Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008983

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease with a multifactorial etiology. A multitarget treatment that modulates multifaceted biological functions might be more effective than a single-target approach. Here, the therapeutic efficacy of combination treatment using anti-Aß antibody NP106 and curcumin analog TML-6 versus monotherapy was investigated in an APP/PS1 mouse model of AD. Our data demonstrate that both combination treatment and monotherapy attenuated brain Aß and improved the nesting behavioral deficit to varying degrees. Importantly, the combination treatment group had the lowest Aß levels, and insoluble forms of Aß were reduced most effectively. The nesting performance of APP/PS1 mice receiving combination treatment was better than that of other APP/PS1 groups. Further findings indicate that enhanced microglial Aß phagocytosis and lower levels of proinflammatory cytokines were concurrent with the aforementioned effects of NP106 in combination with TML-6. Intriguingly, combination treatment also normalized the gut microbiota of APP/PS1 mice to levels resembling the wild-type control. Taken together, combination treatment outperformed NP106 or TML-6 monotherapy in ameliorating Aß pathology and the nesting behavioral deficit in APP/PS1 mice. The superior effect might result from a more potent modulation of microglial function, cerebral inflammation, and the gut microbiota. This innovative treatment paradigm confers a new avenue to develop more efficacious AD treatments.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/deficiencia , Anticuerpos Monoclonales/farmacología , Curcumina/farmacología , Presenilina-1/deficiencia , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores , Curcumina/análogos & derivados , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Inmunohistoquímica , Ratones , Ratones Noqueados , Microbiota/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Terapia Molecular Dirigida , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/patología
2.
J Leukoc Biol ; 110(6): 1005-1022, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34494312

RESUMEN

Chronic neuroinflammation has been shown to exert adverse influences on the pathology of Alzheimer's disease (AD), associated with the release of abundant proinflammatory mediators by excessively activated microglia, causing synaptic dysfunction, neuronal degeneration, and memory deficits. Thus, the prevention of microglial activation-associated neuroinflammation is important target for deterring neurodegenerative disorders. Peony seed oil (PSO) is a new food resource, rich in α-linolenic acid, the precursor of long chain omega-3 polyunsaturated fatty acids, including docosahexaenoic acid and eicosapentaenoic acid, which exhibit anti-inflammatory properties by altering cell membrane phospholipid fatty acid compositions, disrupting lipid rafts, and inhibiting the activation of the proinflammatory transcription factor NF-κB. However, few studies have examined the anti-neuroinflammatory effects of PSO in AD, and the relevant molecular mechanisms remain unclear. Presenilin1/2 conditional double knockout (PS cDKO) mice display obvious AD-like phenotypes, such as neuroinflammatory responses, synaptic dysfunction, and cognitive deficits. Here, we assessed the potential neuroprotective effects of PSO against neuroinflammation-mediated cognitive deficits in PS cDKO using behavioral tests and molecular biologic analyses. Our study demonstrated that PSO suppressed microglial activation and neuroinflammation through the down-regulation of proinflammatory mediators, such as inducible NOS, COX-2, IL-1ß, and TNF-α, in the prefrontal cortex and hippocampus of PS cDKO mice. Further, PSO significantly lessened memory impairment by reversing hyperphosphorylated tau and synaptic proteins deficits in PS cDKO mice. Importantly, PSO's therapeutic effects on cognitive deficits were due to inhibiting neuroinflammatory responses mediated by NF-κB signaling pathway. Taken together, PSO may represent an effective dietary supplementation to restrain the neurodegenerative processes of AD.


Asunto(s)
Enfermedad de Alzheimer , Antiinflamatorios/farmacología , Disfunción Cognitiva , Microglía/efectos de los fármacos , Aceites de Plantas/farmacología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Inflamación , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Paeonia , Presenilina-1/deficiencia , Presenilina-2/deficiencia , Semillas , Transducción de Señal/efectos de los fármacos
3.
Neural Plast ; 2021: 8814616, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33505459

RESUMEN

Neuroinflammation is considered as one of the crucial pathogenesis in promoting neurodegenerative progress of Alzheimer's disease (AD). As complementary and alternative therapy, electroacupuncture (EA) stimulation has been widely used in clinical practice for anti-inflammation. However, whether EA promotes the cognitive deficits resulting from neuroinflammation in AD remains unclear. In this study, the presenilin 1 and 2 conditional double knockout (PS cDKO) mice, exhibited a series of AD-like pathology, robust neuroinflammatory responses, and memory deficits, were used to evaluate the potential neuroprotective effect of EA at Baihui (GV 20) and Shenting (GV 24) by behavioral testing, electrophysiology recording, and molecular biology analyzing. First, we observed that EA improved memory deficits and impaired synaptic plasticity. Moreover, EA possesses an ability to suppress the hyperphosphorylated tau and robust elevated NLRP3, ASC, Caspase-1, IL-1ß, and IL-18 in PS cDKO mice. Importantly, MCC950, a potent and selective inhibitor of NLPR3 inflammasome, has similar effects on inhibiting the hyperphosphorylated tau and the robust elevated NLRP3 components and neuroinflammatory responses of PS cDKO mice as well as EA treatment. Furthermore, EA treatment is not able to further improve the AD-like phenotypes of PS cDKO mice in combination with the MCC950 administration. Therefore, EA stimulation at GV 20 and GV 24 acupoints may be a potential alternative therapy for deterring cognitive deficits in AD through suppression of NLRP3 inflammasome activation.


Asunto(s)
Disfunción Cognitiva/terapia , Electroacupuntura/métodos , Mediadores de Inflamación/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Presenilina-1/deficiencia , Presenilina-2/deficiencia , Animales , Disfunción Cognitiva/metabolismo , Furanos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Indenos/farmacología , Mediadores de Inflamación/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Presenilina-1/genética , Presenilina-2/genética , Sulfonamidas/farmacología
4.
J Biochem ; 167(5): 463-471, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-31816046

RESUMEN

Alzheimer disease (AD) is the most common neurodegenerative disease worldwide. The pathological hallmark of AD is the presence of senile plaques in the brain, which are accumulations of amyloid-ß peptide (Aß) ending at the 42nd residue (i.e. Aß42), which is produced through multistep cleavage by γ-secretase. Thus, methods to regulate γ-secretase activity to attenuate the production of Aß42 are in urgent demand towards the development of treatments for AD. We and others have demonstrated that γ-secretase activity is affected by its localization and ambient environment. In particular, an increase in Aß42 production is correlated with the intracellular transport of γ-secretase and endosomal maturation-dependent luminal acidification. In this study, we focused on the mechanism by which γ-secretase affects Aß42 production together with alterations in pH. Histidine is known to function as a pH sensor in many proteins, to regulate their activities through the protonation state of the imidazole side chain. Among the histidines facing the luminal side of presenilin (PS) 1, which is the catalytic subunit of γ-secretase, point mutations at H131 had no effect on the Aß42 production ratio in an acidic environment. We also observed an increase in Aß42 ratio when histidine was introduced into N137 of PS2, which is the corresponding residue of H131 in PS1. These results indicated that H131 serves as the pH sensor in PS1, which contains γ-secretase, to regulate Aß42 production depending on the luminal pH. Our findings provide new insights into therapeutic strategies for AD targeting endosomes or the intracellular transport of γ-secretase.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Histidina/metabolismo , Presenilina-1/química , Presenilina-1/metabolismo , Animales , Línea Celular , Femenino , Histidina/genética , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Noqueados , Mutación , Presenilina-1/deficiencia , Presenilina-1/genética , Ratas
5.
Cell Death Dis ; 9(9): 879, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158533

RESUMEN

Autophagy impairment is commonly implicated in the pathological characteristic of Alzheimer's disease (AD). Presenilin 1 (PS1) expression in human brain gradually decreases with age and its mutations account for the most common cases of early-onset familial Alzheimer's disease (FAD). The dominant autophagy phenotypes occur in PS1-knockout and PS1 mutant neurons; it is still unknown whether PS1 deficiency causes serious autophagy impairment in neural stem cells (NSCs). Herein, we generated the heterozygote and homozygote of PS1 knockout in human induced pluripotent stem cells (iPSCs) via CRISPR/Cas9-based gene editing and differentiated them into human NSCs. In these human PS1-deficient NSCs, reduced autophagosome formation and downregulated expression of autophagy-lysosome pathway (ALP)-related mRNAs, as well as proteins were observed. Mechanistically, ERK/CREB inhibition and GSK3ß activation had key roles in reducing TFEB expression in PS1-knockout NSCs. Pharmacological inhibition of GSK3ß upregulated the expression of TFEB and ALP-related proteins in PS1-knockout NSCs, whereas this effect could be blocked by CREB inhibition. These findings demonstrate that PS1 deficiency causes autophagy suppression in human NSCs via downregulating ERK/CREB signaling.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Autofagia/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Presenilina-1/deficiencia , Autofagosomas/metabolismo , Autofagosomas/fisiología , Encéfalo/metabolismo , Encéfalo/fisiología , Sistemas CRISPR-Cas/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Lisosomas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Mutación/fisiología , Células-Madre Neurales/fisiología , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
6.
PLoS One ; 13(8): e0200752, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30089166

RESUMEN

Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) where pathology is thought to be regulated by autoreactive T cells of the Th1 and Th17 phenotype. In this study we sought to understand the functions of Presenilin 1 (PSEN1) in regulating T cell effector responses in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. PSEN1 is the catalytic subunit of γ-secretase a multimolecular protease that mediates intramembranous proteolysis. γ-secretase is known to regulate several pathways of immune importance. Here we examine the effects of disrupting PSEN1 functions on EAE and T effector differentiation using small molecule inhibitors of γ-secretase (GSI) and T cell-specific conditional knockout mice (PSEN1 cKO). Surprisingly, blocking PSEN1 function by GSI treatment or PSEN1 cKO had little effect on the development or course of MOG35-55-induced EAE. In vivo GSI administration reduced the number of myelin antigen-specific T cells and suppressed Th1 and Th17 differentiation following immunization. In vitro, GSI treatment inhibited Th1 differentiation in neutral but not IL-12 polarizing conditions. Th17 differentiation was also suppressed by the presence of GSI in all conditions and GSI-treated Th17 T cells failed to induce EAE following adoptive transfer. PSEN cKO T cells showed reduced Th1 and Th17 differentiation. We conclude that γ-secretase and PSEN1-dependent signals are involved in T effector responses in vivo and potently regulate T effector differentiation in vitro, however, they are dispensable for EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Presenilina-1/genética , Células TH1/metabolismo , Células Th17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dibenzazepinas/farmacología , Dibenzazepinas/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Presenilina-1/deficiencia , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología
7.
Mol Neurobiol ; 55(6): 5232-5242, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28879407

RESUMEN

MicroRNAs are small non-coding RNAs that function as regulators of gene expression. The altered expression of microRNAs influences the pathogenesis of Alzheimer's disease. Many researchers have focused on studies based on the relatively distinctive etiology of familial Alzheimer's disease due to the absence of risk factors in the pathogenesis of sporadic Alzheimer's disease. Although there is a limitation in Alzheimer's disease studies, both Alzheimer's disease types have a common risk factor-aging. No study to date has examined the aging factor in Alzheimer's disease animal models with microRNAs. To investigate the effect of aging on the changes in microRNA expressions in the Alzheimer's disease animal model, we selected 37 hippocampal microRNAs whose expression in 12- and 18-month aged mice changed significantly using microRNA microarray. On the basis of bioinformatics databases, 30 hippocampal microRNAs and their putative targets of PSEN1/PSEN2 double knockout mice were included in 28 pathways such as the wnt signaling pathway and ubiquitin-mediated proteolysis pathway. Cortical microRNAs and its putative targets involved in pathological aging were included in only four pathways such as the heparin sulfate biosynthesis. The altered expressions of these hippocampal microRNAs were associated to the imbalance between neurotoxic and neuroprotective functions and seemed to affect neurodegeneration in PSEN1/PSEN2 double knockout mice more severely than in wild-type mice. This microRNA profiling suggests that microRNAs play potential roles in the normal aging process, as well as in the Alzheimer's disease process.


Asunto(s)
Envejecimiento/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Presenilina-1/deficiencia , Presenilina-2/deficiencia , Animales , Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Ratones Noqueados , MicroARNs/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-2/genética , Presenilina-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
J Cell Physiol ; 233(2): 1548-1557, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28617969

RESUMEN

Recently, PSEN1 has been reported to have mutations in dilated cardiomyopathy pedigrees. However, the function and mechanism of PSEN1 in cardiomyopathy remains unresolved. Here, we established four types of genetically modified mice to determine the function of PSEN1 in cardiac development and pathology. PSEN1 null mutation resulted in perinatal death, retardation of heart growth, ventricular dilatation, septum defects, and valvular thickening. PSEN1 knockout in adults led to decreased muscle fibers, widened sarcomere Z lines and reduced lengths of sarcomeres in cardiomyocytes. Cardiovascular loss of function of PSEN1 induced by Sm22a-Cre or Myh6-Cre/ER/tamoxifen also resulted in severe ultrastructural abnormalities, such as relaxed gap junctions between neighboring cardiomyocytes. Functionally, cardiovascular deletion of PSEN1 caused spontaneous mortality from birth to adulthood and led to diastolic heart dysfunction, including decreased volume of the left ventricle at the end-systolic and end-diastolic stages. Additionally, in a myocardial ischemia model, deletion of PSEN1 in the cardiovascular system first protected mice by inducing adaptive hypertrophy but ultimately resulted in severe heart failure. Furthermore, a collection of genes was abnormally expressed in the hearts of cardiac-specific PSEN1 knockout mice. They were enriched in cell proliferation, calcium regulation, and so on. Taken together, dynamic regulation and abnormal function of PSEN1 underlie the pathogenesis of cardiovascular diseases due to ultrastructural abnormality of cardiomyocytes.


Asunto(s)
Eliminación de Gen , Cardiopatías Congénitas/fisiopatología , Presenilina-1/deficiencia , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Animales , Diástole , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones Noqueados , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Fenotipo , Presenilina-1/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología
9.
Glia ; 65(4): 569-580, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28130845

RESUMEN

Astrocytes play crucial roles in brain homeostasis and are emerging as regulatory elements of neuronal and synaptic physiology by responding to neurotransmitters with Ca2+ elevations and releasing gliotransmitters that activate neuronal receptors. Aging involves neuronal and astrocytic alterations, being considered risk factor for neurodegenerative diseases. Most evidence of the astrocyte-neuron signaling is derived from studies with young animals; however, the features of astrocyte-neuron signaling in adult and aging brain remain largely unknown. We have investigated the existence and properties of astrocyte-neuron signaling in physiologically and pathologically aging mouse hippocampal and cortical slices at different lifetime points (0.5 to 20 month-old animals). We found that astrocytes preserved their ability to express spontaneous and neurotransmitter-dependent intracellular Ca2+ signals from juvenile to aging brains. Likewise, resting levels of gliotransmission, assessed by neuronal NMDAR activation by glutamate released from astrocytes, were largely preserved with similar properties in all tested age groups, but DHPG-induced gliotransmission was reduced in aged mice. In contrast, gliotransmission was enhanced in the APP/PS1 mouse model of Alzheimer's disease, indicating a dysregulation of astrocyte-neuron signaling in pathological conditions. Disruption of the astrocytic IP3 R2 mediated-signaling, which is required for neurotransmitter-induced astrocyte Ca2+ signals and gliotransmission, boosted the progression of amyloid plaque deposits and synaptic plasticity impairments in APP/PS1 mice at early stages of the disease. Therefore, astrocyte-neuron interaction is a fundamental signaling, largely conserved in the adult and aging brain of healthy animals, but it is altered in Alzheimer's disease, suggesting that dysfunctions of astrocyte Ca2+ physiology may contribute to this neurodegenerative disease. GLIA 2017 GLIA 2017;65:569-580.


Asunto(s)
Envejecimiento , Astrocitos/fisiología , Encéfalo/citología , Comunicación Celular/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Acetilcolina/farmacología , Adenosina Trifosfato/farmacología , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Calcio/metabolismo , Comunicación Celular/efectos de los fármacos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Presenilina-1/deficiencia , Presenilina-1/genética , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética
10.
Nature ; 552(7685): 355-361, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29293211

RESUMEN

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Microglía/metabolismo , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presenilina-1/deficiencia , Presenilina-1/genética , Dominios Proteicos , Memoria Espacial/fisiología
11.
Sci Rep ; 6: 26874, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27240887

RESUMEN

Notch signaling is known to control early pancreatic differentiation through Ngn3 repression. In later stages, downstream of Notch, the Presenilins are still required to maintain the endocrine fate allocation. Amongst their multiple targets, it remains unclear which one actually controls the maintenance of the fate of the early islets. Conditional deletions of the Notch effector RBP-Jκ with lineage tracing in Presenilin-deficient endocrine progenitors, demonstrated that this factor is central to the control of the fate through a non-canonical Notch mechanism. RBP-Jκ mice exhibit normal islet morphogenesis and function, however, a fraction of the progenitors fails to differentiate and develop into disorganized masses resembling acinar to ductal metaplasia and chronic pancreatitis. A subsequent deletion of RBP-Jκ in forming ß-cells led to the transdifferentiation into the other endocrine cells types, indicating that this factor still mediates the maintenance of the fate within the endocrine lineage itself. These results highlight the dual importance of Notch signaling for the endocrine lineage. Even after Ngn3 expression, Notch activity is required to maintain both fate and maturation of the Ngn3 progenitors. In a subset of the cells, these alterations of Notch signaling halt their differentiation and leads to acinar to ductal metaplasia.


Asunto(s)
Células Enteroendocrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Islotes Pancreáticos/metabolismo , Morfogénesis/genética , Células Madre Embrionarias de Ratones/metabolismo , Células Acinares/citología , Células Acinares/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Embrión de Mamíferos , Células Enteroendocrinas/citología , Femenino , Genes Reporteros , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Integrasas/genética , Integrasas/metabolismo , Islotes Pancreáticos/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Presenilina-1/deficiencia , Presenilina-1/genética , Presenilina-2/deficiencia , Presenilina-2/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal
12.
Nat Commun ; 6: 8997, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26646366

RESUMEN

Alzheimer's disease (AD) is characterized by the transition of amyloid-ß (Aß) monomers into toxic oligomers and plaques. Given that Aß abnormality typically precedes the development of clinical symptoms, an agent capable of disaggregating existing Aß aggregates may be advantageous. Here we report that a small molecule, 4-(2-hydroxyethyl)-1-piperazinepropanesulphonic acid (EPPS), binds to Aß aggregates and converts them into monomers. The oral administration of EPPS substantially reduces hippocampus-dependent behavioural deficits, brain Aß oligomer and plaque deposits, glial γ-aminobutyric acid (GABA) release and brain inflammation in an Aß-overexpressing, APP/PS1 transgenic mouse model when initiated after the development of severe AD-like phenotypes. The ability of EPPS to rescue Aß aggregation and behavioural deficits provides strong support for the view that the accumulation of Aß is an important mechanism underlying AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Precursor de Proteína beta-Amiloide/deficiencia , Hipocampo/efectos de los fármacos , Piperazinas/administración & dosificación , Placa Amiloide/metabolismo , Presenilina-1/deficiencia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Presenilina-1/genética
13.
Sci Rep ; 5: 17042, 2015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26608390

RESUMEN

The presenilin 1 (PSEN1) L271V mutation causes early-onset familial Alzheimer's disease by disrupting the alternative splicing of the PSEN1 gene, producing some transcripts harboring the L271V point mutation and other transcripts lacking exon 8 (PS1(∆exon8)). We previously reported that PS1 L271V increased amyloid beta (Aß) 42/40 ratios, while PS1(∆exon8) reduced Aß42/40 ratios, indicating that the former and not the exon 8 deletion transcript is amyloidogenic. Also, PS1(∆exon8) did not rescue Aß generation in PS1/2 double knockout cells indicating its identity as a severe loss-of-function splice form. PS1(∆exon8) is generated physiologically raising the possibility that we had identified the first physiological inactive PS1 isoform. We studied PS1(∆exon8) in vivo by crossing PS1(∆exon8) transgenics with either PS1-null or Dutch APP(E693Q) mice. As a control, we crossed APP(E693Q) with mice expressing a deletion in an adjacent exon (PS1(∆exon9)). PS1(∆exon8) did not rescue embryonic lethality or Notch-deficient phenotypes of PS1-null mice displaying severe loss of function in vivo. We also demonstrate that this splice form can interact with wildtype PS1 using cultured cells and co-immunoprecipitation (co-IP)/bimolecular fluorescence complementation. Further co-IP demonstrates that PS1(∆exon8) interacts with nicastrin, participating in the γ-secretase complex formation. These data support that catalytically inactive PS1(∆exon8) is generated physiologically and participates in protein-protein interactions.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Exones/genética , Glicoproteínas de Membrana/metabolismo , Presenilina-1/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Animales , Encéfalo/metabolismo , Embrión de Mamíferos/metabolismo , Retículo Endoplásmico/metabolismo , Fluorescencia , Células HEK293 , Humanos , Inmunoprecipitación , Ratones Noqueados , Actividad Motora , Fenotipo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1/deficiencia , Presenilina-1/metabolismo , Unión Proteica , Eliminación de Secuencia/genética , Transgenes
14.
Behav Brain Res ; 281: 32-42, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25510196

RESUMEN

In addition to cognitive impairments, deficits in non-cognitive behaviors are also common neurological sequelae in Alzheimer's disease and its animal models. Hesperidin, a flavanone glycoside found abundantly in citrus fruits, was orally given (100 mg/kg body weight) to 5-month-old transgenic APP/PS1 mice, a mouse model of cerebral amyloidosis for Alzheimer's disease. After a relatively short-term treatment of 10 days, hesperidin significantly restored deficits in non-cognitive nesting ability and social interaction. Further immunohistochemical analysis showed significantly attenuated ß-amyloid deposition, plaque associated APP expression, microglial activation and TGF-ß immunoreactivity in brains of APP/PS1 mice, which suggests that ameliorated behavioral impairments might be attributable to reduced Aß deposition and attenuated neuro-inflammatory reaction. Additionally, efficient anti-inflammatory effects of hesperidin were confirmed in vitro. Our findings suggest that hesperidin might be a potential candidate for the treatment of AD or even other neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/patología , Hesperidina/farmacología , Desempeño Psicomotor/efectos de los fármacos , Conducta Social , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/inmunología , Placa Amiloide/tratamiento farmacológico , Presenilina-1/deficiencia , Presenilina-1/genética , Factor de Crecimiento Transformador beta/inmunología , Resultado del Tratamiento
15.
J Neurosci ; 34(48): 15912-22, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25429133

RESUMEN

Mutations in the presenilin (PSEN1 and PSEN2) genes are linked to familial Alzheimer's disease (AD) and cause loss of its essential function. Complete inactivation of presenilins in excitatory neurons of the adult mouse cerebral cortex results in progressive memory impairment and age-dependent neurodegeneration, recapitulating key features of AD. In this study, we examine the effects of varying presenilin dosage on cortical neuron survival by generating presenilin-1 conditional knock-out (PS1 cKO) mice carrying two, one, or zero copies of the PS2 gene. We found that PS1 cKO;PS2(+/-) mice at 16 months exhibit marked neurodegeneration in the cerebral cortex with ∼17% reduction of cortical volume and neuron number, as well as astrogliosis and microgliosis compared with ∼50% reduction of cortical volume and neuron number in PS1 cKO;PS2(-/-) mice. Moreover, there are more apoptotic neurons labeled by activated caspase-3 immunoreactivity and TUNEL assay in PS1 cKO;PS2(+/-) mice at 16 months, whereas apoptotic neurons are increased in the PS1 cKO;PS2(-/-) cerebral cortex at 4 months. The accumulation of the C-terminal fragments of the amyloid precursor protein is inversely correlated with PS dosage. Interestingly, levels of PS2 are higher in the cerebral cortex of PS1 cKO mice, suggesting a compensatory upregulation that may provide protection against neurodegeneration in these mice. Together, our findings show that partial to complete loss of presenilin activity causes progressively more severe neurodegeneration in the mouse cerebral cortex during aging, suggesting that impaired presenilin function by PSEN mutations may lead to neurodegeneration and dementia in AD.


Asunto(s)
Envejecimiento/metabolismo , Corteza Cerebral/metabolismo , Neuronas/metabolismo , Presenilina-1/deficiencia , Presenilina-2/deficiencia , Envejecimiento/patología , Animales , Supervivencia Celular/fisiología , Corteza Cerebral/patología , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/patología
16.
Mitochondrion ; 17: 106-15, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25034304

RESUMEN

A water-soluble formulation of CoQ10 (WS-CoQ10) was shown to stabilize mitochondria and prevent oxidative stress-induced neuronal death. Presenilin-1 (PS-1)-mutated Alzheimer's Disease (AD) fibroblasts (PSAF) were used for studying the effects of PS-1 mutation. PS-1 mutation correlated to increased reactive oxygen species (ROS) production and stress induced premature senescence (SIPS) in PSAF; WS-CoQ10 treatment decreased ROS generation, increased population doublings, and postponed SIPS. Treated PSAF had higher PCNA expression, and lower levels of MnSOD, p21, p16Ink4A, and Rb. WS-CoQ10 caused the resumption of autophagy in PSAF. Thus, WS-CoQ10 as inhibitor of SIPS and ameliorator of autophagy could be an effective prophylactic/therapeutic agent for AD.


Asunto(s)
Envejecimiento , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Presenilina-1/deficiencia , Estrés Fisiológico , Ubiquinona/análogos & derivados , Vitaminas/metabolismo , Enfermedad de Alzheimer , Células Cultivadas , Femenino , Humanos , Masculino , Ubiquinona/metabolismo
17.
J Biol Chem ; 289(29): 20182-91, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24878959

RESUMEN

Although soluble species of the amyloid-ß peptide Aß42 correlate with disease symptoms in Alzheimer disease, little is known about the biological activities of amyloid-ß (Aß). Here, we show that Aß peptides varying in lengths from 38 to 43 amino acids are internalized by cultured neuroblastoma cells and can be found in the nucleus. By three independent methods, we demonstrate direct detection of nuclear Aß42 as follows: (i) biochemical analysis of nuclear fractions; (ii) detection of biotin-labeled Aß in living cells by confocal laser scanning microscopy; and (iii) transmission electron microscopy of Aß in cultured cells, as well as brain tissue of wild-type and transgenic APPPS1 mice (overexpression of amyloid precursor protein and presenilin 1 with Swedish and L166P mutations, respectively). Also, this study details a novel role for Aß42 in nuclear signaling, distinct from the amyloid precursor protein intracellular domain. Chromatin immunoprecipitation showed that Aß42 specifically interacts as a repressor of gene transcription with LRP1 and KAI1 promoters. By quantitative RT-PCR, we confirmed that mRNA levels of the examined candidate genes were exclusively decreased by the potentially neurotoxic Aß42 wild-type peptide. Shorter peptides (Aß38 or Aß40) and other longer peptides (nontoxic Aß42 G33A substitution or Aß43) did not affect mRNA levels. Overall, our data indicate that the nuclear translocation of Aß42 impacts gene regulation, and deleterious effects of Aß42 in Alzheimer disease pathogenesis may be influenced by altering the expression profiles of disease-modifying genes.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Transporte Activo de Núcleo Celular , Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Neuronas/metabolismo , Neuronas/ultraestructura , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Presenilina-1/deficiencia , Presenilina-1/genética , Presenilina-1/metabolismo , Multimerización de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Electricidad Estática
18.
Brain ; 137(Pt 7): 1958-70, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24860142

RESUMEN

A growing number of PSEN1 mutations have been associated with dementia with Lewy bodies and familial Alzheimer's disease with concomitant α-synuclein pathology. The objective of this study was to determine if PSEN1 plays a direct role in the development of α-synuclein pathology in these diseases. Using mass spectrometry, immunoelectron microscopy and fluorescence lifetime image microscopy based on Forster resonance energy transfer (FLIM-FRET) we identified α-synuclein as a novel interactor of PSEN1 in wild-type mouse brain tissue. The interaction of α-synuclein with PSEN1 was detected in post-mortem brain tissue from cognitively normal cases and was significantly increased in tissue from cases with dementia with Lewy bodies and familial Alzheimer's disease associated with known PSEN1 mutations. We confirmed an increased interaction of PSEN1 and α-synuclein in cell lines expressing well characterized familial Alzheimer's disease PSEN1 mutations, L166P and delta exon 9, and demonstrated that PSEN1 mutations associate with increased membrane association and accumulation of α-synuclein. Our data provides evidence of a molecular interaction of PSEN1 and α-synuclein that may explain the clinical and pathophysiological overlap seen in synucleinopathies, including Parkinson's disease, dementia with Lewy bodies, and some forms of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Presenilina-1/metabolismo , alfa-Sinucleína/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Células CHO , Células Cultivadas , Corteza Cerebral , Cricetulus , Femenino , Glutatión Transferasa/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Microscopía Inmunoelectrónica , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Presenilina-1/deficiencia , Presenilina-1/genética
19.
J Neurosci ; 34(9): 3320-39, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24573290

RESUMEN

Overexpression and/or abnormal cleavage of amyloid precursor protein (APP) are linked to Alzheimer's disease (AD) development and progression. However, the molecular mechanisms regulating cellular levels of APP or its processing, and the physiological and pathological consequences of altered processing are not well understood. Here, using mouse and human cells, we found that neuronal damage induced by UV irradiation leads to specific APP, APLP1, and APLP2 decline by accelerating their secretase-dependent processing. Pharmacological inhibition of endosomal/lysosomal activity partially protects UV-induced APP processing implying contribution of the endosomal and/or lysosomal compartments in this process. We found that a biological consequence of UV-induced γ-secretase processing of APP is impairment of APP axonal transport. To probe the functional consequences of impaired APP axonal transport, we isolated and analyzed presumptive APP-containing axonal transport vesicles from mouse cortical synaptosomes using electron microscopy, biochemical, and mass spectrometry analyses. We identified a population of morphologically heterogeneous organelles that contains APP, the secretase machinery, molecular motors, and previously proposed and new residents of APP vesicles. These possible cargoes are enriched in proteins whose dysfunction could contribute to neuronal malfunction and diseases of the nervous system including AD. Together, these results suggest that damage-induced APP processing might impair APP axonal transport, which could result in failure of synaptic maintenance and neuronal dysfunction.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/efectos de la radiación , Axones/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Neuronas/citología , Rayos Ultravioleta , Precursor de Proteína beta-Amiloide/deficiencia , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Axones/ultraestructura , Células Cultivadas , Embrión de Mamíferos , Hipocampo/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroblastoma/patología , Neuronas/efectos de la radiación , Presenilina-1/deficiencia , Presenilina-2/deficiencia , Transfección
20.
Mol Neurobiol ; 49(1): 10-27, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23794287

RESUMEN

Mutations in presenilin-1 (PS1) are tightly associated with early-onset familial Alzheimer's disease (FAD), which is characterized by extracellular amyloid plaques and the accumulation of intracellular Tau. In addition to being the catalytic subunit of γ-secretase, PS1 has been shown to regulate diverse cellular functions independent of its proteolytic activity. We found that cells deficient in PS1 exhibit reduced levels of p62 protein, a cargo-receptor shuttling Tau for degradation. The downregulation of PS1 led to a significant decrease in both the protein and mRNA transcript of p62, concomitant with attenuated p62 promoter activity. This PS1-dependent regulation of p62 expression was mediated through an Akt/AP-1 pathway independent of the proteolytic activity of PS1/γ-secretase. This p62-mediated Tau degradation was significantly impaired in PS1-deficient cells, which can be rescued by ectopic expression of either p62 or wild-type PS1 but not mutant PS1 containing FAD-linked mutations. Our study suggests a novel function for PS1 in modulating p62 expression to control the proteostasis of Tau.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Regulación de la Expresión Génica , Presenilina-1/fisiología , Proteolisis , Proteínas tau/antagonistas & inhibidores , Proteínas tau/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mutación , Fenotipo , Presenilina-1/deficiencia , Presenilina-1/genética , Proteína Sequestosoma-1 , Pez Cebra/genética , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA