Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38328119

RESUMEN

As the most abundant glial cells in the CNS, astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress have remained elusive. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stresser, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, neurotoxic stress induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechnistically, YTHDF2 RIP-sequencing identified MAP2K4 ( MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed Mn-exposed astrocytes mediates proinflammatory response by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves a key upstream 'molecular switch' controlling SEK1( MAP2K4 )-JNK-cJUN proinflammatory signaling in astrocytes.

2.
Int J Mol Sci ; 23(18)2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36142718

RESUMEN

As a prevalent progressive neurodegenerative disorder, Parkinson's disease (PD) is characterized by the neuropathological hallmark of the loss of nigrostriatal dopaminergic (DAergic) innervation and the appearance of Lewy bodies with aggregated α-synuclein. Although several familial forms of PD have been reported to be associated with several gene variants, most cases in nature are sporadic, triggered by a complex interplay of genetic and environmental risk factors. Numerous epidemiological studies during the past two decades have shown positive associations between PD and several environmental factors, including exposure to neurotoxic pesticides/herbicides and heavy metals as well as traumatic brain injury. Other environmental factors that have been implicated as potential risk factors for PD include industrial chemicals, wood pulp mills, farming, well-water consumption, and rural residence. In this review, we summarize the environmental toxicology of PD with the focus on the elaboration of chemical toxicity and the underlying pathogenic mechanisms associated with exposure to several neurotoxic chemicals, specifically 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, paraquat (PQ), dichloro-diphenyl-trichloroethane (DDT), dieldrin, manganese (Mn), and vanadium (V). Our overview of the current findings from cellular, animal, and human studies of PD provides information for possible intervention strategies aimed at halting the initiation and exacerbation of environmentally linked PD.


Asunto(s)
Herbicidas , Síndromes de Neurotoxicidad , Enfermedad de Parkinson , Plaguicidas , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , DDT , Dieldrín/metabolismo , Herbicidas/metabolismo , Humanos , Manganeso/metabolismo , Mitocondrias/metabolismo , Enfermedades Neuroinflamatorias , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo , Paraquat , Enfermedad de Parkinson/metabolismo , Plaguicidas/metabolismo , Plaguicidas/toxicidad , Factores de Riesgo , Rotenona/metabolismo , Tricloroetanos/metabolismo , Vanadio/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
Int J Biochem Cell Biol ; 147: 106225, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35550926

RESUMEN

Despite the growing recognition that gastrointestinal (GI) dysfunction is prevalent in Parkinson's disease (PD) and occurs as a major prodromal symptom of PD, its cellular and molecular mechanisms remain largely unknown. Among the various types of GI cells, enteric glial cells (EGCs), which resemble astrocytes in structure and function, play a critical role in the pathophysiology of many GI diseases including PD. Thus, we investigated how EGCs respond to the environmental pesticides rotenone (Rot) and tebufenpyrad (Tebu) in cell and animal models to better understand the mechanism underlying GI abnormalities. Both Rot and Tebu induce dopaminergic neuronal cell death through complex 1 inhibition of the mitochondrial respiratory chain. We report that exposing a rat enteric glial cell model (CRL-2690 cells) to these pesticides increased mitochondrial fission and reduced mitochondrial fusion by impairing MFN2 function. Furthermore, they also increased mitochondrial superoxide generation and impaired mitochondrial ATP levels and basal respiratory rate. Measurement of LC3, p62 and lysosomal assays revealed impaired autolysosomal function in ECGs during mitochondrial stress. Consistent with our recent findings that mitochondrial dysfunction augments inflammation in astrocytes and microglia, we found that neurotoxic pesticide exposure also enhanced the production of pro-inflammatory factors in EGCs in direct correlation with the loss in mitochondrial mass. Finally, we show that pesticide-induced mitochondrial defects functionally impaired smooth muscle velocity, acceleration, and total kinetic energy in a mixed primary culture of the enteric nervous system (ENS). Collectively, our studies demonstrate for the first time that exposure to environmental neurotoxic pesticides impairs mitochondrial bioenergetics and activates inflammatory pathways in EGCs, further augmenting mitochondrial dysfunction and pro-inflammatory events to induce gut dysfunction. Our findings have major implications in understanding the GI-related pathogenesis and progression of environmentally linked PD.


Asunto(s)
Enfermedad de Parkinson , Plaguicidas , Animales , Eje Cerebro-Intestino , Inflamación/inducido químicamente , Mitocondrias , Neuroglía , Enfermedad de Parkinson/etiología , Plaguicidas/toxicidad , Ratas , Rotenona/toxicidad
4.
Front Neurosci ; 16: 836605, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281490

RESUMEN

The human gut microbiota is a complex, dynamic, and highly diverse community of microorganisms. Beginning as early as in utero fetal development and continuing through birth to late-stage adulthood, the crosstalk between the gut microbiome and brain is essential for modulating various metabolic, neurodevelopmental, and immune-related pathways. Conversely, microbial dysbiosis - defined as alterations in richness and relative abundances - of the gut is implicated in the pathogenesis of several chronic neurological and neurodegenerative disorders. Evidence from large-population cohort studies suggests that individuals with neurodegenerative conditions have an altered gut microbial composition as well as microbial and serum metabolomic profiles distinct from those in the healthy population. Dysbiosis is also linked to psychiatric and gastrointestinal complications - comorbidities often associated with the prodromal phase of Parkinson's disease (PD) and Alzheimer's disease (AD). Studies have identified potential mediators that link gut dysbiosis and neurological disorders. Recent findings have also elucidated the potential mechanisms of disease pathology in the enteric nervous system prior to the onset of neurodegeneration. This review highlights the functional pathways and mechanisms, particularly gut microbe-induced chronic inflammation, protein misfolding, propagation of disease-specific pathology, defective protein clearance, and autoimmune dysregulation, linking gut microbial dysbiosis and neurodegeneration. In addition, we also discuss how pathogenic transformation of microbial composition leads to increased endotoxin production and fewer beneficial metabolites, both of which could trigger immune cell activation and enteric neuronal dysfunction. These can further disrupt intestinal barrier permeability, aggravate the systemic pro-inflammatory state, impair blood-brain barrier permeability and recruit immune mediators leading to neuroinflammation and neurodegeneration. Continued biomedical advances in understanding the microbiota-gut-brain axis will extend the frontier of neurodegenerative disorders and enable the utilization of novel diagnostic and therapeutic strategies to mitigate the pathological burden of these diseases.

5.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34494552

RESUMEN

Mitochondrial dysfunction is a major pathophysiological contributor to the progression of Parkinson's disease (PD); however, whether it contributes to epigenetic dysregulation remains unknown. Here, we show that both chemically and genetically driven mitochondrial dysfunctions share a common mechanism of epigenetic dysregulation. Under both scenarios, lysine 27 acetylation of likely variant H3.3 (H3.3K27ac) increased in dopaminergic neuronal models of PD, thereby opening that region to active enhancer activity via H3K27ac. These vulnerable epigenomic loci represent potential transcription factor motifs for PD pathogenesis. We further confirmed that mitochondrial dysfunction induces H3K27ac in ex vivo and in vivo (MitoPark) neurodegenerative models of PD. Notably, the significantly increased H3K27ac in postmortem PD brains highlights the clinical relevance to the human PD population. Our results reveal an exciting mitochondrial dysfunction-metabolism-H3K27ac-transcriptome axis for PD pathogenesis. Collectively, the mechanistic insights link mitochondrial dysfunction to epigenetic dysregulation in dopaminergic degeneration and offer potential new epigenetic intervention strategies for PD.


Asunto(s)
Encéfalo/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Mutación , Estrés Oxidativo , Enfermedad de Parkinson/genética , Acetilación , Animales , Encéfalo/patología , Células Cultivadas , ADN/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Procesamiento Proteico-Postraduccional , Factores de Transcripción/genética
6.
Environ Health Perspect ; 129(8): 87005, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34410835

RESUMEN

BACKGROUND: Chronic environmental exposure to manganese (Mn) can cause debilitating damage to the central nervous system. However, its potential toxic effects on the enteric nervous system (ENS) have yet to be assessed. OBJECTIVE: We examined the effect of Mn on the ENS using both cell and animal models. METHOD: Rat enteric glial cells (EGCs) and mouse primary enteric cultures were exposed to increasing concentrations of Mn and cell viability and mitochondrial health were assessed using various morphological and functional assays. C57BL/6 mice were exposed daily to a sublethal dose of Mn (15mg/kg/d) for 30 d. Gut peristalsis, enteric inflammation, gut microbiome profile, and fecal metabolite composition were assessed at the end of exposure. RESULTS: EGC mitochondria were highly susceptible to Mn neurotoxicity, as evidenced by lower mitochondrial mass, adenosine triphosphate-linked respiration, and aconitase activity as well as higher mitochondrial superoxide, upon Mn exposure. Minor differences were seen in the mouse model: specifically, longer intestinal transit times and higher levels of colonic inflammation. CONCLUSION: Based on our findings from this study, Mn preferentially induced mitochondrial dysfunction in a rat EGC line and in vivo resulted in inflammation in the ENS. https://doi.org/10.1289/EHP7877.


Asunto(s)
Sistema Nervioso Entérico , Microbioma Gastrointestinal , Animales , Manganeso/toxicidad , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Ratas
7.
Front Aging Neurosci ; 13: 661505, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276337

RESUMEN

A classical hallmark of Parkinson's disease (PD) pathogenesis is the accumulation of misfolded alpha-synuclein (αSyn) within Lewy bodies and Lewy neurites, although its role in microglial dysfunction and resultant dopaminergic (DAergic) neurotoxicity is still elusive. Previously, we identified that protein kinase C delta (PKCδ) is activated in post mortem PD brains and experimental Parkinsonism and that it participates in reactive microgliosis; however, the relationship between PKCδ activation, endoplasmic reticulum stress (ERS) and the reactive microglial activation state in the context of α-synucleinopathy is largely unknown. Herein, we show that oxidative stress, mitochondrial dysfunction, NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, and PKCδ activation increased concomitantly with ERS markers, including the activating transcription factor 4 (ATF-4), serine/threonine-protein kinase/endoribonuclease inositol-requiring enzyme 1α (p-IRE1α), p-eukaryotic initiation factor 2 (eIF2α) as well as increased generation of neurotoxic cytokines, including IL-1ß in aggregated αSynagg-stimulated primary microglia. Importantly, in mouse primary microglia-treated with αSynagg we observed increased expression of Thioredoxin-interacting protein (TXNIP), an endogenous inhibitor of the thioredoxin (Trx) pathway, a major antioxidant protein system. Additionally, αSynagg promoted interaction between NLRP3 and TXNIP in these cells. In vitro knockdown of PKCδ using siRNA reduced ERS and led to reduced expression of TXNIP and the NLRP3 activation response in αSynagg-stimulated mouse microglial cells (MMCs). Additionally, attenuation of mitochondrial reactive oxygen species (mitoROS) via mito-apocynin and amelioration of ERS via the eIF2α inhibitor salubrinal (SAL) reduced the induction of the ERS/TXNIP/NLRP3 signaling axis, suggesting that mitochondrial dysfunction and ERS may act in concert to promote the αSynagg-induced microglial activation response. Likewise, knockdown of TXNIP by siRNA attenuated the αSynagg-induced NLRP3 inflammasome activation response. Finally, unilateral injection of αSyn preformed fibrils (αSynPFF) into the striatum of wild-type mice induced a significant increase in the expression of nigral p-PKCδ, ERS markers, and upregulation of the TXNIP/NLRP3 inflammasome signaling axis prior to delayed loss of TH+ neurons. Together, our results suggest that inhibition of ERS and its downstream signaling mediators TXNIP and NLRP3 might represent novel therapeutic avenues for ameliorating microglia-mediated neuroinflammation in PD and other synucleinopathies.

8.
Cells ; 9(8)2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759670

RESUMEN

Astrocytic dysfunction has been implicated in Parkinson's disease (PD) pathogenesis. While the Tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/Fn14 signaling axis is known to play a role in PD-like neuropathology, the molecular mechanisms that govern this process remain poorly understood. Herein, we show that TWEAK levels are elevated in PD serum compared to controls. Moreover, using both U373 human astrocyte cells and primary mouse astrocytes, we demonstrate that TWEAK induces mitochondrial oxidative stress as well as protein kinase C delta (PKCδ) and signal transducer and activator of transcription 3 (STAT3) activation, accompanied by NLRC4 inflammasome activation and upregulation and release of proinflammatory cytokines, including IL-1ß, TNF-α, and IL-18. Mechanistically, TWEAK-induced PKCδ activation enhances the STAT3/NLRC4 signaling pathway and other proinflammatory mediators through a mitochondrial oxidative stress-dependent mechanism. We further show that PKCδ knockdown and mito-apocynin, a mitochondrial antioxidant, suppress TWEAK-induced proinflammatory NLRC4/STAT3 signaling and cellular oxidative stress response. Notably, we validated our in vitro findings in an MPTP mouse model of PD and in mice receiving intrastriatal administration of TWEAK. These results indicate that TWEAK is a key regulator of astroglial reactivity and illustrate a novel mechanism by which mitochondrial oxidative stress may influence dopaminergic neuronal survival in PD.


Asunto(s)
Astrocitos/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Citocina TWEAK/metabolismo , Inflamasomas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteína Quinasa C-delta/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Astrocitos/patología , Supervivencia Celular , Células Cultivadas , Citocina TWEAK/sangre , Citocina TWEAK/genética , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/patología , Proteína Quinasa C-delta/genética , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor de TWEAK/metabolismo
9.
Front Neurosci ; 13: 654, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293375

RESUMEN

Occupational or environmental exposure to manganese (Mn) can lead to the development of "Manganism," a neurological condition showing certain motor symptoms similar to Parkinson's disease (PD). Like PD, Mn toxicity is seen in the central nervous system mainly affecting nigrostriatal neuronal circuitry and subsequent behavioral and motor impairments. Since the first report of Mn-induced toxicity in 1837, various experimental and epidemiological studies have been conducted to understand this disorder. While early investigations focused on the impact of high concentrations of Mn on the mitochondria and subsequent oxidative stress, current studies have attempted to elucidate the cellular and molecular pathways involved in Mn toxicity. In fact, recent reports suggest the involvement of Mn in the misfolding of proteins such as α-synuclein and amyloid, thus providing credence to the theory that environmental exposure to toxicants can either initiate or propagate neurodegenerative processes by interfering with disease-specific proteins. Besides manganism and PD, Mn has also been implicated in other neurological diseases such as Huntington's and prion diseases. While many reviews have focused on Mn homeostasis, the aim of this review is to concisely synthesize what we know about its effect primarily on the nervous system with respect to its role in protein misfolding, mitochondrial dysfunction, and consequently, neuroinflammation and neurodegeneration. Based on the current evidence, we propose a 'Mn Mechanistic Neurotoxic Triad' comprising (1) mitochondrial dysfunction and oxidative stress, (2) protein trafficking and misfolding, and (3) neuroinflammation.

10.
J Neuroimmune Pharmacol ; 14(4): 595-607, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30879240

RESUMEN

Chronic and debilitating neurodegenerative diseases, such as Parkinson's disease (PD), impose an immense medical, emotional, and economic burden on patients and society. Due to a complex interaction between genetic and environmental risk factors, the etiology of PD remains elusive. However, the cumulative evidence emerging from clinical and experimental research over the last several decades has identified mitochondrial dysfunction, oxidative stress, neuroinflammation, and dysregulated protein degradation as the main drivers of PD neurodegeneration. The genome-editing system CRISPR (clustered regularly interspaced short palindromic repeats) has recently transformed the field of biotechnology and biomedical discovery and is poised to accelerate neurodegenerative disease research. It has been leveraged to generate PD animal models, such as Parkin, DJ-1, and PINK1 triple knockout miniature pigs. CRISPR has also allowed the deeper understanding of various PD gene interactions, as well as the identification of novel apoptotic pathways associated with neurodegenerative processes in PD. Furthermore, its application has been used to dissect neuroinflammatory pathways involved in PD pathogenesis, such as the PKCδ signaling pathway, as well as the roles of novel compensatory or protective pathways, such as Prokineticin-2 signaling. This review aims to highlight the historical milestones in the evolution of this technology and attempts to illustrate its transformative potential in unraveling disease mechanisms as well as in the development of innovative treatment strategies for PD. Graphical Abstract.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/fisiología , Edición Génica/tendencias , Terapia Genética/tendencias , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , Animales , Proteína 9 Asociada a CRISPR/inmunología , Edición Génica/métodos , Terapia Genética/métodos , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/terapia , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/terapia , Enfermedad de Parkinson/inmunología
11.
Sci Signal ; 12(572)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862700

RESUMEN

The aggregation of α-synuclein (αSyn) is considered a key pathophysiological feature of certain neurodegenerative disorders, collectively termed synucleinopathies. Given that a prion-like, cell-to-cell transfer of misfolded αSyn has been recognized in the spreading of αSyn pathology in synucleinopathies, we investigated the biological mechanisms underlying the propagation of the disease with respect to environmental neurotoxic stress. Considering the potential role of the divalent metal manganese (Mn2+) in protein aggregation, we characterized its effect on αSyn misfolding and transmission in experimental models of Parkinson's disease. In cultured dopaminergic neuronal cells stably expressing wild-type human αSyn, misfolded αSyn was secreted through exosomes into the extracellular medium upon Mn2+ exposure. These exosomes were endocytosed through caveolae into primary microglial cells, thereby mounting neuroinflammatory responses. Furthermore, Mn2+-elicited exosomes exerted a neurotoxic effect in a human dopaminergic neuronal model (LUHMES cells). Moreover, bimolecular fluorescence complementation (BiFC) analysis revealed that Mn2+ accelerated the cell-to-cell transmission of αSyn, resulting in dopaminergic neurotoxicity in a mouse model of Mn2+ exposure. Welders exposed to Mn2+ had increased misfolded αSyn content in their serum exosomes. Stereotaxically delivering αSyn-containing exosomes, isolated from Mn2+-treated αSyn-expressing cells, into the striatum initiated Parkinsonian-like pathological features in mice. Together, these results indicate that Mn2+ exposure promotes αSyn secretion in exosomal vesicles, which subsequently evokes proinflammatory and neurodegenerative responses in both cell culture and animal models.


Asunto(s)
Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Exosomas/metabolismo , Manganeso/toxicidad , Enfermedad de Parkinson Secundaria/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Exosomas/patología , Manganeso/farmacología , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Priones/metabolismo , Agregación Patológica de Proteínas/inducido químicamente , Agregación Patológica de Proteínas/patología
12.
Neurotoxicology ; 64: 204-218, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28539244

RESUMEN

Chronic manganese (Mn) exposure induces neurotoxicity, which is characterized by Parkinsonian symptoms resulting from impairment in the extrapyramidal motor system of the basal ganglia. Mitochondrial dysfunction and oxidative stress are considered key pathophysiological features of Mn neurotoxicity. Recent evidence suggests astrocytes as a major target of Mn neurotoxicity since Mn accumulates predominantly in astrocytes. However, the primary mechanisms underlying Mn-induced astroglial dysfunction and its role in metal neurotoxicity are not completely understood. In this study, we examined the interrelationship between mitochondrial dysfunction and astrocytic inflammation in Mn neurotoxicity. We first evaluated whether Mn exposure alters mitochondrial bioenergetics in cultured astrocytes. Metabolic activity assessed by MTS assay revealed an IC50 of 92.68µM Mn at 24h in primary mouse astrocytes (PMAs) and 50.46µM in the human astrocytic U373 cell line. Mn treatment reduced mitochondrial mass, indicative of impaired mitochondrial function and biogenesis, which was substantiated by the significant reduction in mRNA of mitofusin-2, a protein that serves as a ubiquitination target for mitophagy. Furthermore, Mn increased mitochondrial circularity indicating augmented mitochondrial fission. Seahorse analysis of bioenergetics status in Mn-treated astrocytes revealed that Mn significantly impaired the basal mitochondrial oxygen consumption rate as well as the ATP-linked respiration rate. The effect of Mn on mitochondrial energy deficits was further supported by a reduction in ATP production. Mn-exposed primary astrocytes also exhibited a severely quiescent energy phenotype, which was substantiated by the inability of oligomycin to increase the extracellular acidification rate. Since astrocytes regulate immune functions in the CNS, we also evaluated whether Mn modulates astrocytic inflammation. Mn exposure in astrocytes not only stimulated the release of proinflammatory cytokines, but also exacerbated the inflammatory response induced by aggregated α-synuclein. The novel mitochondria-targeted antioxidant, mito-apocynin, significantly attenuated Mn-induced inflammatory gene expression, further supporting the role of mitochondria dysfunction and oxidative stress in mediating astrogliosis. Lastly, intranasal delivery of Mn in vivo elevated GFAP and depressed TH levels in the olfactory bulbs, clearly supporting the involvement of astrocytes in Mn-induced dopaminergic neurotoxicity. Collectively, our study demonstrates that Mn drives proinflammatory events in astrocytes by impairing mitochondrial bioenergetics.


Asunto(s)
Astrocitos/efectos de los fármacos , Encefalitis/inducido químicamente , Manganeso/toxicidad , Mitocondrias/efectos de los fármacos , Animales , Astrocitos/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Encefalitis/metabolismo , Concentración 50 Inhibidora , Masculino , Ratones Endogámicos C57BL , Mitocondrias/metabolismo
13.
Am J Pathol ; 187(7): 1459-1472, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28606756

RESUMEN

Traumatic brain injury due to blast exposure is currently the most prevalent of war injuries. Although secondary ocular blast injuries due to flying debris are more common, primary ocular blast exposure resulting from blast wave pressure has been reported among survivors of explosions, but with limited understanding of the resulting retinal pathologies. Using a compressed air-driven shock tube system, adult male and female C57BL/6 mice were exposed to blast wave pressure of 300 kPa (43.5 psi) per day for 3 successive days, and euthanized 30 days after injury. We assessed retinal tissues using immunofluorescence for glial fibrillary acidic protein, microglia-specific proteins Iba1 and CD68, and phosphorylated tau (AT-270 pThr181 and AT-180 pThr231). Primary blast wave pressure resulted in activation of Müller glia, loss of photoreceptor cells, and an increase in phosphorylated tau in retinal neurons and glia. We found that 300-kPa blasts yielded no detectable cognitive or motor deficits, and no neurochemical or biochemical evidence of injury in the striatum or prefrontal cortex, respectively. These changes were detected 30 days after blast exposure, suggesting the possibility of long-lasting retinal injury and neuronal inflammation after primary blast exposure.


Asunto(s)
Traumatismos por Explosión/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Ondas de Choque de Alta Energía/efectos adversos , Proteínas de Microfilamentos/metabolismo , Enfermedades de la Retina/fisiopatología , Heridas y Lesiones/fisiopatología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Traumatismos por Explosión/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Retina/lesiones , Enfermedades de la Retina/metabolismo , Factores de Tiempo , Heridas y Lesiones/metabolismo , Proteínas tau/metabolismo
14.
J Vis Exp ; (122)2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28447995

RESUMEN

Microglia are the primary responders to central nervous system insults; however, much remains unknown about their role in regulating neuroinflammation. Microglia are mesodermal cells that function similarly to macrophages in surveying inflammatory stress. The classical (M1-type) and alternative (M2-type) activations of macrophages have also been extended to microglia in an effort to better understand the underlying interplay these phenotypes have in neuroinflammatory conditions such as Parkinson's, Alzheimer's, and Huntington's Diseases. In vitro experimentation utilizing primary microglia offers rapid and reliable results that may be extended to the in vivo environment. Although this is a clear advantage over in vivo experimentation, isolating microglia while achieving adequate yields of optimal purity has been a challenge. Common methods currently in use either suffer from low recovery, low purity, or both. Herein, we demonstrate a refinement of the column-free CD11b magnetic separation method that achieves a high cell recovery and enhanced purity in half the amount of time. We propose this optimized method as a highly useful model of primary microglial isolation for the purposes of studying neuroinflammation and neurodegeneration.


Asunto(s)
Antígeno CD11b/química , Separación Celular/métodos , Microglía/fisiología , Recuento de Células , Humanos , Magnetismo
15.
J Neurosci ; 33(21): 9097-103, 2013 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23699520

RESUMEN

The inferior olive (IO) is considered a crucial component of the eyeblink conditioning network. The cerebellar learning hypothesis proposes that the IO provides the cerebellum with a teaching signal that is required for the acquisition and maintenance of conditioned eyeblinks. Supporting this concept, previous experiments showed that lesions or inactivation of the IO blocked CR acquisition. However, these studies were not conclusive. The drawback of the methods used by those studies is that they not only blocked task-related signals, but also completely shut down the spontaneous activity within the IO, which affects the rest of the eyeblink circuits in a nonspecific manner. We hypothesized that more selective blocking of task-related IO signals could be achieved by using injections of glutamate antagonists, which reduce, but do not eliminate, the spontaneous activity in the IO. We expected that if glutamate-mediated IO signals are required for learning, then blocking these signals during training sessions should prevent conditioned response (CR) acquisition. To test this prediction, rabbits were trained to acquire conditioned eyeblinks to a mild vibrissal airpuff as the conditioned stimulus while injections of the glutamate antagonist γ-d-glutamylglycine were administered to the IO. Remarkably, even though this treatment suppressed CRs during training sessions, the postacquisition retention test revealed that CR acquisition had not been abolished. The ability to acquire CRs with IO unconditioned stimulus signals that were blocked or severely suppressed suggests that mechanisms responsible for CR acquisition are extremely resilient and probably less dependent on IO-task-related signals than previously thought.


Asunto(s)
Condicionamiento Palpebral/fisiología , Glutamatos/metabolismo , Núcleo Olivar/fisiología , Estimulación Acústica/efectos adversos , Animales , Condicionamiento Palpebral/efectos de los fármacos , Dipéptidos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Núcleo Olivar/efectos de los fármacos , Conejos , Retención en Psicología/efectos de los fármacos
16.
Behav Brain Res ; 226(1): 189-96, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21933685

RESUMEN

Classical conditioning of the eyeblink response in the rabbit is a form of motor learning whereby the animal learns to respond to an initially irrelevant conditioned stimulus (CS). It is thought that acquired conditioned responses (CRs) are adaptive because they protect the eye in anticipation of potentially harmful events. This protective mechanism is surprisingly inefficient because the acquisition of CRs requires extensive training - a condition that is unlikely to occur in nature. We hypothesized that the rate of conditioning in rabbits could depend on CS modality and that stimulating mystacial vibrissae as the CS could produce CR acquisition faster than the traditional auditory or visual stimulation. We tested this hypothesis by conditioning naïve rabbits in the delay paradigm using a weak airpuff CS (vCS) directed to the ipsilateral mystacial vibrissae. We found that the trigeminal vCS yields significantly faster CR acquisition. We next examined if vCS-evoked CRs are dependent on the intermediate cerebellum in the same fashion as CRs evoked by the traditional auditory CS. We found that vibrissal CRs could be abolished by inactivating the cerebellar interposed nuclei (IN) with muscimol. In addition, injections of picrotoxin in the IN shortened the onset latency of vibrissal CRs. These findings suggest that the tone and vCS-evoked CRs share similar cerebellar dependency.


Asunto(s)
Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Condicionamiento Palpebral/fisiología , Animales , Masculino , Conejos , Vibrisas/fisiología
17.
Brain Res ; 1045(1-2): 175-84, 2005 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15910776

RESUMEN

It is well established that the intermediate cerebellum is involved in the acquisition of classically conditioned eyeblink responses (CRs). Recent studies that inactivated the interposed nuclei (IN) demonstrated that blocking the intermediate cerebellum also interrupts CR extinction. Is this extinction deficit related to interrupting the information flow to efferent targets of the IN? To address this question, we inactivated axons of IN neurons in the brachium conjunctivum (BC). This treatment blocked the output of the intermediate cerebellum without directly affecting neurons in the deep cerebellar nuclei. Rabbits were trained in a delay classical conditioning paradigm, using a tone as the conditioned stimulus (CS) and a corneal air puff as the unconditioned stimulus (US). Then, the BC was microinjected with a sodium channel blocker, tetrodotoxin, during 4 extinction sessions in which rabbits were presented only with the CS. Tests performed after the 4-day injection period revealed that CRs did not extinguish in BC inactivation sessions but extinguished at a normal rate in the absence of the drug. CRs were then re-acquired. These data show that the normal flow of information along axons of cerebellar nuclear cells is required for CR extinction.


Asunto(s)
Tronco Encefálico/fisiología , Núcleos Cerebelosos/fisiología , Condicionamiento Palpebral/fisiología , Vías Eferentes/fisiología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Tronco Encefálico/efectos de los fármacos , Núcleos Cerebelosos/efectos de los fármacos , Condicionamiento Palpebral/efectos de los fármacos , Vías Eferentes/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Modelos Animales , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Estimulación Física , Conejos , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA