Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(12)2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37371088

RESUMO

The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aß1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-ß oligomers (AßOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AßOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AßOs from ground zero is supported by Aß's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AßOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Córtex Entorrinal/metabolismo , Proteínas Priônicas/metabolismo
2.
Pharm Res ; 39(7): 1497-1507, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35704250

RESUMO

PURPOSE: We have recently demonstrated the brain-delivery of an Amyloid-ß oligomer (Aßo)-binding peptide-therapeutic fused to the BBB-crossing single domain antibody FC5. The bi-functional fusion protein, FC5-mFc-ABP (KG207-M) lowered both CSF and brain Aß levels after systemic dosing in transgenic mouse and rat models of Alzheimer's disease (AD). For development as a human therapeutic, we have humanized and further engineered the fusion protein named KG207-H. The purpose of the present study was to carry out comparative PK/PD studies of KG207-H in wild type rat and beagle dogs (middle-aged and older) to determine comparability of systemic PK and CSF exposure between rodent species and larger animals with more complex brain structure such as dogs. METHOD: Beagle dogs were used in this study as they accumulate cerebral Aß with age, as seen in human AD patients, and can serve as a model of sporadic AD. KG207-H (5 to 50 mg/kg) was administered intravenously and serum and CSF samples were serially collected for PK studies and to assess target engagement. KG207-H and Aß levels were quantified using multiplexed selected reaction monitoring mass spectrometry. RESULTS: After systemic dosing, KG207-H demonstrated similar serum pharmacokinetics in rats and dogs. KG207-H appeared in the CSF in a time- and dose-dependent manner with similar kinetics, indicating CNS exposure. Further analyses revealed a dose-dependent inverse relationship between CSF KG207-H and Aß levels in both species indicating target engagement. CONCLUSION: This study demonstrates translational attributes of BBB-crossing Aß-targeting biotherapeutic KG207-H in eliciting a pharmacodynamic response, from rodents to larger animal species.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Cães , Camundongos , Camundongos Transgênicos , Ratos
3.
J Cereb Blood Flow Metab ; 42(5): 788-801, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34378436

RESUMO

In vivo biomarker abnormalities provide measures to monitor therapeutic interventions targeting amyloid-ß pathology as well as its effects on downstream processes associated with Alzheimer's disease pathophysiology. Here, we applied an in vivo longitudinal study design combined with imaging and cerebrospinal fluid biomarkers, mirroring those used in human clinical trials to assess the efficacy of a novel brain-penetrating anti-amyloid fusion protein treatment in the McGill-R-Thy1-APP transgenic rat model. The bi-functional fusion protein consisted of a blood-brain barrier crossing single domain antibody (FC5) fused to an amyloid-ß oligomer-binding peptide (ABP) via Fc fragment of mouse IgG (FC5-mFc2a-ABP). A five-week treatment with FC5-mFc2a-ABP (loading dose of 30 mg/Kg/iv followed by 15 mg/Kg/week/iv for four weeks) substantially reduced brain amyloid-ß levels as measured by positron emission tomography and increased the cerebrospinal fluid amyloid-ß42/40 ratio. In addition, the 5-week treatment rectified the cerebrospinal fluid neurofilament light chain concentrations, resting-state functional connectivity, and hippocampal atrophy measured using magnetic resonance imaging. Finally, FC5-mFc2a-ABP (referred to as KG207-M) treatment did not induce amyloid-related imaging abnormalities such as microhemorrhage. Together, this study demonstrates the translational values of the designed preclinical studies for the assessment of novel therapies based on the clinical biomarkers providing tangible metrics for designing early-stage clinical trials.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Biomarcadores , Estudos Longitudinais , Camundongos , Tomografia por Emissão de Pósitrons , Ratos
4.
PLoS One ; 16(5): e0250568, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33970919

RESUMO

The development of effective therapies as well as early, molecular diagnosis of Alzheimer's disease is impeded by the lack of understanding of the underlying pathological mechanisms. Metabolomics studies of body fluids as well as brain tissues have shown major changes in metabolic profiles of Alzheimer's patients. However, with analysis performed at the late stages of the disease it is not possible to distinguish causes and consequence. The mouse model APP/PS1 expresses a mutant amyloid precursor protein resulting in early Amyloid ß (Aß) accumulation as well as many resulting physiological changes including changes in metabolic profile and metabolism. Analysis of metabolic profile of cerebrospinal fluid (CSF) and blood of APP/PS1 mouse model can provide information about metabolic changes in these body fluids caused by Aß accumulation. Using our novel method for analysis of correlation and mathematical ranking of significant correlations between metabolites in CSF and blood, we have explored changes in metabolite correlation and connectedness in APP/PS1 and wild type mice. Metabolites concentration and correlation changes in CSF, blood and across the blood brain barrier determined in this work are affected by the production of amyloid plaque. Metabolite changes observed in the APP/PS1 mouse model are the response to the mutation causing plaque formation, not the cause for the plaque suggesting that they are less relevant in the context of early treatment and prevention then the metabolic changes observed only in humans.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Líquido Cefalorraquidiano/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Metaboloma , Presenilina-1/genética , Soro/metabolismo , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Animais , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
5.
Neurochem Int ; 101: 22-29, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27693452

RESUMO

We have previously shown in SH-SY5Y human neuroblastoma cells that the expressions of basal (75 kDa) and high molecular weight (HMW; 85 kDa) isoforms of the p75 neurotrophic receptor (p75NTR) are stimulated by amyloid-ß peptide1-42 oligomers (AßOs) via the insulin-like growth factor-1 receptor (IGF-1R). On the other hand, it is known that AßOs inhibit insulin receptor (IR) signaling. The purpose of the present study was to determine the involvement of IR signaling in the regulation of p75 neurotrophin receptor (p75NTR) protein isoform expression in cultured SH-SY5Y cells and in hippocampi from late-stage human Alzheimer's disease (AD) brains. Interestingly, insulin induced the expression of basal and HMW p75NTR isoforms in SH-SY5Y cells, suggesting the presence of cross-talk between the IR and IGF-1R for the regulation of p75NTR expression. Reducing IR signaling with an IR kinase inhibitor (AG 1024) or IR-targeted siRNAs increased HMW p75NTR expression and reduced tyrosine receptor kinase-A (Trk-A) expression as well as postsynaptic density protein 95 (PSD95) expression in SH-SY5Y cells. Both basal and HMW p75NTR isoforms were increased in the hippocampi of post-mortem late-stage human AD brains (relative to non-AD brains), and the protein expression of HMW p75NTR was negatively associated with Trk-A expression, PSD95 expression, and IR expression. Thus, increased p75NTR expression, specifically an increased p75NTR-to-Trk-A ratio, is likely to play a role in synaptic loss and neuronal cell death in late-stage AD. Collectively, these findings suggest that increased expression of the p75NTR due to IR signaling inhibition by AßOs might be involved in the pathology of AD.


Assuntos
Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptor de Insulina/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular Tumoral , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/metabolismo , Transdução de Sinais/fisiologia
6.
Cells ; 4(3): 253-74, 2015 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-26184316

RESUMO

Dentate-gyral granule cells in the hippocampus plus dentate gyrus memory-recording/retrieving machine, unlike most other neurons in the brain, are continuously being generated in the adult brain with the important task of separating overlapping patterns of data streaming in from the outside world via the entorhinal cortex. This "adult neurogenesis" is driven by tools in the mature granule cell's cilium. Here we report our discovery of leptin's LepRb receptor in this cilium. In addition, we discuss how ciliary LepRb signaling might be involved with ciliary p75NTR and SSTR3 receptors in adult neurogenesis and memory formation as well as attenuation of Alzheimer's neuropathology by reducing the production of its toxic amyloid-ß-derived drivers.

7.
Curr Pharm Biotechnol ; 16(8): 696-706, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25941885

RESUMO

The "amyloid cascade hypothesis" posits that an extracellular build-up of amyloid-ß oligomers (Aß-os) and polymers (fibrils) subsequently inducing toxic hyperphosphorylated (p)-Tau oligomers (p-Tau-os) and neurofibrillary tangles starts the sporadic late-onset Alzheimer's disease (LOAD) in the aged lateral entorhinal cortex. Conversely, mutated genes cause a diffuse cerebral Aßs/Aß-os overproduction promoting early-onset familiar AD (EOFAD). Surplus exogenous Aß-os exert toxic actions at several levels. They reach the nuclei of human astrocyte-neurons teams (ANTs) to enhance the transcription of Aß precursor protein (APP) and ß-secretase/BACE1 genes. The overexpressed APP and BACE1 proteins act in concert with γ-secretase to overproduce endogenous Aßs/Aß-os, of which a few enter the nuclei to upkeep Aßs overproduction, while the rest gather in the cytoplasm, damage mitochondria, and are oversecreted. Simultaneously, extracellular Aß-os bind the ANTs' calcium-sensing receptors (CaSRs) activating signalings that hinder the proteolysis and hence favor the surplus hoarding/secretion of Aßs/Aß-os. Overreleased Aß-os spread, reach growing numbers of adjacent ANTs to recruit them to overproduce/oversecrete further Aß-os amounts via the just mentioned mechanisms. Alongside, Aß•CaSR signalings elicit a noxious overproduction/overrelease of nitric oxide (NO) and vascular endothelial growth factor (VEGF)-A from ANTs' astrocytes. While astrocytes survive the toxic onslaught, neurons die. Thus, AD progression is driven by ceaselessly self-sustaining neurotoxic cycles, which engender first Aß-os and later p-Tau-os that cooperatively destroy increasingly wider cognition-related cortical areas. Notably, a highly selective allosteric CaSR antagonist (calcilytic), like NPS 2143, does preserve human cortical postnatal HCN-1A neurons viability notwithstanding the presence of exogenous Aß-os by suppressing the otherwise elicited oversecretion and spread of newly synthesized Aß-os. Therefore, if given at minimal cognitive impairment or earlier stages, calcilytics could halt AD progression and preserve the patients' cortical neurons, cognitive abilities, and eventually life.


Assuntos
Doença de Alzheimer/prevenção & controle , Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/metabolismo , Humanos , Proteínas tau/metabolismo
8.
Neuroscientist ; 21(1): 9-29, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24740577

RESUMO

Evidence has begun emerging for the "contagious" and destructive Aß42 (amyloid-beta42) oligomers and phosphorylated Tau oligomers as drivers of sporadic Alzheimer's disease (AD), which advances along a pathway starting from the brainstem or entorhinal cortex and leading to cognition-related upper cerebral cortex regions. Seemingly, Aß42 oligomers trigger the events generating the neurotoxic Tau oligomers, which may even by themselves spread the characteristic AD neuropathology. It has been assumed that only neurons make and spread these toxic drivers, whereas their associated astrocytes are just janitorial bystanders/scavengers. But this view is likely to radically change since normal human astrocytes freshly isolated from adult cerebral cortex can be induced by exogenous Aß25-35, an Aß42 proxy, to make and secrete increased amounts of endogenous Aß42. Thus, it would seem that the steady slow progression of AD neuropathology along specific cognition-relevant brain networks is driven by both Aß42 and phosphorylated Tau oligomers that are variously released from increasing numbers of "contagion-stricken" members of tightly coupled neuron-astrocyte teams. Hence, we surmise that stopping the oversecretion and spread of the two kinds of "contagious" oligomers by such team members, perhaps via a specific CaSR (Ca(2+)-sensing receptor) antagonist like NPS 2143, might effectively treat AD.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Humanos , Neurônios/patologia , Fosforilação
9.
Neuromolecular Med ; 16(4): 645-57, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24948534

RESUMO

The excess vascular endothelial growth factor (VEGF) produced in the Alzheimer's disease (AD) brain can harm neurons, blood vessels, and other components of the neurovascular units (NVUs). But could astrocytes partaking in networks of astrocyte-neuron teams and connected to blood vessels of NVUs contribute to VEGF production? We have shown with cultured cerebral cortical normal (i.e., untransformed) adult human astrocytes (NAHAs) that exogenous amyloid-ß peptides (Aßs) stimulate the astrocytes to make and secrete large amounts of Aßs and nitric oxide by a mechanism mediated through the calcium-sensing receptor (CaSR). Here, we report that exogenous Aßs stimulate the NAHAs to produce and secrete even VEGF-A through a CaSR-mediated mechanism. This is indicated by the ability of Aßs to specifically bind the CaSR, and the capability of a CaSR activator, the "calcimimetic" NPS R-568, to imitate, and of the CaSR antagonist, "calcilytic" NPS 2143, to inhibit, the Aßs stimulation of VEGF-A production and secretion by the NAHAs. Thus, Aßs that accumulate in the AD brain may make the astrocytes that envelop and functionally collaborate with neurons into multi-agent AD-driving "machines" via a CaSR signaling mechanism(s). These observations suggest the possibility that CaSR allosteric antagonists such as NPS 2143 might impede AD progression.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Astrócitos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Detecção de Cálcio/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adulto , Regulação Alostérica , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Compostos de Anilina/farmacologia , Astrócitos/metabolismo , Cálcio/agonistas , Comunicação Celular , Células Cultivadas , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Naftalenos/farmacologia , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Fenetilaminas , Propilaminas , Ligação Proteica , Receptores de Detecção de Cálcio/antagonistas & inibidores , Lobo Temporal/citologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Biochem Biophys Res Commun ; 445(3): 656-60, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24569075

RESUMO

The synthetic ~5 kDa ABP (amyloid-ß binding peptide) consists of a region of the 228 kDa human pericentrioloar material-1 (PCM-1) protein that selectively and avidly binds in vitro Aß1-42 oligomers, believed to be key co-drivers of Alzheimer's disease (AD), but not monomers (Chakravarthy et al., (2013) [3]). ABP also prevents Aß1-42 from triggering the apoptotic death of cultured human SHSY5Y neuroblasts, likely by sequestering Aß oligomers, suggesting that it might be a potential AD therapeutic. Here we support this possibility by showing that ABP also recognizes and binds Aß1-42 aggregates in sections of cortices and hippocampi from brains of AD transgenic mice and human AD patients. More importantly, ABP targets Aß1-42 aggregates when microinjected into the hippocampi of the brains of live AD transgenic mice.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Autoantígenos/química , Autoantígenos/farmacologia , Encéfalo/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Peptídeos/química , Peptídeos/farmacologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Animais , Autoantígenos/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas de Ciclo Celular/administração & dosagem , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Peptídeos/administração & dosagem , Ligação Proteica
11.
Biochim Biophys Acta ; 1832(10): 1634-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23628734

RESUMO

The "amyloid-ß (Aß) hypothesis" posits that accumulating Aß peptides (Aßs) produced by neurons cause Alzheimer's disease (AD). However, the Aßs contribution by the more numerous astrocytes remains undetermined. Previously we showed that fibrillar (f)Aß25-35, an Aß42 proxy, evokes a surplus endogenous Aß42 production/accumulation in cortical adult human astrocytes. Here, by using immunocytochemistry, immunoblotting, enzymatic assays, and highly sensitive sandwich ELISA kits, we investigated the effects of fAß25-35 and soluble (s)Aß25-35 on Aß42 and Aß40 accumulation/secretion by human cortical astrocytes and HCN-1A neurons and, since the calcium-sensing receptor (CaSR) binds Aßs, their modulation by NPS 2143, a CaSR allosteric antagonist (calcilytic). The fAß25-35-exposed astrocytes and surviving neurons produced, accumulated, and secreted increased amounts of Aß42, while Aß40 also accrued but its secretion was unchanged. Accordingly, secreted Aß42/Aß40 ratio values rose for astrocytes and neurons. While slightly enhancing Aß40 secretion by fAß25-35-treated astrocytes, NPS 2143 specifically suppressed the fAß25-35-elicited surges of endogenous Aß42 secretion by astrocytes and neurons. Therefore, NPS 2143 addition always kept Aß42/Aß40 values to baseline or lower levels. Mechanistically, NPS 2143 decreased total CaSR protein complement, transiently raised proteasomal chymotrypsin activity, and blocked excess NO production without affecting the ongoing increases in BACE1/ß-secretase and γ-secretase activity in fAß25-35-treated astrocytes. Compared to fAß25-35, sAß25-35 also stimulated Aß42 secretion by astrocytes and neurons and NPS 2143 specifically and wholly suppressed this effect. Therefore, since NPS 2143 thwarts any Aß/CaSR-induced surplus secretion of endogenous Aß42 and hence further vicious cycles of Aß self-induction/secretion/spreading, calcilytics might effectively prevent/stop the progression to full-blown AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/efeitos dos fármacos , Naftalenos/farmacologia , Neurônios/efeitos dos fármacos , Receptores de Detecção de Cálcio/antagonistas & inibidores , Adulto , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/fisiologia , Astrócitos/metabolismo , Biopterinas/análogos & derivados , Biopterinas/farmacologia , Células Cultivadas , Humanos , Naftalenos/uso terapêutico , Neurônios/metabolismo
12.
J Neurochem ; 126(3): 415-24, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23432034

RESUMO

We have recently reported that a ~19-kDa polypeptide, rPK-4, is a protein kinase Cs inhibitor that is 89% homologous to the 1171-1323 amino acid region of the 228-kDa human pericentriolar material-1 (PCM-1) protein (Chakravarthy et al. 2012). We have now discovered that rPK-4 binds oligomeric amyloid-ß peptide (Aß)1-42 with high affinity. Most importantly, a PCM-1-selective antibody co-precipitated Aß and amyloid ß precursor protein (AßPP) from cerebral cortices and hippocampi from AD (Alzheimer's disease) transgenic mice that produce human AßPP and Aß1-42 , suggesting that PCM-1 may interact with amyloid precursor protein/Aß in vivo. We have identified rPK-4's Aß-binding domain using a set of overlapping synthetic peptides. We have found with ELISA, dot-blot, and polyacrylamide gel electrophoresis techniques that a ~ 5 kDa synthetic peptide, amyloid binding peptide (ABP)-p4-5 binds Aß1-42 at nM levels. Most importantly, ABP-p4-5, like rPK-4, appears to preferentially bind Aß1-42 oligomers, believed to be the toxic AD-drivers. As expected from these observations, ABP-p4-5 prevented Aß1-42 from killing human SH-SY5Y neuroblastoma cells via apoptosis. These findings indicate that ABP-p4-5 is a possible candidate therapeutic for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína
13.
Int J Mol Med ; 31(1): 3-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23124509

RESUMO

The pathophysiological mechanisms underlying the onset and inexorable progression of the late­onset form of Alzheimer's disease (AD) are still the object of controversy. This review takes stock of some most recent advancements of this field concerning the complex roles played by the amyloid­ß (Aß)­binding p75 neurotrophin receptor (p75NTR) and calcium­sensing receptor (CaSR) and by the primary cilia in AD. Apart from their physiological roles, p75NTR is more intensely expressed in the hippocampus of human AD brains and Aß­bound p75NTR triggers cell death, whereas Aß­bound CaSR signalling induces the de novo synthesis and release of nitric oxide (NO), vascular endothelial growth factor (VEGF)­A and Aß peptides (Aßs), particularly on the part of normal adult human astrocytes. The latter effect could significantly increase the pool of Aß­ and NO­producing nerve cells favouring the progressive spread of a self­sustaining and self­reinforcing 'infectious' mechanism of neural and vascular (i.e. blood-brain barrier) cell damage. Interestingly, primary cilia concentrate p75NTR receptors in their membranes and are abnormally structured/damaged in transgenic (Tg) AD­model mice, which could impact on the adult neurogenesis occurring in the dentate gyrus's subgranular zone (SGZ) that is necessary for new memory encoding, thereby favouring typical AD cognitive decline. Altogether, these findings may pave the way to novel therapeutic approaches to AD, particularly in its mild cognitive impairment (MCI) and pre­MCI stages of development.


Assuntos
Doença de Alzheimer/fisiopatologia , Astrócitos/metabolismo , Cílios/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Morte Celular , Giro Denteado/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Transdução de Sinais
14.
Biochem Biophys Res Commun ; 427(1): 218-22, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22995307

RESUMO

The hippocampal dentate gyrus is one of the two sites of continuous neurogenesis in adult rodents and humans. Virtually all dentate granule cells have a single immobile cilium with a microtubule spine or axoneme covered with a specialized cell membrane loaded with receptors such as the somatostatin receptor 3 (SSTR3), and the p75 neurotrophin receptor (p75(NTR)). The signals from these receptors have been reported to stimulate neuroprogenitor proliferation and the post-mitotic maturation of newborn granule cells into functioning granule cells. We have found that in 6-24-months-old triple transgenic Alzheimer's disease model mice (3xTg-AD) producing both Aß(1-42) and the mutant human tau protein tau(P301L,) the dentate granule cells still had immunostainable SSTR3- and p75(NTR)-bearing cilia but they were only half the length of the immunostained cilia in the corresponding wild-type mice. However, the immunostainable length of the granule cell cilia was not reduced either in 2xTg-AD mice accumulating large amounts of Aß(1-42) or in mice accumulating only a mutant human tau protein. Thus it appears that a combination of Aß(1-42) and tau protein accumulation affects the levels of functionally important receptors in 3xTg-AD mice. These observations raise the important possibility that structural and functional changes in granule cell cilia might have a role in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Giro Denteado/metabolismo , Fragmentos de Peptídeos/biossíntese , Proteínas tau/biossíntese , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Biomarcadores/metabolismo , Cílios/metabolismo , Giro Denteado/patologia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Receptores de Fator de Crescimento Neural/metabolismo , Receptores de Somatostatina/metabolismo , Coloração e Rotulagem , Proteínas tau/genética
15.
J Neurol Sci ; 322(1-2): 137-40, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22841885

RESUMO

Alzheimer's disease (AD) is the most common human neurodegenerative ailment, the most prevalent (>95%) late-onset type of which has a still uncertain etiology. The progressive decline of cognitive functions, dementia, and physical disabilities of AD is caused by synaptic losses that progressively disconnect key neuronal networks in crucial brain areas, like the hippocampus and temporoparietal cortex, and critically impair language, sensory processing, memory, and conscious thought. AD's two main hallmarks are fibrillar amyloid-ß (fAß) plaques in extracellular spaces and intracellular accumulation of fAß peptides and neurofibrillary tangles (NFTs). It is still undecided whether either or both these AD hallmarks cause or result from the disease. Recently, the dysregulation of calcium homeostasis has been advanced as a novel cause of AD. In this case, a suitable candidate of AD driver would be the Aß peptides-binding/activated calcium-sensing receptor (CaSR), whose intracellular signalling is triggered by Aß peptides. In this review, we briefly discuss CaSR's roles in normal adult human astrocytes (NAHAs) and their possible impacts on AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Doença de Alzheimer/complicações , Astrócitos/metabolismo , Astrócitos/patologia , Transtornos Cognitivos/etiologia , Humanos , Emaranhados Neurofibrilares/patologia , Placa Amiloide
16.
Biochem Biophys Res Commun ; 424(1): 147-51, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22732405

RESUMO

We had previously identified a protein kinase C (PKC) inhibitor in murine neuroblastoma cells (Chakravarthy et al. [1]). Similar PKC inhibitory activity was also found in adult rat brain. Using polyclonal antibodies raised against the partially purified PKC inhibitor from rat brain as bait, we isolated a putative brain PKC inhibitor using a T-7 phage display system expressing human brain cDNA library. After enriching the phage population expressing the putative PKC inhibitor with four rounds of biopanning using ELISA and in vitro PKC binding assays, we identified a phage clone that expressed a product with significant PKC inhibitory activity. We have cloned and expressed this cDNA in a bacterial system and purified the recombinant protein. This polypeptide (174 amino acids) is highly homologous to a region of the 228-kDa PCM-1, the human pericentriolar material 1 protein. We have mapped this polypeptide's PKC-inhibitory domain and shown its PKC inhibitory activity in vitro. However, it will need to be determined whether the full-length PCM-1 protein possesses PKC inhibitory activity in vivo, and if so, how this might contribute to PCM-1's recently demonstrated roles in ciliogenesis and neurogenesis.


Assuntos
Autoantígenos/química , Proteínas de Ciclo Celular/química , Peptídeos/química , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Sequência de Aminoácidos , Animais , Autoantígenos/genética , Autoantígenos/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacologia , Biblioteca Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos/genética , Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
17.
J Alzheimers Dis ; 31(3): 493-506, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22635104

RESUMO

The p75 neurotrophin receptor (p75NTR) has been thought to play a critical role in amyloid-ß peptide (Aß)-mediated neurodegeneration and Aß metabolism in Alzheimer's disease (AD) brains. Our previous report showed that membrane-associated p75NTR protein expression was significantly increased in the hippocampi of two different strains of transgenic AD mice and was associated with the age-dependent elevation of Aß1-42 levels. Here, we provide evidence that the Aß1-42 oligomers known as ADDLs (Aß-derived diffusible ligands) induce p75NTR protein expression through insulin-like growth factor 1 receptor (IGF-1R) phosphorylation in SH-SY5Y human neuroblastoma cells. An in vivo microinjection study demonstrated that microinjected ADDLs increased the p75NTR protein expression by 1.4-fold in the ipsilateral hippocampus compared to the contralateral hippocampus. In addition, ADDLs microinjected into mouse hippocampi facilitated IGF-1R phosphorylation within 30 min and the co-administration of picropodophyllin, an IGF-1R kinase inhibitor, blocked ADDLs-induced p75NTR expression. We examined the possible involvement of IGF-1R in the increased p75NTR protein expression in the hippocampi of 6-month-old AßPPswe/PS1dE9 AD model mice that had accumulated significant amounts of Aß1-42 and showed significantly higher p75NTR expression than age-matched wild-type mice. We found that IGF-1R phosphorylation in these transgenic mice was higher than that in the wild-type mice. These findings indicate that Aß1-42 oligomers stimulate the p75NTR protein expression in the hippocampus through IGF-1R signaling. Thus, Aß1-42 oligomers-mediated IGF-1R activation may trigger an increase in p75NTR protein expression in the hippocampus of AD brain during the early stages of disease development.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Regulação da Expressão Gênica , Hipocampo/metabolismo , Fragmentos de Peptídeos/fisiologia , Receptor IGF Tipo 1/fisiologia , Receptores de Fator de Crescimento Neural/biossíntese , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Hipocampo/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
18.
J Alzheimers Dis ; 30(3): 675-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22451321

RESUMO

The pan-specific p75 neurotrophin receptor (p75(NTR)) is believed to play an important role in the pathogenesis of Alzheimer's disease (AD). It is involved in mediating amyloid-ß (Aß) toxicity and stimulating amyloidogenesis. In addition, we have recently shown that stimulating cultured SH-SY5Y human neuroblastoma cells with Aß(42) increases the level of membrane-associated p75(NTR) and that Aß(42)-accumation in two strains of transgenic AD model mice is accompanied by an increased level of hippocampal membrane-associated p75(NTR) (Chakravarthy et al. J Alzheimers Dis 19, 915-925, 2010). This raised an important question whether accumulating Aß(42) in human AD is also accompanied by an increased hippocampal membrane-associated p75(NTR). In this study, using polyclonal and monoclonal antibodies against the p75(NTR) receptor's intra- and extracellular domains, we show that indeed the mean level of membrane-associated p75(NTR) in the hippocampal formation is significantly higher (~two-fold, p < 0.03) in human AD brains than in identical samples of hippocampal formation in age-matched non-AD human brains. The possible relation of this elevated hippocampal p75(NTR) to AD cognitive decline is discussed.


Assuntos
Doença de Alzheimer/metabolismo , Membrana Celular/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Linhagem Celular Tumoral , Membrana Celular/patologia , Feminino , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Hipocampo/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Células Tumorais Cultivadas
19.
J Alzheimers Dis ; 24(2): 335-47, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21258151

RESUMO

Astrocytes in amyloid-ß (Aß)42-accumulating human brains afflicted with Alzheimer's disease (AD) upregulate vascular endothelial growth factor (VEGF)-A synthesis and also become loaded with Aß42. We have already shown that Aß(25-35) (surrogate of Aß42)-induced VEGF-A production in 'normoxic' cultures of early passage normal human cerebral astrocytes (NAHAs) is mediated by the stabilization of VEGF gene-stimulating hypoxia-inducible factor (HIF)-1α and nuclear translocation of HIF-1α•HIF-1ß complexes. We have now found that treating these NAHAs with Aß(25-35) also stimulates them to make Aß42 (appearing in immunoblots as several bands with M(r)'s from 8 kDa upwards), whose levels peak at 48 h (2.8-fold versus 0 h, p < 0.001) and then start falling slowly. This rise of Aß42 peptide production coincides with a transiently increased flow of HIF-1α (therefore HIF-1α•HIF-1ß complexes; at 24 h, 1.5-fold versus 0 h, p < 0.001) into the nucleus and transient surges first of ß-secretase (BACE-1/ß-S) mRNA expression (1.2-fold versus 0 h, p = 0.013) and activity peaking at 24-h (1.4-fold versus 0 h, p = 0.001), and then of γ-secretase (γ-S) activity cresting at 48 h (1.6-fold versus 0 h, p < 0.001) that cleave the Aß42 peptides from amyloid-ß protein precursor. Since the genes encoding components of these two secretases have the same HIF-1α•HIF-1ß-responsive elements in their promoters as the VEGF gene, these observations suggest that the Aß42 released from neurons in the AD brain can recruit associated astrocytes via HIF-1α•HIF-1ß signaling into the pool of Aß42-producing cells. In other words, Aß42 begets Aß42 in NAHAs.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Astrócitos/efeitos dos fármacos , Córtex Cerebral/citologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Adolescente , Adulto , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Análise de Variância , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Fragmentos de Peptídeos/antagonistas & inibidores , RNA Mensageiro/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
20.
Biochem Biophys Res Commun ; 401(3): 458-62, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20875398

RESUMO

The densely ciliated granule cell layer of the adult murine hippocampal dentate gyrus is one of two sites of adult neurogenesis. The granule cells have already been proven to localize their SSTR3 (somatostatin receptor 3) receptors to their so-called primary cilia. Here we show for the first time that 70-90% of these cells in 7-18 months-old wild-type and 3×Tg-AD (Alzheimer disease transgenic) mice also load p75(NTR) receptors into the structures containing SSTR3, i.e., their primary cilia. On the other hand, p75(NTR')s TrkA co-receptors were not localized to cilia but conventionally distributed throughout the cell surface. Significantly fewer cells (20-40%) in the hippocampal CA1 and CA3 regions and cerebral cortex have p75(NTR) containing cilia. While we don't know what the impact of the cilial localization of p75(NTR) on dentate gyral adult neurogenesis and memory encoding might be, the cilia's amyloid ß-activatable p75(NTR) receptors could be damaging or lethal to the hippocampal functioning of amyloid ß-accumulating Alzheimer brain.


Assuntos
Doença de Alzheimer/metabolismo , Giro Denteado/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Região CA3 Hipocampal/metabolismo , Cílios/metabolismo , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...