Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
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.
Neuroscience ; 379: 177-188, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29572166

RESUMEN

Human amyloid ß1-42 (hAß1-42) peptides are known to self-aggregate into oligomers that contribute to the degeneration of neurons and development of Alzheimer's disease (AD) pathology. Unlike humans, rodents do not develop AD, possibly due to differences in three amino acids (R5G, Y10F and H13R) within the hydrophilic N-terminal domain of Aß1-42. This is partly supported by evidence that hAß1-42 is more prone to fibrillization and has a higher cellular toxicity than rodent Aß1-42 (rAß1-42). Mutagenesis studies, however, have shown that correlation between fibrillization potential and toxicity is not always direct. Thus, to understand better how N-terminal mutations can affect hAß1-42 toxicity through oligomerization, we evaluated fibrillization kinetics, oligomer sizes and toxicity profiles of double mutant (human toward rodent) Aß1-42. Additionally, we tested the mutant peptides in combination with hAß1-42, to assess effects on hAß1-42 aggregation/toxicity. Our results clearly show that double mutations to humanize rAß1-42 result in a significantly reduced efficiency of fibril formation, as determined by Thioflavin-T aggregation assays and confirmed with electron micrographic studies. Interestingly, the mutants are still able to aggregate into oligomers, which are predominantly larger than those comprised of hAß1-42. Our cell viability experiments further showed a rank order of oligomer toxicity of hAß1-42 > rAß1-42 ≫ mutant Aß1-42, suggesting that toxicity can be influenced by N-terminal Aß1-42 mutations via reduction of fibril formation and/or alteration of oligomer size. These results, taken together, confirm that N-terminal mutations can affect Aß fibril and oligomer formation with reduced toxicity despite lying outside the core amyloid region of Aß peptide.


Asunto(s)
Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Humanos , Cinética , Estructura Molecular , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/toxicidad , Ratas Sprague-Dawley
3.
Mol Neurobiol ; 54(1): 748-754, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26768593

RESUMEN

Accumulation of amyloid-ß peptide (Aß) is a pathological hallmark of Alzheimer's disease (AD). We have previously demonstrated that electrophysiological and neurotoxic effects of Aß and human amylin are expressed via the amylin receptor. Recently, pramlintide, a synthetic analog of amylin, has been reported to improve cognitive function in transgenic AD mouse models. In this study, we examined the effects of pramlintide on Aß1-42 and human amylin-evoked depression of long-term potentiation (LTP) at Schaeffer collateral-CA1 hippocampal synapses. In mouse hippocampal brain slices, field excitatory postsynaptic potentials (fEPSPs) were recorded from the stratum radiatum layer of the CA1 area in response to electrical stimulation of Schaeffer collateral afferents and LTP induced by 3-theta-burst stimulation (TBS) protocol. Aß1-42 (50 nM) and human amylin (50 nM), but not Aß42-1 (50 nM), depressed LTP. Pre-application of pramlintide (250 nM) blocked Aß- and human amylin-induced reduction of LTP without affecting baseline transmission or LTP. We also examined the effects of pramlintide on LTP in transgenic mice (TgCRND8) that over-express amyloid precursor protein. In contrast to wild-type controls, where robust LTP was observed, 10- to 12-month-old TgCRND8 mice show blunted LTP. In TgCRND8 mice, basal LTP is enhanced by application of pramlintide. Our data indicate that pramlintide acts as a functional amylin receptor antagonist to reverse the effects of Aß1-42 and human amylin on LTP and also increases LTP in transgenic mice that demonstrate increased ambient brain amyloid levels. Amylin receptor antagonists may thus serve as potentially useful therapeutic agents in treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Potenciación a Largo Plazo/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Secuencia de Aminoácidos , Péptidos beta-Amiloides/toxicidad , Animales , Femenino , Hipocampo/fisiología , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Fragmentos de Péptidos/toxicidad
4.
Mol Psychiatry ; 18(8): 889-97, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22850627

RESUMEN

Latrepirdine (Dimebon) is a pro-neurogenic, antihistaminic compound that has yielded mixed results in clinical trials of mild to moderate Alzheimer's disease, with a dramatically positive outcome in a Russian clinical trial that was unconfirmed in a replication trial in the United States. We sought to determine whether latrepirdine (LAT)-stimulated amyloid precursor protein (APP) catabolism is at least partially attributable to regulation of macroautophagy, a highly conserved protein catabolism pathway that is known to be impaired in brains of patients with Alzheimer's disease (AD). We utilized several mammalian cellular models to determine whether LAT regulates mammalian target of rapamycin (mTOR) and Atg5-dependent autophagy. Male TgCRND8 mice were chronically administered LAT prior to behavior analysis in the cued and contextual fear conditioning paradigm, as well as immunohistological and biochemical analysis of AD-related neuropathology. Treatment of cultured mammalian cells with LAT led to enhanced mTOR- and Atg5-dependent autophagy. Latrepirdine treatment of TgCRND8 transgenic mice was associated with improved learning behavior and with a reduction in accumulation of Aß42 and α-synuclein. We conclude that LAT possesses pro-autophagic properties in addition to the previously reported pro-neurogenic properties, both of which are potentially relevant to the treatment and/or prevention of neurodegenerative diseases. We suggest that elucidation of the molecular mechanism(s) underlying LAT effects on neurogenesis, autophagy and behavior might warranty the further study of LAT as a potentially viable lead compound that might yield more consistent clinical benefit following the optimization of its pro-neurogenic, pro-autophagic and/or pro-cognitive activities.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Autofagia/efectos de los fármacos , Cognición/efectos de los fármacos , Indoles/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Indoles/uso terapéutico , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , alfa-Sinucleína/metabolismo
5.
Mol Neurobiol ; 47(1): 37-63, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22983915

RESUMEN

Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder believed to be initiated by accumulation of amyloid ß (Aß)-related peptides derived from proteolytic processing of amyloid precursor protein (APP). Research over the past two decades provided a mechanistic link between cholesterol and AD pathogenesis. Genetic polymorphisms in genes regulating the pivotal points in cholesterol metabolism have been suggested to enhance the risk of developing AD. Altered neuronal membrane cholesterol level and/or subcellular distribution have been implicated in aberrant formation, aggregation, toxicity, and degradation of Aß-related peptides. However, the results are somewhat contradictory and we still do not have a complete understanding on how cholesterol can influence AD pathogenesis. In this review, we summarize our current understanding on the role of cholesterol in regulating the production/function of Aß-related peptides and also examine the therapeutic potential of regulating cholesterol homeostasis in the treatment of AD pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Animales , Encéfalo/metabolismo , Encéfalo/patología , Homeostasis , Humanos , Datos de Secuencia Molecular
6.
Neuroscience ; 169(2): 781-6, 2010 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-20493928

RESUMEN

Estrogen (17beta-estradiol) plays key regulatory roles in a variety of physiological and biological processes. Several lines of evidence also support its role as a protective factor in Alzheimer's disease; however, the basis of this effect is unclear. Here we show that an early-onset Alzheimer's disease transgenic mouse model expressing the double-mutant form of human amyloid precursor protein (APP); Swedish (K670N/M671L) and Indiana (V717F) undergoing treatment with 17beta-estradiol show significantly lower levels of APP processing through beta-secretase and enhanced alpha-secretase processing resulting in marked reductions of APP-CTFbeta, Abeta42 and plaque burden, along with increased levels of the non-amyloidogenic sAPPalpha. Moreover, 17beta-estradiol resulted in elevated brain levels of transthyretin, which inhibits aggregation of Abeta into plaques; though the insulin-degrading enzyme, which breaks down Abeta, was significantly reduced. These results illustrate a multifaceted effect of 17beta-estradiol on the biochemical basis of Alzheimer's disease, through effects on APP processing, Abeta levels and factors that affect its clearance and aggregation. Overall, these results support the need for further long-term longitudinal studies to elucidate consequences of menopause as well as hormone therapy on Alzheimer's disease, and explore its potential as a therapeutic avenue for the disease.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Estradiol/farmacología , Estrógenos/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Estradiol/sangre , Estradiol/uso terapéutico , Estrógenos/sangre , Estrógenos/uso terapéutico , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Fármacos Neuroprotectores/uso terapéutico , Placa Amiloide/efectos de los fármacos , Placa Amiloide/patología
7.
Neurobiol Aging ; 30(1): 54-70, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17561313

RESUMEN

The insulin-like growth factor-II/mannose-6-phosphate (IGF-II/M6P) receptor participates in the trafficking of lysosomal enzymes from the trans-Golgi network or the cell surface to lysosomes. In Alzheimer's disease (AD) brains, marked up-regulation of the lysosomal system in vulnerable neuronal populations has been correlated with altered metabolic functions. To establish whether IGF-II/M6P receptors and lysosomal enzymes are altered in the brain of transgenic mice harboring different familial AD mutations, we measured the levels and distribution of the receptor and lysosomal enzymes cathepsins B and D in select brain regions of transgenic mice overexpressing either mutant presenilin 1 (PS1; PS1(M146L+L286V)), amyloid precursor protein (APP; APP(KM670/671NL+V717F)) or APP+PS1 (APP(KM670/671NL+V717F)+PS1(M146L+L286V)) transgenes. Our results revealed that levels and expression of the IGF-II/M6P receptor and lysosomal enzymes are increased in the hippocampus and frontal cortex of APP and APP+PS1, but not in PS1, transgenic mouse brains compared with wild-type controls. The changes were more prominent in APP+PS1 than in APP single transgenic mice. Additionally, all beta-amyloid-containing neuritic plaques in the hippocampal and cortical regions of APP and APP+PS1 transgenic mice were immunopositive for both lysosomal enzymes, whereas only a subset of the plaques displayed IGF-II/M6P receptor immunoreactivity. These results suggest that up-regulation of the IGF-II/M6P receptor and lysosomal enzymes in neurons located in vulnerable regions reflects an altered functioning of the endosomal-lysosomal system which may be associated with the increased intracellular and/or extracellular A beta deposits observed in APP and APP+PS1 transgenic mouse brains.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Encéfalo/metabolismo , Catepsina B/metabolismo , Catepsina D/metabolismo , Lisosomas/enzimología , Placa Amiloide/patología , Presenilina-1/genética , Receptor IGF Tipo 2/metabolismo , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distribución Tisular , Regulación hacia Arriba
8.
Nat Med ; 8(11): 1263-9, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12379850

RESUMEN

Immunization of transgenic mouse models of Alzheimer disease using amyloid-beta peptide (Abeta) reduces both the Alzheimer disease-like neuropathology and the spatial memory impairments of these mice. However, a therapeutic trial of immunization with Abeta42 in humans was discontinued because a few patients developed significant meningo-encephalitic cellular inflammatory reactions. Here we show that beneficial effects in mice arise from antibodies selectively directed against residues 4-10 of Abeta42, and that these antibodies inhibit both Abeta fibrillogenesis and cytotoxicity without eliciting an inflammatory response. These findings provide the basis for improved immunization antigens as well as attempts to design small-molecule mimics as alternative therapies.


Asunto(s)
Vacunas contra el Alzheimer/uso terapéutico , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos/uso terapéutico , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular
9.
Exp Neurol ; 175(2): 398-406, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12061869

RESUMEN

Most autosomal dominant forms of Alzheimer disease (AD) are related to missense mutations in the human presenilin (PS) 1 gene. Although the underlying mechanisms associated with pathophysiology of AD have yet to be clearly established, pathogenic mutations in the PS1 gene influence the processing of beta-amyloid precursor protein, leading to increased production and deposition of highly fibrillogenic amyloid beta(1-42) peptide in the brains of AD patients. As cognitive dysfunction in AD is associated with a dramatic loss of cholinergic innervation particularly in the hippocampus and neocortex, we investigated learning and cholinergic neurochemistry in transgenic mice expressing pathogenic mutant L286V or wild-type(wt) human PS1 transgenes. Relative to wt, the L286V PS1 transgenic mice exhibited reduced sensorimotor activity and marked deterioration of object memory between 3 and 5 h after the first encounter. Activity of the biosynthetic enzyme choline acetyltransferase was not altered in the hippocampus, frontoparietal cortex, or striatum of mutant transgenic mice relative to wt transgenic or littermate nontransgenic controls. No differences in the densities of M1/[3H]pirenzepine, M2/[3H]AF-DX 384, or alpha(7) nicotinic/125I-alpha-bungarotoxin receptor binding sites were evident in any brain regions among L286V PS1 transgenic, wt PS1 transgenic, and littermate nontransgenic controls. These results suggest that overexpression of a mutated PS1 gene induces a subtle alteration in object memory without affecting cholinergic neurochemistry.


Asunto(s)
Acetilcolina/metabolismo , Enfermedad de Alzheimer/fisiopatología , Proteínas de la Membrana/genética , Reconocimiento en Psicología/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Colina O-Acetiltransferasa/metabolismo , Expresión Génica , Aprendizaje por Laberinto/fisiología , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Equilibrio Postural/fisiología , Presenilina-1 , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Transgenes
10.
Neurobiol Aging ; 23(2): 187-94, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11804702

RESUMEN

Presenilin 1-null mice die at birth from brain and skeletal developmental deformities due to disrupted Notch signaling. Presenilin 1-null mice also have severely reduced gamma-secretase cleavage of betaAPP. The assumption has been that facilitation of Notch signaling and betaAPP processing by presenilin 1 are analogous functions. Here we describe a presenilin 1-targetted mouse model that expresses extremely low levels ( approximately 1% of normal) of mutant PS1-M146L. Homozygous mice have significantly reduced viability due to a Notch-like phenotype. The animals that survive have severe axial skeletal deformities and markedly diminished gamma-secretase activity and accumulation of betaAPP-C100, but no obvious abnormalities in brain development. These results suggest that, in mice, a marked reduction of PS1-facilitated gamma-secretase activity is not detrimental to normal brain development.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/crecimiento & desarrollo , Endopeptidasas/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/genética , Animales , Especificidad de Anticuerpos , Ácido Aspártico Endopeptidasas , Western Blotting , Encéfalo/anatomía & histología , Encéfalo/patología , Ensayo de Inmunoadsorción Enzimática , Marcación de Gen , Genotipo , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Fenotipo , Presenilina-1 , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Médula Espinal/patología
11.
Proc Natl Acad Sci U S A ; 98(26): 15288-93, 2001 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-11734625

RESUMEN

Doppel (Dpl) is a paralog of the mammalian prion protein (PrP); it is abundant in testes but expressed at low levels in the adult central nervous system. In two Prnp-deficient (Prnp(0/0)) mouse lines (Ngsk and Rcm0), Dpl overexpression correlated with ataxia and death of cerebellar neurons. To determine whether Dpl overexpression, rather than the dysregulation of genes neighboring the Prn gene complex, was responsible for the ataxic syndrome, we placed the mouse Dpl coding sequence under the control of the Prnp promoter and produced transgenic (Tg) mice on the Prnp(0/0)-ZrchI background (hereafter referred to as ZrchI). ZrchI mice exhibit neither Dpl overexpression nor cerebellar degeneration. In contrast, Tg(Dpl)ZrchI mice showed cerebellar granule and Purkinje cell loss; the age of onset of ataxia was inversely proportional to the levels of Dpl protein. Crosses of Tg mice overexpressing wild-type PrP with two lines of Tg(Dpl)ZrchI mice resulted in a phenotypic rescue of the ataxic syndrome, while Dpl overexpression was unchanged. Restoration of PrP expression also rendered the Tg(Dpl) mice susceptible to prion infection, with incubation times indistinguishable from non-Tg controls. Whereas the rescue of Dpl-induced neurotoxicity by coexpression of PrP argues for an interaction between the PrP and Dpl proteins in vivo, the unaltered incubation times in Tg mice overexpressing Dpl in the central nervous system suggest that Dpl is unlikely to be involved in prion formation.


Asunto(s)
Cerebelo/patología , Priones/fisiología , Animales , Ataxia/genética , Cerebelo/anatomía & histología , Proteínas Ligadas a GPI , Ratones , Ratones Transgénicos , Fenotipo , Priones/genética , Regiones Promotoras Genéticas
12.
Biochem Cell Biol ; 79(5): 613-28, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11716303

RESUMEN

The prion protein gene Prnp encodes PrPSc, the major structural component of prions, infectious pathogens causing a number of disorders including scrapie and bovine spongiform encephalopathy (BSE). Missense mutations in the human Prnp gene, PRNP, cause inherited prion diseases such as familial Creutzfeldt-Jakob Disease. In uninfected animals, Prnp encodes a GPI-anchored protein denoted PrPC, and in prion infections, PrPC is converted to PrPSc by templated refolding. Although Prnp is conserved in mammalian species, attempts to verify interactions of putative PrP-binding proteins by genetic means have proven frustrating in that two independent lines of Prnp gene ablated mice (Prnp0/0 mice: ZrchI and Npu) lacking PrPC remain healthy throughout development. This indicates that PrPC serves a function that is not apparent in a laboratory setting or that other molecules have overlapping functions. Shuttling or sequestration of synaptic Cu(II) via binding to N-terminal octapeptide residues and (or) signal transduction involving the fyn kinase are possibilities currently under consideration. A new point of entry into the issue of prion protein function has emerged from identification of a paralog, Prnd, with 25% coding sequence identity to Prnp. Prnd lies downstream of Prnp and encodes the Dpl protein. Like PrPC, Dpl is presented on the cell surface via a GPI anchor and has three alpha-helices: however, it lacks the conformationally plastic and octapeptide repeat domains present in its well-known relative. Interestingly, Dpl is overexpressed in two other lines of Prnp0/0 mice (Ngsk and Rcm0) via intergenic splicing events. These lines of Prnp0/0 mice exhibit ataxia and apoptosis of cerebellar cells, indicating that ectopic synthesis of Dpl protein is toxic to CNS neurons: this inference has now been confirmed by the construction of transgenic mice expressing Dpl under the direct control of the PrP promoter. Remarkably, Dpl-programmed ataxia is rescued by wt Prnp transgenes. The interaction between the Prnp and Prnd genes in mouse cerebellar neurons may have a physical correlate in competition between Dpl and PrPC within a common biochemical pathway that, when misregulated, leads to apoptosis.


Asunto(s)
Enfermedades por Prión/genética , Priones/genética , Animales , Muerte Celular , Síndrome de Creutzfeldt-Jakob/genética , Humanos , Ratones , Modelos Genéticos , Modelos Moleculares , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Polimorfismo Genético , Enfermedades por Prión/metabolismo , Priones/metabolismo , Unión Proteica , Scrapie/genética
13.
Amyloid ; 8 Suppl 1: 10-9, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11676284

RESUMEN

Amyloid plaques are the principal features of Alzheimers disease (AD) pathology and are considered to be a major factor in the disease process. These fibrillar deposits are composed primarily of the 40-42 residue amyloid-beta (Abeta) peptide which is a proteolytic product of a larger membrane precursor protein. Electron microscopy and X-ray diffraction have revealed that the mature amyloid fibrils are assembled as a highly beta-sheet polymer that has a well-defined protofilament quaternary structure. This organization is observed for amyloid fibrils from a wide variety of disorders and appears to represent a structural superfamily. Amyloid plaques also contain a number of other components such as proteoglycans that contain highly sulfated glycosaminoglycan (GAG) chains. These amyloid-associated elements may contribute to the aggregation and/or stabilization of Abeta as insoluble fibrils. We have recently developed an aggressive model for Abeta plaque formation in transgenic mice that exhibits an "early-onset" phenotype. Immunocytochemistry has demonstrated that even with this rapid progression, Abeta deposits within the neuropil and cerebrovascular system all co-localize with heparan sulfate proteoglycans (HSPG). These findings indicate a number of structural features that can be targeted as potential sites for the development of amyloid inhibitors. In addition, the use of small compounds that interfere with the proteoglycan-amyloid pathway may be effective therapeutic agents that can be assessed through the use of these transgenic models.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Sustancias Macromoleculares , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fuerza Atómica , Estructura Molecular , Proteoglicanos/metabolismo , Difracción de Rayos X
14.
Gene ; 275(1): 1-18, 2001 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-11574147

RESUMEN

The prion protein gene, Prnp, encodes PrP(Sc), the major structural component of prions, infectious pathogens causing a number of disorders including scrapie and bovine spongiform encephalopathy (or BSE). Missense mutations in the human Prnp gene cause inherited prion diseases such as familial Creutzfeldt-Jakob disease. In uninfected animals Prnp encodes a glycophosphatidylinositol (GPI)-anchored protein denoted PrP(C) and in prion infections PrP(C) is converted to PrP(Sc) by templated refolding. Though Prnp is conserved in mammalian species, attempts to verify interactions of putative PrP binding proteins by genetic means have proven frustrating and the ZrchI and Npu lines of Prnp gene-ablated mice (Prnp(0/0) mice) lacking PrP(C) remain healthy throughout development. This indicates that PrP(C) serves a function that is not apparent in a laboratory setting or that other molecules have overlapping functions. Current possibilities involve shuttling or sequestration of synaptic Cu(II) via binding to N-terminal octapeptide residues and/or signal transduction involving the fyn kinase. A new point of entry into the issue of prion protein function has emerged from identification of a paralogue, Prnd, with 24% coding sequence identity to Prnp. Prnd lies downstream of Prnp and encodes the doppel (Dpl) protein. Like PrP(C), Dpl is presented on the cell surface via a GPI anchor and has three alpha-helices: however, it lacks the conformationally plastic and octapeptide repeat domains present in its well-known relative. Interestingly, Dpl is overexpressed in the Ngsk and Rcm0 lines of Prnp(0/0) mice via intergenic splicing events. These lines of Prnp(0/0) mice exhibit ataxia and apoptosis of cerebellar cells, indicating that ectopic synthesis of Dpl protein is toxic to central nervous system neurons: this inference has now been confirmed by the construction of transgenic mice expressing Dpl under the direct control of the PrP promoter. Remarkably, Dpl-programmed ataxia is rescued by wild-type Prnp transgenes. The interaction between the Prnp and Prnd genes in mouse cerebellar neurons may have a physical correlate in competition between Dpl and PrP(C) within a common biochemical pathway that when mis-regulated leads to apoptosis.


Asunto(s)
Proteínas PrPC/genética , Priones/genética , Animales , Secuencia de Bases , Proteínas Ligadas a GPI , Regulación de la Expresión Génica , Genes/genética , Humanos , Modelos Biológicos , Mutación , Enfermedades por Prión/genética
15.
Physiol Behav ; 73(5): 873-86, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11566220

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by a progressive loss of cognitive function. Despite considerable progress, a complete description of the molecular pathology of this disease has yet to be elucidated. In this respect, the need for an animal model that develops some or all aspects of this uniquely human disease in a reproducible fashion is crucial for the development and testing of potential treatments. A valid animal model for AD should exhibit (1) progressive AD-like neuropathology and (2) cognitive deficits, and (3) should be verified in several laboratories. Transgenic models should be able to (4) discern pathogenic effects of familial forms (FAD) mutations from those of transgene overexpression. Models derived from microinjection of FAD mutant alleles should (5) encompass more than one Tg line. At present, however, no model that replicates all of these desirable features exists. In this review, we discuss transgenic mouse models with well-characterized AD-like neuropathology that show some form of cognitive impairment. We argue that conclusions drawn from a limited selection of cross-sectional experiments should be verified in longitudinally designed experiments. Future studies should attempt to establish a closer relationship between molecular pathology and the degree of cognitive impairment. While exact replication of AD in mice may not attainable (due to phylogenetic differences and fundamental differences in behavioral ecology), rigorous comparative analysis of cognitive behavior observed in various mouse models of AD should provide a framework for better understanding of molecular mechanisms underlying cognitive impairment observed in AD patients.


Asunto(s)
Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Ratones Transgénicos/genética , Anciano , Péptidos beta-Amiloides/genética , Animales , Conducta Animal/fisiología , Encéfalo/patología , Genética Conductual , Humanos , Ratones
16.
Biochem Soc Symp ; (67): 89-100, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11447843

RESUMEN

Missense mutations in presenilin 1 (PS1) and presenilin 2 (PS2) are associated with early-onset familial Alzheimer's disease which displays an accelerated deposition of amyloid plaques and neurofibrillary tangles. Presenilins are multi-spanning transmembrane proteins which localize primarily to the endoplasmic reticulum and the Golgi compartments. We have previously demonstrated that PS1 exists as a high-molecular-mass complex that is likely to contain several functional ligands. Potential binding proteins were screened by the yeast two-hybrid system using the cytoplasmically orientated PS1 loop domain which was shown to interact strongly with members of the armadillo family of proteins, including beta-catenin, p0071 and a novel neuron-specific plakophilin-related armadillo protein (NPRAP). Armadillo proteins can have dual functions that encompass the stabilization of cellular junctions/synapses and the mediation of signal transduction pathways. Our observations suggest that PS1 may contribute to both aspects of armadillo-related pathways involving neurite outgrowth and nuclear translocation of beta-catenin upon activation of the wingless (Wnt) pathway. Alzheimer's disease (AD)-related presenilin mutations exhibit a dominant gain of aberrant function resulting in the prevention of beta-catenin translocation following Wnt signalling. These findings indicate a functional role for PS1 in signalling and suggest that mistrafficking of selected presenilin ligands may be a potential mechanism in the genesis of AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Proteínas de la Membrana/fisiología , Transactivadores , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Proteínas del Citoesqueleto/fisiología , Humanos , Sustancias Macromoleculares , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Modelos Neurológicos , Estructura Molecular , Mutación Missense , Presenilina-1 , Presenilina-2 , Procesamiento Proteico-Postraduccional , Transducción de Señal , beta Catenina
17.
J Biol Chem ; 276(24): 21562-70, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11279122

RESUMEN

We have created early-onset transgenic (Tg) models by exploiting the synergistic effects of familial Alzheimer's disease mutations on amyloid beta-peptide (Abeta) biogenesis. TgCRND8 mice encode a double mutant form of amyloid precursor protein 695 (KM670/671NL+V717F) under the control of the PrP gene promoter. Thioflavine S-positive Abeta amyloid deposits are present at 3 months, with dense-cored plaques and neuritic pathology evident from 5 months of age. TgCRND8 mice exhibit 3,200-4,600 pmol of Abeta42 per g brain at age 6 months, with an excess of Abeta42 over Abeta40. High level production of the pathogenic Abeta42 form of Abeta peptide was associated with an early impairment in TgCRND8 mice in acquisition and learning reversal in the reference memory version of the Morris water maze, present by 3 months of age. Notably, learning impairment in young mice was offset by immunization against Abeta42 (Janus, C., Pearson, J., McLaurin, J., Mathews, P. M., Jiang, Y., Schmidt, S. D., Chishti, M. A., Horne, P., Heslin, D., French, J., Mount, H. T. J., Nixon, R. A., Mercken, M., Bergeron, C., Fraser, P. E., St. George-Hyslop, P., and Westaway, D. (2000) Nature 408, 979-982). Amyloid deposition in TgCRND8 mice was enhanced by the expression of presenilin 1 transgenes including familial Alzheimer's disease mutations; for mice also expressing a M146L+L286V presenilin 1 transgene, amyloid deposits were apparent by 1 month of age. The Tg mice described here suggest a potential to investigate aspects of Alzheimer's disease pathogenesis, prophylaxis, and therapy within short time frames.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Amiloidosis/genética , Encéfalo/patología , Trastornos del Conocimiento/genética , Envejecimiento , Sustitución de Aminoácidos , Amiloide/análisis , Amiloide/genética , Precursor de Proteína beta-Amiloide/análisis , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/patología , Amiloidosis/psicología , Animales , Encéfalo/crecimiento & desarrollo , Trastornos del Conocimiento/patología , Cruzamientos Genéticos , Femenino , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Mapeo Restrictivo
18.
Proc Natl Acad Sci U S A ; 98(5): 2352-7, 2001 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-11226243

RESUMEN

The downstream prion-like protein (doppel, or Dpl) is a paralog of the cellular prion protein, PrP(C). The two proteins have approximately 25% sequence identity, but seem to have distinct physiologic roles. Unlike PrP(C), Dpl does not support prion replication; instead, overexpression of Dpl in the brain seems to cause a completely different neurodegenerative disease. We report the solution structure of a fragment of recombinant mouse Dpl (residues 26-157) containing a globular domain with three helices and a small amount of beta-structure. Overall, the topology of Dpl is very similar to that of PrP(C). Significant differences include a marked kink in one of the helices in Dpl, and a different orientation of the two short beta-strands. Although the two proteins most likely arose through duplication of a single ancestral gene, the relationship is now so distant that only the structures retain similarity; the functions have diversified along with the sequence.


Asunto(s)
Enfermedades Neurodegenerativas/etiología , Proteínas PrPC/fisiología , Priones/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Cartilla de ADN , Disulfuros/química , Proteínas Ligadas a GPI , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Enfermedades Neurodegenerativas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Proteínas PrPC/química , Priones/química , Priones/genética , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
19.
Curr Neurol Neurosci Rep ; 1(5): 451-7, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11898556

RESUMEN

Alzheimer's disease (AD) is characterized by deterioration in mental function leading to dementia, deposition of amyloid plaques and neurofibrillary tangles (NFTs), and neuronal loss. The major component of plaques is the amyloid-beta peptide (A beta), whereas NFTs are assemblies of hyperphosphorylated forms of the microtubule-associated protein tau. Electron microscopy of NFTs reveals structures known as paired helical filaments (PHFs). In familial AD (FAD), mutations in three distinct genes drive A beta synthesis by favoring endoproteolytic secretase cleavages that liberate A beta from the Alzheimer beta-amyloid precursor protein (APP). This suggests that excess A beta initiates a pathogenic cascade in humans that culminates in all the pathologic and cellular hallmarks of AD. Building upon the knowledge of FAD mutations, incremental technical advances have now allowed reproduceable creation of APP transgenic mice that exhibit AD-like amyloid pathology and A beta burdens. These transgenic mouse lines also exhibit deficits in spatial reference and working memory, with immunization against A beta abrogating both AD-associated phenotypes. Besides establishing a proof of principle for A beta-directed therapies, these findings suggest a potential to identify individual elements in the pathogenic pathway that lead to cognitive dysfunction. Furthermore, transgenic APP mice with potent amyloid deposition will likely form a beach-head to capture the final elements of AD neuropathology--cell loss and NFTs composed of PHFs--that are missing from current transgenic models.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Modelos Animales de Enfermedad , Ratones Transgénicos , Animales , Humanos , Ratones
20.
Proc Natl Acad Sci U S A ; 97(23): 12822-7, 2000 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-11070093

RESUMEN

A direct pathophysiological role of Familial Alzheimer's Disease (FAD)-associated Presenilin 1 (PS1) mutations in neuronal vulnerability remains a controversial matter. We evaluated the relationship between PS1 and excitotoxicity in four different experimental models of neurotoxicity by using primary neurons from (i) transgenic (tg) mice overexpressing a human FAD-linked PS1 variant (L286V mutation), (ii) tg mice overexpressing human wild-type (wt) PS1, (iii) PS1 knockout mice, and (iv) wt mice in which PS1 gene expression was knocked down by antisense treatment. We found that primary neurons overexpressing mutated PS1 showed an increased vulnerability to both excitotoxic and hypoxic-hypoglycemic damage when compared with neurons obtained from either mice overexpressing human wt PS1 or in wt mice. In addition, reduced excitotoxic damage was obtained in neurons in which PS1 expression was absent or diminished. Data obtained in in vivo experimental models of excitotoxicity partially supported the in vitro observations. Accelerated neuronal death was demonstrated in the hippocampus of mice overexpressing mutated PS1 after peripheral administration of kainic acid in comparison with wt animals. However, measurement of the infarct volume after middle cerebral artery occlusion did not show significant difference between the two animal groups. The results altogether suggest that expression of FAD-linked PS1 variants increases the vulnerability of neurons to a specific type of damage in which excitotoxicity plays a relevant role. In addition, they support the view that reduction of endogenous PS1 expression results in neuroprotection.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Proteínas de la Membrana/metabolismo , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Humanos , Infarto de la Arteria Cerebral Media/fisiopatología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa , Neuronas/citología , Neuronas/patología , Neuronas/fisiología , Presenilina-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...