Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuropathol Appl Neurobiol ; 46(7): 686-706, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32716575

RESUMEN

BACKGROUND: Evidence suggests that amyloid ß (Aß) peptides play an important role in the degeneration of neurons during the development of Alzheimer's disease (AD), the prevalent cause of dementia affecting the elderly. The endosomal-lysosomal system, which acts as a major site for Aß metabolism, has been shown to exhibit abnormalities in vulnerable neurons of the AD brain, reflected by enhanced levels/expression of lysosomal enzymes including cathepsin D (CatD). At present, the implication of CatD in selective neuronal vulnerability in AD pathology remains unclear. METHODS: We evaluated the role of CatD in the degeneration of neurons in Aß-treated cultures, transgenic AD mouse model (that is 5xFAD) and post mortem AD brain samples. RESULTS: Our results showed that Aß1-42 -induced toxicity in cortical cultured neurons is associated with impaired lysosomal integrity, enhanced levels of carbonylated proteins and tau phosphorylation. The cellular and cytosolic levels/activity of CatD are also elevated in cultured neurons following exposure to Aß peptide. Additionally, we observed that CatD cellular and subcellular levels/activity are increased in the affected cortex, but not in the unaffected cerebellum, of 5xFAD mice and post mortem AD brains. Interestingly, treatment of cultured neurons with nanoparticles PLGA, which targets lysosomal system, attenuated Aß toxicity by reducing the levels of carbonylated proteins, tau phosphorylation and the level/distribution/activity of CatD. CONCLUSION: Our study reveals that increased cytosolic level/activity of CatD play an important role in determining neuronal vulnerability in AD. Additionally, native PLGA can protect neurons against Aß toxicity by restoring lysosomal membrane integrity, thus signifying its implication in attenuating AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Catepsina D/metabolismo , Lisosomas/metabolismo , Neuronas/patología , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fragmentos de Péptidos/metabolismo , Proteínas tau/metabolismo
2.
Nat Cell Biol ; 2(12): 863-70, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11146649

RESUMEN

Presenilin 1 (PS1), a polytopic membrane protein, has a critical role in the trafficking and proteolysis of a selected set of transmembrane proteins. The vast majority of individuals affected with early onset familial Alzheimer's disease (FAD) carry missense mutations in PS1. Two studies have suggested that loss of PS1 function, or expression of FAD-linked PS1 variants, compromises the mammalian unfolded-protein response (UPR), and we sought to evaluate the potential role of PS1 in the mammalian UPR. Here we show that that neither the endoplasmic reticulum (ER) stress-induced accumulation of BiP and CHOP messenger RNA, nor the activation of ER stress kinases IRE1alpha and PERK, is compromised in cells lacking both PS1 and PS2 or in cells expressing FAD-linked PS1 variants. We also show that the levels of BiP are not significantly different in the brains of individuals with sporadic Alzheimer's disease or PS1-mediated FAD to levels in control brains. Our findings provide evidence that neither loss of PS1 and PS2 function, nor expression of PS1 variants, has a discernable impact on ER stress-mediated induction of the several established 'readouts' of the UPR pathway.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Portadoras/genética , Proteínas de Choque Térmico , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/genética , Factores de Transcripción/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Línea Celular , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Variación Genética , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Presenilina-1 , Presenilina-2 , Desnaturalización Proteica , Pliegue de Proteína , Proteínas/química , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción CHOP , Regulación hacia Arriba
3.
Nat Med ; 3(7): 756-60, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9212102

RESUMEN

Mutations in the presenilin 1 (PS1) and presenilin 2 (PS2) genes can cause Alzheimer's disease in affected members of the majority of early-onset familial Alzheimer's disease (FAD) pedigrees. PS1 encodes an ubiquitously expressed, eight transmembrane protein. PS1 is endoproteolytically processed to an amino-terminal derivative (approximately 27-28 kDa) and a carboxy-terminal derivative (approximately 17-18 kDa). These polypeptides accumulate to saturable levels in the brains of transgenic mice, independent of the expression of PS1 holoprotein. We now document that, in the brains of transgenic mice, the absolute amounts of accumulated N- and C-terminal derivatives generated from the FAD-linked PS1 variants in which Glu replaces Ala at codon 246 (A246E) or Leu replaces Met at codon 146 (M146L) accumulate to a significantly higher degree (approximately 40-50%) than the fragments derived from wild-type PS1. Moreover, the FAD-linked deltaE9 PS1 variant, a polypeptide that is not subject to endoproteolytic cleavage in vivo, also accumulates in greater amounts than the fragments generated from wild-type human PS1. Thus, the metabolism of PS1 variants linked to FAD is fundamentally different from that of wild-type PS1 in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Actinas/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Corteza Cerebral/metabolismo , Variación Genética , Hipocampo/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Transgénicos , Mutación Puntual , Presenilina-1
4.
Nat Med ; 4(4): 447-51, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9546791

RESUMEN

Alzheimer's disease (AD) is characterized by the accumulation of cerebral plaques composed of 40- and 42-amino acid beta-amyloid (Abeta) peptides, and autosomal dominant forms of AD appear to cause disease by promoting brain Abeta accumulation. Recent studies indicate that postmenopausal estrogen replacement therapy may prevent or delay the onset of AD. Here we present evidence that physiological levels of 17beta-estradiol reduce the generation of Abeta by neuroblastoma cells and by primary cultures of rat, mouse and human embryonic cerebrocortical neurons. These results suggest a mechanism by which estrogen replacement therapy can delay or prevent AD.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/biosíntesis , Corteza Cerebral/citología , Estradiol/farmacología , Neuronas/fisiología , Enfermedad de Alzheimer , Animales , Células Cultivadas , Técnicas de Cocultivo , Embrión de Mamíferos , Feto , Humanos , Ratones , Neuroblastoma , Neuronas/citología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/biosíntesis , Ratas , Proteínas Recombinantes/biosíntesis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección , Células Tumorales Cultivadas
5.
Glia ; 58(11): 1267-81, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20607864

RESUMEN

Niemann-Pick type C (NPC) disease is an autosomal recessive neurodegenerative disorder characterized by intracellular accumulation of cholesterol and glycosphingolipids in many tissues including the brain. The disease is caused by mutations of either NPC1 or NPC2 gene and is accompanied by a severe loss of neurons in the cerebellum, but not in the hippocampus. NPC pathology exhibits some similarities with Alzheimer's disease, including increased levels of amyloid beta (Abeta)-related peptides in vulnerable brain regions, but very little is known about the expression of amyloid precursor protein (APP) or APP secretases in NPC disease. In this article, we evaluated age-related alterations in the level/distribution of APP and its processing enzymes, beta- and gamma-secretases, in the hippocampus and cerebellum of Npc1(-/-) mice, a well-established model of NPC pathology. Our results show that levels and expression of APP and beta-secretase are elevated in the cerebellum prior to changes in the hippocampus, whereas gamma-secretase components are enhanced in both brain regions at the same time in Npc1(-/-) mice. Interestingly, a subset of reactive astrocytes in Npc1(-/-) mouse brains expresses high levels of APP as well as beta- and gamma-secretase components. Additionally, the activity of beta-secretase is enhanced in both the hippocampus and cerebellum of Npc1(-/-) mice at all ages, while the level of C-terminal APP fragments is increased in the cerebellum of 10-week-old Npc1(-/-) mice. These results, taken together, suggest that increased level and processing of APP may be associated with the development of pathology and/or degenerative events observed in Npc1(-/-) mouse brains.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Cerebelo/metabolismo , Hipocampo/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología , Procesamiento Proteico-Postraduccional/genética , Proteínas/genética , Proteínas/metabolismo , Envejecimiento/genética , Envejecimiento/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Cerebelo/enzimología , Cerebelo/patología , Modelos Animales de Enfermedad , Activación Enzimática/genética , Hipocampo/enzimología , Hipocampo/patología , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/enzimología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo
6.
Neuron ; 28(2): 461-73, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11144356

RESUMEN

Amyloid precursor protein (APP) generates the beta-amyloid peptide, postulated to participate in the neurotoxicity of Alzheimer's disease. We report that APP and APLP bind to heme oxygenase (HO), an enzyme whose product, bilirubin, is antioxidant and neuroprotective. The binding of APP inhibits HO activity, and APP with mutations linked to the familial Alzheimer's disease (FAD) provides substantially greater inhibition of HO activity than wild-type APP. Cortical cultures from transgenic mice expressing Swedish mutant APP have greatly reduced bilirubin levels, establishing that mutant APP inhibits HO activity in vivo. Oxidative neurotoxicity is markedly greater in cerebral cortical cultures from APP Swedish mutant transgenic mice than wild-type cultures. These findings indicate that augmented neurotoxicity caused by APP-HO interactions may contribute to neuronal cell death in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Precursor de Proteína beta-Amiloide/análogos & derivados , Precursor de Proteína beta-Amiloide/metabolismo , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Neuronas/enzimología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Bilirrubina/metabolismo , Unión Competitiva/genética , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Hemo-Oxigenasa 1 , Hemina/toxicidad , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Mutación , Neuronas/efectos de los fármacos , Neuronas/patología , Estrés Oxidativo/genética , Estructura Terciaria de Proteína/genética , Transfección , Técnicas del Sistema de Dos Híbridos
7.
Neuron ; 17(5): 1023-30, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8938133

RESUMEN

Mutations in a gene encoding a multitransmembrane protein, termed presenilin 1 (PS1), are causative in the majority of early-onset cases of AD. To determine the topology of PS1, we utilized two strategies: first, we tested whether putative transmembranes are sufficient to export a protease-sensitive substrate across a lipid bilayer; and second, we examined the binding of antibodies to specific PS1 epitopes in cultured cells selectively permeabilized with the pore-forming toxin, streptolysin-O. We document that the "loop," N-terminal, and C-terminal domains of PS1 are oriented toward the cytoplasm.


Asunto(s)
Proteínas de la Membrana/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Células CHO/química , Células CHO/fisiología , Células COS/química , Células COS/fisiología , Cricetinae , Citoplasma/química , Exones/genética , Humanos , Proteínas de la Membrana/genética , Mutación/fisiología , Presenilina-1 , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
8.
Neuron ; 21(5): 1213-21, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9856475

RESUMEN

We have examined the trafficking and metabolism of the beta-amyloid precursor protein (APP), an APP homolog (APLP1), and TrkB in neurons that lack PS1. We report that PS1-deficient neurons fail to secrete Abeta, and that the rate of appearance of soluble APP derivatives in the conditioned medium is increased. Remarkably, carboxyl-terminal fragments (CTFs) derived from APP and APLP1 accumulate in PS1-deficient neurons. Hence, PS1 plays a role in promoting intramembrane cleavage and/or degradation of membrane-bound CTFs. Moreover, the maturation of TrkB and BDNF-inducible TrkB autophosphorylation is severely compromised in neurons lacking PS1. We conclude that PS1 plays an essential role in modulating trafficking and metabolism of a selected set of membrane and secretory proteins in neurons.


Asunto(s)
Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Feto , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/citología , Presenilina-1
9.
Neuron ; 17(1): 181-90, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8755489

RESUMEN

The majority of early-onset cases of familial Alzheimer's disease (FAD) are linked to mutations in two related genes, PS1 and PS2, located on chromosome 14 and 1, respectively. Using two highly specific antibodies against nonoverlapping epitopes of the PS1-encoded polypeptide, termed presenilin 1 (PS1), we document that the preponderant PS1-related species that accumulate in cultured mammalian cells, and in the brains of rodents, primates, and humans are approximately 27-28 kDa N-terminal and approximately 16-17 kDa C-terminal derivatives. Notably, a FAD-linked PS1 variant that lacks exon 9 is not subject to endoproteolytic cleavage. In brains of transgenic mice expressing human PS1, approximately 17 kDa and approximately 27 kDa PS1 derivatives accumulate to saturable levels, and at approximately 1:1 stoichiometry, independent of transgene-derived mRNA. We conclude that PS1 is subject to endoproteolytic processing in vivo.


Asunto(s)
Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Secuencia de Bases , Encéfalo/metabolismo , Células Cultivadas , Chlorocebus aethiops , Humanos , Ratones , Ratones Transgénicos , Sondas Moleculares/genética , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Presenilina-1
10.
Neuron ; 17(5): 1005-13, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8938131

RESUMEN

Mutations in the presenilin 1 (PS1) and presenilin 2 genes cosegregate with the majority of early-onset familial Alzheimer's disease (FAD) pedigrees. We now document that the Abeta1-42(43)/Abeta1-40 ratio in the conditioned media of independent N2a cell lines expressing three FAD-linked PS1 variants is uniformly elevated relative to cells expressing similar levels of wild-type PS1. Similarly, the Abeta1-42(43)/Abeta1-40 ratio is elevated in the brains of young transgenic animals coexpressing a chimeric amyloid precursor protein (APP) and an FAD-linked PS1 variant compared with brains of transgenic mice expressing APP alone or transgenic mice coexpressing wild-type human PS1 and APP. These studies provide compelling support for the view that one mechanism by which these mutant PS1 cause AD is by increasing the extracellular concentration of Abeta peptides terminating at 42(43), species that foster Abeta deposition.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Proteínas de la Membrana/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Química Encefálica/genética , Medios de Cultivo Condicionados , Expresión Génica/fisiología , Humanos , Ratones , Ratones Transgénicos , Mutación/fisiología , Neuroblastoma , Presenilina-1 , Proteínas Recombinantes de Fusión/fisiología , Transfección , Células Tumorales Cultivadas/química , Células Tumorales Cultivadas/fisiología
11.
Nat Neurosci ; 2(11): 984-8, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10526337

RESUMEN

The genetic lesion underlying familial British dementia (FBD), an autosomal dominant neurodegenerative disorder, is a T-A transversion at the termination codon of the BRI gene. The mutant gene encodes BRI-L, the precursor of ABri peptides that accumulate in amyloid deposits in FBD brain. We now report that both BRI-L and its wild-type counterpart, BRI, were constitutively processed by the proprotein convertase, furin, resulting in the secretion of carboxyl-terminal peptides that encompass all or part of ABri. Elevated levels of peptides were generated from the mutant BRI precursor. Electron microscopic studies revealed that synthetic ABri peptides assembled into irregular, short fibrils. Collectively, our results support the view that enhanced furin-mediated processing of mutant BRI generates fibrillogenic peptides that initiate the pathogenesis of FBD.


Asunto(s)
Amiloide/biosíntesis , Demencia/genética , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Neurofibrillas/metabolismo , Fragmentos de Péptidos/biosíntesis , Subtilisinas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Furina , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Glicoproteínas de Membrana , Proteínas de la Membrana , Ratones , Microscopía Electrónica , Células Tumorales Cultivadas , Reino Unido
12.
Mol Neurobiol ; 55(7): 5757-5766, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29052144

RESUMEN

Amyloid precursor protein (APP) is cleaved by a set of proteases including α-/ß-/γ- and recently identified η-secretases, generating C-terminal fragments (CTFs) of varying lengths and amyloid ß (Aß) peptides, which are considered to play a pivotal role in Alzheimer's disease (AD) pathogenesis. Cellular cholesterol content/distribution can regulate the production/clearance of APP metabolites and hence modify AD pathology. To determine the functional relation between endosomal-lysosomal (EL) cholesterol sequestration and APP metabolism, we used our recently developed mouse N2a-ANPC cells that overexpress Swedish mutant human APP in the absence of cholesterol-trafficking Niemann-Pick type C1 (Npc1) protein. Here, we report that neither increased levels nor EL cholesterol sequestration altered APP holoprotein levels but caused the intracellular accumulation of APP α-/ß-/η-CTFs and Aß1-40/42 peptides. The levels of APP-cleaved products increased as a function of extracellular serum concentration in N2a-ANPC cells, which are more vulnerable to death than the control cells. Additionally, we show that pH of the lysosomal vesicles in N2a-ANPC cells shifted to a less acidic range with increasing serum concentrations, thus making them less efficient functionally. Interestingly, the addition of cholesterol to the culture media not only increased the levels of cellular cholesterol and APP-cleaved products but also rendered the cells more vulnerable to toxicity. Collectively, our results suggest that extracellular cholesterol concentration in serum under conditions of Npc1 deficiency can influence intracellular cholesterol content/distribution and lysosomal efficacy, triggering the accumulation of toxic APP-cleaved products, eventually leading to cell death.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Espacio Extracelular/metabolismo , Proteína Niemann-Pick C1/deficiencia , Suero/metabolismo , Animales , Línea Celular , Supervivencia Celular , Colesterol/metabolismo , Ratones , Proteína Niemann-Pick C1/metabolismo
13.
Mol Cell Biol ; 38(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29530923

RESUMEN

Amyloid ß (Aß) peptide, derived from amyloid precursor protein (APP), plays a critical role in the development of Alzheimer's disease. Current evidence indicates that altered levels or subcellular distribution of cholesterol can regulate Aß production and clearance, but it remains unclear how cholesterol sequestration within the endosomal-lysosomal (EL) system can influence APP metabolism. Thus, we evaluated the effects of U18666A, which triggers cholesterol redistribution within the EL system, on mouse N2a cells expressing different levels of APP in the presence or absence of extracellular cholesterol and lipids provided by fetal bovine serum (FBS). Our results reveal that U18666A and FBS differentially increase the levels of APP and its cleaved products, the α-, ß-, and η-C-terminal fragments, in N2a cells expressing normal levels of mouse APP (N2awt), higher levels of human wild-type APP (APPwt), or "Swedish" mutant APP (APPsw). The cellular levels of Aß1-40/Aß1-42 were markedly increased in U18666A-treated APPwt and APPsw cells. Our studies further demonstrate that APP and its cleaved products are partly accumulated in the lysosomes, possibly due to decreased clearance. Finally, we show that autophagy inhibition plays a role in mediating U18666A effects. Collectively, these results suggest that altered levels and distribution of cholesterol and lipids can differentially regulate APP metabolism depending on the nature of APP expression.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Androstenos/farmacología , Colesterol/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Enfermedad de Alzheimer/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Células Cultivadas , Endosomas/efectos de los fármacos , Humanos , Lisosomas/efectos de los fármacos
14.
Curr Biol ; 9(24): 1493-6, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10607593

RESUMEN

Studies in invertebrates have indicated a functional requirement for presenilin (PS) genes in the Notch pathway [1-5]. One model of Notch signal transduction suggests that proteolysis releases an activated Notch fragment that migrates to the nucleus and regulates gene transcription in concert with CBF1/Su(H)/lag1 (CSL) proteins [6-9]. Recent studies suggest that PS genes control the proteolysis and nuclear access of the Notch intracellular domain [3,4,10,11], offering a basis for the functional interaction of PS and Notch genes [12]. Here, we report that Notch1 signaling elicited by the ligand Delta1 was quantitatively unchanged in PS1-deficient primary embryonic fibroblasts (PEFs). Notch1 signals were measured by both the activation of the hairy/enhancer of split (HES1) promoter and by the antagonism of MyoD-induced muscle creatine kinase (MCK) promoter activity. A membrane-tethered ligand-independent Notch1 construct also showed full efficacy in both assays, despite its presumed requirement for cleavage. Although signaling through Notch1 persisted in PS1-deficient cells, we found a marked reduction in the appearance of a complex of a cleaved, intracellular Notch fragment (NICD) and a CSL protein, as previously reported [6] [10]. These studies reveal that PS1 is not required for ligand-dependent Notch signaling, and that PS1 and PS2 may be redundant. Our data also suggest that the identified NICD fragment may not be necessary for Notch signal transduction [9].


Asunto(s)
Proteínas de la Membrana/fisiología , Receptores de Superficie Celular , Factores de Transcripción , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica , Proteínas de Homeodominio/fisiología , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteína MioD/fisiología , Presenilina-1 , Presenilina-2 , Regiones Promotoras Genéticas , Receptor Notch1 , Transducción de Señal , Factor de Transcripción HES-1
15.
Mol Neurobiol ; 54(4): 2636-2658, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26993302

RESUMEN

The insulin-like growth factor II/mannose 6-phosphate (IGF-II/M6P) receptor is a multifunctional single transmembrane glycoprotein. Recent studies have advanced our understanding of the structure, ligand-binding properties, and trafficking of the IGF-II/M6P receptor. This receptor has been implicated in a variety of important cellular processes including growth and development, clearance of IGF-II, proteolytic activation of enzymes, and growth factor precursors, in addition to its well-known role in the delivery of lysosomal enzymes. The IGF-II/M6P receptor, distributed widely in the central nervous system, has additional roles in mediating neurotransmitter release and memory enhancement/consolidation, possibly through activating IGF-II-related intracellular signaling pathways. Recent studies suggest that overexpression of the IGF-II/M6P receptor may have an important role in regulating the levels of transcripts and proteins involved in the development of Alzheimer's disease (AD)-the prevalent cause of dementia affecting the elderly population in our society. It is reported that IGF-II/M6P receptor overexpression can increase the levels/processing of amyloid precursor protein leading to the generation of ß-amyloid peptide, which is associated with degeneration of neurons and subsequent development of AD pathology. Given the significance of the receptor in mediating the transport and functioning of the lysosomal enzymes, it is being considered for therapeutic delivery of enzymes to the lysosomes to treat lysosomal storage disorders. Notwithstanding these results, additional studies are required to validate and fully characterize the function of the IGF-II/M6P receptor in the normal brain and its involvement in various neurodegenerative disorders including AD. It is also critical to understand the interaction between the IGF-II/M6P receptor and lysosomal enzymes in neurodegenerative processes, which may shed some light on developing approaches to detect and prevent neurodegeneration through the dysfunction of the receptor and the endosomal-lysosomal system.


Asunto(s)
Cationes/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Receptor IGF Tipo 2/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Humanos , Lisosomas/metabolismo , Receptor IGF Tipo 2/química
16.
Mol Cell Biol ; 35(14): 2368-84, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25939386

RESUMEN

Amyloid ß (Aß) peptides originating from amyloid precursor protein (APP) in the endosomal-lysosomal compartments play a critical role in the development of Alzheimer's disease (AD), the most common type of senile dementia affecting the elderly. Since insulin-like growth factor II (IGF-II) receptors facilitate the delivery of nascent lysosomal enzymes from the trans-Golgi network to endosomes, we evaluated their role in APP metabolism and cell viability using mouse fibroblast MS cells deficient in the murine IGF-II receptor and corresponding MS9II cells overexpressing the human IGF-II receptors. Our results show that IGF-II receptor overexpression increases the protein levels of APP. This is accompanied by an increase of ß-site APP-cleaving enzyme 1 levels and an increase of ß- and γ-secretase enzyme activities, leading to enhanced Aß production. At the cellular level, IGF-II receptor overexpression causes localization of APP in perinuclear tubular structures, an increase of lipid raft components, and increased lipid raft partitioning of APP. Finally, MS9II cells are more susceptible to staurosporine-induced cytotoxicity, which can be attenuated by ß-secretase inhibitor. Together, these results highlight the potential contribution of IGF-II receptor to AD pathology not only by regulating expression/processing of APP but also by its role in cellular vulnerability.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Receptor IGF Tipo 2/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Endosomas/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Lisosomas/metabolismo , Ratones , Microscopía Fluorescente , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Interferencia de ARN , Receptor IGF Tipo 2/genética , Estaurosporina/farmacología , Transfección
17.
Neurobiol Aging ; 18(6): 661-9, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9461064

RESUMEN

Amyloid precursor protein (APP) is a member of a larger gene family including amyloid precursor-like proteins (APLP), APLP2 and APLP1. To examine the function of APLP2 in vivo, we generated APLP2 knockout (KO) mice. They are of normal size, fertile, and appear healthy up to 22 months of age. We observed no impaired axonal outgrowth of olfactory sensory neurons following bulbectomy, suggesting against an important role for APLP2 alone in this process. Because APLP2 and APP are highly homologous and may serve similar functions in vivo, we generated mice with targeted APLP2 and APP alleles. Approximately 80% of double KO mice die within the first week after birth, suggesting that APLP2 and APP are required for early postnatal development. The surviving approximately 20% of double KO mice are 20-30% reduced in weight and show difficulty in righting, ataxia, spinning behavior, and a head tilt, suggesting a deficit in balance and/or strength. Adult double KO mice mate poorly, despite apparent normal ovarian and testicular development. Otherwise, double KO mice appear healthy up to 13 months of age. We conclude, that APLP2 and APP can substitute for each other functionally.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Conducta Animal/fisiología , Animales , Animales Recién Nacidos , Ataxia/genética , Ataxia/psicología , Blastocisto/fisiología , Encéfalo/patología , Marcación de Gen , Inmunohistoquímica , Esperanza de Vida , Ratones , Ratones Noqueados , Regeneración Nerviosa/fisiología , Bulbo Olfatorio/fisiología , Equilibrio Postural/fisiología , Conducta Sexual Animal/fisiología
18.
FEBS Lett ; 319(3): 271-6, 1993 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-8458421

RESUMEN

Mitogen withdrawal triggers myogenic differentiation in skeletal myoblasts in culture. We have examined the expression of the proto-oncogene c-jun during this process in mouse C2C12 myoblasts. c-jun belongs to a family of immediate early genes whose expression is activated in cultured cells in response to the addition of serum growth factors. Interestingly, expression of c-jun was maintained in mouse C2C12 and rat L6 myoblasts undergoing myogenic differentiation under low-serum conditions. Previously it has been reported that expression of c-jun is downregulated during differentiation of C2 cells. However, our results using C2C12 cells, a subclone of the C2 line, show that c-jun mRNA, protein and the activator-protein 1 (AP-1) DNA-binding activity were easily detected in proliferating myoblasts and differentiated myotubes. Although overexpression of c-jun has been shown to block myogenic differentiation in C2 cells, results presented here suggest that expression of c-jun at physiological levels may not interfere with skeletal myogenesis.


Asunto(s)
Regulación de la Expresión Génica , Genes jun , Músculos/citología , Proteínas Proto-Oncogénicas c-jun/genética , Animales , Secuencia de Bases , Western Blotting , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/metabolismo , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/genética , Oligodesoxirribonucleótidos/química , ARN Mensajero/genética , Ratas
19.
Invest Ophthalmol Vis Sci ; 39(2): 292-300, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9477985

RESUMEN

PURPOSE: The authors previously showed that the mRNA encoding the amyloid precursor-like protein 2 (APLP2) was one of several genes with upregulated expression in healing corneal epithelium. To elucidate the physiological role of APLP2 in corneal epithelial cells, the expression, distribution, and posttranslational modification of APLP2 were investigated. METHODS: Alternative splicing was assessed by reverse transcription-polymerase chain reaction; translational expression and posttranslational modification were assessed by chondroitinase digestion and Western blotting. The differential distribution of APLP2 in corneal epithelium was examined by in situ hybridization and immunohistochemical analyses. RESULTS: In rat corneal epithelium, the majority of APLP2 mRNA generated from two alternative splicing sites was found to contain the exon encoding a Kunitz protease inhibitor domain in the first site but to lack the exon encoding a 12-amino acid insert in the second site. The absence of the 12-amino acid insert indicated that APLP2 could be modified by the addition of a chondroitin sulfate (CS) glycosaminoglycan chain. The CS proteoglycan nature of APLP2 was verified by chondroitinase digestion. After wounding, APLP2 mRNA and polypeptides were increased markedly in the basal epithelial cells that were actively migrating. Furthermore, APLP2 was observed in the denuded wound bed immediately adjacent to the leading edge of migratory cells and under the epithelial sheet after wound closure. CONCLUSIONS: The wound-induced, basal-cell-specific APLP2 expression correlates with epithelial cell migration. The spatial and temporal expression of Kunitz protease inhibitor-containing, CS-modified APLP2 in healing corneal epithelium is consistent with its hypothesized role(s) in mediating reorganization of the extracellular matrix and dynamic cell-matrix adhesion during reepithelialization.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Epitelio Corneal/metabolismo , Proteínas del Tejido Nervioso/fisiología , Cicatrización de Heridas , Empalme Alternativo , Precursor de Proteína beta-Amiloide/genética , Animales , Aprotinina/genética , Western Blotting , Movimiento Celular/fisiología , Cartilla de ADN/química , Electroforesis en Gel de Agar , Técnica del Anticuerpo Fluorescente Indirecta , Hibridación in Situ , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Ratas , Cicatrización de Heridas/fisiología
20.
J Mol Neurosci ; 17(2): 183-92, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11816791

RESUMEN

Understanding mechanisms involved in the production of Abeta has long been the central focus of cell biologists engaged in molecular AD research. The discovery of two genes that encode homologous polytopic membrane proteins termed Presenilins (PS), has lead to several exciting recent findings on the proteolytic processes responsible for generating the COOH-terminus of Abeta. What we now know is that PS proteins play an important role in Abeta production and are considered one of the therapeutic targets. Here I have reviewed the vast literature on the biology of PS, especially focusing on PS endoproteolysis and the accumulation of stable PS derivatives that are likely the functional units.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Caspasas/metabolismo , Dominio Catalítico/genética , Humanos , Proteínas de la Membrana/genética , Péptido Hidrolasas/genética , Presenilina-1 , Presenilina-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA