0.72, 0.81, 0.88, and 0.85, respectively. Furthermore, we found that some signature proteins (e.g., Complement C3 and albumin) were associated with cognitive score and AD diagnosis in both cohorts. CONCLUSIONS: Our results add further evidence for a role of DKK regulation of Wnt signaling in AD and suggest that DKK1 induced signature proteins obtained in vitro could reflect theATNframework as well as predict disease severity and progression in vivo.


Asunto(s)
Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/patología , Péptidos y Proteínas de Señalización Intercelular/sangre , Anciano , Enfermedad de Alzheimer/genética , Biomarcadores/sangre , Femenino , Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Persona de Mediana Edad
6.
Brain ; 142(10): 3243-3264, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31504240

RESUMEN

Neuroinflammation and microglial activation are significant processes in Alzheimer's disease pathology. Recent genome-wide association studies have highlighted multiple immune-related genes in association with Alzheimer's disease, and experimental data have demonstrated microglial proliferation as a significant component of the neuropathology. In this study, we tested the efficacy of the selective CSF1R inhibitor JNJ-40346527 (JNJ-527) in the P301S mouse tauopathy model. We first demonstrated the anti-proliferative effects of JNJ-527 on microglia in the ME7 prion model, and its impact on the inflammatory profile, and provided potential CNS biomarkers for clinical investigation with the compound, including pharmacokinetic/pharmacodynamics and efficacy assessment by TSPO autoradiography and CSF proteomics. Then, we showed for the first time that blockade of microglial proliferation and modification of microglial phenotype leads to an attenuation of tau-induced neurodegeneration and results in functional improvement in P301S mice. Overall, this work strongly supports the potential for inhibition of CSF1R as a target for the treatment of Alzheimer's disease and other tau-mediated neurodegenerative diseases.


Asunto(s)
Imidazoles/farmacología , Microglía/efectos de los fármacos , Piridinas/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Enfermedad de Alzheimer/patología , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Imidazoles/metabolismo , Ratones , Ratones Transgénicos , Microglía/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neurogénesis , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Piridinas/metabolismo , Receptores de GABA/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Tauopatías/tratamiento farmacológico , Proteínas tau/genética
7.
Cells ; 8(5)2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072055

RESUMEN

As genome-wide association studies (GWAS) have grown in size, the number of genetic variants that have been associated per disease has correspondingly increased. Despite this increase in the number of single-nucleotide polymorphisms (SNPs) identified per disease, their biological interpretation has in many cases remained elusive. To address this, we have combined GWAS results with orthogonal sources of evidence, namely the current knowledge of molecular pathways; real-world clinical data from six million patients; RNA expression across tissues from Alzheimer's disease (AD) patients, and purpose-built rodent models for experimental validation. In more detail, first we show that when examined at a pathway level, analysis of all GWAS studies groups AD in a cluster with disorders of immunity and inflammation. Using clinical data, we show that the degree of comorbidity of these diseases with AD correlates with the strength of their genetic association with molecular participants in the Janus kinases/signal transducer and activator of transcription (JAK-STAT) pathway. Using four independent RNA expression datasets we then find evidence for the altered regulation of JAK-STAT pathway genes in AD. Finally, we use both in vitro and in vivo rodent models to demonstrate that Aß induces gene expression of the key drivers of this pathway, providing experimental evidence to validate these data-driven observations. These results therefore nominate JAK-STAT anomalies as a prominent aetiopathological event in AD and hence a potential target for therapeutic development, and moreover demonstrate a de novo multi-modal approach to derive information from rapidly increasing genomic datasets.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Quinasas Janus/metabolismo , Terapia Molecular Dirigida , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Anciano , Enfermedad de Alzheimer/sangre , Péptidos beta-Amiloides/toxicidad , Animales , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Inmunidad/genética , Masculino , Modelos Biológicos , Morbilidad , Neurotoxinas/toxicidad , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Regulación hacia Arriba/efectos de los fármacos
8.
Front Neurosci ; 13: 164, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30872998

RESUMEN

Clusterin (CLU) or APOJ is a multifunctional glycoprotein that has been implicated in several physiological and pathological states, including Alzheimer's disease (AD). With a prominent extracellular chaperone function, additional roles have been discussed for clusterin, including lipid transport and immune modulation, and it is involved in pathways common to several diseases such as cell death and survival, oxidative stress, and proteotoxic stress. Although clusterin is normally a secreted protein, it has also been found intracellularly under certain stress conditions. Multiple hypotheses have been proposed regarding the origin of intracellular clusterin, including specific biogenic processes leading to alternative transcripts and protein isoforms, but these lines of research are incomplete and contradictory. Current consensus is that intracellular clusterin is most likely to have exited the secretory pathway at some point or to have re-entered the cell after secretion. Clusterin's relationship with amyloid beta (Aß) has been of great interest to the AD field, including clusterin's apparent role in altering Aß aggregation and/or clearance. Additionally, clusterin has been more recently identified as a mediator of Aß toxicity, as evidenced by the neuroprotective effect of CLU knockdown and knockout in rodent and human iPSC-derived neurons. CLU is also the third most significant genetic risk factor for late onset AD and several variants have been identified in CLU. Although the exact contribution of these variants to altered AD risk is unclear, some have been linked to altered CLU expression at both mRNA and protein levels, altered cognitive and memory function, and altered brain structure. The apparent complexity of clusterin's biogenesis, the lack of clarity over the origin of the intracellular clusterin species, and the number of pathophysiological functions attributed to clusterin have all contributed to the challenge of understanding the role of clusterin in AD pathophysiology. Here, we highlight clusterin's relevance to AD by discussing the evidence linking clusterin to AD, as well as drawing parallels on how the role of clusterin in other diseases and pathways may help us understand its biological function(s) in association with AD.

9.
Transl Psychiatry ; 8(1): 179, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30232325

RESUMEN

In Alzheimer's disease (AD), the canonical Wnt inhibitor Dickkopf-1 (Dkk1) is induced by ß-amyloid (Aß) and shifts the balance from canonical towards non-canonical Wnt signalling. Canonical (Wnt-ß-catenin) signalling promotes synapse stability, while non-canonical (Wnt-PCP) signalling favours synapse retraction; thus Aß-driven synapse loss is mediated by Dkk1. Here we show that the Amyloid Precursor Protein (APP) co-activates both arms of Wnt signalling through physical interactions with Wnt co-receptors LRP6 and Vangl2, to bi-directionally modulate synapse stability. Furthermore, activation of non-canonical Wnt signalling enhances Aß production, while activation of canonical signalling suppresses Aß production. Together, these findings identify a pathogenic-positive feedback loop in which Aß induces Dkk1 expression, thereby activating non-canonical Wnt signalling to promote synapse loss and drive further Aß production. The Swedish familial AD variant of APP (APPSwe) more readily co-activates non-canonical, at the expense of canonical Wnt activity, indicating that its pathogenicity likely involves direct effects on synapses, in addition to increased Aß production. Finally, we report that pharmacological inhibition of the Aß-Dkk1-Aß positive feedback loop with the drug fasudil can restore the balance between Wnt pathways, prevent dendritic spine withdrawal in vitro, and reduce Aß load in vivo in mice with advanced amyloid pathology. These results clarify a relationship between Aß accumulation and synapse loss and provide direction for the development of potential disease-modifying treatments.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Sinapsis/patología , Vía de Señalización Wnt , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo
10.
Front Neurosci ; 12: 504, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30090055

RESUMEN

Our understanding of the molecular processes underlying Alzheimer's disease (AD) is still limited, hindering the development of effective treatments, and highlighting the need for human-specific models. Advances in identifying components of the amyloid cascade are progressing, including the role of the protein clusterin in mediating ß-amyloid (Aß) toxicity. Mutations in the clusterin gene (CLU), a major genetic AD risk factor, are known to have important roles in Aß processing. Here we investigate how CLU mediates Aß-driven neurodegeneration in human induced pluripotent stem cell (iPSC)-derived neurons. We generated a novel CLU-knockout iPSC line by CRISPR/Cas9-mediated gene editing to investigate Aß-mediated neurodegeneration in cortical neurons differentiated from wild type and CLU knockout iPSCs. We measured response to Aß using an imaging assay and measured changes in gene expression using qPCR and RNA sequencing. In wild type neurons imaging indicated that neuronal processes degenerate following treatment with Aß25-35 peptides and Aß1-42 oligomers, in a dose dependent manner, and that intracellular levels of clusterin are increased following Aß treatment. However, in CLU knockout neurons Aß exposure did not affect neurite length, suggesting that clusterin is an important component of the amyloid cascade. Transcriptomic data were analyzed to elucidate the pathways responsible for the altered response to Aß in neurons with the CLU deletion. Four of the five genes previously identified as downstream to Aß and Dickkopf-1 (DKK1) proteins in an Aß-driven neurotoxic pathway in rodent cells were also dysregulated in human neurons with the CLU deletion. AD and lysosome pathways were the most significantly dysregulated pathways in the CLU knockout neurons, and pathways relating to cytoskeletal processes were most dysregulated in Aß treated neurons. The absence of neurodegeneration in the CLU knockout neurons in response to Aß compared to the wild type neurons supports the role of clusterin in Aß-mediated AD pathogenesis.

11.
Alzheimers Dement ; 14(3): 306-317, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29055813

RESUMEN

INTRODUCTION: Synapse loss is the structural correlate of the cognitive decline indicative of dementia. In the brains of Alzheimer's disease sufferers, amyloid ß (Aß) peptides aggregate to form senile plaques but as soluble peptides are toxic to synapses. We previously demonstrated that Aß induces Dickkopf-1 (Dkk1), which in turn activates the Wnt-planar cell polarity (Wnt-PCP) pathway to drive tau pathology and neuronal death. METHODS: We compared the effects of Aß and of Dkk1 on synapse morphology and memory impairment while inhibiting or silencing key elements of the Wnt-PCP pathway. RESULTS: We demonstrate that Aß synaptotoxicity is also Dkk1 and Wnt-PCP dependent, mediated by the arm of Wnt-PCP regulating actin cytoskeletal dynamics via Daam1, RhoA and ROCK, and can be blocked by the drug fasudil. DISCUSSION: Our data add to the importance of aberrant Wnt signaling in Alzheimer's disease neuropathology and indicate that fasudil could be repurposed as a treatment for the disease.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Péptidos beta-Amiloides/metabolismo , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Sinapsis/metabolismo , Vía de Señalización Wnt , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacocinética , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Femenino , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Fármacos Neuroprotectores/farmacocinética , Nootrópicos/farmacocinética , Cultivo Primario de Células , ARN Mensajero/metabolismo , Ratas , Sinapsis/efectos de los fármacos , Sinapsis/patología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología
12.
Redox Biol ; 13: 444-451, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28704727

RESUMEN

Failure to translate successful neuroprotective preclinical data to a clinical setting in Alzheimer's disease (AD) indicates that amyloidopathy and tauopathy alone provide an incomplete view of disease. We have tested here the relevance of additional homeostatic deviations that result from loss of activity of transcription factor NRF2, a crucial regulator of multiple stress responses whose activity declines with ageing. A transcriptomic analysis demonstrated that NRF2-KO mouse brains reproduce 7 and 10 of the most dysregulated pathways of human ageing and AD brains, respectively. Then, we generated a mouse that combines amyloidopathy and tauopathy with either wild type (AT-NRF2-WT) or NRF2-deficiency (AT-NRF2-KO). AT-NRF2-KO brains presented increased markers of oxidative stress and neuroinflammation as well as higher levels of insoluble phosphorylated-TAU and Aß*56 compared to AT-NRF2-WT mice. Young adult AT-NRF2-KO mice exhibited deficits in spatial learning and memory and reduced long term potentiation in the perforant pathway. This study demonstrates the relevance of normal homeostatic responses that decline with ageing, such as NRF2 activity, in the protection against proteotoxic, inflammatory and oxidative stress and provide a new strategy to fight AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factor 2 Relacionado con NF-E2/genética , Transcriptoma , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Humanos , Potenciación a Largo Plazo , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Biochem J ; 455(1): 15-25, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23815625

RESUMEN

Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, ß-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteína Adaptadora de Señalización CRADD/genética , Caspasa 2/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas/genética , Activación Transcripcional/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Proteína Adaptadora de Señalización CRADD/metabolismo , Caspasa 2/metabolismo , Feto , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Neuronas/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
14.
Nat Commun ; 4: 1939, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23748737

RESUMEN

Caspases have critical roles in Alzheimer's disease pathogenesis. Here we show that caspase-2 is required for the cognitive decline seen in human amyloid precursor protein transgenic mice (J20). The age-related changes in behaviour and dendritic spine density observed in these mice are absent when they lack caspase-2, in spite of similar levels of amyloid beta (Aß) deposition and inflammation. A similar degree of protection is observed in cultured hippocampal neurons lacking caspase-2, which are immune to the synaptotoxic effects of Aß. Our studies suggest that caspase-2 is a critical mediator in the activation of the RhoA/ROCK-II signalling pathway, leading to the collapse of dendritic spines. We propose that this is controlled by an inactive caspase-2/RhoA/ROCK-II complex localized in dendrites, which dissociates in the presence of Aß, allowing for their activation and entry in the spine. These findings directly implicate caspase-2 as key driver of synaptic dysfunction in Alzheimer's disease and offer novel therapeutic targets.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Conducta Animal/fisiología , Caspasa 2/metabolismo , Espinas Dendríticas/enzimología , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Astrocitos/metabolismo , Astrocitos/patología , Western Blotting , Caspasa 2/deficiencia , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Hipocampo/patología , Humanos , Inmunoprecipitación , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Placa Amiloide/patología , Transporte de Proteínas/efectos de los fármacos , Ratas , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
15.
Biochem J ; 444(3): 591-9, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22515271

RESUMEN

Caspase 2 was initially identified as a neuronally expressed developmentally down-regulated gene (HUGO gene nomenclature CASP2) and has been shown to be required for neuronal death induced by several stimuli, including NGF (nerve growth factor) deprivation and Aß (ß-amyloid). In non-neuronal cells the PIDDosome, composed of caspase 2 and two death adaptor proteins, PIDD (p53-inducible protein with a death domain) and RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, has been proposed as the caspase 2 activation complex, although the absolute requirement for the PIDDosome is not clear. To investigate the requirement for the PIDDosome in caspase-2-dependent neuronal death, we have examined the necessity for each component in induction of active caspase 2 and in execution of caspase-2-dependent neuronal death. We find that both NGF deprivation and Aß treatment of neurons induce active caspase 2 and that induction of this activity depends on expression of RAIDD, but is independent of PIDD expression. We show that treatment of wild-type or PIDD-null neurons with Aß or NGF deprivation induces formation of a complex of caspase 2 and RAIDD. We also show that caspase-2-dependent execution of neurons requires RAIDD, not PIDD. Caspase 2 activity can be induced in neurons from PIDD-null mice, and NGF deprivation or Aß use caspase 2 and RAIDD to execute death of these neurons.


Asunto(s)
Proteína Adaptadora de Señalización CRADD/biosíntesis , Caspasa 2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/biosíntesis , Neuronas/enzimología , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática/fisiología , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Ratas , Ratas Sprague-Dawley
16.
Ann Neurol ; 69(5): 819-30, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21472769

RESUMEN

OBJECTIVE: The goal of this study was to investigate the role of endogenous amyloid-ß peptide (Aß) in healthy brain. METHODS: Long-term potentiation (LTP), a type of synaptic plasticity that is thought to be associated with learning and memory, was examined through extracellular field recordings from the CA1 region of hippocampal slices, whereas behavioral techniques were used to assess contextual fear memory and reference memory. Amyloid precursor protein (APP) expression was reduced through small interfering RNA (siRNA) technique. RESULTS: We found that both antirodent Aß antibody and siRNA against murine APP reduced LTP as well as contextual fear memory and reference memory. These effects were rescued by the addition of human Aß42, suggesting that endogenously produced Aß is needed for normal LTP and memory. Furthermore, the effect of endogenous Aß on plasticity and memory was likely due to regulation of transmitter release, activation of α7-containing nicotinic acetylcholine receptors, and Aß42 production. INTERPRETATION: Endogenous Aß42 is a critical player in synaptic plasticity and memory within the normal central nervous system. This needs to be taken into consideration when designing therapies aiming at reducing Aß levels to treat Alzheimer disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos/farmacología , Conducta Animal/efectos de los fármacos , Biofisica/métodos , Estimulación Eléctrica/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/fisiología , Hipocampo/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/metabolismo , ARN Interferente Pequeño/farmacología
17.
PLoS One ; 6(2): e16991, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21347323

RESUMEN

BACKGROUND: Impaired insulin signalling is increasingly thought to contribute to Alzheimer's disease (AD). The ε4 isoform of the APOE gene is the greatest genetic risk factor for sporadic, late onset AD, and is also associated with risk for type 2 diabetes mellitus (T2DM). Neuropathological studies reported the highest number of AD lesions in brain tissue of ε4 diabetic patients. However other studies assessing AD pathology amongst the diabetic population have produced conflicting reports and have failed to show an increase in AD-related pathology in diabetic brain. The thiazolidinediones (TZDs), peroxisome proliferator-activated receptor gamma agonists, are peripheral insulin sensitisers used to treat T2DM. The TZD, pioglitazone, improved memory and cognitive functions in mild to moderate AD patients. Since it is not yet clear how apoE isoforms influence the development of T2DM and its progression to AD, we investigated amyloid beta and tau pathology in APOE knockout mice, carrying human APOEε3 or ε4 transgenes after diet-induced insulin resistance with and without pioglitazone treatment. METHODS: Male APOE knockout, APOEε3-transgenic and APOEε4-transgenic mice, together with background strain C57BL6 mice were kept on a high fat diet (HFD) or low fat diet (LFD) for 32 weeks, or were all fed HFD for 32 weeks and during the final 3 weeks animals were treated with pioglitazone or vehicle. RESULTS: All HFD animals developed hyperglycaemia with elevated plasma insulin. Tau phosphorylation was reduced at 3 epitopes (Ser396, Ser202/Thr205 and Thr231) in all HFD, compared to LFD, animals independent of APOE genotype. The introduction of pioglitazone to HFD animals led to a significant reduction in tau phosphorylation at the Ser202/Thr205 epitope in APOEε3 animals only. We found no changes in APP processing however the levels of soluble amyloid beta 40 was reduced in APOE knockout animals treated with pioglitazone.


Asunto(s)
Alelos , Apolipoproteínas E/genética , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hipoglucemiantes/farmacología , Tiazolidinedionas/farmacología , Proteínas tau/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Genotipo , Humanos , Hiperinsulinismo/etiología , Resistencia a la Insulina/genética , Masculino , Ratones , Fosforilación/efectos de los fármacos , Fosforilación/genética , Pioglitazona , Solubilidad , Proteínas tau/química
18.
J Alzheimers Dis ; 19(3): 885-94, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20157244

RESUMEN

Multiple studies suggest that cystatin C (CysC) has a role in Alzheimer's disease (AD) and a decrease in CysC secretion is linked to the disease in patients with a polymorphism in the CysC gene. CysC binds amyloid-beta (Abeta) and inhibits formation of Abeta fibrils and oligomers both in vitro and in mouse models of amyloid deposition. Here we studied the effect of CysC on cultured primary hippocampal neurons and a neuronal cell line exposed to either oligomeric or fibrillar cytotoxic forms of Abeta. The extracellular addition of the secreted human CysC together with preformed either oligomeric or fibrillar Abeta increased cell survival. While CysC inhibits Abeta aggregation, it does not dissolve preformed Abeta fibrils or oligomers. Thus, CysC has multiple protective effects in AD, by preventing the formation of the toxic forms of Abeta and by direct protection of neuronal cells from Abeta toxicity. Therapeutic manipulation of CysC levels, resulting in slightly higher concentrations than physiological could protect neuronal cells from cell death in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Cistatina C/fisiología , Neuronas/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Animales , Apoptosis , Western Blotting , Células Cultivadas , Cistatina C/genética , Cistatina C/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Ratones , Neurotoxinas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
19.
J Neurosci ; 29(4): 1011-6, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19176810

RESUMEN

Neurodegenerative illnesses such as Parkinson and Alzheimer disease are an increasingly prevalent problem in aging societies, yet no therapies exist that retard or prevent neurodegeneration. Dominant missense mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson disease (PD), but the mechanisms by which mutant forms of LRRK2 disrupt neuronal function and cause cell death remain poorly understood. We report that LRRK2 interacts with the death adaptor Fas-associated protein with death domain (FADD), and that in primary neuronal culture LRRK2-mediated neurodegeneration is prevented by the functional inhibition of FADD or depletion of caspase-8, two key elements of the extrinsic cell death pathway. This pathway is activated by disease-triggering mutations, which enhance the LRRK2-FADD association and the consequent recruitment and activation of caspase-8. These results establish a direct molecular link between a mutant PD gene and the activation of programmed cell death signaling, and suggest that FADD/caspase-8 signaling contributes to LRRK2-induced neuronal death.


Asunto(s)
Apoptosis/fisiología , Caspasa 8/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Portadoras/genética , Línea Celular Transformada , Péptidos de Penetración Celular , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mutagénesis Sitio-Dirigida/métodos , Neuronas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/farmacología , Transfección/métodos
20.
Biochem J ; 415(2): 165-82, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18800967

RESUMEN

Dysregulation of life and death at the cellular level leads to a variety of diseases. In the nervous system, aberrant neuronal death is an outstanding feature of neurodegenerative diseases. Since the discovery of the caspase family of proteases, much effort has been made to determine how caspases function in disease, including neurodegenerative diseases. Although many papers have been published examining caspases in neuronal death and disease, the pathways have not been fully clarified. In the present review, we examine the potential players in the death pathways, the current tools for examining these players and the models for studying neurological disease. Alzheimer's disease, the most common neurodegenerative disorder, and cerebral ischaemia, the most common cause of neurological death, are used to illustrate our current understanding of death signalling in neurodegenerative diseases. A better understanding of the neuronal death pathways would provide targets for the development of therapeutic interventions for these diseases.


Asunto(s)
Apoptosis/fisiología , Enfermedades del Sistema Nervioso/fisiopatología , Neuronas/citología , Transducción de Señal , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Caspasas/metabolismo , Humanos , Modelos Biológicos , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...