Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39077965

RESUMEN

INTRODUCTION: The degeneration of cortical layers is associated with cognitive decline in Alzheimer's disease (AD). Current therapies for AD are not disease-modifying, and, despite substantial efforts, research and development for AD has faced formidable challenges. In addition, cellular senescence has emerged as a significant contributor to therapy resistance. METHODS: Human iPSC-derived cortical neurons were cultured on microelectrode arrays to measure long-term potentiation (LTP) noninvasively. Neurons were treated with pathogenic amyloid-ß (Aß) to analyze senescence and response to therapeutic molecules. RESULTS: Microphysiological recordings revealed Aß dampened cortical LTP activity and accelerated neuronal senescence. Aging neurons secreted inflammatory factors previously detected in brain, plasma, and cerebral spinal fluid of AD patients, in which drugs modulated senescence-related factors. DISCUSSION: This platform measures and records neuronal LTP activity in response to Aß and therapeutic molecules in real-time. Efficacy data from similar platforms have been accepted by the FDA for neurodegenerative diseases, expediting regulatory submissions. HIGHLIGHTS: This work developed a progerontic model of amyloid-ß (Aß)-driven cortical degeneration. This work measured neuronal LTP and correlated function with aging biomarkers. Aß is a driver of neuronal senescence and cortical degeneration. Molecules rescued neuronal function but did not halt Aß-driven senescence. Therapeutic molecules modulated secretion of inflammatory factors by aging neurons.

2.
Glia ; 71(2): 245-258, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36106533

RESUMEN

Fractalkine (FKN) is a membrane-bound chemokine that can be cleaved by proteases such as ADAM 10, ADAM 17, and cathepsin S to generate soluble fragments. Studies using different forms of the soluble FKN yield conflicting results in vivo. These observations prompted us to investigate the function and pharmacology of two commonly used isoforms of FKN, a human full-length soluble FKN (sFKN), and a human chemokine domain only FKN (cdFKN). Both are prevalent in the literature and are often assumed to be functionally equivalent. We observed that recombinant sFKN and cdFKN exhibit similar potencies in a cell-based cAMP assay, but binding affinity for CX3CR1 was modestly different. There was a 10-fold difference in potency between sFKN and cdFKN when assessing their ability to stimulate ß-arrestin recruitment. Interestingly, high concentrations of FKN, regardless of cleavage variant, were ineffective at reducing pro-inflammatory microglial activation and may induce a pro-inflammatory response. This effect was observed in mouse and rat primary microglial cells as well as microglial cell lines. The inflammatory response was exacerbated in aged microglia, which is known to exhibit age-related inflammatory phenotypes. We observed the same effects in Cx3cr1-/- primary microglia and therefore speculate that an alternative FKN receptor may exist. Collectively, these data provide greater insights into the function and pharmacology of these common FKN reagents, which may clarify conflicting reports and urge greater caution in the selection of FKN peptides for use in in vitro and in vivo studies and the interpretation of results obtained using these differing peptides.


Asunto(s)
Quimiocina CX3CL1 , Microglía , Ratones , Ratas , Humanos , Animales , Anciano , Quimiocina CX3CL1/metabolismo , Microglía/metabolismo , Proteolisis , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Línea Celular
3.
J Neurosci ; 35(44): 14842-60, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26538654

RESUMEN

Tau accumulation remains one of the closest correlates of neuronal loss in Alzheimer's disease. In addition, tau associates with several other neurodegenerative diseases, collectively known as tauopathies, in which clinical phenotypes manifest as cognitive impairment, behavioral disturbances, and motor impairment. Polyamines act as bivalent regulators of cellular function and are involved in numerous biological processes. The regulation of the polyamines system can become dysfunctional during disease states. Arginase 1 (Arg1) and nitric oxide synthases compete for l-arginine to produce either polyamines or nitric oxide, respectively. Herein, we show that overexpression of Arg1 using adeno-associated virus (AAV) in the CNS of rTg4510 tau transgenic mice significantly reduced phospho-tau species and tangle pathology. Sustained Arg1 overexpression decreased several kinases capable of phosphorylating tau, decreased inflammation, and modulated changes in the mammalian target of rapamycin and related proteins, suggesting activation of autophagy. Arg1 overexpression also mitigated hippocampal atrophy in tau transgenic mice. Conversely, conditional deletion of Arg1 in myeloid cells resulted in increased tau accumulation relative to Arg1-sufficient mice after transduction with a recombinant AAV-tau construct. These data suggest that Arg1 and the polyamine pathway may offer novel therapeutic targets for tauopathies.


Asunto(s)
Arginasa/biosíntesis , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Tauopatías/enzimología , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Arginasa/genética , Células HeLa , Hipocampo/enzimología , Hipocampo/patología , Humanos , Ratones , Ratones Transgénicos , Tauopatías/genética , Proteínas tau/genética
4.
Mol Ther ; 23(1): 17-23, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25195598

RESUMEN

In Parkinson's disease, α-synuclein is known to activate microglia and this activation has been proposed as one of the mechanisms of neurodegeneration. There are several signals produced by neurons that have an anti-inflammatory action on microglia, including CX3CL1 (fractalkine). We have shown that a soluble form of CX3CL1 is required to reduce neuron loss in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice and that fractalkine agonism can reduce neuron loss in a 6-hydroxydopamine lesion model. Here, we show that fractalkine can reduce α-synuclein-mediated neurodegeneration in rats. Rats that received fractalkine showed abrogated loss of tyrosine hydroxylase and Neu-N staining. This was replicated in animals where we expressed fractalkine from astrocytes with the glial fibrillary acid protein (GFAP) promoter. Interestingly, we did not observe a reduction in MHCII expression suggesting that soluble fractalkine is likely altering the microglial state to a more neuroprotective one rather than reducing antigen presentation.


Asunto(s)
Quimiocina CX3CL1/genética , Terapia Genética/métodos , Enfermedad de Parkinson Secundaria/terapia , Trastornos Parkinsonianos/terapia , alfa-Sinucleína/genética , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Presentación de Antígeno , Astrocitos/metabolismo , Astrocitos/patología , Quimiocina CX3CL1/agonistas , Quimiocina CX3CL1/metabolismo , Dependovirus/genética , Regulación de la Expresión Génica , Vectores Genéticos , Proteína Ácida Fibrilar de la Glía , Antígenos de Histocompatibilidad Clase II/genética , Masculino , Ratones , Microglía/metabolismo , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Oxidopamina , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/metabolismo , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Sustancia Negra/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/metabolismo
5.
Mol Cell Neurosci ; 67: 46-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26048366

RESUMEN

Calorie restriction (CR) has been shown to increase lifespan and delay aging phenotypes in many diverse eukaryotic species. In mouse models of Alzheimer's disease (AD), CR has been shown to decrease amyloid-beta and hyperphosphorylated tau levels and preserve cognitive function. Overexpression of human mutant tau protein has been shown to induce deficits in mitochondrial electron transport chain complex I activity. Therefore, experiments were performed to determine the effects of 4-month CR on brain mitochondrial function in Tg4510 mice, which express human P301L tau. Expression of mutant tau led to decreased ADP-stimulated respiratory rates, but not uncoupler-stimulated respiratory rates. The membrane potential was also slightly higher in mitochondria from the P301L tau mice. As shown previously, tau expression decreased mitochondrial complex I activity. The decreased complex I activity, decreased ADP-stimulated respiratory rate, and increased mitochondrial membrane potential occurring in mitochondria from Tg4510 mice were not restored by CR. However, the CR diet did result in a genotype independent decrease in mitochondrial F0F1-ATPase activity. This decrease in F0F1-ATPase activity was not due to lowered levels of the alpha or beta subunits of F0F1-ATPase. The possible mechanisms through which CR reduces the F0F1-ATPase activity in brain mitochondria are discussed.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Restricción Calórica , Mitocondrias/metabolismo , ATPasas de Translocación de Protón/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Animales , Encéfalo/metabolismo , Respiración de la Célula , Potencial de la Membrana Mitocondrial , Ratones , Proteínas tau/metabolismo
6.
J Neuroinflammation ; 11: 152, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25183004

RESUMEN

BACKGROUND: Abnormal tau hyperphosphorylation and its accumulation into intra-neuronal neurofibrillary tangles are linked to neurodegeneration in Alzheimer's disease and similar tauopathies. One strategy to reduce accumulation is through immunization, but the most immunogenic tau epitopes have so far remained unknown. To fill this gap, we immunized mice with recombinant tau to build a map of the most immunogenic tau epitopes. METHODS: Non-transgenic and rTg4510 tau transgenic mice aged 5 months were immunized with either human wild-type tau (Wt, 4R0N) or P301L tau (4R0N). Each protein was formulated in Quil A adjuvant. Sera and splenocytes of vaccinated mice were collected to assess the humoral and cellular immune responses to tau. We employed a peptide array assay to identify the most effective epitopes. Brain histology was utilized to measure the effects of vaccination on tau pathology and inflammation. RESULTS: Humoral immune responses following immunization demonstrated robust antibody titers (up to 1:80,000 endpoint titers) to each tau species in both mice models. The number of IFN-γ producing T cells and their proliferation were also increased in splenocytes from immunized mice, indicating an increased cellular immune response, and tau levels and neuroinflammation were both reduced. We identified five immunogenic motifs within either the N-terminal (9-15 and 21-27 amino acids), proline rich (168-174 and 220-228 amino acids), or the C-terminal regions (427-438 amino acids) of the wild-type and P301L tau protein sequence. CONCLUSIONS: Our study identifies five previously unknown immunogenic motifs of wild-type and mutated (P301L) tau protein. Immunization with both proteins resulted in reduced tau pathology and neuroinflammation in a tau transgenic model, supporting the efficacy of tau immunotherapy in tauopathy.


Asunto(s)
Mapeo Epitopo , Epítopos/inmunología , Tauopatías/inmunología , Tauopatías/terapia , Vacunación/métodos , Proteínas tau/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos/sangre , Proliferación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/etiología , Encefalitis/inmunología , Encefalitis/terapia , Ensayo de Inmunoadsorción Enzimática , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Saponinas de Quillaja , Saponinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Tauopatías/complicaciones , Tauopatías/patología , Proteínas tau/genética
7.
Res Sq ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38826367

RESUMEN

Preclinical methods are needed for screening potential Alzheimer's disease (AD) therapeutics that recapitulate phenotypes found in the Mild Cognitive Impairment (MCI) stage or even before this stage of the disease. This would require a phenotypic system that reproduces cognitive deficits without significant neuronal cell death to mimic the clinical manifestations of AD during these stages. A potential functional parameter to be monitored is long-term potentiation (LTP), which is a correlate of learning and memory, that would be one of the first functions effected by AD onset. Mature human iPSC-derived cortical neurons and primary astrocytes were co-cultured on microelectrode arrays (MEA) where surface chemistry was utilized to create circuit patterns connecting two adjacent electrodes to model LTP function. LTP maintenance was significantly reduced in the presence of Amyloid-Beta 42 (Aß42) oligomers compared to the controls, however, co-treatment with AD therapeutics (Donepezil, Memantine, Rolipram and Saracatinib) corrected Aß42 induced LTP impairment. The results presented here illustrate the significance of the system as a validated platform that can be utilized to model and study MCI AD pathology, and potentially for the pre-MCI phase before the occurrence of significant cell death. It also has the potential to become an ideal platform for high content therapeutic screening for other neurodegenerative diseases.

8.
J Neuroinflammation ; 10: 86, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23866683

RESUMEN

BACKGROUND: The chemokine (C-C motif) ligand 2 (CCL2) is a monocyte chemoattractant protein that mediates macrophage recruitment and migration during peripheral and central nervous system (CNS) inflammation. METHODS: To determine the impact of CCL2 in inflammation in vivo and to elucidate the CCL2-induced polarization of activated brain microglia, we delivered CCL2 into the brains of wild-type mice via recombinant adeno-associated virus serotype 9 (rAAV-9) driven by the chicken ß-actin promoter. We measured microglial activation using histological and chemical measurement and recruitment of monocytes using histology and flow cytometry. RESULTS: The overexpression of CCL2 in the CNS induced significant activation of brain resident microglia. CD45 and major histocompatibility complex class II immunoreactivity significantly increased at the sites of CCL2 administration. Histological characterization of the microglial phenotype revealed the elevation of "classically activated" microglial markers, such as calgranulin B and IL-1ß, as well as markers associated with "alternative activation" of microglia, including YM1 and arginase 1. The protein expression profile in the hippocampus demonstrated markedly increased levels of IL-6, GM-CSF and eotaxin (CCL-11) in response to CCL2, but no changes in the levels of other cytokines, including TNF-α and IFN-γ. Moreover, real-time PCR analysis confirmed increases in mRNA levels of gene transcripts associated with neuroinflammation following CCL2 overexpression. Finally, we investigated the chemotactic properties of CCL2 in vivo by performing adoptive transfer of bone marrow-derived cells (BMDCs) isolated from donor mice that ubiquitously expressed green fluorescent protein. Flow cytometry and histological analyses indicated that BMDCs extravasated into brain parenchyma and colabeled with microglial markers. CONCLUSION: Taken together, our results suggest that CCL2 strongly activates resident microglia in the brain. Both pro- and anti-inflammatory activation of microglia were prominent, with no bias toward the M1 or M2 phenotype in the activated cells. As expected, CCL2 overexpression actively recruited circulating monocytes into the CNS. Thus, CCL2 expression in mouse brain induces microglial activation and represents an efficient method for recruitment of peripheral macrophages.


Asunto(s)
Química Encefálica/fisiología , Quimiocina CCL2/fisiología , Activación de Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Traslado Adoptivo , Animales , Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/biosíntesis , Citocinas/biosíntesis , Dependovirus/genética , Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos
9.
Neurodegener Dis ; 11(4): 165-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22796753

RESUMEN

BACKGROUND: We aimed to investigate the influence of oligomeric forms of ß-amyloid (Aß) and the influence of the duration of exposure on the development of tau phosphorylation. METHODS: Aß oligomers were injected intracranially either acutely into 5-month-old rTg4510 mice and tissue was collected 3 days later, or chronically into 3-month-old mice and tissue was collected 2 months later. Several forms of phosphorylated tau (p-tau), GSK3 (glycogen synthase kinase-3) and microglial and astrocyte activation were measured. RESULTS: Acute injections of Aß oligomers had no effect on p-tau epitopes but did result in elevation of phosphorylated/activated GSK3 (pGSK3). Chronic infusion of Aß oligomers into the right hippocampus resulted in 3- to 4-fold elevations in several p-tau isoforms with no changes in total tau levels. A significant elevation in pGSK3 accompanied these changes. Microglial staining with CD68 paralleled the increase in tau phosphorylation, however, CD45 staining was unaffected by Aß. Control experiments revealed that the infusion of Aß from the minipumps was largely complete by 10 days after implantation. Thus, the elevation in p-tau 2 months after implantation implies that the changes are quite persistent. CONCLUSION: Soluble Aß(1-42) oligomers have long-lasting effects on tau phosphorylation in the rTg4510 model, possibly due to elevations in GSK3. These data suggest that even brief elevations in Aß production, may have enduring impact on the risk for tauopathy.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Hipocampo/efectos de los fármacos , Microglía/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Tauopatías/metabolismo
10.
Geroscience ; 44(1): 173-194, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34410588

RESUMEN

C-terminal cleaved tau at D421 (∆D421-tau) accumulates in the brains of Alzheimer's disease (AD) patients. However, it is unclear how tau truncation, an understudied tau post-translational modification, contributes to AD pathology and progression. Utilizing an adeno-associated virus (AAV) gene delivery-based approach, we overexpressed full-length tau (FL-tau) and ∆D421-tau in 4- and 12-month-old mice for 4 months to study the neuropathological impact of accumulation in young adult (8-month) and middle-aged (16-month) mice. Overall, we show that independent of the tau species, age was an important factor facilitating tau phosphorylation, oligomer formation, and deposition into silver-positive tangles. However, mice overexpressing ∆D421-tau exhibited a distinct phosphorylation profile to those overexpressing FL-tau and increased tau oligomerization in the middle-age group. Importantly, overexpression of ∆D421-tau, but not FL-tau in middle-aged mice, resulted in pronounced cognitive impairments and hippocampal long-term potentiation deficits. While both FL-tau and ∆D421-tau induced neuronal loss in mice with age, ∆D421-tau led to significant neuronal loss in the CA3 area of the hippocampus and medial entorhinal cortex compared to FL-tau. Based on our data, we conclude that age increases the susceptibility to neuronal degeneration associated with ΔD421-tau accumulation. Our findings suggest that ΔD421-tau accumulation contributes to synaptic plasticity and cognitive deficits, thus representing a potential target for tau-associated pathologies.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/genética , Animales , Cognición , Disfunción Cognitiva/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal
11.
Stem Cell Reports ; 17(1): 96-109, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34942087

RESUMEN

The maturation and functional characteristics of human induced pluripotent stem cell (hiPSC)-cortical neurons has not been fully documented. This study developed a phenotypic model of hiPSC-derived cortical neurons, characterized their maturation process, and investigated its application for disease modeling with the integration of multi-electrode array (MEA) technology. Immunocytochemistry analysis indicated early-stage neurons (day 21) were simultaneously positive for both excitatory (vesicular glutamate transporter 1 [VGlut1]) and inhibitory (GABA) markers, while late-stage cultures (day 40) expressed solely VGlut1, indicating a purely excitatory phenotype without containing glial cells. This maturation process was further validated utilizing patch clamp and MEA analysis. Particularly, induced long-term potentiation (LTP) successfully persisted for 1 h in day 40 cultures, but only achieved LTP in the presence of the GABAA receptor antagonist picrotoxin in day 21 cultures. This system was also applied to epilepsy modeling utilizing bicuculline and its correction utilizing the anti-epileptic drug valproic acid.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Potenciales de Acción , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Humanos , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/terapia , Sinapsis/metabolismo
12.
J Neurosci ; 30(29): 9651-8, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20660248

RESUMEN

A major question for gene therapy in brain concerns methods to administer therapeutic genes in a uniform manner over major portions of the brain. A second question in neuroimmunology concerns the extent to which monocytes migrate to the CNS in degenerative disorders. Here we show that CD11b+ cells (largely monocytes) isolated from the bone marrow of GFP (green fluorescent protein)-expressing donors spontaneously home to compacted amyloid plaques in the brain. Injections of these cells as a single pulse show a rapid clearance from circulation (90 min half-life) and tissue residence half-lives of approximately 3 d. The uptake into brain was minimal in nontransgenic mice. In transgenic mice containing amyloid deposits, uptake was dramatically increased and associated with a corresponding decrease in monocyte uptake into peripheral organs compared to nontransgenic littermates. Twice weekly infusions of the CD11b+ bone marrow cells transfected with a genetically engineered form of the protease neprilysin completely arrest amyloid deposition in an aggressively depositing transgenic model. Exploiting the natural homing properties of peripherally derived blood cells to deliver therapeutic genes has the advantages of access to the entire CNS, expression largely restricted to sites of injury, low risk of immune reactivity, and fading of expression if adverse reactions are encountered. These observations support the feasibility of testing autologous monocytes for application of therapeutic genes in human CNS disease. Moreover, these data support the results from bone marrow grafts that circulating CD11b+ cells can enter the CNS without requiring the use of lethal irradiation.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Amiloide/química , Antígeno CD11b/administración & dosificación , Terapia Genética/métodos , Monocitos/trasplante , ATPasas Asociadas con Actividades Celulares Diversas , Enfermedad de Alzheimer/enzimología , Animales , Biomarcadores/análisis , Encéfalo/enzimología , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/análisis , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Monocitos/citología , Neprilisina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo
13.
J Cell Mol Med ; 15(2): 327-38, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19900216

RESUMEN

Mutations in the presenilin-1 (PS1) gene are independent causes of familial Alzheimer's disease (AD). AD patients have dysregulated immunity, and PS1 mutant mice exhibit abnormal systemic immune responses. To test whether immune function abnormality caused by a mutant human PS1 gene (mhPS1) could modify AD-like pathology, we reconstituted immune systems of AD model mice carrying a mutant human amyloid precursor protein gene (mhAPP; Tg2576 mice) or both mhAPP and mhPS1 genes (PSAPP mice) with allogeneic bone marrow cells. Here, we report a marked reduction in amyloid-ß (Aß) levels, ß-amyloid plaques and brain inflammatory responses in PSAPP mice following strain-matched wild-type PS1 bone marrow reconstitution. These effects occurred with immune switching from pro-inflammatory T helper (Th) 1 to anti-inflammatory Th2 immune responses in the periphery and in the brain, which likely instructed microglia to phagocytose and clear Aß in an ex vivo assay. Conversely, Tg2576 mice displayed accelerated AD-like pathology when reconstituted with mhPS1 bone marrow. These data show that haematopoietic cells bearing the mhPS1 transgene exacerbate AD-like pathology, suggesting a novel therapeutic strategy for AD based on targeting PS1 in peripheral immune cells.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/inmunología , Presenilina-1/genética , Presenilina-1/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Enfermedad de Alzheimer/inmunología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Antígeno CD11b/biosíntesis , Citocinas/biosíntesis , Ratones , Ratones Transgénicos , Proteínas Mutantes/metabolismo , Mutación , Placa Amiloide
14.
J Neuroinflammation ; 8: 115, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21906275

RESUMEN

BACKGROUND: Anti-Aß immunotherapy is a promising approach to the prevention and treatment of Alzheimer's disease (AD) currently in clinical trials. There is extensive evidence, both in mice and humans that a significant adverse event is the occurrence of microhemorrhages. Also, vasogenic edema was reported in phase 2 of a passive immunization clinical trial. In order to overcome these vascular adverse effects it is critical that we understand the mechanism(s) by which they occur. METHODS: We have examined the matrix metalloproteinase (MMP) protein degradation system in two previously published anti-Aß immunotherapy studies. The first was a passive immunization study in which we examined 22 month old APPSw mice that had received anti-Aß antibodies for 1, 2 or 3 months. The second is an active vaccination study in which we examined 16 month old APPSw/NOS2-/- mice treated with Aß vaccination for 4 months. RESULTS: There is a significant activation of the MMP2 and MMP9 proteinase degradation systems by anti-Aß immunotherapy, regardless of whether this is delivered through active vaccination or passive immunization. We have characterized this activation by gene expression, protein expression and zymography assessment of MMP activity. CONCLUSIONS: Since the MMP2 and MMP9 systems are heavily implicated in the pathophysiology of intracerbral hemorrhage, these data may provide a potential mechanism of microhemorrhage due to immunotherapy. Increased activity of the MMP system, therefore, is likely to be a major factor in increased microhemorrhage occurrence.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/uso terapéutico , Hemorragia Cerebral , Inmunoterapia/métodos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos adversos , Péptidos beta-Amiloides/inmunología , Animales , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/inmunología , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones , Ratones Transgénicos , Microcirculación , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo
15.
Proc Natl Acad Sci U S A ; 105(9): 3622-7, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18292230

RESUMEN

A hallmark of the pathology of Alzheimer's disease is the accumulation of the microtubule-associated protein tau into fibrillar aggregates. Recent studies suggest that they accumulate because cytosolic chaperones fail to clear abnormally phosphorylated tau, preserving a pool of toxic tau intermediates within the neuron. We describe a mechanism for tau clearance involving a major cellular kinase, Akt. During stress, Akt is ubiquitinated and degraded by the tau ubiquitin ligase CHIP, and this largely depends on the Hsp90 complex. Akt also prevents CHIP-induced tau ubiquitination and its subsequent degradation, either by regulating the Hsp90/CHIP complex directly or by competing as a client protein with tau for binding. Akt levels tightly regulate the expression of CHIP, such that, as Akt levels are suppressed, CHIP levels also decrease, suggesting a potential stress response feedback mechanism between ligase and kinase activity. We also show that Akt and the microtubule affinity-regulating kinase 2 (PAR1/MARK2), a known tau kinase, interact directly. Akt enhances the activity of PAR1 to promote tau hyperphosphorylation at S262/S356, a tau species that is not recognized by the CHIP/Hsp90 complex. Moreover, Akt1 knockout mice have reduced levels of tau phosphorylated at PAR1/MARK2 consensus sites. Hence, Akt serves as a major regulator of tau biology by manipulating both tau kinases and protein quality control, providing a link to several common pathways that have demonstrated dysfunction in Alzheimer's disease.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/deficiencia , Ubiquitina-Proteína Ligasas/deficiencia
16.
Front Immunol ; 12: 628156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34046031

RESUMEN

Brain myeloid cells, include infiltrating macrophages and resident microglia, play an essential role in responding to and inducing neurodegenerative diseases, such as Alzheimer's disease (AD). Genome-wide association studies (GWAS) implicate many AD casual and risk genes enriched in brain myeloid cells. Coordinated arginine metabolism through arginase 1 (Arg1) is critical for brain myeloid cells to perform biological functions, whereas dysregulated arginine metabolism disrupts them. Altered arginine metabolism is proposed as a new biomarker pathway for AD. We previously reported Arg1 deficiency in myeloid biased cells using lysozyme M (LysM) promoter-driven deletion worsened amyloidosis-related neuropathology and behavioral impairment. However, it remains unclear how Arg1 deficiency in these cells impacts the whole brain to promote amyloidosis. Herein, we aim to determine how Arg1 deficiency driven by LysM restriction during amyloidosis affects fundamental neurodegenerative pathways at the transcriptome level. By applying several bioinformatic tools and analyses, we found that amyloid-ß (Aß) stimulated transcriptomic signatures in autophagy-related pathways and myeloid cells' inflammatory response. At the same time, myeloid Arg1 deficiency during amyloidosis promoted gene signatures of lipid metabolism, myelination, and migration of myeloid cells. Focusing on Aß associated glial transcriptomic signatures, we found myeloid Arg1 deficiency up-regulated glial gene transcripts that positively correlated with Aß plaque burden. We also observed that Aß preferentially activated disease-associated microglial signatures to increase phagocytic response, whereas myeloid Arg1 deficiency selectively promoted homeostatic microglial signature that is non-phagocytic. These transcriptomic findings suggest a critical role for proper Arg1 function during normal and pathological challenges associated with amyloidosis. Furthermore, understanding pathways that govern Arg1 metabolism may provide new therapeutic opportunities to rebalance immune function and improve microglia/macrophage fitness.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Arginasa/metabolismo , Encéfalo/enzimología , Perfilación de la Expresión Génica , Microglía/enzimología , Células Mieloides/enzimología , Degeneración Nerviosa , Transcriptoma , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Arginasa/genética , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Redes Reguladoras de Genes , Haploinsuficiencia , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Mutación , Células Mieloides/patología
17.
J Neurochem ; 113(3): 784-95, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20180882

RESUMEN

Aggregation of amyloid-beta (Abeta) in the forebrain of Alzheimer's disease (AD) subjects may disturb the molecular organization of the extracellular microenvironment that modulates neural and synaptic plasticity. Proteoglycans are major components of this extracellular environment. To test the hypothesis that Abeta, or another amyloid precursor protein (APP) dependent mechanism modifies the accumulation and/or turnover of extracellular proteoglycans, we examined whether the expression and processing of brevican, an abundant extracellular, chondroitin sulfate (CS)-bearing proteoglycan, were altered in brains of Abeta-depositing transgenic mice (APPsw - APP gene bearing the Swedish mutation) as a model of AD. The molecular size of CS chains attached to brevican was smaller in hippocampal tissue from APPsw mice bearing Abeta deposits compared to non-transgenic mice, likely because of changes in the CS chains. Also, the abundance of the major proteolytic fragment of brevican was markedly diminished in extracts from several telencephalic regions of APPsw mice compared to non-transgenic mice, yet these immunoreactive fragments appeared to accumulate adjacent to the plaque edge. These results suggest that Abeta or APP exert inhibitory effects on proteolytic cleavage mechanisms responsible for synthesis and turnover of proteoglycans. As proteoglycans stabilize synaptic structure and inhibit molecular plasticity, defective brevican processing observed in Abeta-bearing mice and potentially end-stage human AD, may contribute to deficient neural plasticity.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Espacio Extracelular/metabolismo , Lectinas Tipo C/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Placa Amiloide/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Western Blotting , Química Encefálica/genética , Brevicano , Línea Celular , Sulfatos de Condroitina/biosíntesis , Medios de Cultivo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Transgénicos , Procesamiento Proteico-Postraduccional/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Neuroinflammation ; 7: 56, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20846376

RESUMEN

Inflammation and microglial activation are associated with Alzheimer's disease (AD) pathology. Somewhat surprisingly, injection of a prototypical inflammatory agent, lipopolysaccharide (LPS) into brains of amyloid precursor protein (APP) transgenic mice clears some of the pre-existing amyloid deposits. It is less well understood how brain inflammation modulates tau pathology in the absence of Aß. These studies examined the role of LPS-induced inflammation on tau pathology. We used transgenic rTg4510 mice, which express the P301L mutation (4R0N TauP301L) and initiate tau pathology between 3-5 months of age. First, we found an age-dependent increase in several markers of microglial activation as these rTg4510 mice aged and tau tangles accumulated. LPS injections into the frontal cortex and hippocampus induced significant activation of CD45 and arginase 1 in rTg4510 and non-transgenic mice. In addition, activation of YM1 by LPS was exaggerated in transgenic mice relative to non-transgenic animals. Expression of Ser199/202 and phospho-tau Ser396 was increased in rTg4510 mice that received LPS compared to vehicle injections. However, the numbers of silver-positive neurons, implying presence of more pre- and mature tangles, was not significantly affected by LPS administration. These data suggest that inflammatory stimuli can facilitate tau phosphorylation. Coupled with prior results demonstrating clearance of Aß by similar LPS injections, these results suggest that brain inflammation may have opposing effects on amyloid and tau pathology, possibly explaining the failures (to date) of anti-inflammatory therapies in AD patients.


Asunto(s)
Encefalitis/patología , Lóbulo Frontal/patología , Hipocampo/patología , Microglía/patología , Neuronas/patología , Proteínas tau/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inmunohistoquímica , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Tinción con Nitrato de Plata
19.
J Healthc Inf Manag ; 24(1): 25-30, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20077922

RESUMEN

The core philosophy of health information exchanges is to bring together industry stakeholders to facilitate the movement of actionable healthcare information within or across organizations. One workable model for automating the HIE is the virtual health exchange. It allows an organization to satisfy its electronic data exchange needs, while positioning it for HIO/RHIO and Nationwide Health Information Network inclusion. Four healthcare organizations in the San Francisco bay area offer a unique case study of this emerging model and a distinctive technology strategy using HIPAA-compliant SaaS online connectivity. The article will explore the foundational elements such as platform connectivity solutions and services that meet the federal HIT Policy Committee's approved recommendation of "meaningful use" 2011 criteria for exchange health information. Several ambulatory solutions are integrated into the data and workflow. The Bay Area HIE consists of John Muir Health, a private health system; Hill Physicians Medical Group, an independent physicians' organization; Alta Bates Medical Group, a 600-physician IPA; San Ramon Regional Medical Center, a 123-bed, acute-care hospital; and University of California, San Francisco (UCSF), a large academic health system. Collectively, 2,800-plus physicians, 900,000 patients, numerous reference labs and interoperability among several health IT vendors are involved. Providers and hospitals are exchanging data and 136,000 patients have connected online to their records. Connected physicians have access to online services, e.g., results management, messaging, colleague-to-colleague messaging, referrals, and eprescribing. Patients are offered secure messaging, including preventive care reminders, PHRs, lab results, script renewals, bill payment, appointment requests, referrals and education services. Revealed are resulting care coordination and practice operational improvements.


Asunto(s)
Acceso a la Información , Atención a la Salud/organización & administración , Registro Médico Coordinado , American Recovery and Reinvestment Act , Estudios de Casos Organizacionales , San Francisco , Estados Unidos
20.
Alzheimers Res Ther ; 12(1): 69, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32498691

RESUMEN

Increasing evidence suggests that infection with Sars-CoV-2 causes neurological deficits in a substantial proportion of affected patients. While these symptoms arise acutely during the course of infection, less is known about the possible long-term consequences for the brain. Severely affected COVID-19 cases experience high levels of proinflammatory cytokines and acute respiratory dysfunction and often require assisted ventilation. All these factors have been suggested to cause cognitive decline. Pathogenetically, this may result from direct negative effects of the immune reaction, acceleration or aggravation of pre-existing cognitive deficits, or de novo induction of a neurodegenerative disease. This article summarizes the current understanding of neurological symptoms of COVID-19 and hypothesizes that affected patients may be at higher risk of developing cognitive decline after overcoming the primary COVID-19 infection. A structured prospective evaluation should analyze the likelihood, time course, and severity of cognitive impairment following the COVID-19 pandemic.


Asunto(s)
Betacoronavirus , Encéfalo/virología , Infecciones por Coronavirus/complicaciones , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/virología , Neumonía Viral/complicaciones , Animales , Encéfalo/patología , COVID-19 , Infecciones por Coronavirus/patología , Humanos , Enfermedades Neurodegenerativas/patología , Pandemias , Neumonía Viral/patología , SARS-CoV-2 , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA