Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 39(20): e103791, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32865299

RESUMEN

The link between cholesterol homeostasis and cleavage of the amyloid precursor protein (APP), and how this relationship relates to Alzheimer's disease (AD) pathogenesis, is still unknown. Cellular cholesterol levels are regulated through crosstalk between the plasma membrane (PM), where most cellular cholesterol resides, and the endoplasmic reticulum (ER), where the protein machinery that regulates cholesterol levels resides. The intracellular transport of cholesterol from the PM to the ER is believed to be activated by a lipid-sensing peptide(s) in the ER that can cluster PM-derived cholesterol into transient detergent-resistant membrane domains (DRMs) within the ER, also called the ER regulatory pool of cholesterol. When formed, these cholesterol-rich domains in the ER maintain cellular homeostasis by inducing cholesterol esterification as a mechanism of detoxification while attenuating its de novo synthesis. In this manuscript, we propose that the 99-aa C-terminal fragment of APP (C99), when delivered to the ER for cleavage by γ-secretase, acts as a lipid-sensing peptide that forms regulatory DRMs in the ER, called mitochondria-associated ER membranes (MAM). Our data in cellular AD models indicates that increased levels of uncleaved C99 in the ER, an early phenotype of the disease, upregulates the formation of these transient DRMs by inducing the internalization of extracellular cholesterol and its trafficking from the PM to the ER. These results suggest a novel role for C99 as a mediator of cholesterol disturbances in AD, potentially explaining early hallmarks of the disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Animales , Línea Celular , Colesterol/biosíntesis , Retículo Endoplásmico/genética , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Células Madre Pluripotentes Inducidas , Metabolismo de los Lípidos , Lipidómica , Ratones , Mitocondrias/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-2/genética , Presenilina-2/metabolismo , Dominios Proteicos , ARN Interferente Pequeño , Esfingomielina Fosfodiesterasa/metabolismo
2.
Hum Mol Genet ; 28(11): 1782-1800, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649465

RESUMEN

Charcot-Marie-Tooth disease (CMT) type 2A is a form of peripheral neuropathy, due almost exclusively to dominant mutations in the nuclear gene encoding the mitochondrial protein mitofusin-2 (MFN2). However, there is no understanding of the relationship of clinical phenotype to genotype. MFN2 has two functions: it promotes inter-mitochondrial fusion and mediates endoplasmic reticulum (ER)-mitochondrial tethering at mitochondria-associated ER membranes (MAM). MAM regulates a number of key cellular functions, including lipid and calcium homeostasis, and mitochondrial behavior. To date, no studies have been performed to address whether mutations in MFN2 in CMT2A patient cells affect MAM function, which might provide insight into pathogenesis. Using fibroblasts from three CMT2AMFN2 patients with different mutations in MFN2, we found that some, but not all, examined aspects of ER-mitochondrial connectivity and of MAM function were indeed altered, and correlated with disease severity. Notably, however, respiratory chain function in those cells was unimpaired. Our results suggest that CMT2AMFN2 is a MAM-related disorder but is not a respiratory chain-deficiency disease. The alterations in MAM function described here could also provide insight into the pathogenesis of other forms of CMT.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Retículo Endoplásmico/genética , GTP Fosfohidrolasas/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Adulto , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Retículo Endoplásmico/metabolismo , Metabolismo Energético/genética , Femenino , Fibroblastos/metabolismo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Dinámicas Mitocondriales/genética , Membranas Mitocondriales/metabolismo , Mutación , Fosforilación Oxidativa , Índice de Severidad de la Enfermedad
3.
EMBO J ; 36(22): 3356-3371, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29018038

RESUMEN

In the amyloidogenic pathway associated with Alzheimer disease (AD), the amyloid precursor protein (APP) is cleaved by ß-secretase to generate a 99-aa C-terminal fragment (C99) that is then cleaved by γ-secretase to generate the ß-amyloid (Aß) found in senile plaques. In previous reports, we and others have shown that γ-secretase activity is enriched in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) and that ER-mitochondrial connectivity and MAM function are upregulated in AD We now show that C99, in addition to its localization in endosomes, can also be found in MAM, where it is normally processed rapidly by γ-secretase. In cell models of AD, however, the concentration of unprocessed C99 increases in MAM regions, resulting in elevated sphingolipid turnover and an altered lipid composition of both MAM and mitochondrial membranes. In turn, this change in mitochondrial membrane composition interferes with the proper assembly and activity of mitochondrial respiratory supercomplexes, thereby likely contributing to the bioenergetic defects characteristic of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular , Respiración de la Célula , Retículo Endoplásmico/ultraestructura , Humanos , Membranas Intracelulares/ultraestructura , Ratones , Mitocondrias/ultraestructura , Mutación/genética , Consumo de Oxígeno , Presenilinas/genética , Transporte de Proteínas , Esfingolípidos/metabolismo , Regulación hacia Arriba
4.
J Neurosci ; 39(36): 7074-7085, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31300519

RESUMEN

Maintaining a pool of functional mitochondria requires degradation of damaged ones within the cell. PINK1 is critical in this quality-control process: loss of mitochondrial membrane potential causes PINK1 to accumulate on the mitochondrial surface, triggering mitophagy. However, little is known about how PINK1 is regulated. Recently, we showed that PINK1 content is kept low in healthy mitochondria by continuous ubiquitination and proteasomal degradation of its mature form via a mechanism inconsistent with the proposed N-end rule process. Using both human female and monkey cell lines, we now demonstrate that once generated within the mitochondria, 52 kDa PINK1 adopts a mitochondrial topology most consistent with it being at the mitochondrial-endoplasmic reticulum (ER) interface. From this particular submitochondrial location, PINK1 interacts with components of the ER-associated degradation pathway, such as the E3 ligases gp78 and HRD1, which cooperate to catalyze PINK1 ubiquitination. The valosin-containing protein and its cofactor, UFD1, then target ubiquitinated PINK1 for proteasomal degradation. Our data show that PINK1 in healthy mitochondria is negatively regulated via an interplay between mitochondria and ER, and shed light on how this mitochondrial protein gains access to the proteasome.SIGNIFICANCE STATEMENT Regulation of mitochondrial content of PINK1, a contributor to mitophagy, is an important area of research. Recently, we found that PINK1 content is kept low in healthy mitochondria by continuous ubiquitination and proteasomal degradation. We now extend and refine this novel finding by showing that PINK1 localizes at the mitochondrial-endoplasmic reticulum (ER) interface, from where it interacts with the ER-associated degradation machinery, which catalyzes its ubiquitination and transfer to the proteasome. Thus, these data show that PINK1 in healthy mitochondria is negatively regulated via a mitochondria and ER interplay, and how this mitochondrial protein gains access to the proteasome.


Asunto(s)
Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Proteínas Quinasas/metabolismo , Proteolisis , Ubiquitinación , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Receptores del Factor Autocrino de Motilidad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína que Contiene Valosina/metabolismo
5.
Curr Opin Clin Nutr Metab Care ; 23(2): 68-75, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32004239

RESUMEN

PURPOSE OF REVIEW: The purpose of this brief review is to gain an understanding on the multiple roles that lipids exert on the brain, and to highlight new ideas in the impact of lipid homeostasis in the regulation of synaptic transmission. RECENT FINDINGS: Recent data underline the crucial function of lipid homeostasis in maintaining neuronal function and synaptic plasticity. Moreover, new advances in analytical approaches to study lipid classes and species is opening a new door to understand and monitor how alterations in lipid pathways could shed new light into the pathogenesis of neurodegeneration. SUMMARY: Lipids are one of the most essential elements of the brain. However, our understanding of the role of lipids within the central nervous system is still largely unknown. Identifying the molecular mechanism (s) by which lipids can regulate neuronal transmission represents the next frontier in neuroscience, and a new challenge in our understanding of the brain and the mechanism(s) behind neurological disorders.


Asunto(s)
Encéfalo/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Enfermedades del Sistema Nervioso/metabolismo , Neuronas/metabolismo , Animales , Homeostasis , Humanos , Plasticidad Neuronal
6.
EMBO Rep ; 17(1): 27-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26564908

RESUMEN

In addition to the appearance of senile plaques and neurofibrillary tangles, Alzheimer's disease (AD) is characterized by aberrant lipid metabolism and early mitochondrial dysfunction. We recently showed that there was increased functionality of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a subdomain of the ER involved in lipid and cholesterol homeostasis, in presenilin-deficient cells and in fibroblasts from familial and sporadic AD patients. Individuals carrying the ε4 allele of apolipoprotein E (ApoE4) are at increased risk for developing AD compared to those carrying ApoE3. While the reason for this increased risk is unknown, we hypothesized that it might be associated with elevated MAM function. Using an astrocyte-conditioned media (ACM) model, we now show that ER-mitochondrial communication and MAM function-as measured by the synthesis of phospholipids and of cholesteryl esters, respectively-are increased significantly in cells treated with ApoE4-containing ACM as compared to those treated with ApoE3-containing ACM. Notably, this effect was seen with lipoprotein-enriched preparations, but not with lipid-free ApoE protein. These data are consistent with a role of upregulated MAM function in the pathogenesis of AD and may help explain, in part, the contribution of ApoE4 as a risk factor in the disease.


Asunto(s)
Apolipoproteína E4/metabolismo , Astrocitos/fisiología , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Apolipoproteína E4/química , Apolipoproteína E4/genética , Colesterol/metabolismo , Ésteres del Colesterol/biosíntesis , Medios de Cultivo Condicionados/química , Retículo Endoplásmico/genética , Humanos , Metabolismo de los Lípidos , Lipoproteínas/metabolismo , Ratones , Fosfolípidos/biosíntesis , Activación Transcripcional , Regulación hacia Arriba
7.
EMBO J ; 31(21): 4106-23, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-22892566

RESUMEN

Alzheimer disease (AD) is associated with aberrant processing of the amyloid precursor protein (APP) by γ-secretase, via an unknown mechanism. We recently showed that presenilin-1 and -2, the catalytic components of γ-secretase, and γ-secretase activity itself, are highly enriched in a subcompartment of the endoplasmic reticulum (ER) that is physically and biochemically connected to mitochondria, called mitochondria-associated ER membranes (MAMs). We now show that MAM function and ER-mitochondrial communication-as measured by cholesteryl ester and phospholipid synthesis, respectively-are increased significantly in presenilin-mutant cells and in fibroblasts from patients with both the familial and sporadic forms of AD. We also show that MAM is an intracellular detergent-resistant lipid raft (LR)-like domain, consistent with the known presence of presenilins and γ-secretase activity in rafts. These findings may help explain not only the aberrant APP processing but also a number of other biochemical features of AD, including altered lipid metabolism and calcium homeostasis. We propose that upregulated MAM function at the ER-mitochondrial interface, and increased cross-talk between these two organelles, may play a hitherto unrecognized role in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Embrión de Mamíferos/patología , Fibroblastos/patología , Microdominios de Membrana/patología , Mitocondrias/patología , Membranas Mitocondriales/patología , Presenilina-1/fisiología , Presenilina-2/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Fibroblastos/metabolismo , Humanos , Microdominios de Membrana/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Presenilina-1/antagonistas & inhibidores , Presenilina-2/antagonistas & inhibidores , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares
8.
J Neurosci ; 34(1): 249-59, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24381286

RESUMEN

Familial Parkinson disease is associated with mutations in α-synuclein (α-syn), a presynaptic protein that has been localized not only to the cytosol, but also to mitochondria. We report here that wild-type α-syn from cell lines, and brain tissue from humans and mice, is present not in mitochondria but rather in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a structurally and functionally distinct subdomain of the ER. Remarkably, we found that pathogenic point mutations in human α-syn result in its reduced association with MAM, coincident with a lower degree of apposition of ER with mitochondria, a decrease in MAM function, and an increase in mitochondrial fragmentation compared with wild-type. Although overexpression of wild-type α-syn in mutant α-syn-expressing cells reverted the fragmentation phenotype, neither overexpression of the mitochondrial fusion/MAM-tethering protein MFN2 nor inhibition/ablation of the mitochondrial fission protein DRP1 was able to do so, implying that α-syn operates downstream of the mitochondrial fusion/fission machinery. These novel results indicate that wild-type α-syn localizes to the MAM and modulates mitochondrial morphology, and that these behaviors are impaired by pathogenic mutations in α-syn. We believe that our results have far-reaching implications for both our understanding of α-syn biology and the treatment of synucleinopathies.


Asunto(s)
Retículo Endoplásmico/química , Mitocondrias/química , alfa-Sinucleína/análisis , Animales , Células Cultivadas , Femenino , Células HeLa , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos
9.
Mov Disord ; 30(8): 1026-33, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25952565

RESUMEN

Familial cases of Parkinson's disease (PD) can be associated with overexpression or mutation of α-synuclein, a synaptic protein reported to be localized mainly in the cytosol and mitochondria. We recently showed that wild-type α-synuclein is not present in mitochondria, as previously thought, but rather is located in mitochondrial-associated endoplasmic reticulum membranes. Remarkably, we also found that PD-related mutated α-synuclein results in its reduced association with mitochondria-associated membranes, coincident with a lower degree of apposition of endoplasmic reticulum with mitochondria and an increase in mitochondrial fragmentation, as compared with wild-type. This new subcellular localization of α-synuclein raises fundamental questions regarding the relationship of α-synuclein to mitochondria-associated membranes function, in both normal and pathological states. In this article, we attempt to relate aspects of PD pathogenesis to what is known about mitochondria-associated membranes' behavior and function. We hypothesize that early events occurring in dopaminergic neurons at the level of the mitochondria-associated membranes could cause long-term disturbances that lead to PD.


Asunto(s)
Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Humanos
10.
Stroke ; 45(2): 413-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24385273

RESUMEN

BACKGROUND AND PURPOSE: It has been proposed that the deposition of the ß-amyloid peptide (Aß) in the brain parenchyma and brain blood vessels has deleterious effects. We tested the hypothesis that the levels of plasma Aß are related to the outcome in patients with intracerebral hemorrhage. METHODS: In a multicenter study, we prospectively included patients with spontaneous intracerebral hemorrhage within the first 24 hours after onset. At admission, we measured plasma Aß40 and Aß42 levels using ELISA techniques. Also, we recorded age, sex, vascular risk factors, National Institutes of Health Stroke Scale score, presence of intraventricular hemorrhage, localization, cause, and volume of the hematoma. We obtained the modified Rankin scale and defined a unfavorable outcome as modified Rankin scale >2 at 3 months. Bivariate and multivariate regression analyses were performed. RESULTS: We studied 160 patients (mean age, 73.8±11.3 years; 59.4% of them were men). A favorable outcome was observed in 64 (40%) of the patients. In the bivariate analyses, unfavorable outcome was associated with high age, female sex, diabetes mellitus, presence of intraventricular hemorrhage, high blood glucose, high National Institutes of Health Stroke Scale score, high volume, and high plasma levels of Aß42 and Aß40. The multivariate analysis showed that increased age (odds ratio, 1.07; 95% confidence interval, 1.035-1.21; P<0.0001), high admission National Institutes of Health Stroke Scale score (odds ratio, 1.29, 95% confidence interval, 1.17-1.42; P<0.0001), presence of diabetes mellitus (odds ratio, 4.15; 95% confidence interval, 1.21-14.1; P=0.02), and Aß42 levels >9.7 pg/mL (odds ratio, 4.11; 95% confidence interval, 1.65-10.1; P=0.02) were independently associated with an increased likelihood of an unfavorable outcome. CONCLUSIONS: High levels of plasma Aß42 in patients with acute intracerebral hemorrhage are associated with a poor functional prognosis.


Asunto(s)
Péptidos beta-Amiloides/sangre , Hemorragia Cerebral/sangre , Factores de Edad , Anciano , Anciano de 80 o más Años , Intervalos de Confianza , Interpretación Estadística de Datos , Complicaciones de la Diabetes/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Fragmentos de Péptidos/sangre , Pronóstico , Estudios Prospectivos , Análisis de Regresión , Factores Sexuales , Resultado del Tratamiento
11.
J Neurochem ; 128(2): 330-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24117942

RESUMEN

Autosomal-dominant Alzheimer's disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studying the mechanisms underlying these mutations can provide insight into the pathways that lead to AD pathology. The majority of biochemical studies on APP mutations to-date have focused on comparing mechanisms between mutations at different codons. It has been assumed that amino acid position is a major determinant of protein dysfunction and clinical phenotype. However, the differential effect of mutations at the same codon has not been sufficiently addressed. In the present study we compared the effects of the aggressive ADAD-associated APP I716F mutation with I716V and I716T on APP processing in human neuroglioma and CHO-K1 cells. All APP I716 mutations increased the ratio of Aß42/40 and changed the product line preference of γ-secretase towards Aß38 production. In addition, the APP I716F mutation impaired the ε-cleavage and the fourth cleavage of γ-secretase and led to abnormal APP ß-CTF accumulation at the plasma membrane. Taken together, these data indicate that APP mutations at the same codon can induce diverse abnormalities in APP processing, some resembling PSEN1 mutations. These differential effects could explain the clinical differences observed among ADAD patients bearing different APP mutations at the same position. The amyloid precursor protein (APP) I716F mutation is associated with autosomal dominant Alzheimer's disease with the youngest age-at-onset for the APP locus. Here, we describe that this mutation, when compared to two other familial Alzheimer's disease mutations at the same codon (I716V and I716T), interfered distinctly with γ-secretase cleavage. While all three mutations direct γ-secretase cleavage towards the 48→38 production line, the APP I716F mutation also impaired the ε-cleavage and the fourth cleavage of γ-secretase, resembling a PSEN1 mutation. These features may contribute to the aggressiveness of this mutation.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Adulto , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Membrana Celular/metabolismo , Codón , Cricetinae , Cricetulus , Humanos , Mutación
12.
bioRxiv ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38948777

RESUMEN

The protein alpha-synuclein (αSyn) plays a critical role in the pathogenesis of synucleinopathy, which includes Parkinson's disease and multiple system atrophy, and mounting evidence suggests that lipid dyshomeostasis is a critical phenotype in these neurodegenerative conditions. Previously, we identified that αSyn localizes to mitochondria-associated endoplasmic reticulum membranes (MAMs), temporary functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we have analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data show region-and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed multiples system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn contributes to regulating phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. Our results support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.

13.
Acta Neuropathol ; 125(2): 201-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23224319

RESUMEN

Autosomal-dominant Alzheimer disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studies from families with ADAD have been critical to support the amyloid cascade hypothesis of Alzheimer disease (AD), the basis for the current development of amyloid-based disease-modifying therapies in sporadic AD (SAD). However, whether the pathological changes in APP processing in the CNS in ADAD are similar to those observed in SAD remains unclear. In this study, we measured ß-site APP-cleaving enzyme (BACE) protein levels and activity, APP and APP C-terminal fragments in brain samples from subjects with ADAD carrying APP or PSEN1 mutations (n = 18), patients with SAD (n = 27) and age-matched controls (n = 22). We also measured sAPPß and BACE protein levels, as well as BACE activity, in CSF from individuals carrying PSEN1 mutations (10 mutation carriers and 7 non-carrier controls), patients with SAD (n = 32) and age-matched controls (n = 11). We found that in the brain, the pattern in ADAD was characterized by an increase in APP ß-C-terminal fragment (ß-CTF) levels despite no changes in BACE protein levels or activity. In contrast, the pattern in SAD in the brain was mainly characterized by an increase in BACE levels and activity, with less APP ß-CTF accumulation than ADAD. In the CSF, no differences were found between groups in BACE activity or expression or sAPPß levels. Taken together, these data suggest that the physiopathological events underlying the chronic Aß production/clearance imbalance in SAD and ADAD are different. These differences should be considered in the design of intervention trials in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Sistema Nervioso Central/metabolismo , Adulto , Anciano , Enfermedad de Alzheimer/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/líquido cefalorraquídeo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ácido Aspártico Endopeptidasas/líquido cefalorraquídeo , Ácido Aspártico Endopeptidasas/metabolismo , Western Blotting , Femenino , Heterocigoto , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación/genética , Neuritas/patología , Presenilina-1/líquido cefalorraquídeo , Presenilina-1/genética
14.
NPJ Parkinsons Dis ; 8(1): 52, 2022 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-35468903

RESUMEN

Lipid profiles in biological fluids from patients with Parkinson's disease (PD) are increasingly investigated in search of biomarkers. However, the lipid profiles in genetic PD remain to be determined, a gap of knowledge of particular interest in PD associated with mutant α-synuclein (SNCA), given the known relationship between this protein and lipids. The objective of this research is to identify serum lipid composition from SNCA A53T mutation carriers and to compare these alterations to those found in cells and transgenic mice carrying the same genetic mutation. We conducted an unbiased lipidomic analysis of 530 lipid species from 34 lipid classes in serum of 30 participants with SNCA mutation with and without PD and 30 healthy controls. The primary analysis was done between 22 PD patients with SNCA+ (SNCA+/PD+) and 30 controls using machine-learning algorithms and traditional statistics. We also analyzed the lipid composition of human clonal-cell lines and tissue from transgenic mice overexpressing the same SNCA mutation. We identified specific lipid classes that best discriminate between SNCA+/PD+ patients and healthy controls and found certain lipid species, mainly from the glycerophosphatidylcholine and triradylglycerol classes, that are most contributory to this discrimination. Most of these alterations were also present in human derived cells and transgenic mice carrying the same mutation. Our combination of lipidomic and machine learning analyses revealed alterations in glycerophosphatidylcholine and triradylglycerol in sera from PD patients as well as cells and tissues expressing mutant α-Syn. Further investigations are needed to establish the pathogenic significance of these α-Syn-associated lipid changes.

15.
J Neurosci ; 30(28): 9402-10, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20631169

RESUMEN

Activity-dependent gene expression mediating changes of synaptic efficacy is important for memory storage, but the mechanisms underlying gene transcriptional changes in age-related memory disorders are poorly understood. In this study, we report that gene transcription mediated by the cAMP-response element binding protein (CREB)-regulated transcription coactivator CRTC1 is impaired in neurons and brain from an Alzheimer's disease (AD) transgenic mouse expressing the human beta-amyloid precursor protein (APP(Sw,Ind)). Suppression of CRTC1-dependent gene transcription by beta-amyloid (Abeta) in response to cAMP and Ca(2+) signals is mediated by reduced calcium influx and disruption of PP2B/calcineurin-dependent CRTC1 dephosphorylation at Ser151. Consistently, expression of CRTC1 or active CRTC1 S151A and calcineurin mutants reverse the deficits on CRTC1 transcriptional activity in APP(Sw,Ind) neurons. Inhibition of calcium influx by pharmacological blockade of L-type voltage-gated calcium channels (VGCCs), but not by blocking NMDA or AMPA receptors, mimics the decrease on CRTC1 transcriptional activity observed in APP(Sw,Ind) neurons, whereas agonists of L-type VGCCs reverse efficiently these deficits. Consistent with a role of CRTC1 on Abeta-induced synaptic and memory dysfunction, we demonstrate a selective reduction of CRTC1-dependent genes related to memory (Bdnf, c-fos, and Nr4a2) coinciding with hippocampal-dependent spatial memory deficits in APP(Sw,Ind) mice. These findings suggest that CRTC1 plays a key role in coupling synaptic activity to gene transcription required for hippocampal-dependent memory, and that Abeta could disrupt cognition by affecting CRTC1 function.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Corteza Cerebral/metabolismo , Memoria/fisiología , Neuronas/metabolismo , Factores de Transcripción/genética , Transcripción Genética/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Conducta Animal/fisiología , Western Blotting , Calcineurina/metabolismo , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Inmunoprecipitación de Cromatina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Transgénicos , Neuronas/citología , Fosforilación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo
16.
J Parkinsons Dis ; 11(3): 1141-1155, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814463

RESUMEN

BACKGROUND: The role of the lipidome as a biomarker for Parkinson's disease (PD) is a relatively new field that currently only focuses on PD diagnosis. OBJECTIVE: To identify a relevant lipidome signature for PD severity markers. METHODS: Disease severity of 149 PD patients was assessed by the Unified Parkinson's Disease Rating Scale (UPDRS) and the Montreal Cognitive Assessment (MoCA). The lipid composition of whole blood samples was analyzed, consisting of 517 lipid species from 37 classes; these included all major classes of glycerophospholipids, sphingolipids, glycerolipids, and sterols. To handle the high number of lipids, the selection of lipid species and classes was consolidated via analysis of interrelations between lipidomics and disease severity prediction using the random forest machine-learning algorithm aided by conventional statistical methods. RESULTS: Specific lipid classes dihydrosphingomyelin (dhSM), plasmalogen phosphatidylethanolamine (PEp), glucosylceramide (GlcCer), dihydro globotriaosylceramide (dhGB3), and to a lesser degree dihydro GM3 ganglioside (dhGM3), as well as species dhSM(20:0), PEp(38:6), PEp(42:7), GlcCer(16:0), GlcCer(24:1), dhGM3(22:0), dhGM3(16:0), and dhGB3(16:0) contribute to PD severity prediction of UPDRS III score. These, together with age, age at onset, and disease duration, also contribute to prediction of UPDRS total score. We demonstrate that certain lipid classes and species interrelate differently with the degree of severity of motor symptoms between men and women, and that predicting intermediate disease stages is more accurate than predicting less or more severe stages. CONCLUSION: Using machine-learning algorithms and methodologies, we identified lipid signatures that enable prediction of motor severity in PD. Future studies should focus on identifying the biological mechanisms linking GlcCer, dhGB3, dhSM, and PEp with PD severity.


Asunto(s)
Lipidómica , Enfermedad de Parkinson , Biomarcadores , Femenino , Humanos , Lípidos , Aprendizaje Automático , Masculino , Enfermedad de Parkinson/diagnóstico , Índice de Severidad de la Enfermedad
17.
J Neuropathol Exp Neurol ; 68(1): 73-82, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19104444

RESUMEN

We describe a Spanish family in which 3 of 4 siblings had dementia with Lewy bodies, 2 of them starting at age 26 years and the other at 29 years. The father has recently been diagnosed with Lewy body disease, with onset at 77 years. Neuropathological examination of the brain of the index patient disclosed unusual features characterized by diffuse Lewy body disease and generalized neurofibrillary tangle pathology but with no amyloid deposits in any region. Moreover, Lewy body pathology colocalized with neurofibrillary tangles in most affected neurons. Mutation screening that included all coding exons of presenilin 1 (PSEN1), presenilin 2 (PSEN2), alpha-synuclein (SNCA), beta-synuclein (SNCB), microtubule-associated protein tau (MAPT), leucine-rich repeat kinase 2 (LRRK2), glucocerebrosidase (GBA), and exons 16 and 17 of the amyloid precursor protein (APP) genes did not identify any mutation. Genome-wide single nucleotide polymorphism was performed in 4 family members and ruled out any pathogenic duplication or deletion in the entire genome. In summary, we report a unique family with pathologically confirmed early-onset dementia with Lewy bodies with widespread tau and alpha-synuclein deposition. The absence of mutations in genes known to cause Lewy body disease suggests that a novel locus or loci are implicated in this neurodegenerative disease.


Asunto(s)
Encéfalo/patología , Salud de la Familia , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/patología , Ovillos Neurofibrilares , Tauopatías/genética , Tauopatías/patología , Adulto , Anciano de 80 o más Años , Encéfalo/metabolismo , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Enfermedad por Cuerpos de Lewy/complicaciones , Imagen por Resonancia Magnética , Masculino , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Presenilinas/genética , Presenilinas/metabolismo , Tauopatías/complicaciones , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
18.
J Neurochem ; 110(1): 220-30, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19457132

RESUMEN

It has been suggested that cellular cholesterol levels can modulate the metabolism of the amyloid precursor protein (APP) but the underlying mechanism remains controversial. In the current study, we investigate in detail the relationship between cholesterol reduction, APP processing and gamma-secretase function in cell culture studies. We found that mild membrane cholesterol reduction led to a decrease in Abeta(40) and Abeta(42) in different cell types. We did not detect changes in APP intracellular domain or Notch intracellular domain generation. Western blot analyses showed a cholesterol-dependent decrease in the APP C-terminal fragments and cell surface APP. Finally, we applied a fluorescence resonance energy transfer (FRET)-based technique to study APP-Presenilin 1 (PS1) interactions and lipid rafts in intact cells. Our data indicate that cholesterol depletion reduces association of APP into lipid rafts and disrupts APP-PS1 interaction. Taken together, our results suggest that mild membrane cholesterol reduction impacts the cleavage of APP upstream of gamma-secretase and appears to be mediated by changes in APP trafficking and partitioning into lipid rafts.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Colesterol/deficiencia , Microdominios de Membrana/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Regulación hacia Abajo/fisiología , Humanos , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1/metabolismo , Estructura Terciaria de Proteína/fisiología , Transporte de Proteínas/fisiología
19.
J Clin Invest ; 129(1): 34-45, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30601141

RESUMEN

Mitochondrial respiratory deficiencies have been observed in numerous neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. For decades, these reductions in oxidative phosphorylation (OxPhos) have been presumed to trigger an overall bioenergetic crisis in the neuron, resulting in cell death. While the connection between respiratory defects and neuronal death has never been proven, this hypothesis has been supported by the detection of nonspecific mitochondrial DNA mutations in these disorders. These findings led to the notion that mitochondrial respiratory defects could be initiators of these common neurodegenerative disorders, instead of being consequences of a prior insult, a theory we believe to be misconstrued. Herein, we review the roots of this mitochondrial hypothesis and offer a new perspective wherein mitochondria are analyzed not only from the OxPhos point of view, but also as a complex organelle residing at the epicenter of many metabolic pathways.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Mitocondrias/metabolismo , Modelos Neurológicos , Neuronas/metabolismo , Fosforilación Oxidativa , Enfermedad de Parkinson/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Muerte Celular , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Humanos , Mitocondrias/genética , Mitocondrias/patología , Mutación , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
20.
Mov Disord ; 27(3): 343, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22512003
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA