Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 122: 185-201, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39142420

RESUMEN

Amyloid-ß (Aß) and hyperphosphorylated tau protein are targets for Alzheimer's Disease (AD) immunotherapies, which are generally focused on single epitopes within Aß or tau. However, due to the complexity of both Aß and tau in AD pathogenesis, a multipronged approach simultaneously targeting multiple epitopes of both proteins could overcome limitations of monotherapies. Herein, we propose an active AD immunotherapy based on a nanoparticle vaccine comprising two Aß peptides (1-14 and pyroglutamate pE3-14) and three tau peptides (centered on phosphorylated pT181, pT217 and pS396/404). These correspond to both soluble and aggregated targets and are displayed on the surface of immunogenic liposomes in an orientation that maintains reactivity with epitope-specific monoclonal antibodies. Intramuscular immunization of mice with individual epitopes resulted in minimally cross-reactive antibody induction, while simultaneous co-display of 5 antigens ("5-plex") induced antibodies against all epitopes without immune interference. Post-immune sera recognized plaques and neurofibrillary tangles from human AD brain tissue. Vaccine administration to 3xTg-AD mice using a prophylactic dosing schedule inhibited tau and amyloid pathologies and resulted in improved cognitive function. Immunization was well tolerated and did not induce antigen-specific cellular responses or persistent inflammatory responses in the peripheral or central nervous system. Antibody levels could be reversed by halting monthly vaccinations. Altogether, these results indicate that active immune therapies based on nanoparticle formulations of multiple Aß and tau epitopes warrant further study for treating early-stage AD.

2.
Mol Biol Rep ; 49(5): 3955-3964, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35235160

RESUMEN

PURPOSE: Cellular responses following cerebral ischemia/reperfusion injury are critical to recovery and survival after ischemic stroke. Understanding of these cellular responses can help the design of therapies to protect brain tissue and promote recovery after stroke. One of these cellular responses may be mediated by the AKT (protein kinase B) signal transduction pathway. This study was aimed to investigate the cerebral ischemia-induced alterations of AKT signaling and the upstream molecular pathways. METHODS: We modeled cerebral ischemia by middle cerebral artery occlusion in 2-3-month-old male C57BL/6J mice and then analyze the brain samples by using quantitative Western blots and phosphorylation/activation-dependent kinase antibodies. Cerebral ischemia was confirmed by staining of brain slices with 1% 2,3,5-triphenyltetrazolium chloride (TTC) and Nissl, as well as neurological assessments of the mice 24 h after ischemia-reperfusion surgery. RESULTS: We found marked downregulation of AKT within 12 h of cerebral ischemia/reperfusion, which leads to overactivation of glycogen synthase kinase-3ß (GSK-3ß). Furthermore, we found that the downregulation of AKT was mediated by downregulation of mTORC2 (the complex 2 of the mechanistic target of rapamycin) instead of its common upstream kinases, phosphatidylinositol 3-kinase and phosphoinositide-dependent kinase-1. CONCLUSION: Our findings provide new insight into the cellular responses to ischemia/reperfusion brain injury and will help develop new treatments targeting the AKT signaling pathway for the treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica , Glucógeno Sintasa Quinasa 3 beta , Accidente Cerebrovascular Isquémico , Proteínas Proto-Oncogénicas c-akt , Daño por Reperfusión , Serina-Treonina Quinasas TOR , Animales , Isquemia Encefálica/metabolismo , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
3.
Int J Mol Sci ; 23(23)2022 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-36499564

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that eventually leads to dementia and death of the patient. Currently, no effective treatment is available that can slow or halt the progression of the disease. The gut microbiota can modulate the host immune system in the peripheral and central nervous system through the microbiota-gut-brain axis. Growing evidence indicates that gut microbiota dysbiosis plays an important role in the pathogenesis of AD, and modulation of the gut microbiota may represent a new avenue for treating AD. Immunotherapy targeting Aß and tau has emerged as the most promising disease-modifying therapy for the treatment of AD. However, the underlying mechanism of AD immunotherapy is not known. Importantly, preclinical and clinical studies have highlighted that the gut microbiota exerts a major influence on the efficacy of cancer immunotherapy. However, the role of the gut microbiota in AD immunotherapy has not been explored. We found that immunotherapy targeting tau can modulate the gut microbiota in an AD mouse model. In this article, we focused on the crosstalk between the gut microbiota, immunity, and AD immunotherapy. We speculate that modulation of the gut microbiota induced by AD immunotherapy may partially underlie the efficacy of the treatment.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Animales , Ratones , Enfermedad de Alzheimer/patología , Disbiosis/terapia , Modelos Animales de Enfermedad , Sistema Nervioso Central/patología
4.
J Neural Transm (Vienna) ; 122(4): 607-17, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25233799

RESUMEN

Intraneuronal accumulation of abnormally hyperphosphorylated tau in the brain is a histopathological hallmark of Alzheimer's disease and a family of related neurodegenerative disorders collectively called tauopathies. At present there is no effective treatment available for these progressive neurodegenerative diseases which are clinically characterized by dementia in mid to old-age. Here we report the treatment of 14-17-months-old 3xTg-AD mice with tau antibodies 43D (tau 6-18) and 77E9 (tau 184-195) to the N-terminal projection domain of tau or mouse IgG as a control by intraperitoneal injection once a week for 4 weeks, and the effects of the passive immunization on reduction of hyperphosphorylated tau, Aß accumulation and cognitive performance in these animals. We found that treatment with tau antibodies 43D and 77E9 reduced total tau level, decreased tau hyperphosphorylated at Ser199, Ser202/Thr205 (AT8), Thr205, Ser262/356 (12E8), and Ser396/404 (PHF-1) sites, and a trend to reduce Aß pathology. Most importantly, targeting N-terminal tau especially by 43D (tau 6-18) improved reference memory in the Morris water maze task in 3xTg-AD mice. We did not observe any abnormality in general physical characteristics of the treated animals with either of the two antibodies during the course of this study. Taken together, our studies demonstrate for the first time (1) that passive immunization targeting normal tau can effectively clear the hyperphosphorylated protein and possibly reduce Aß pathology from the brain and (2) that targeting N-terminal projection domain of tau containing amino acid 6-18 is especially beneficial. Thus, targeting selective epitopes of N-terminal domain of tau may present a novel effective therapeutic opportunity for Alzheimer disease and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/terapia , Inmunización Pasiva/métodos , Proteínas tau/inmunología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos/administración & dosificación , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Aprendizaje por Laberinto , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Fosforilación , Placa Amiloide/etiología , Placa Amiloide/patología , Placa Amiloide/terapia , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
5.
J Biol Chem ; 288(33): 23875-83, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23843462

RESUMEN

The homeostasis of protein metabolism is maintained and regulated by the rates of protein biosynthesis and degradation in living systems. Alterations of protein degradation may regulate protein biosynthesis through a feedback mechanism. Whether a change in protein biosynthesis modulates protein degradation has not been reported. In this study, we found that inhibition of protein biosynthesis induced phosphorylation/activation of AKT and led to phosphorylation of AKT target substrates, including FoxO1, GSK3α/ß, p70S6K, AS160, and the E3 ubiquitin ligase MDM2. Phosphorylation of ribosomal protein S6 was also modulated by inhibition of protein biosynthesis. The AKT phosphorylation/activation was mediated mainly through the PI3K pathway because it was blocked by the PI3K inhibitor LY294002. The activated AKT phosphorylated MDM2 at Ser(166) and promoted degradation of the tumor suppressor p53. These findings suggest that inhibition of protein biosynthesis can alter degradation of some proteins through activation of AKT. This study reveals a novel regulation of protein degradation and calls for caution in blocking protein biosynthesis to study the half-life of proteins.


Asunto(s)
Biosíntesis de Proteínas , Proteolisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Cromonas , Cicloheximida/farmacología , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Morfolinas , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal , Sirolimus/farmacología , Especificidad por Sustrato/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
6.
Neurobiol Dis ; 71: 110-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25046994

RESUMEN

Besides the presence of amyloid beta (Aß) plaques and neurofibrillary tangles, neurogenesis and synaptic plasticity are markedly impaired in Alzheimer's disease (AD) possibly contributing to cognitive impairment. In this context, neurotrophic factors serve as a promising therapeutic approach via utilization of regenerative capacity of brain to shift the balance from neurodegeneration to neural regeneration. However, besides more conventional "bystander" effect, to what extent can neurotrophic compounds affect underlying AD pathology remains questionable. Here we investigated the effect of chronic oral treatment with a ciliary neurotrophic factor (CNTF) derived peptidergic compound, P021 (Ac-DGGL(A)G-NH2), on disease pathology both at moderate and severe stages in a transgenic mouse model of AD. 3xTg-AD and wild type female mice were treated for 12months with P021 or vehicle diet starting at 9-10months of age. A significant reduction in abnormal hyperphosphorylation and accumulation of tau at known major AD neurofibrillary pathology associated sites was observed. The effect of P021 on Aß pathology was limited to a significant decrease in soluble Aß levels and a trend towards reduction in Aß plaque load in CA1 region of hippocampus, consistent with reduction in Aß generation and not clearance. This disease modifying effect was probably via increased brain derived neurotrophic factor (BDNF) expression mediated decrease in glycogen synthase kinase-3-ß (GSK3ß) activity we found in P021 treated 3xTg-AD mice. P021 treatment also rescued deficits in cognition, neurogenesis, and synaptic plasticity in 3xTg-AD mice. These findings demonstrate the potential of the neurotrophic peptide mimetic as a disease modifying therapy for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antipsicóticos/administración & dosificación , Encéfalo/efectos de los fármacos , Factor Neurotrófico Ciliar/administración & dosificación , Administración Oral , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Antipsicóticos/sangre , Antipsicóticos/química , Barrera Hematotesticular/efectos de los fármacos , Barrera Hematotesticular/fisiología , Células Cultivadas , Factor Neurotrófico Ciliar/sangre , Factor Neurotrófico Ciliar/química , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Presenilina-1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Proteínas tau/genética
7.
J Alzheimers Dis Rep ; 8(1): 615-626, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38746631

RESUMEN

Background: Diabetes mellitus (DM) increases the risk for cognitive impairment and Alzheimer's disease (AD). Diabetic ketoacidosis (DKA), a serious complication of DM, may also cause brain damage and further AD, but the underlying molecular mechanisms remain unclear. Objective: Our objective was to understand how DKA can promote neurodegeneration in AD. Methods: We induced DKA in rats through intraperitoneal injection of streptozotocin, followed by starvation for 48 hours and investigated AD-related brain alterations focusing on tau phosphorylation. Results: We found that DKA induced hyperphosphorylation of tau protein at multiple sites associated with AD. Studies of tau kinases and phosphatases suggest that the DKA-induced hyperphosphorylation of tau was mainly mediated through activation of c-Jun N-terminal kinase and downregulation of protein phosphatase 2A. Disruption of the mTOR-AKT (the mechanistic target of rapamycin-protein kinase B) signaling pathway and increased levels of synaptic proteins were also observed in the brains of rats with DKA. Conclusions: These results shed some light on the mechanisms by which DKA may increase the risk for AD.

8.
Exp Neurol ; 362: 114337, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36717015

RESUMEN

Tau pathology is essential in the pathogenesis of Alzheimer's disease (AD) and related tauopathies. Tau immunotherapy aimed at reducing the progression of tau pathology provides a potential therapeutic strategy for treating these diseases. By screening monoclonal antibodies 43D, 63B, 39E10, and 77G7 that recognize epitopes ranging from tau's N-terminus to C-terminus, we found the 77G7, which targets the microtubule-binding domain promoted tau clearance in a dose-dependent manner by entering neuronal cells in vitro. Intra-cerebroventricular injection of 77G7 antibody reduced tau levels in the wild-type FVB mouse brain. Without influencing the levels of detergent-insoluble and aggregated tau, intravenous injection of 77G7 reduced tau hyperphosphorylation in the brain and improved novel object recognition but not spatial learning and memory in 15-18-month-old 3xTg-AD mice. These studies suggest that epitopes recognized by tau antibodies are crucial for the efficacy of immunotherapy. Immunization with antibody 77G7 provides a novel potential opportunity for tau-directed immunotherapy of AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Ratones , Animales , Enfermedad de Alzheimer/patología , Proteínas tau/metabolismo , Fosforilación , Ratones Transgénicos , Anticuerpos Monoclonales/farmacología , Inmunización Pasiva , Epítopos , Modelos Animales de Enfermedad
9.
Genes (Basel) ; 14(2)2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36833432

RESUMEN

Background Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and autism. Gene therapy may offer an efficient method to ameliorate the symptoms of this disorder. Methods An AAVphp.eb-hSyn-mFMR1IOS7 vector and an empty control were injected into the tail vein of adult Fmr1 knockout (KO) mice and wildtype (WT) controls. The KO mice were injected with 2 × 1013 vg/kg of the construct. The control KO and WT mice were injected with an empty vector. Four weeks following treatment, the animals underwent a battery of tests: open field, marble burying, rotarod, and fear conditioning. The mouse brains were studied for levels of the Fmr1 product FMRP. Results: No significant levels of FMRP were found outside the CNS in the treated animals. The gene delivery was highly efficient, and it exceeded the control FMRP levels in all tested brain regions. There was also improved performance in the rotarod test and partial improvements in the other tests in the treated KO animals. Conclusion: These experiments demonstrate efficient, brain-specific delivery of Fmr1 via peripheral administration in adult mice. The gene delivery led to partial alleviation of the Fmr1 KO phenotypical behaviors. FMRP oversupply may explain why not all behaviors were significantly affected. Since AAV.php vectors are less efficient in humans than in the mice used in the current experiment, studies to determine the optimal dose using human-suitable vectors will be necessary to further demonstrate feasibility.


Asunto(s)
Barrera Hematoencefálica , Síndrome del Cromosoma X Frágil , Humanos , Animales , Ratones , Ratones Noqueados , Barrera Hematoencefálica/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Estudios de Factibilidad , Terapia Genética
10.
J Alzheimers Dis Rep ; 6(1): 245-255, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35719714

RESUMEN

Background: The triple transgenic mouse model of Alzheimer's disease (3×Tg-AD) has gained popularity in Alzheimer's research owing to the progressive development of both amyloid-ß and tau pathologies in its brain. Prior handling-habituation, a necessary preparation procedure that reduces anxiety and stress in rodents, was seldom described in the literature involving these mice and needs to be addressed. Objective: We sought to determine whether 3×Tg-AD mice differ from B6;129 genetic control mice in terms of tameness and prior habituation to handling. Methods: We devised hand-staying and hand-boarding assays to evaluate tameness in 3×Tg-AD and B6;129 genetic control mice at 2.5, 7, and 11.5 months of age, representing cognitively pre-symptomatic, early symptomatic and advanced symptomatic stages of the disease, respectively. We monitored the progress of handling-habituation across 8-15 daily handling sessions and assessed the animal behaviors in elevated plus maze. Results: We found that 3×Tg-AD mice were markedly tamer than age-matched control mice at the baseline. Whereas it took 2-3 days for 3×Tg-AD mice to reach the criteria for full tameness, it took an average of 7-9 days for young genetic control mice to do so. Prior handling-habituation enhanced risk assessment and coping strategy in mice in elevated plus maze. Completely handling-habituated mice exhibited comparable anxiety indices in the maze regardless of genotype and age. Conclusion: These findings collectively point to inherently heightened tameness and accelerated handling-habituation in 3×Tg-AD mice on a B6;129 genetic background. These traits should be carefully considered when behaviors are compared between 3×Tg-AD and the genetic control mice.

11.
Front Aging Neurosci ; 14: 837649, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35222001

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that eventually leads to dementia and death of the patient. Despite the enormous amounts of resources and efforts for AD drug development during the last three decades, no effective treatments have been developed that can slow or halt the progression of the disease. Currently available drugs for treating AD can only improve clinical symptoms temporarily with moderate efficacies. In recent years, the scientific community has realized these challenges and reconsidered the future directions of AD drug development. The most significant recent changes in AD drug development strategy include shifting from amyloid-based targets to other targets, such as tau, and efforts toward better designs for clinical trials. However, most AD drug development is still focused on a single mechanism or target, which is the conventional strategy for drug development. Although multifactorial mechanisms and, on this basis, multi-target strategies have been proposed in recent years, this approach has not been widely recognized and accepted by the mainstream of AD drug development. Here, we emphasize the multifactorial mechanisms of AD and discuss the urgent need for a paradigm shift in AD drug development from a single target to multiple targets, either with the multi-target-directed ligands approach or the combination therapy approach. We hope this article will increase the recognition of the multifactorial nature of AD and promote this paradigm shift. We believe that such a shift will facilitate successful development of effective AD therapies.

12.
CNS Neurosci Ther ; 28(12): 2245-2259, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36114722

RESUMEN

INTRODUCTION: Neurofibrillary tangle (NFT) of hyperphosphorylated tau is a hallmark of Alzheimer's disease (AD) and related tauopathies. Tau lesion starts in the trans-entorhinal cortex, from where it spreads to limbic regions, followed by neocortical areas. The regional distribution of NFTs associates with the progression of AD. Accumulating evidence suggests that proteopathic tau can seed tau aggregation in a prion-like fashion in vitro and in vivo. Inhibition of tau seeding activity could provide a potential therapeutic opportunity to block the propagation of tau pathology in AD and related tauopathies. AIMS: In the present study, we investigated the role of 77G7, a monoclonal tau antibody to the microtubule-binding repeats, in repressing the seeding activity of proteopathic tau. RESULTS: We found that 77G7 had a higher affinity toward aggregated pathological tau fractions than un-aggregated tau derived from AD brain. 77G7 inhibited the internalization of tau aggregates by cells, blocked AD O-tau to capture normal tau, and to seed tau aggregation in vitro and in cultured cells. Tau pathology induced by hippocampal injection of AD O-tau in 3xTg-AD mice was suppressed by mixing 77G7 with AD O-tau. Intravenous administration of 77G7 ameliorated site-specific hyperphosphorylation of tau induced by AD O-tau in the hippocampi of Tg/hTau mice. CONCLUSION: These findings indicate that 77G7 can effectively suppress the seeding activity of AD O-tau and thus could be developed as a potential immunotherapeutic drug to inhibit the propagation of tau pathology in AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Animales , Ratones , Proteínas tau/metabolismo , Tauopatías/metabolismo , Tauopatías/patología , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Anticuerpos Monoclonales , Microtúbulos/metabolismo , Microtúbulos/patología
13.
Mol Pharm ; 8(5): 1687-97, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21812426

RESUMEN

Overexpression of HER2/neu, a transmembrane tyrosine kinase acting as a coreceptor for other EGFR family members, is well-known to be associated with a poor prognosis in cancer. In the present study, we observed that emodin AMAD, a novel emodin azide methyl anthraquinone derivative, extracted from nature's giant knotweed rhizome of traditional Chinese herbs, potently decreased Her2/neu protein in dose- and time-dependent manners and also inhibited the downstream MAPK and PI3K-Akt signaling pathway. Intriguingly, reverse transcription-PCR and protein turnover assay revealed that the decrease of Her2/neu was independent of mRNA level but primarily owing to its protein stability. Meanwhile, proteasome inhibitor MG132 but not lysosome inhibitor chloroquine could restore Her2/neu and polyubiquitination of Her2/neu was augmented during emodin AMAD treatment. Furthermore, immunofluorescence study with anti-Her2/neu antibody showed that emodin AMAD disturbed the subcellular distribution of Her2/neu, with decreased location in the plasma membrane. Molecular docking studies predicted that AMAD can interact with the ATP-binding pocket of both Hsp90 and Her2/neu. Importantly, coimmunoprecipitation and immunofluorescence study revealed that emodin AMAD markedly impaired the binding between Hsp90 and Her2/neu and could bind to both Hsp90 and Her2/neu as reinforced by molecular modeling studies. In addition, combination of emodin AMAD treatment and siRNA against Her2 synergistically inhibited proliferation and induced apoptosis. Taken together, these data suggest that blockade of Her2/neu binding to Hsp90 and following proteasomal degradation of Her2/neu were involved in emodin AMAD-induced apoptosis in Her2/neu-overexpressing cancer cells. Our results provide suggestions that emodin AMAD could be promising as a new targeting therapeutic strategy in the treatment of Her2/neu-overexpressing cancers.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos Fitogénicos/farmacología , Emodina/análogos & derivados , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antraquinonas/administración & dosificación , Antraquinonas/química , Antraquinonas/uso terapéutico , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Azidas/química , Azidas/farmacología , Línea Celular Tumoral , Emodina/química , Emodina/farmacología , Emodina/uso terapéutico , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Inhibidores de Proteasoma , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Distribución Aleatoria , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Alzheimers Dis ; 82(2): 631-646, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34057082

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by impairments in synaptic plasticity and cognitive performance. Cognitive dysfunction and loss of neuronal plasticity are known to begin decades before the clinical diagnosis of the disease. The important influence of congenital genetic mutations on the early development of AD provides a novel opportunity to initiate treatment during early development to prevent the Alzheimer-like behavior and synaptic dysfunction. OBJECTIVE: To explore strategies for early intervention to prevent Alzheimer's disease. METHODS: In the present study, we investigated the effect of treatment during early development with a ciliary neurotrophic factor (CNTF) derived peptidergic compound, P021 (Ac-DGGLAG-NH2) on cognitive function and synaptic plasticity in 3xTg-AD transgenic mouse model of AD. 3xTg-AD and genetic background-matched wild type female mice were treated from birth to postnatal day 120 with P021 in diet or as a control with vehicle diet, and cognitive function and molecular markers of neuroplasticity were evaluated. RESULTS: P021 treatment during early development prevented cognitive impairment and increased expressions of pCREB and BDNF that activated downstream various signaling cascades such as PLC/PKC, MEK/ERK and PI3K/Akt, and ameliorated synaptic protein deficit in 4-month-old 3xTg-AD mice. CONCLUSION: These findings indicate that treatment with the neurotrophic peptide mimetic such as P021 during early development can be an effective therapeutic strategy to rescue synaptic deficit and cognitive impairment in familial AD and related tauopathies.


Asunto(s)
Adamantano/análogos & derivados , Enfermedad de Alzheimer , Disfunción Cognitiva , Plasticidad Neuronal , Oligopéptidos/farmacología , Adamantano/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/psicología , Animales , Cognición/efectos de los fármacos , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Modelos Animales de Enfermedad , Intervención Médica Temprana/métodos , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/farmacología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Periodo Posparto , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
15.
J Alzheimers Dis ; 81(1): 273-286, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814439

RESUMEN

BACKGROUND: Abnormal hyperphosphorylation of microtubule-associated protein tau plays a pivotal role in Alzheimer's disease (AD). We previously found that O-GlcNAcylation inversely correlates to hyperphosphorylation of tau in AD brain, and downregulation of brain O-GlcNAcylation promotes tau hyperphosphorylation and AD-like neurodegeneration in mice. OBJECTIVE: Herein we investigated the effect of increasing O-GlcNAcylation by using intermittent dosing with low doses of a potent novel O-GlcNAcase (OGA) inhibitor on AD-like brain changes and cognitive function in a mouse model of sporadic AD (sAD) induced by intracerebroventricular (ICV) injection of streptozotocin (STZ). METHODS: STZ was injected into the lateral ventricle of C57BL/6J mice. From the second day, Thiamme2-G (TM2G) or saline, as a vehicle control, was orally administered to the ICV-STZ mice three times per week for five weeks. A separate group of ICV-saline mice treated with saline was used as a baseline control. Behavioral tests, including open field and novel object recognition, were conducted three weeks after the first dose of the TM2G or saline. Protein O-GlcNAcylation, tau hyperphosphorylation, synaptic proteins, and neuroinflammation in the mouse brain were assessed by western blotting. RESULTS: ICV-STZ caused decreased protein O-GlcNAcylation. Enhancement of O-GlcNAcylation to moderate levels by using low-dose OGA inhibitor in ICV-STZ mice prevented STZ-induced body weight loss, rescued cognitive impairments, and restored AD-like pathologies, including hyperphosphorylation of tau and abnormalities in synaptic proteins and neuroinflammation. CONCLUSION: These findings suggest that moderately increasing protein O-GlcNAcylation by using low doses of OGA inhibitor may be a suitable therapeutic strategy for sAD.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , Proteínas tau/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cognición/fisiología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Ratones , Fosforilación/efectos de los fármacos , Reconocimiento en Psicología/fisiología
16.
J Alzheimers Dis ; 75(4): 1339-1349, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32417779

RESUMEN

BACKGROUND: Evidence from clinical studies and basic research has shown a strong correlation between Alzheimer's disease (AD) and type 2 diabetes. Tau, a neuronal microtubule-associated protein, is hyperphosphorylated and aggregated into neurofibrillary tangles in the AD brain. However, the expression of tau in pancreas is under debate. OBJECTIVE: We determined the expression of tau in mouse pancreas. METHODS: We used western blots, immunoprecipitation, and immunohistochemical staining to analyze pancreatic expression of tau in mice. RESULTS: We found that neither total tau nor phosphorylated tau was detectable in the mouse pancreas by western blots. Immunostaining with pan tau antibodies R134d and Tau-5 revealed bright and dense varicosities in the pancreatic islets and the exocrine pancreas. These varicosities were immunoreactive to synapsin 1, a presynaptic marker which can outline autonomic nerve profiles in pancreas, exhibiting complete colocalization with tau. Importantly, endocrine cells in islets did not exhibit specific immunoreactivity to any of pan tau antibodies tested, nor did the exocrine cells. CONCLUSION: In the mouse pancreas, we found that tau is exclusively expressed in autonomic nerve fibers, but there is no detectable expression in endocrine cells in the islet.


Asunto(s)
Vías Autónomas/metabolismo , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Proteínas tau/metabolismo , Animales , Islotes Pancreáticos/inervación , Ratones Endogámicos C57BL , Ratones Transgénicos , Páncreas/inervación , Fosforilación
17.
J Alzheimers Dis ; 75(4): 1377-1390, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32417781

RESUMEN

BACKGROUND: Abnormally hyperphosphorylated tau is the major protein of neurofibrillary tangles in Alzheimer's disease. Insulin activates PI3K-AKT signaling and regulates tau phosphorylation. Impaired brain insulin signaling is involved in Alzheimer's disease pathogenesis. However, the effect of peripheral insulin on tau phosphorylation is controversial. OBJECTIVE: In the present study, we determined the effect of peripheral insulin administration on tau phosphorylation in brain. METHODS: We intraperitoneally injected a super physiological dose of insulin to mice and analyzed PI3K-AKT signaling and tau phosphorylation in brains by western blots. RESULTS: We found that peripherally administered insulin activated the PI3K-AKT signaling pathway immediately in the liver, but not in the brain. Tau phosphorylation in the mouse brain was found to be first decreased (15 min) and then increased (30 min and 60 min) after peripheral insulin administration and these changes correlated inversely with body temperature and the level of brain protein O-GlcNAcylation. Maintaining body temperature of mice post peripheral insulin administration prevented the insulin/hypoglycemia-induced tau hyperphosphorylation after peripheral insulin administration. CONCLUSION: These findings suggest that peripheral insulin can induce tau hyperphosphorylation through both hypothermia and downregulation of brain protein O-GlcNAcylation during hypoglycemia.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Insulina/administración & dosificación , Proteínas tau/metabolismo , Animales , Temperatura Corporal/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
18.
J Alzheimers Dis ; 74(3): 851-864, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32116254

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common cause of dementia. Studies indicate that neuroinflammation plays an important role in the pathophysiology of AD. High-mobility group box 1 (HMGB1) is an important chromatin protein. It can be secreted by immune cells and passively released from damaged cells to promote inflammation. HMGB1 also can recruit stem cells and promote their proliferation and tissue repairing. However, the role of HMGB1 in the progression of AD is currently unknown. OBJECTIVE: The aims were to investigate the effect of HMGB1 on the AD-related pathologies and cognitive function using 3×Tg-AD mouse model. METHODS: Female 5-month-old 3×Tg-AD mice were intracerebroventricularly injected with 4.5 µg of HMGB1 or with saline as a control. The levels of interesting protein were assessed by western blots or immunofluorescence. The effect of HMGB1 on the cognitive function was evaluated by one-trial novel object recognition test and Morris water maze. RESULTS: Intracerebroventricular injection of recombinant HMGB1 ameliorated cognitive impairment in 5-6-month-old 3×Tg-AD mice. The levels of synapsin 1, synaptophysin, MAP2, NeuN, and phosphorylated CREB were increased in HMGB1-treated 3×Tg-AD mouse brains. HMGB1 decreased intracellular amyloid-ß level but did not affect tau phosphorylation. HMGB1 treatment also promoted neurogenesis in the dentate gyrus and increased the level of GFAP in the 3×Tg-AD mouse brains. CONCLUSION: These results reveal a novel function of HMGB1 in enhancing neuroplasticity and improving cognitive function in 3×Tg-AD mice.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Disfunción Cognitiva/prevención & control , Proteína HMGB1/uso terapéutico , Nootrópicos/uso terapéutico , Péptidos beta-Amiloides/metabolismo , Animales , Química Encefálica/efectos de los fármacos , Cognición , Disfunción Cognitiva/psicología , Femenino , Proteína HMGB1/administración & dosificación , Humanos , Inyecciones Intraventriculares , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Nootrópicos/administración & dosificación , Fosforilación , Reconocimiento en Psicología/efectos de los fármacos , Proteínas Recombinantes , Proteínas tau/metabolismo
19.
PLoS One ; 15(4): e0231090, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32240260

RESUMEN

Sevoflurane and isoflurane are among the most commonly used general anesthetics for children including infants, but their impact on metabolism, especially on blood glucose level, in children is not well understood. We investigated the impacts of anesthesia of neonatal (7-8 days old) and adult (2-3 months old) mice with the inhalational anesthetics 2.5% sevoflurane or 1.5% isoflurane, or the injectable anesthetics propofol (150 mg/kg) or avertin (375 mg/kg), for up to 6 hours. We found that sevoflurane and isoflurane induced severe hypoglycemia in neonatal mice and that this phenomenon was specific to the inhalational anesthetics because the injectable anesthetics propofol and avertin did not induce hypoglycemia. Surprisingly, the inhalational anesthesia induced hyperglycemia instead in adult mice. We also demonstrated that the inhalational anesthesia-induced hypoglycemia was a major cause of death for the neonatal mice receiving intranasal administration of saline prior to anesthesia. These studies revealed severe hypoglycemia in neonatal mice during anesthesia with sevoflurane or isoflurane. If this phenomenon also occurs in human, our findings would warrant closely monitoring blood glucose level and maintaining it in the normal range in infants receiving inhalational anesthesia.


Asunto(s)
Hipoglucemia/inducido químicamente , Isoflurano/efectos adversos , Sevoflurano/efectos adversos , Periodo de Recuperación de la Anestesia , Anestesia por Inhalación , Anestésicos por Inhalación/efectos adversos , Anestésicos Intravenosos/efectos adversos , Animales , Animales Recién Nacidos , Glucemia/metabolismo , Etanol/análogos & derivados , Etanol/farmacología , Hipoglucemia/sangre , Insulina/sangre , Ratones Endogámicos C57BL , Propofol/farmacología
20.
Alzheimers Res Ther ; 12(1): 102, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32854771

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder of middle-aged to old individuals. The pathophysiological process of AD is believed to begin many years before the emergence of clinical symptoms. The important influence of congenital genetic aberrations on the development of AD provides a novel opportunity to initiate prenatal to early postnatal pharmacological treatment to address the role of this critical period of brain development in the disease. METHODS: We investigated for the first time the effect of oral treatment during prenatal to early postnatal development with a neurotrophic compound, P021 (Ac-DGGLAG-NH2), on neurobehavior and AD-like pathology in 3xTg-AD, a transgenic mouse model of AD. The transgenic and control wild-type female mice were treated from prenatal day 8 to postnatal day 21 with a custom-made diet containing P021 or a vehicle diet, followed by a standard diet. AD-type cognitive function and pathological features were studied during adulthood and old age. RESULTS: The P021 treatment rescued cognitive deficits at 4 months, reduced abnormal hyperphosphorylation and accumulation of tau at known major AD neurofibrillary pathology-associated sites, and decreased Aß plaque load at 22 months in 3xTg-AD mice. Prenatal to early postnatal treatment with P021 also ameliorated certain markers of postsynaptic deficits, including PSD-95 levels and CREB activity, and decreased one measure of neuroinflammation, GFAP level in the brain at 4 and 22 months in 3xTg mice. CONCLUSIONS: These findings suggest that neurotrophic impairment during early development can be one of the etiopathogenic factors of AD and that the neurotrophic peptide mimetic is a potential early prevention strategy for this disease.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide , Embarazo , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA