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












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 3518, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568697

RESUMEN

Lipid peroxidation-initiated ferroptosis is an iron-dependent mechanism of programmed cell death taking place in neurological diseases. Here we show that a condensed benzo[b]thiazine derivative small molecule with an arylthiazine backbone (ADA-409-052) inhibits tert-Butyl hydroperoxide (TBHP)-induced lipid peroxidation (LP) and protects against ferroptotic cell death triggered by glutathione (GSH) depletion or glutathione peroxidase 4 (GPx4) inhibition in neuronal cell lines. In addition, ADA-409-052 suppresses pro-inflammatory activation of BV2 microglia and protects N2a neuronal cells from cell death induced by pro-inflammatory RAW 264.7 macrophages. Moreover, ADA-409-052 efficiently reduces infarct volume, edema and expression of pro-inflammatory genes in a mouse model of thromboembolic stroke. Targeting ferroptosis may be a promising therapeutic strategy in neurological diseases involving severe neuronal death and neuroinflammation.


Asunto(s)
Muerte Celular/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/fisiología , Ferroptosis/fisiología , Glutatión/metabolismo , Hierro/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuroprotección/efectos de los fármacos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/farmacología
2.
Aging Dis ; 5(2): 76-87, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24729933

RESUMEN

Cerebral ischemia is a risk factor for Alzheimer's disease (AD). Moreover, recent evidence indicates that it is a two-way street as the incidence rate of stroke is significantly higher in AD patients than those without the disease. Here we investigated the interaction of ischemic brain insults and AD in 9-month-old ApdE9 mice, which show full-blown accumulation of Aß deposits and microgliosis in the brain. Permanent occlusion of the middle cerebral artery (pMCAo) resulted in 36% larger infarct in ApdE9 mice compared to their wild-type (wt) controls. This was not due to differences in endothelium-dependent vascular reactivity. Treatment with human intravenous immunoglobulin (IVIG) reduced the infarct volumes and abolished the increased vulnerability of ApdE9 mice to pMCAo induced brain ischemia. When the mice were exposed to global brain ischemia (GI), an insult of hippocampal cells, ApdE9 mice showed increased neuronal loss in CA2 and CA3 subregions compared to their wt controls. GI was associated with increased microgliosis, astrogliosis, infiltration of blood-derived monocytic cells, and neurogenesis without clear differences between the genotypes. IVIG treatment prevented the GI-induced neuron loss in hippocampal CA1 and CA3 regions in ApdE9 mice. IVIG treatment increased microgliosis in wt but not in ApdE9 mice. Finally, GI induced 60% reduction in the hippocampal Aß burden in ApdE9 mice, which was not affected by IVIG treatment. The results indicate that the AD pathology with Aß deposits and microgliosis increases ischemic vulnerability in various brain areas. Moreover, IVIG treatment may be beneficial especially in patients suffering from both acute ischemic insult and AD.

3.
J Cell Mol Med ; 16(5): 1060-73, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21777378

RESUMEN

Accumulation of amyloid ß (Aß) is a major hallmark in Alzheimer's disease (AD). Bone marrow derived monocytic cells (BMM) have been shown to reduce Aß burden in mouse models of AD, alleviating the AD pathology. BMM have been shown to be more efficient phagocytes in AD than the endogenous brain microglia. Because BMM have a natural tendency to infiltrate into the injured area, they could be regarded as optimal candidates for cell-based therapy in AD. In this study, we describe a method to obtain monocytic cells from BM-derived haematopoietic stem cells (HSC). Mouse or human HSC were isolated and differentiated in the presence of macrophage colony stimulating factor (MCSF). The cells were characterized by assessing the expression profile of monocyte markers and cytokine response to inflammatory stimulus. The phagocytic capacity was determined with Aß uptake assay in vitro and Aß degradation assay of natively formed Aß deposits ex vivo and in a transgenic APdE9 mouse model of AD in vivo. HSC were lentivirally transduced with enhanced green fluorescent protein (eGFP) to determine the effect of gene modification on the potential of HSC-derived cells for therapeutic purposes. HSC-derived monocytic cells (HSCM) displayed inflammatory responses comparable to microglia and peripheral monocytes. We also show that HSCM contributed to Aß reduction and could be genetically modified without compromising their function. These monocytic cells could be obtained from human BM or mobilized peripheral blood HSC, indicating a potential therapeutic relevance for AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Células Madre Hematopoyéticas/fisiología , Monocitos/fisiología , Monocitos/trasplante , Péptidos beta-Amiloides/metabolismo , Animales , Separación Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de los fármacos , Fagocitosis/efectos de los fármacos
4.
Glia ; 59(11): 1643-57, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21826742

RESUMEN

Astrocytes and microglia are able to degrade potentially neurotoxic ß-amyloid (Aß) deposits typical for Alzheimer's disease (AD) pathology. Contrary to microglia, astrocytes degrade human Aß from tissue sections in vitro without any additional stimulation, but it has remained unclear whether transplanted astrocytes are able to clear deposited human Aß in vivo. We transplanted adult mouse astrocytes into the hippocampi of transgenic mice mimicking AD and observed their fate, effects on microglial responses, and Aß clearance. After 2-months follow-up time, we discovered a significant reduction in Aß burden compared with AD mice infused with PBS only. The remaining Aß deposits were fragmented and most of the Aß immunoreactivity was seen within the transplanted astrocytes. Concomitant to Aß reduction, both CD68 and CD45 immunoreactivities were significantly upregulated but phagocytic microglia were often surrounding and engulfing Aß burdened, TUNEL-positive astrocytes rather than co-localizing with Aß alone. Astrocytes are known to degrade Aß also by secreting proteases involved in Aß catabolism. To study the contribution of neprilysin (NEP), angiotensin-converting enzyme-1 (ACE-1), and endothelin-converting enzyme-2 (ECE-2) in human Aß clearance, we utilized an ex vivo assay to demonstrate that adult astrocytes respond to human Aß by upregulating NEP expression. Further, incubation of adult astrocytes with known inhibitors of NEP, ACE-1, or ECE-2 significantly inhibited the removal of human Aß from the tissue suggesting an important role for these proteases in Aß clearance by adult astrocytes ex vivo.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Astrocitos/trasplante , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Apoptosis/fisiología , Ácido Aspártico Endopeptidasas/metabolismo , Proteínas de Unión al Calcio , Proteínas de Unión al ADN/metabolismo , Enzimas Convertidoras de Endotelina , Femenino , Gliosis/patología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Antígenos Comunes de Leucocito/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Microglía/metabolismo , Neprilisina/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Inhibidores de Proteasas/farmacología
5.
J Neuroinflammation ; 7: 90, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21138577

RESUMEN

BACKGROUND: Purified intravenous immunoglobulin (IVIG) obtained from the plasma of healthy humans is indicated for the treatment of primary immunodeficiency disorders associated with defects in humoral immunity. IVIG contains naturally occurring auto-antibodies, including antibodies (Abs) against ß-amyloid (Aß) peptides accumulating in the brains of Alzheimer's disease (AD) patients. IVIG has been shown to alleviate AD pathology when studied with mildly affected AD patients. Although its mechanisms-of-action have been broadly studied, it remains unresolved how IVIG affects the removal of natively formed brain Aß deposits by primary astrocytes and microglia, two major cell types involved in the neuroinflammatory responses. METHODS: We first determined the effect of IVIG on Aß toxicity in primary neuronal cell culture. The mechanisms-of-action of IVIG in reduction of Aß burden was analyzed with ex vivo assay. We studied whether IVIG solubilizes natively formed Aß deposits from brain sections of APP/PS1 mice or promotes Aß removal by primary glial cells. We determined the role of lysosomal degradation pathway and Aß Abs in the IVIG-promoted reduction of Aß. Finally, we studied the penetration of IVIG into the brain parenchyma and interaction with brain deposits of human Aß in a mouse model of AD in vivo. RESULTS: IVIG was protective against Aß toxicity in a primary mouse hippocampal neuron culture. IVIG modestly inhibited the fibrillization of synthetic Aß1-42 but did not solubilize natively formed brain Aß deposits ex vivo. IVIG enhanced microglia-mediated Aß clearance ex vivo, with a mechanism linked to Aß Abs and lysosomal degradation. The IVIG-enhanced Aß clearance appears specific for microglia since IVIG did not affect Aß clearance by astrocytes. The cellular mechanisms of Aß clearance we observed have potential relevance in vivo since after peripheral administration IVIG penetrated to mouse brain tissue reaching highest concentrations in the hippocampus and bound selectively to Aß deposits in co-localization with microglia. CONCLUSIONS: Our results demonstrate that IVIG promotes recognition and removal of natively formed brain Aß deposits by primary microglia involving natural Aß Abs in IVIG. These findings may have therapeutic relevance in vivo as IVIG penetrates through the blood-brain barrier and specifically binds to Aß deposits in brain parenchyma.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/toxicidad , Inmunoglobulinas Intravenosas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Astrocitos/patología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Hipocampo/citología , Hipocampo/patología , Humanos , Inmunoglobulinas Intravenosas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Microglía/patología , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/inmunología , Placa Amiloide/patología , Presenilina-1/genética , Presenilina-1/metabolismo
6.
Glia ; 58(8): 889-900, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20155817

RESUMEN

Alzheimer's disease (AD) is a dementing neurodegenerative disorder without a cure. The abnormal parenchymal accumulation of beta-amyloid (Abeta) is associated with inflammatory reactions involving microglia and astrocytes. Increased levels of Abeta and Abeta deposition in the brain are thought to cause neuronal dysfunction and underlie dementia. Microglia, the brain resident cells of monocytic origin, have a potential ability to phagocytose Abeta but they also react to Abeta by increased production of proinflammatory toxic agents. Microglia originate from hemangioblastic mesoderm during early embryonic stages and from bone marrow (BM)-derived monocytic cells that home the brain throughout the neonatal stage of development. Recent studies indicate that BM or blood-derived monocytes are recruited to the diseased AD brain, associate with the Abeta depositions, and are more efficient phagocytes of Abeta compared with resident microglia. The clearance of Abeta deposition by these cells has been recently under intensive investigation and can occur through several different mechanisms. Importantly, peripheral monocytic cells of patients with AD appear to be deficient in clearing Abeta. This review will summarize the findings on the role of blood-derived cells in AD and discuss their therapeutic potential for treating patients suffering from this devastating disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Encéfalo , Monocitos/fisiología , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Humanos , Modelos Biológicos , Fagocitosis/inmunología
7.
Proc Natl Acad Sci U S A ; 106(38): 16505-10, 2009 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-19805328

RESUMEN

The amyloid hypothesis of Alzheimer's disease (AD) postulates that amyloid-beta (Abeta) deposition and neurotoxicity play a causative role in AD; oxidative injury is thought to be central in the pathogenesis. An endogenous defense system against oxidative stress is induced by binding of the transcription factor nuclear factor E2-related factor 2 (Nrf2) to the antioxidant response element (ARE) enhancer sequence. The Nrf2-ARE pathway is activated in response to reactive oxygen species to trigger the simultaneous expression of numerous protective enzymes and scavengers. To exploit the Nrf2-ARE pathway therapeutically, we delivered Nrf2 bilaterally into the hippocampus of 9-month-old transgenic AD mice (APP/PS1 mice) using a lentiviral vector encoding human Nrf2. The data indicate that significant reductions in spatial learning deficits of aged APP/PS1 mice in a Morris Water Maze can be achieved by modulating levels of Nrf2 in the brain. Memory improvement in APP/PS1 mice after Nrf2 transduction shifts the balance between soluble and insoluble Abeta toward an insoluble Abeta pool without concomitant change in total brain Abeta burden. Nrf2 gene transfer is associated with a robust reduction in astrocytic but not microglial activation and induction of Nrf2 target gene heme oxygenase 1, indicating overall activation of the Nrf2-ARE pathway in hippocampal neurons 6 months after injection. Results warrant further exploration of the Nrf2-ARE pathway for treatment of AD and suggest that the Nrf2-ARE pathway may represent a potential therapeutic strategy to pursue in AD in humans, particularly in view of the multiple mechanisms by which Nrf2 can exert its protective effects.


Asunto(s)
Enfermedad de Alzheimer/terapia , Terapia Genética/métodos , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Percepción Espacial/fisiología
8.
Glia ; 56(16): 1767-79, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18649403

RESUMEN

Bone marrow (BM)-derived monocytes contribute to the development of microglial reaction around beta-amyloid (Abeta) plaques in Alzheimer's disease (AD) and possibly clear Abeta. Therefore, it is of great importance to separate the proinflammatory actions of monocytic cells from Abeta phagocytic effects. We used minocycline (mino) to systemically downregulate microglial activation and studied proliferation, expression of markers for activated microglia, and Abeta removal in vitro and in vivo. Mino did not affect proliferation or phagocytic activity of BM-derived cells toward Abeta in vitro. Intrahippocampal LPS injection used to induce inflammation and increase recruitment of BM cells from periphery, reduced Abeta burden in BM-transplanted AD transgenic mice. All engrafted cells expressed CD45, approximately 50% expressed Iba-1, and <0.5% of these cells expressed CD3e. About 40% of the engrafted cells were mitotically active. LPS increased immunoreactivity for Iba-1, MHC II, a marker of antigen presenting cells, and CD68, a marker of lysosomal activity. The endogenous microglia largely contributed to these LPS-induced immunoreactivities. Mino reduced the engraftment of BM-derived cells and blocked the LPS-induced MHC II and Iba-1 immunoreactivities, but did not prevent the increased CD68-immunoreactivity or the reduced Abeta burden. Importantly, mino did not block the association of eGFP-positive cells with Abeta deposits and the percentage of mitotically active BM-derived cells. In conclusion, mino reduces overall inflammatory potential of BM-derived monocytic cells without preventing their phagocytic activity. The separation of harmful activation of microglia/monocytic cells from their Abeta clearing mechanism may hold important therapeutic potential.


Asunto(s)
Enfermedad de Alzheimer/terapia , Trasplante de Médula Ósea/métodos , Microglía/efectos de los fármacos , Minociclina/farmacología , Monocitos/efectos de los fármacos , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Antibacterianos/farmacología , Biomarcadores/análisis , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Encefalitis/tratamiento farmacológico , Encefalitis/fisiopatología , Femenino , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/fisiología , Humanos , Mediadores de Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Microglía/fisiología , Minociclina/uso terapéutico , Monocitos/metabolismo , Monocitos/trasplante , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología
9.
Glia ; 56(2): 154-63, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18004725

RESUMEN

Alzheimer's disease (AD) is one of the most devastating neurodegenerative disorders. The neuropathological hallmarks include extracellular senile plaques consisting of deposited beta-amyloid (Abeta) peptides and intraneuronal neurofibrillary tangles. Neuroinflammation and activation of astrocytes are also well-established features of AD neuropathology; however, the relationships between astrocytes and Abeta deposition remain unclear. Previous studies have shown that adult mouse astrocytes internalize and degrade Abeta deposits in brain sections prepared from human amyloid precursor protein (APP) transgenic mice. In the present study, we demonstrate that cultured adult, but not neonatal mouse astrocytes, respond morphologically and degrade Abeta deposits present in human AD brain. We also transplanted astrocytes isolated from enhanced green fluorescent protein expressing adult and neonatal mice into the hippocampi of human Abeta plaque-bearing transgenic APPSwe+PS1dE9 (APdE9) mice and their wild-type littermates and followed the migration and localization of these astrocytes by confocal microscopy upto 7 days after transplantation. Posttransplantation the astrocytes localized as aggregates or thin strings of many cells within the hippocampi of APdE9 and wild-type mice and showed limited migration from the injection site. Interestingly, most of the transplanted astrocytes were found near Abeta deposits in the hippocampi of APdE9 mice. In contrast to findings in ex vivo degradation assay, confocal microscopy revealed that both adult and neonatal transplanted astrocytes internalized human Abeta immunoreactive material in vivo. These results support the role of astrocytes as active Abeta clearing cells in the CNS that may have important implications for future development of therapeutic strategies for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/cirugía , Péptidos beta-Amiloides/metabolismo , Astrocitos/fisiología , Trasplante de Células/métodos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Recién Nacidos , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Hipocampo/metabolismo , Humanos , Imagenología Tridimensional , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal/métodos , Mutación , Presenilina-1/genética
10.
Nat Neurosci ; 10(4): 436-43, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17369826

RESUMEN

Excitotoxic neuronal death contributes to many neurological disorders, and involves calcium influx and stress-activated protein kinases (SAPKs) such as p38alpha. There is indirect evidence that the small Rho-family GTPases Rac and cdc42 are involved in neuronal death subsequent to the withdrawal of nerve growth factor (NGF), whereas Rho is involved in the inhibition of neurite regeneration and the release of the amyloidogenic Abeta(42) peptide. Here we show that Rho is activated in rat neurons by conditions that elevate intracellular calcium and in the mouse cerebral cortex during ischemia. Rho is required for the rapid glutamate-induced activation of p38alpha and ensuing neuronal death. The ability of RhoA to activate p38alpha was not expected, and it was specific to primary neuronal cultures. The expression of active RhoA alone not only activated p38alpha but also induced neuronal death that was sensitive to the anti-apoptotic protein Bcl-2, showing that RhoA was sufficient to induce the excitotoxic pathway. Therefore, Rho is an essential component of the excitotoxic cell death pathway.


Asunto(s)
Calcio/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Neuronas/fisiología , Proteína de Unión al GTP rhoA/fisiología , ADP Ribosa Transferasas/metabolismo , ADP Ribosa Transferasas/farmacología , Animales , Animales Recién Nacidos , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacología , Encéfalo/citología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Lateralidad Funcional , Ácido Glutámico/farmacología , Isquemia/metabolismo , Isquemia/patología , Ratones , Modelos Moleculares , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Transfección/métodos , Proteína de Unión al GTP cdc42/metabolismo
11.
Proc Natl Acad Sci U S A ; 103(23): 8852-7, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16723396

RESUMEN

beta-Amyloid (Abeta) polypeptide plays a critical role in the pathogenesis of Alzheimer's disease (AD), which is characterized by progressive decline of cognitive functions, formation of Abeta deposits and neurofibrillary tangles, and loss of neurons. Increased genetic production or direct intracerebral administration of Abeta in animal models results in Abeta deposition, gliosis, and impaired cognitive functions. Whether aging renders the brain prone to Abeta and whether inflammation is required for Abeta-induced learning deficits is unclear. We show that intraventricular infusion of Abeta1-42 results in learning deficits in 9-month-old but not 2.5-month-old mice. Deficits that become detectable 12 weeks after the infusion are associated with a slight reduction in Cu,Zn superoxide dismutase activity but do not correlate with Abeta deposition and are not associated with gliosis. In rats, Abeta infusion induced learning deficits that were detectable 6 months after the infusion. Approximately 20% of the Abeta immunoreactivity in rats was associated with astrocytes. NMR spectrum analysis of the animals cerebrospinal fluid revealed a strong reduction trend in several metabolites in Abeta-infused rats, including lactate and myo-inositol, supporting the idea of dysfunctional astrocytes. Even a subtle increase in brain Abeta1-42 concentration may disrupt normal metabolism of astrocytes, resulting in altered neuronal functions and age-related development of learning deficits independent of Abeta deposition and inflammation.


Asunto(s)
Envejecimiento/fisiología , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/farmacología , Discapacidades para el Aprendizaje/inducido químicamente , Aprendizaje por Laberinto/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , Animales , Encéfalo/citología , Encéfalo/enzimología , Encéfalo/patología , Inflamación/metabolismo , Infusiones Intravenosas , Discapacidades para el Aprendizaje/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Endogámicas SHR
12.
Brain Res Brain Res Rev ; 48(2): 240-50, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15850663

RESUMEN

Progressive memory impairment, beta-amyloid (Abeta) plaques associated with local inflammation, neurofibrillary tangles, and loss of neurons in selective brain areas are hallmarks of Alzheimer's disease (AD). Although beta-amyloid precursor protein (APP) and Abeta have a central role in the etiology of AD, it is not clear which forms of APP or Abeta are responsible for the neuronal vulnerability in AD brain. Brain ischemia, another cause of dementia in the elderly, has recently been recognized to contribute to the pathogenesis of AD and individuals with severe cognitive decline and possibly underlying AD are at increased risk for ischemic events in the brain. Moreover, the epsilon4 allele of apolipoprotein E (ApoE) is a risk factor for both AD and poor outcome following brain ischemia and hemorrhage. Several factors and molecular mechanisms that lower the threshold of neuronal death in models of AD have recently been described. Among these neuroinflammation seems to play an important role. The development and maturation of both AD neuropathology and ischemic lesions in the central nervous system are characterized by activation of glial cells and upregulation of inflammatory mediators. Indeed, anti-inflammatory approaches have proven to be beneficial in the prevention and treatment of AD-like neuropathology and ischemic injuries in vivo. This review summarizes some of the findings suggesting that neuronal overexpression of human APP renders the brain more vulnerable to ischemic injury and describes the factors that are involved in increased neuronal susceptibility to ischemic stroke.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Isquemia Encefálica/complicaciones , Inflamación/complicaciones , Neuronas/fisiología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Aspirina/metabolismo , Isquemia Encefálica/metabolismo , Humanos , Inflamación/metabolismo , Factores de Riesgo
13.
J Cereb Blood Flow Metab ; 25(4): 460-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15674236

RESUMEN

Minocycline is protective in models of transient middle cerebral artery occlusion (MCAO). We studied whether minocycline and doxycycline, another tetracycline derivative, provide protection in permanent MCAO. Because minocycline inhibits matrix metalloprotease-9 (MMP-9), we also compared minocycline's protective effect in wild type (wt) and MMP-9 knock-out (ko) mice. Wt FVB/N, Balb/C, and two lines of MMP-9 ko and their wt C57Bl/6 control mice were subjected to 24- or 72-hour permanent MCAO. Drug administration was started either 12 hours before or 2 hours after the onset of MCAO. Infarct size was determined by triphenyltetrazolium staining or T2-weighted MRI. Zymography was used to study the expression of MMPs. In wt strains, tetracycline treatments started before MCAO reduced the infarct size by 25% to 50%, whereas the treatment started after MCAO was not protective. Minocycline inhibited ischemia-provoked pro-MMP-9 induction in wt mice, but was not protective in MMP-9 ko mice. Pro-MMP-2 was induced by MCAO in wt and MMP-9 ko mice. MCAO-induced pro-MMP-2 was downregulated by minocycline treatment in wt mice but remained in MMP-9 ko mice at the same level as in saline-treated wt mice. Tetracyclines are protective in permanent MCAO when the treatment is started before the insult. Minocycline may provide protection by interfering with MMPs.


Asunto(s)
Antibacterianos/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Metaloproteinasa 9 de la Matriz/deficiencia , Minociclina/uso terapéutico , Animales , Isquemia Encefálica/patología , Corteza Cerebral/patología , Regulación hacia Abajo/efectos de los fármacos , Doxiciclina/uso terapéutico , Infarto de la Arteria Cerebral Media/patología , Masculino , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
14.
Neurobiol Dis ; 18(1): 134-42, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15649704

RESUMEN

The role of microglia recruited from bone marrow (BM) into the CNS during the progression of Alzheimer's disease (AD) is poorly understood. To investigate whether beta-amyloid (Abeta) associated microglia are derived from blood monocytes, we transplanted BM cells from enhanced green fluorescent protein expressing mice into young or old transgenic AD mice and determined the engraftment of BM-derived cells into the brain and their relative distribution near Abeta deposits. When young transgenic mice were transplanted before the onset of AD-like pathology and the brains analyzed 6.5 months later, the number of engrafted cells was significantly higher than in age-matched wild type mice. Moreover, the number of BM-derived cells associated with Abeta was significantly higher than in old transgenic mice transplanted after the establishment of AD-like pathology. Local inflammation caused by intrahippocampal lipopolysaccharide injection significantly increased the engraftment of BM-derived cells in old AD mice and decreased the hippocampal Abeta burden. These results suggest that infiltration of BM-derived monocytic cells into the brain contributes to the development of microglial reaction in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Células de la Médula Ósea/fisiología , Movimiento Celular/fisiología , Gliosis/fisiopatología , Microglía/fisiología , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Encefalitis/patología , Encefalitis/fisiopatología , Femenino , Gliosis/patología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Lipopolisacáridos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microglía/patología , Presenilina-1 , Proteínas Recombinantes de Fusión/genética
15.
Proc Natl Acad Sci U S A ; 102(4): 1211-6, 2005 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-15657137

RESUMEN

Apolipoprotein E (apoE) alleles are important genetic risk factors for Alzheimer's disease (AD), with the epsilon4 allele increasing and the epsilon2 allele decreasing risk for developing AD. ApoE has been shown to influence brain amyloid-beta peptide (Abeta) and amyloid burden, both in humans and in transgenic mice. Here we show that direct intracerebral administration of lentiviral vectors expressing the three common human apoE isoforms differentially alters hippocampal Abeta and amyloid burden in the PDAPP mouse model of AD. Expression of apoE4 in the absence of mouse apoE increases hippocampal Abeta(1-42) levels and amyloid burden. By contrast, expression of apoE2, even in the presence of mouse apoE, markedly reduces hippocampal Abeta burden. Our data demonstrate rapid apoE isoform-dependent effects on brain Abeta burden in a mouse model of AD. Gene delivery of apoE2 may prevent or reduce brain Abeta burden and the subsequent development of neuritic plaques.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Modelos Animales de Enfermedad , Terapia Genética , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteína E2 , Encéfalo/metabolismo , Hipocampo/metabolismo , Humanos , Lentivirus/genética , Ratones , Ratones Transgénicos
16.
Nat Med ; 10(7): 719-26, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15195085

RESUMEN

We have previously shown that apolipoprotein E (Apoe) promotes the formation of amyloid in brain and that astrocyte-specific expression of APOE markedly affects the deposition of amyloid-beta peptides (Abeta) in a mouse model of Alzheimer disease. Given the capacity of astrocytes to degrade Abeta, we investigated the potential role of Apoe in this astrocyte-mediated degradation. In contrast to cultured adult wild-type mouse astrocytes, adult Apoe(-/-) astrocytes do not degrade Abeta present in Abeta plaque-bearing brain sections in vitro. Coincubation with antibodies to either Apoe or Abeta, or with RAP, an antagonist of the low-density lipoprotein receptor family, effectively blocks Abeta degradation by astrocytes. Phase-contrast and confocal microscopy show that Apoe(-/-) astrocytes do not respond to or internalize Abeta deposits to the same extent as do wild-type astrocytes. Thus, Apoe seems to be important in the degradation and clearance of deposited Abeta species by astrocytes, a process that may be impaired in Alzheimer disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/fisiología , Astrocitos/metabolismo , Animales , Agregación Celular , Supervivencia Celular , Células Cultivadas , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Ratones , Ratones Endogámicos C57BL
17.
Stroke ; 35(4): 987-91, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14988572

RESUMEN

BACKGROUND AND PURPOSE: Activation of transcription factor nuclear factor-kappaB (NF-kappaB) may induce expression of either proinflammatory/apoptotic genes or antiapoptotic genes. Because a considerable number of middle cerebral artery occlusions (MCAOs) in humans are not associated with reperfusion during the first 24 hours, the role of NF-kappaB after permanent MCAO (pMCAO) was investigated. METHODS: Mice transgenic for a NF-kappaB-driven beta-globin reporter were exposed to pMCAO, and the expression of the reporter gene was quantified with real-time polymerase chain reaction. Mice lacking the p50 subunit of NF-kappaB and wild-type controls were exposed to pMCAO with or without treatment with pyrrolidinedithiocarbamate (PDTC), an NF-kappaB inhibitor. Brain sections of human stroke patients were immunostained for the activated NF-kappaB. RESULTS: pMCAO increased NF-kappaB transcriptional activity to 260% (36.9+/-4.5 compared with 14.4+/-2.6; n=10; P<0.01) in the brain; this NF-kappaB activation was completely blocked by PDTC (17.2+/-2.6; n=9; P<0.05). In p50-/- mice, pMCAO resulted in 41% (18+/-3.2 mm3; n=12) smaller infarcts compared with wild-type controls (32.9+/-3.8 mm3; n=9; P<0.05), which was comparable to the protection achieved with PDTC in wild-type mice (19.6+/-4.2 mm3; n=8). Pro-DTC, a PDTC analogue that does not cross the blood-brain barrier, had no effect, even though Pro-DTC and PDTC were equally protective in vitro. During the first 2 days of human stroke, NF-kappaB was activated in neurons in the penumbral areas. CONCLUSIONS: NF-kappaB is induced in neurons during human stroke, and activation of NF-kappaB in the brain may contribute to infarction in pMCAO.


Asunto(s)
Isquemia Encefálica/complicaciones , Infarto de la Arteria Cerebral Media/etiología , FN-kappa B/fisiología , Adulto , Anciano , Animales , Células Cultivadas , Globinas/genética , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Subunidad p50 de NF-kappa B , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Pirrolidinas/farmacología , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/metabolismo , Tiocarbamatos/farmacología
18.
Glia ; 40(2): 175-183, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12379905

RESUMEN

Although microglial cells are thought to play a beneficial role in the regeneration and plasticity of the central nervous system (CNS), recent studies have indicated that at least some molecules released by microglia may be harmful in acute brain insults and neurodegenerative diseases. Therefore, the pathways mediating the synthesis and release of these neurotoxic compounds are of importance. p38 and p44/42 families of mitogen-activated protein kinases (MAPKs) in microglia respond strongly to various extracellular stimuli, such as ATP, thrombin, and beta-amyloid, a peptide thought to be responsible for the neuropathology in Alzheimer's disease. In this review we describe in vivo evidence implicating that p38 and p44/42 MAPKs may play a critical role in harmful microglial activation in acute brain injury, such as stroke, and in more chronic neurodegenerative diseases, such as Alzheimer's disease. We also clarify the extracellular signals responsible for activation of p38 and p44/42 MAPK in microglia and review the responses so far reported to be mediated by these kinases.


Asunto(s)
Microglía/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Lesiones Encefálicas/enzimología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Lipopolisacáridos/farmacología , Proteína Quinasa 3 Activada por Mitógenos , Enfermedades Neurodegenerativas/enzimología , Accidente Cerebrovascular/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos
19.
Stroke ; 33(7): 1905-10, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12105373

RESUMEN

BACKGROUND AND PURPOSE: Epidemiological findings and experimental data on transgenic mice show that Alzheimer's disease-related changes render the brain more susceptible to ischemic damage. We studied whether the previously observed vulnerability in mice overexpressing the 751-amino-acid isoform of human amyloid precursor protein (APP751) is regulated by human apolipoprotein E (apoE) alleles, which determine the relative risk for Alzheimer's disease and the susceptibility to various forms of acute brain damage. METHODS: Aged apoE knock out (KO) mice, mice overexpressing APP751 in the apoE KO background and mice expressing either human apoE3 or apoE4 and APP751 in the apoE KO background were exposed to permanent occlusion of the middle cerebral artery (MCA). Infarct volumes were quantified from T2-weighted magnetic resonance images 24 hours after the MCA occlusion. Local cortical blood flow was monitored by laser Doppler flowmetry. Ischemia-induced microgliosis was detected by immunohistochemistry. RESULTS: Overexpression of human APP751 significantly increased the infarct volumes in apoE KO mice. Furthermore, this APP751-induced ischemic vulnerability was attenuated by the coexpression of either human apoE isoform. MCA occlusion resulted in a similar relative reduction in cortical blood flow in all mouse groups. Vascular anatomy showed no variation in the MCA territory between the groups. Instead, the expression of human apoE isoforms reduced the ischemia-induced microgliosis. CONCLUSIONS: Expression of either the human apoE3 or apoE4 isoform protects against the increased ischemic vulnerability observed in aged mice overexpressing human APP751, probably by modulating the inflammatory response induced by MCA occlusion.


Asunto(s)
Precursor de Proteína beta-Amiloide/farmacología , Apolipoproteínas E/biosíntesis , Isquemia Encefálica/genética , Regulación hacia Abajo/fisiología , Predisposición Genética a la Enfermedad , Envejecimiento/metabolismo , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Velocidad del Flujo Sanguíneo , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Circulación Cerebrovascular/genética , Modelos Animales de Enfermedad , Gliosis/patología , Humanos , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Flujometría por Láser-Doppler , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/farmacología
20.
Proc Natl Acad Sci U S A ; 99(3): 1610-5, 2002 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-11818564

RESUMEN

beta-amyloid (A beta), derived form the beta-amyloid precursor protein (APP), is important for the pathogenesis of Alzheimer's disease (AD), which is characterized by progressive decline of cognitive functions, formation of A beta plaques and neurofibrillary tangles, and loss of neurons. However, introducing a human wild-type or mutant APP gene to rodent models of AD does not result in clear neurodegeneration, suggesting that contributory factors lowering the threshold of neuronal death may be present in AD. Because brain ischemia has recently been recognized to contribute to the pathogenesis of AD, we studied the effect of focal brain ischemia in 8- and 20-month-old mice overexpressing the 751-amino acid isoform of human APP. We found that APP751 mice have higher activity of p38 mitogen-activated protein kinase (p38 MAPK) in microglia, the main immune effector cells within the brain, and increased vulnerability to brain ischemia when compared with age-matched wild-type mice. These characteristics are associated with enhanced microglial activation and inflammation but not with altered regulation of cerebral blood flow, as assessed by MRI and laser Doppler flowmetry. Suppression of inflammation with aspirin or inhibition of p38 MAPK with a selective inhibitor, SD-282, abolishes the increased neuronal vulnerability in APP751 transgenic mice. SD-282 also suppresses the expression of inducible nitric-oxide synthase and the binding activity of activator protein 1. These findings elucidate molecular mechanisms of neuronal injury in AD and suggest that antiinflammatory compounds preventing activation of p38 MAPK in microglia may reduce neuronal vulnerability in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Isquemia Encefálica/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Aspirina/farmacología , Presión Sanguínea , Encéfalo/patología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Dióxido de Carbono/sangre , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/farmacología , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Péptidos y Proteínas de Señalización Intracelular , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Microglía/patología , Microglía/fisiología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Oxígeno/sangre , Presión Parcial , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción AP-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...