Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Intervalo de año de publicación
1.
Front Neuroanat ; 18: 1339308, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601797

RESUMEN

The house wren shows complex song, and the rufous-tailed hummingbird has a simple song. The location of vocal brain areas supports the song's complexity; however, these still need to be studied. The astrocytic population in songbirds appears to be associated with change in vocal control nuclei; however, astrocytic distribution and morphology have not been described in these species. Consequently, we compared the distribution and volume of the vocal brain areas: HVC, RA, Area X, and LMAN, cell density, and the morphology of astrocytes in the house wren and the rufous-tailed hummingbird. Individuals of the two species were collected, and their brains were analyzed using serial Nissl- NeuN- and MAP2-stained tissue scanner imaging, followed by 3D reconstructions of the vocal areas; and GFAP and S100ß astrocytes were analyzed in both species. We found that vocal areas were located close to the cerebral midline in the house wren and a more lateralized position in the rufous-tailed hummingbird. The LMAN occupied a larger volume in the rufous-tailed hummingbird, while the RA and HVC were larger in the house wren. While Area X showed higher cell density in the house wren than the rufous-tailed hummingbird, the LMAN showed a higher density in the rufous-tailed hummingbird. In the house wren, GFAP astrocytes in the same bregma where the vocal areas were located were observed at the laminar edge of the pallium (LEP) and in the vascular region, as well as in vocal motor relay regions in the pallidum and mesencephalon. In contrast, GFAP astrocytes were found in LEP, but not in the pallidum and mesencephalon in hummingbirds. Finally, when comparing GFAP astrocytes in the LEP region of both species, house wren astrocytes exhibited significantly more complex morphology than those of the rufous-tailed hummingbird. These findings suggest a difference in the location and cellular density of vocal circuits, as well as morphology of GFAP astrocytes between the house wren and the rufous-tailed hummingbird.

2.
Front Aging Neurosci ; 15: 1194203, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37744400

RESUMEN

Introduction: Proteolytic processing of amyloid protein precursor by ß-site secretase enzyme (BACE1) is dependent on the cellular lipid composition and is affected by endomembrane trafficking in dementia and Alzheimer's disease (AD). Stearoyl-CoA desaturase 1 (SCD1) is responsible for the synthesis of fatty acid monounsaturation (MUFAs), whose accumulation is strongly associated with cognitive dysfunction. Methods: In this study, we analyzed the relationship between BACE1 and SCD1 in vivo and in vitro neurodegenerative models and their association in familial AD (FAD), sporadic AD (SAD), and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) using microscopy, biochemical, and mass SPECT approach. Results: Our findings showed that BACE1 and SCD1 immunoreactivities were increased and colocalized in astrocytes of the hippocampus in a rat model of global cerebral ischemia (2-VO). A synergistic effect of double BACE1/SCD1 silencing on the recovery of motor and cognitive functions was obtained. This neuroprotective regulation involved the segregation of phospholipids (PLs) associated with polyunsaturated fatty acids in the hippocampus, cerebrospinal fluid, and serum. The double silencing in the sham and ischemic groups was stronger in the serum, inducing an inverse ratio between total phosphatydilcholine (PC) and lysophosphatidylcholine (LPC), represented mainly by the reduction of PC 38:4 and PC 36:4 and an increase in LPC 16:0 and LPC 18:0. Furthermore, PC 38:4 and PC:36:4 levels augmented in pathological conditions in in vitro AD models. BACE1 and SCD1 increases were confirmed in the hippocampus of FAD, SAD, and CADASIL. Conclusion: Therefore, the findings suggest a novel convergence of BACE-1 and SCD1 in neurodegeneration, related to pro-inflammatory phospholipids.

3.
Brain Pathol ; 33(2): e13119, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36130084

RESUMEN

In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aß. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/patología , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Homocigoto , Mutación , Encéfalo/patología , Péptidos beta-Amiloides/metabolismo
4.
Nutr Neurosci ; 25(6): 1159-1172, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33164710

RESUMEN

Introduction: Obesity is a public health problem that is associated with cerebrovascular diseases, such as ischemic stroke. The coexistence of obesity with cerebral ischemia has been suggested to be considerably detrimental to the neurological system. Objective: Hence, in this study, we evaluated the long-term effects of a 20% high fructose diet (HFD) and global cerebral ischemia on neurological, cognitive and emotional performance in three-month-old male Wistar rats. Results: Our results demonstrated that fructose intake led to increases in body weight and blood glucose, as well as reduced insulin sensitivity. The co-morbidity of fructose intake and cerebral ischemia resulted to hyperlipidemia, as well as increases in liver and adipocyte damage, which worsened neurological performance and resulted in alterations in learning and emotional skills at two weeks post-ischemia. No significant biochemical changes in autophagy and plasticity markers at the late stage of ischemia were observed. Conclusion: These results suggested that obesity causes a lasting effect on metabolic disorders that can contribute to increased neurological impairment after cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Enfermedades Metabólicas , Síndrome Metabólico , Animales , Glucemia/metabolismo , Isquemia Encefálica/complicaciones , Dieta , Dieta Alta en Grasa , Fructosa , Masculino , Obesidad/etiología , Ratas , Ratas Wistar
5.
Front Cell Neurosci ; 15: 643717, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790098

RESUMEN

Glutamate excitotoxicity triggers overactivation of CDK5 and increases calcium influx in neural cells, which promotes dendritic retraction, spine loss, increased mitochondrial calcium from the endoplasmic reticulum, and neuronal death. Our previous studies showed that CDK5 knockdown (KD) in astrocytes improves neurovascular integrity and cognitive functions and exerts neuroprotective effects. However, how CDK5-targeted astrocytes affect calcium regulation and whether this phenomenon is associated with changes in neuronal plasticity have not yet been analyzed. In this study, CDK5 KD astrocytes transplanted in CA3 remained at the injection site without proliferation, regulated calcium in the CA1 hippocampal region after excitotoxicity by glutamate in ex vivo hippocampal slices, improving synapsin and PSD95 clustering. These CDK5 KD astrocytes induced astrocyte stellation and neuroprotection after excitotoxicity induced by glutamate in vitro. Also, these effects were supported by CDK5 inhibition (CDK5i) in vitro through intracellular stabilization of calcium levels in astrocytes. Additionally, these cells in cocultures restored calcium homeostasis in neurons, redistributing calcium from somas to dendrites, accompanied by dendrite branching, higher dendritic spines and synapsin-PSD95 clustering. In summary, induction of calcium homeostasis at the CA1 hippocampal area by CDK5 KD astrocytes transplanted in the CA3 area highlights the role of astrocytes as a cell therapy target due to CDK5-KD astrocyte-mediated synaptic clustering, calcium spreading regulation between both areas, and recovery of the intracellular astrocyte-neuron calcium imbalance and plasticity impairment generated by glutamate excitotoxicity.

6.
Front Cell Neurosci ; 15: 656832, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025357

RESUMEN

Dysfunction in the neurovascular unit (NVU) is a key component in the progressive deterioration of Alzheimer's disease (AD) and is critical in vascular dementia. Recent studies have shown that inflammation plays early and perhaps causal roles in the pathogenesis of AD related to NVU damage, possibly in part by overactivating the aspartic acid protease activity of ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1), which until now has almost solely been studied in the context of the ß-amyloid cascade. In this study, we analyzed the relationship of BACE1 with astrocytes and blood vessels in human brains with sporadic and familial dementia [Autosomal dominant cerebral arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), sporadic Alzheimer's disease (SAD), and familial Alzheimer's disease (FAD)] and how BACE1 inhibition affects astrocytes and endothelial cells under conditions of glutamate toxicity. Our results show increased BACE1, PHF (Paired helical filaments)-tau and GFAP (Glial Fibrillary Acid Protein) immunoreactivity (IR) in the CA1 hippocampal regions of FAD and SAD brains. Furthermore, BACE1 immunoprecipitated with GFAP in tissue samples from all study cases, but their immunofluorescence close to (10 µm3) or overlapping blood vessels was only increased in FAD and SAD brains, and PHF-tau was present around the vessels mainly in FAD brains. Interestingly, the increased BACE1 levels were associated with reactive astrocytes, characterized by morphological changes and upregulation of GFAP under pathological and stressful conditions, and endothelial disruption by glutamate excitotoxicity, and these effects were reversed by BACE1 inhibition; further, BACE1-inhibited astrocytes protected endothelial cell integrity by preserving zonula occludens-1 (ZO-1) distribution and decreasing the expression of inflammatory markers. Taken together, these findings suggest that BACE1 dysregulation in astrocytes may have a role in the alterations in NVU integrity implicated in neurodegeneration.

7.
Front Aging Neurosci ; 13: 593927, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679370

RESUMEN

Astrocytes are specialized glial cells that are essential components of the neurovascular unit (NVU) and are involved in neurodevelopment, brain maintenance and repair, and neurodegeneration. Astrocytes mediate these processes by releasing cellular mediators such as extracellular vesicles (EVs). EVs are vehicles of cell-cell communication and have been proposed as mediators of damage in AD. However, the transcellular mechanism by which Alzheimer disease (AD) astrocytes impair the function of NVU components is poorly understood. Therefore, we evaluated the effects of adult PS1-KI and 3xTg-AD astrocyte conditioned media (CM) and EVs on NVU components (neuroglia and endothelium) in vitro. Additionally, SAD and FAD astrocyte-derived EVs (A-EVs) were characterized, and we evaluated their effects on NVU in cocultured cells in vitro and on intrahippocampal CA1 cells in vivo. Surprisingly, cultured 3xTg-AD astrocytes showed increased glial fibrillary acidic protein (GFAP) reactivity compared to PS1-KI astrocytes, which denotes astrocytic hyperreactivity. CM from adult mice 3xTg-AD astrocytes increased cell-cell gaps between endothelial cells, filopodia-like dendritic protrusions in neurons and neuronal and endothelial cell death. 3xTg-AD A-EVs induced neurotoxicity and increased astrocyte GFAP reactivity. Cultured human postmortem astrocytes from AD patients also increased GFAP reactivity and EVs release. No differences in the size or number of A-EVs were detected between AD and control samples; however, both SAD and FAD A-EVs showed increased expression of the surface marker aquaporin 4. A-EVs induced cytotoxicity and astrocyte hyperactivation: specifically, FAD A-EVs induced neuroglial cytotoxicity and increased gaps between the endothelium, while SAD A-EVs mainly altered the endothelium. Similarly, both AD A-EVs increased astrocyte GS reactivity and vascular deterioration in vivo. We associated this finding with perivascular reactive astrocytes and vascular deterioration in the human AD brain. In summary, these results suggest that AD A-EVs impair neuroglial and vascular components.

8.
J Alzheimers Dis ; 82(s1): S141-S161, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33016916

RESUMEN

The neurovascular unit (NVU) is responsible for synchronizing the energetic demand, vasodynamic changes, and neurochemical and electrical function of the brain through a closed and interdependent interaction of cell components conforming to brain tissue. In this review, we will focus on cyclin-dependent kinase 5 (CDK5) as a molecular pivot, which plays a crucial role in the healthy function of neurons, astrocytes, and the endothelium and is implicated in the cross-talk of cellular adhesion signaling, ion transmission, and cytoskeletal remodeling, thus allowing the individual and interconnected homeostasis of cerebral parenchyma. Then, we discuss how CDK5 overactivation affects the integrity of the NVU in Alzheimer's disease (AD) and cognitive impairment; we emphasize how CDK5 is involved in the excitotoxicity spreading of glutamate and Ca2+ imbalance under acute and chronic injury. Additionally, we present pharmacological and gene therapy strategies for producing partial depletion of CDK5 activity on neurons, astrocytes, or endothelium to recover neuroplasticity and neurotransmission, suggesting that the NVU should be the targeted tissue unit in protective strategies. Finally, we conclude that CDK5 could be effective due to its intervention on astrocytes by its end feet on the endothelium and neurons, acting as an intermediary cell between systemic and central communication in the brain. This review provides integrated guidance regarding the pathogenesis of and potential repair strategies for AD.


Asunto(s)
Astrocitos/metabolismo , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Silenciador del Gen/fisiología , Acoplamiento Neurovascular/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Ensayos Clínicos como Asunto/métodos , Silenciador del Gen/efectos de los fármacos , Humanos , Acoplamiento Neurovascular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165797, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32302650

RESUMEN

Sporadic Alzheimer's disease (SAD) is the most common form of dementia, and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most frequent hereditary ischemic small vessel disease of the brain. Relevant biomarkers or specific metabolic signatures could provide powerful tools to manage these diseases. Therefore, the main goal of this study was to compare the postmortem frontal cortex gray matter, white matter and cerebrospinal fluid (CSF) between a cognitively healthy group and CADASIL and SAD groups. We evaluated 352 individual lipids, belonging to 13 lipid classes/subclasses, using mass spectrometry, and the lipid profiles were subjected to multivariate analysis to discriminate between the dementia groups (CADASIL and SAD) and healthy controls. The main lipid molecular species showing greater discrimination by partial least squares-discriminant analysis (PLS-DA) and a higher significance multivariate correlation (sMC) index were as follows: phosphatidylserine (PS) PS(44:7) and lysophosphatidylethanolamine (LPE) LPE(18:2) in gray matter (GM); phosphatidylethanolamine (PE) PE(32:2) and phosphatidylcholine PC PC(44:6) in white matter (WM), and ether PE (ePE) ePE(38:2) and ether PC (ePC) ePC(34:3) in CSF. Common phospholipid molecular species were obtained in both dementias, such as PS(44:7) and lyso PC (LPC) LPC(22:5) in GM, PE(32:2) in WM and phosphatidic acid (PA) PA(38:5) and PC(42:7) in CFS. Our exploratory study suggests that phospholipids (PLs) involved in neurotransmission alteration, connectivity impairment and inflammation response in GM, WM and CSF are a transversal phenomenon affecting dementias such as CADASIL and SAD independent of the etiopathogenesis, thus providing a possible common prodromal phospholipidic biomarker of dementia.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , CADASIL/metabolismo , Lóbulo Frontal/metabolismo , Sustancia Gris/metabolismo , Tejido Parenquimatoso/metabolismo , Fosfolípidos/metabolismo , Sustancia Blanca/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/patología , Autopsia , Biomarcadores/análisis , CADASIL/diagnóstico , CADASIL/patología , Estudios de Casos y Controles , Análisis Discriminante , Femenino , Lóbulo Frontal/patología , Sustancia Gris/patología , Humanos , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Análisis Multivariante , Tejido Parenquimatoso/patología , Fosfolípidos/química , Fosfolípidos/clasificación , Fosfolípidos/aislamiento & purificación , Sustancia Blanca/patología
10.
J Neurol Sci ; 408: 116547, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31683050

RESUMEN

Multiple studies on cerebral ischemia have been performed in animal models to propose different strategies of neuroprotection that mitigate either the early or late consequences of the disease. These therapies have been successful in reducing the volume of infarction, the proinflammatory cascade, and the amount of free radicals, as well as reversing markers of neurodegeneration, among other events. However, when those strategies are translated to clinical studies, their effectiveness is not reproduced. This review will focus on highlighting some of the main limitations of the animal models of stroke that lead to unsuccessful translational therapies and the common risk factors in humans that should be carefully considered in the experimental design of future studies to generate a more realistic spatiotemporal physiopathology and improve therapeutic efficacy in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/terapia , Modelos Animales de Enfermedad , Accidente Cerebrovascular/terapia , Investigación Biomédica Traslacional/métodos , Animales , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/epidemiología , Humanos , Hipertensión/diagnóstico , Hipertensión/epidemiología , Hipertensión/terapia , Neuroprotección/fisiología , Obesidad/diagnóstico , Obesidad/epidemiología , Obesidad/terapia , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/genética , Investigación Biomédica Traslacional/tendencias , Resultado del Tratamiento
11.
PLoS One ; 14(10): e0223578, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31596896

RESUMEN

INTRODUCTION: Bexarotene, a retinoid X receptor agonist, improves cognition in murine models of Alzheimer's disease (AD). This study evaluated the effects of bexarotene on pathological and electrophysiological changes in very old triple transgenic AD mice (3xTg-AD mice). METHODS: 24-month-old 3xTg-AD mice were treated with bexarotene (100 mg/kg/day for 30 days). The Morris water maze was used to evaluate spatial memory; immunofluorescence and confocal microscopy were used to evaluate pathological changes; and in vivo electrophysiological recordings were used to evaluate basal transmission and plasticity in the commissural CA3-CA1 pathway. RESULTS: In addition to cognitive improvement, bexarotene-treated 3xTg-AD mice were found to have 1) reductions of astrogliosis and reactive microglia both in cortex and hippocampus; 2) increased ApoE expression restricted to CA1; 3) increased number of cells co-labeled with ApoE and NeuN; 4) recovery of NeuN expression, suggesting neuronal protection; and, 5) recovery of basal synaptic transmission and synaptic plasticity. DISCUSSION: These results indicate that bexarotene-induced improvement in cognition is due to multiple changes that contribute to recovery of synaptic plasticity.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Bexaroteno/uso terapéutico , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Alzheimer/genética , Animales , Apolipoproteínas E/metabolismo , Bexaroteno/farmacología , Proteínas de Unión al ADN/metabolismo , Femenino , Gliosis , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Plasticidad Neuronal , Fármacos Neuroprotectores/farmacología , Transmisión Sináptica
12.
Eur J Pharmacol ; 857: 172420, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31136761

RESUMEN

Stroke is the second cause of death and first cause of physical disability around the world; it affects the brain parenchyma through oxygen deficiency and spreads excitotoxicity. The complexity of the disease has made it difficult to find effective therapies. It is necessary to identify new treatments that effectively act within the narrow therapeutic window but also offer long-term protection poststroke. Our previous work found that oral linalool reversed the hippocampal and peripheral pro-inflammatory phospholipidomic biomarkers in ischemic rats; based on these observations, the "proof of concept" was to demonstrate that intranasal administration of linalool has a faster delivery to the central nervous system to protect it after focal ischemia in Wistar rats. The ischemic animals treated with linalool (25 mg/kg) showed a decrease in infarct volume at 24 h and seven days, and the treated animals had better neurological and motor skills at both poststroke times. Additionally, one month after daily intranasal administration of linalool, the ischemic rats showed improved relearning performance in the Morris water maze test. They also exhibited a reduction in microgliosis and decreased COX2, IL-1Beta and Nrf2 markers in the cerebral cortex and hippocampus. In astrocyte and microglial cultures, linalool reduced pro-inflammation and had a potent effect on microglial cells, generating Nrf2 subcellular redistribution under glutamate excitotoxicity conditions. Together, our findings indicate an acute and chronic recovery after ischemia induced by a daily intranasal puff of linalool, which mainly acts on microglial populations with anti-inflammatory actions.


Asunto(s)
Monoterpenos Acíclicos/administración & dosificación , Monoterpenos Acíclicos/farmacología , Isquemia Encefálica/patología , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Monoterpenos Acíclicos/farmacocinética , Administración Intranasal , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Isquemia Encefálica/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Microglía/metabolismo , Microglía/patología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacocinética , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Distribución Tisular
13.
Biomed Pharmacother ; 110: 482-492, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30530228

RESUMEN

In Alzheimer's disease (AD), amyloid beta (Aß) plaques initiates a cascade of pathological events where the overactivation of N-methyl-d-aspartate receptors (NMDA) by excess glutamate (Glu) triggers oxidative processes that lead to the activation of microglial cells, inflammation, and finally neuronal death. Amaryllidaceae alkaloids exert neuroprotective activities against different neurotoxin-induced injuries in vitro, and although their biological potential is well demonstrated, their neuroprotective activity has not been reported in an in vivo model of AD. The aim of our study was to determine the in vitro and in vivo neuroprotective potential of standardized alkaloidal fractions of Zephyranthes carinata. In this work, the neuroprotective effect of two alkaloidal fractions extracted from Z. carinata (bulbs and leaves) was analyzed in an in vitro excitotoxicity model in order to select the most promising one for subsequent evaluation in a triple transgenic mouse model of AD (3xTg-AD). We found that Z. carinata bulbs protect neurons against a Glu-mediated toxic stimulus in vitro, as evidenced by the decrease in apoptotic nuclei, the reduction in the lipid peroxidation product malondialdehyde and the conservation of dendritic structures. The effects of intraperitoneal administration of Z. carinata bulbs (10 mg/kg) every 12 h for 1 month on 3xTg-AD (18 months old) showed improved learning and spatial memory. Histopathologically, the alkaloidal fraction-treated 3xTg-AD mice exhibited a significant reduction in tauopathy and astrogliosis, as well as a significant decrease in the proinflammatory marker COX-2 and an increase in pAkt. The results suggest that Z. carinata bulbs provide neuroprotective effects both in vitro and in 3xTg-AD mice by intervening in the inflammatory processes, regulating the aggregation of pair helical filaments (PHFs) and activating survival pathways.


Asunto(s)
Alcaloides/uso terapéutico , Enfermedad de Alzheimer/prevención & control , Amaryllidaceae , Aprendizaje por Laberinto/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Alcaloides/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Neuroprotección/fisiología , Distribución Aleatoria , Ratas , Ratas Wistar , Resultado del Tratamiento
14.
Biomedica ; 38(3): 388-397, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335244

RESUMEN

INTRODUCTION: Cerebral ischemia is the third cause of death risk in Colombia and the first cause of physical disability worldwide. Different studies on the silencing of the cyclin-dependent kinase 5 (CDK5) have shown that reducing its activity is beneficial in ischemic contexts. However, its effect on neural cell production after cerebral ischemia has not been well studied yet. OBJECTIVE: To evaluate CDK5 silencing on the production of neurons and astrocytes after a focal cerebral ischemia in rats. MATERIALS AND METHODS: We used 40 eight-week-old male Wistar rats. Both sham and ischemia groups were transduced at CA1 hippocampal region with an adeno-associated viral vector using a noninterfering (shSCRmiR) and an interfering sequence for CDK5 (shCDK5miR). We injected 50 mg/kg of bromodeoxyuridine intraperitoneally from hour 24 to day 7 post-ischemia. We assessed the neurological abilities during the next 15 days and we measured the immunoreactivity of bromodeoxyuridine (BrdU), doublecortin (DCX), NeuN, and glial fibrillary acid protein (GFAP) from day 15 to day 30 post-ischemia. RESULTS: Our findings showed that CDK5miR-treated ischemic animals improved their neurological score and presented increased BrdU+ cells 15 days after ischemia, which correlated with higher DCX and lower GFAP fluorescence intensities, and, although mature neurons populations did not change, GFAP immunoreactivity was still significantly reduced at 30 days post-ischemia in comparison with untreated ischemic groups. CONCLUSION: CDK5miR therapy generated the neurological recovery of ischemic rats associated with the induction of immature neurons proliferation and the reduction of GFAP reactivity at short and longterm post-ischemia.


Asunto(s)
Isquemia Encefálica/terapia , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Terapia Genética , Vectores Genéticos/uso terapéutico , Terapia Molecular Dirigida , Neurogénesis/genética , Neuroglía/fisiología , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Animales , Astrocitos/patología , Biomarcadores , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Estenosis Carotídea , Quinasa 5 Dependiente de la Ciclina/genética , Replicación del ADN , Dependovirus/genética , Proteína Doblecortina , Evaluación de Medicamentos , Terapia Genética/métodos , Ligadura , Masculino , Terapia Molecular Dirigida/métodos , Neuronas/patología , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Wistar
15.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3696-3707, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30251689

RESUMEN

Inflammation has been suggested to play early and perhaps causative roles in Alzheimer's disease (AD) pathogenesis possibly in part by the overactivation of the aspartic acid protease named ß-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), which is responsible for the ß-amyloid cascade. We have described that BACE1 is involved in the lysophosphatidylethanolamine (LPE) (18:1/20:4/22:6) upregulation associated with tauopathy and inflammation signaling (cPLA2/arachidonic acid/COX2) in a triple transgenic model of Alzheimer's disease, where BACE1 silencing reversed the imbalanced profile and produced cognitive function improvement. In this study, we analyze the role of cPLA2 and desaturases (SCD1, FAD6) in the BACE1 knockdown-induced protective action under a glutamate excitotoxicity model. Glutamate (125 µM) produced hyperphosphorylation of tau in cortical primary cultures along with increased apoptotic nuclei, LDH release, and cPLA2 expression, which were all reversed by BACE1-KD. This beneficial effect was reinforced by the silencing of cPLA2 but attenuated by the reduction in SCD1 and partially attenuated by the reduction in FAD6. Inversely, overexpression SCD1 and FAD6 recovered the neuroprotective effect produced by BACE1-KD, which was not achieved by the overexpression of each desaturase alone. These findings suggest that the hyperphosphorylation of tau and the creation of a pro-inflammatory cell environment are blocked in a desaturase-dependent manner by targeting BACE1.


Asunto(s)
Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Fosfolipasas A2 Grupo IV/metabolismo , Estearoil-CoA Desaturasa/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apoptosis/genética , Ácido Aspártico Endopeptidasas/genética , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Potenciales Postsinápticos Excitadores , Técnicas de Silenciamiento del Gen , Ácido Glutámico/metabolismo , Fosfolipasas A2 Grupo IV/genética , Neuronas/metabolismo , Fosforilación/genética , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Estearoil-CoA Desaturasa/genética , Proteínas tau/toxicidad
16.
Biomédica (Bogotá) ; 38(3): 388-397, jul.-set. 2018. graf
Artículo en Inglés | LILACS | ID: biblio-973992

RESUMEN

Abstract Introduction: Cerebral ischemia is the third cause of death risk in Colombia and the first cause of physical disability worldwide. Different studies on the silencing of the cyclin-dependent kinase 5 (CDK5) have shown that reducing its activity is beneficial in ischemic contexts. However, its effect on neural cell production after cerebral ischemia has not been well studied yet. Objective: To evaluate CDK5 silencing on the production of neurons and astrocytes after a focal cerebral ischemia in rats. Materials and methods: We used 40 eight-week-old male Wistar rats. Both sham and ischemia groups were transduced at CA1 hippocampal region with an adeno-associated viral vector using a noninterfering (shSCRmiR) and an interfering sequence for CDK5 (shCDK5miR). We injected 50 mg/kg of bromodeoxyuridine intraperitoneally from hour 24 to day 7 post-ischemia. We assessed the neurological abilities during the next 15 days and we measured the immunoreactivity of bromodeoxyuridine (BrdU), doublecortin (DCX), NeuN, and glial fibrillary acid protein (GFAP) from day 15 to day 30 post-ischemia. Results: Our findings showed that CDK5miR-treated ischemic animals improved their neurological score and presented increased BrdU+ cells 15 days after ischemia, which correlated with higher DCX and lower GFAP fluorescence intensities, and, although mature neurons populations did not change, GFAP immunoreactivity was still significantly reduced at 30 days post-ischemia in comparison with untreated ischemic groups. Conclusion: CDK5miR therapy generated the neurological recovery of ischemic rats associated with the induction of immature neurons proliferation and the reduction of GFAP reactivity at short and longterm post-ischemia.


Resumen Introducción. La isquemia cerebral es la tercera causa de riesgo de muerte en Colombia y la primera causa de discapacidad física en el mundo. En diversos estudios en los que se silenció la cinasa 5 dependiente de la ciclina (CDK5) se ha demostrado que la reducción de su actividad es beneficiosa frente a la isquemia. Sin embargo, su efecto sobre la neurogénesis después de la isquemia no se ha dilucidado suficientemente. Objetivo. Evaluar el silenciamiento de la CDK5 en la neurogénesis y la gliogénesis después de la isquemia cerebral focal en ratas. Materiales y métodos. Se usaron 40 machos de rata Wistar de ocho semanas de edad. Los grupos de control y los isquémicos sometidos a transducción en la región del hipocampo CA1, se inyectaron intraperitonealmente por estereotaxia con 50 mg/kg de bromodesoxiuridina (BrdU) a partir de las 24 horas y hasta el día 7 después de la isquemia, con un vector viral asociado a adenovirus usando una secuencia no interferente (SCRmiR) y una interferente (CDK5miR). Se evaluó la capacidad neurológica durante los quince días siguientes y se detectó la capacidad de inmunorreacción para la BrdU, la proteína doblecortina (DCX), los núcleos neuronales (NeuN), y la proteína fibrilar acídica de la glía (Glial Fibrillary Acidic Protein, GFAP) a los 15 y 30 días de la isquemia. Resultados. Los animales isquémicos tratados con CDK5miR mejoraron su puntuación neurológica y presentaron un incremento de la BrdU+ a los 15 días de la isquemia, lo cual se correlacionó con una mayor intensidad de la DCX+ y una menor de la GFAP+. No hubo modificación de los NeuN+, pero sí una reducción significativa de la GFAP+ a los 30 días de la isquemia en los animales tratados comparados con los animales isquémicos no tratados. Conclusión. La terapia con CDK5miR generó la recuperación neurológica de ratas isquémicas asociada con la inducción de la neurogénesis y el control de la capacidad de reacción de la proteína GFAP a corto y largo plazo después de la isquemia.


Asunto(s)
Animales , Masculino , Ratas , Terapia Genética , Isquemia Encefálica/terapia , Neuroglía/fisiología , ARN Interferente Pequeño/uso terapéutico , Interferencia de ARN , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Neurogénesis/genética , Terapia Molecular Dirigida , Vectores Genéticos/uso terapéutico , Biomarcadores , Terapia Genética/métodos , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Astrocitos/patología , Estenosis Carotídea , Ratas Wistar , Dependovirus/genética , ARN Interferente Pequeño/administración & dosificación , Replicación del ADN , Evaluación de Medicamentos , Quinasa 5 Dependiente de la Ciclina/genética , Terapia Molecular Dirigida/métodos , Proteína Doblecortina , Ligadura , Neuronas/patología
17.
Biomedica ; 38(0): 69-76, 2018 05 01.
Artículo en Español | MEDLINE | ID: mdl-29809330

RESUMEN

INTRODUCTION: Alzheimer's disease is the most common form of dementia. It is characterized by histopathological hallmarks such as senile plaques and neurofibrillary tangles, as well as a concomitant activation of microglial cells and astrocytes that release pro-inflammatory mediators such as IL-1ß, iNOS, and COX-2, leading to neuronal dysfunction and death. OBJECTIVE: To evaluate the effect of quercetin on the inflammatory response in the CA1 area of the hippocampus in a 3xTg-AD male and female mice model. MATERIALS AND METHODS: Animals were injected intraperitoneally with quercetin every 48 hours during three months, and we conducted histological and biochemical studies. RESULTS: We found that in quercetin-treated 3xTg-AD mice, reactive microglia and fluorescence intensity of Aß aggregates significantly decreased. GFAP, iNOS, and COX-2 immunoreactivity also decreased and we observed a clear tendency in the reduction of IL-1ß in hippocampal lysates. CONCLUSION: Our work suggests an anti-inflammatory effect of quercetin in the CA1 hippocampal region of aged triple transgenic Alzheimer's disease mice.


Asunto(s)
Enfermedad de Alzheimer , Antioxidantes/uso terapéutico , Región CA1 Hipocampal/efectos de los fármacos , Inflamación/tratamiento farmacológico , Quercetina/uso terapéutico , Factores de Edad , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos
18.
Biomédica (Bogotá) ; 38(supl.1): 62-69, mayo 2018. graf
Artículo en Inglés | LILACS | ID: biblio-950955

RESUMEN

Introduction: Alzheimer's disease is the most common form of dementia. It is characterized by histopathological hallmarks such as senile plaques and neurofibrillary tangles, as well as a concomitant activation of microglial cells and astrocytes that release pro-inflammatory mediators such as IL-1ß, iNOS, and COX-2, leading to neuronal dysfunction and death. Objective: To evaluate the effect of quercetin on the inflammatory response in the CA1 area of the hippocampus in a 3xTg-AD male and female mice model. Materials and methods: Animals were injected intraperitoneally with quercetin every 48 hours during three months, and we conducted histological and biochemical studies. Results: We found that in quercetin-treated 3xTg-AD mice, reactive microglia and fluorescence intensity of Aß aggregates significantly decreased. GFAP, iNOS, and COX-2 immunoreactivity also decreased and we observed a clear tendency in the reduction of IL-1ß in hippocampal lysates. Conclusion: Our work suggests an anti-inflammatory effect of quercetin in the CA1 hippocampal region of aged triple transgenic Alzheimer's disease mice.


Introducción. La enfermedad de Alzheimer es la forma más común de demencia; se caracteriza por la presencia de marcadores histopatológicos, como las placas seniles y los ovillos neurofibrilares, así como por una activación concomitante de células microgliales y astrocitos que liberan mediadores proinflamatorios, como IL-1ß, iNOS y COX-2, lo cual conduce a la disfunción y la muerte neuronal. Objetivo. Evaluar el efecto de la quercetina sobre la reacción inflamatoria en el área CA1 del hipocampo en un modelo de ratones 3xTg-AD. Materiales y métodos. Los animales se inyectaron intraperitonealmente con quercetina cada 48 horas durante tres meses, y se hicieron estudios histológicos y bioquímicos. Resultados. Se encontró que en los animales 3xTg-AD tratados con quercetina, la microglía reactiva y la intensidad de fluorescencia de los agregados Aß disminuyeron significativamente, y que hubo una menor reacción de GFAP, iNOS y COX-2, así como una clara tendencia a la reducción de la IL-1 ß en lisados de hipocampo. Conclusión. Los resultados del estudio sugieren un efecto antiinflamatorio de la quercetina en la región CA1 del hipocampo en un modelo en ratón triple trasgénico para la enfermedad de Alzheimer.


Asunto(s)
Quercetina , Enfermedad de Alzheimer , Astrocitos , Microglía
19.
Neuropharmacology ; 135: 555-571, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29680773

RESUMEN

Phospholipid alterations in the brain are associated with progressive neurodegeneration and cognitive impairment after acute and chronic injuries. Various types of treatments have been evaluated for their abilities to block the progression of the impairment, but effective treatments targeting long-term post-stroke alterations are not available. In this study, we analyzed changes in the central and peripheral phospholipid profiles in ischemic rats and determined whether a protective monoterpene, Linalool, could modify them. We used an in vitro model of glutamate (125 µM) excitotoxicity and an in vivo global ischemia model in Wistar rats. Linalool (0.1 µM) protected neurons and astrocytes by reducing LDH release and restoring ATP levels. Linalool was administered orally at a dose of 25 mg/kg every 24 h for a month, behavioral tests were performed, and a lipidomic analysis was conducted using mass spectrometry. Animals treated with Linalool displayed faster neurological recovery than untreated ischemic animals, accompanied by better motor and cognitive performances. These results were confirmed by the significant reduction in astrogliosis, microgliosis and COX-2 marker, involving a decrease of 24:0 free fatty acid in the hippocampus. The altered profiles of phospholipids composed of mono and polyunsaturated fatty acids (PC 36:1; 42:1 (24:0/18:1)/LPC 22:6)/LPE 22:6) in the ischemic hippocampus and the upregulation of PI 36:2 and other LCFA (long chain fatty acids) in the serum of ischemic rats were prevented by the monoterpene. Based on these data, alterations in the central and peripheral phospholipid profiles after long-term was attenuated by oral Linalool, promoting a phospholipid homeostasis, related to the recovery of brain function.


Asunto(s)
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Monoterpenos/farmacología , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/farmacología , Fosfolípidos/metabolismo , Monoterpenos Acíclicos , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Ácido Glutámico/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/patología , Distribución Aleatoria , Ratas Wistar , Recuperación de la Función/efectos de los fármacos
20.
Life Sci ; 203: 54-65, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29679699

RESUMEN

AIMS: Determine a relationship between the neuroprotective activity and the antioxidant capacity of the Amaryllidaceae alkaloids in a model of Glu excitotoxicity in rat cortical neurons. MATERIALS AND METHODS: Was evaluated several alkaloidal fractions isolated from Amaryllidaceae species, a family known to contain neuroprotective alkaloids, in a model of Glu excitotoxicity in rat cortical neurons. In addition, several mechanisms of antioxidant activity were used, and a theoretical study of the antioxidants was performed. KEY FINDINGS: The results of this study suggest that a possible neuroprotective mode of action of the alkaloidal fractions of Eucharis bonplandii (Kunth) Traub bulbs, Eucharis caucana Meerow bulbs, and Clivia miniata Regel leaves, is through their antioxidant activity and ability to stabilize free radicals generated from an excitotoxic process mediated by Glu. The chemical structure characterization and antioxidant activity of the fractions suggest that the phenol and enol groups in the structures of the alkaloids are critical for the stabilization of ROS and RNS. Additionally, the pair of free electrons on the N is spatially close to a hydroxyl group, which benefits the cleavage of this group and, consequently, the stabilization of the generated O. SIGNIFICANCE: The versatility of the structures of the studied Amaryllidaceae alkaloids suggests that they have potential as neuroprotective agents against an oxidative stimulus.


Asunto(s)
Alcaloides de Amaryllidaceae/farmacología , Antioxidantes/farmacología , Embrión de Mamíferos/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Mamíferos/citología , Estructura Molecular , Neuronas/patología , Oxidación-Reducción , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...