Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Neurochem ; 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37381043

RESUMEN

The receptor for advanced glycation end products (RAGE) is a transmembrane receptor that belongs to the immunoglobulin superfamily and is extensively associated with chronic inflammation in non-transmissible diseases. As chronic inflammation is consistently present in neurodegenerative diseases, it was largely assumed that RAGE could act as a critical modulator of neuroinflammation in Parkinson's disease (PD), similar to what was reported for Alzheimer's disease (AD), where RAGE is postulated to mediate pro-inflammatory signaling in microglia by binding to amyloid-ß peptide. However, accumulating evidence from studies of RAGE in PD models suggests a less obvious scenario. Here, we review physiological aspects of RAGE and address the current questions about the potential involvement of this receptor in the cellular events that may be critical for the development and progression of PD, exploring possible mechanisms beyond the classical view of the microglial activation/neuroinflammation/neurodegeneration axis that is widely assumed to be the general mechanism of RAGE action in the adult brain.

2.
J Neurochem ; 2023 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-37661637

RESUMEN

The receptor for advanced glycation end products (RAGE) is a protein of the immunoglobulin superfamily capable of regulating inflammation. Considering the role of this receptor in the initiation and establishment of neuroinflammation, and the limited understanding of the function of RAGE in the maintenance of this condition, this study describes the effects of RAGE inhibition in the brain, through an intranasal treatment with the antagonist FPS-ZM1, in an animal model of chronic neuroinflammation induced by acute intraperitoneal injection of lipopolysaccharide (LPS). Seventy days after LPS administration (2 mg/kg, i.p.), Wistar rats received, intranasally, 1.2 mg of FPS-ZM1 over 14 days. On days 88 and 89, the animals were submitted to the open-field test and were killed on day 90 after the intraperitoneal injection of LPS. Our results indicate that blockade of encephalic RAGE attenuates LPS-induced chronic neuroinflammation in different brain regions. Furthermore, we found that intranasal FPS-ZM1 administration reduced levels of gliosis markers, RAGE ligands, and α-synuclein in the substantia nigra pars compacta. Additionally, the treatment also reversed the increase in S100 calcium-binding protein B (RAGE ligand) in the cerebrospinal fluid and the cognitive-behavioral deficits promoted by LPS-less time spent in the central zone of the open-field arena (more time in the lateral zones), decreased total distance traveled, and increased number of freezing episodes. In summary, our study demonstrates the prominent role of RAGE in the maintenance of a chronic neuroinflammatory state triggered by a single episode of systemic inflammation and also points to possible future RAGE-based therapeutic approaches to treat conditions in which chronic neuroinflammation and increased α-synuclein levels could play a relevant role, such as in Parkinson's disease.

3.
J Biol Chem ; 297(2): 100979, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34303703

RESUMEN

Schistosomiasis, a neglected tropical disease caused by trematodes of the Schistosoma genus, affects over 250 million people around the world. This disease has been associated with learning and memory deficits in children, whereas reduced attention levels, impaired work capacity, and cognitive deficits have been observed in adults. Strongly correlated with poverty and lack of basic sanitary conditions, this chronic endemic infection is common in Africa, South America, and parts of Asia and contributes to inhibition of social development and low quality of life in affected areas. Nonetheless, studies on the mechanisms involved in the neurological impairment caused by schistosomiasis are scarce. Here, we used a murine model of infection with Schistosoma mansoni in which parasites do not invade the central nervous system to evaluate the consequences of systemic infection on neurologic function. We observed that systemic infection with S. mansoni led to astrocyte and microglia activation, expression of oxidative stress-induced transcription factor Nrf2, oxidative damage, Tau phosphorylation, and amyloid-ß peptide accumulation in the prefrontal cortex of infected animals. We also found impairment in spatial learning and memory as evaluated by the Morris water maze task. Administration of anthelmintic (praziquantel) and antioxidant (N-acetylcysteine plus deferoxamine) treatments was effective in inhibiting most of these phenotypes, and the combination of both treatments had a synergistic effect to prevent such changes. These data demonstrate new perspectives toward the understanding of the pathology and possible therapeutic approaches to counteract long-term effects of systemic schistosomiasis on brain function.


Asunto(s)
Astrocitos/patología , Microglía/patología , Enfermedades Neurodegenerativas/patología , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis mansoni/complicaciones , Acetilcisteína/farmacología , Animales , Antihelmínticos/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/farmacología , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Praziquantel/farmacología , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/patología , Sideróforos/farmacología
4.
Chemosphere ; 351: 141239, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272134

RESUMEN

Mercury (Hg) and vitamin A (VitA) are two environmental factors with potential health impacts, especially during pregnancy and early childhood. Fish and seafood may present elevated levels of methylmercury (MeHg), the major Hg derivative, and VitA. This study aimed to evaluate the transgenerational effects of exposure to MeHg and/or VitA on epigenetic and toxicological parameters in a Wistar rat model. Our findings revealed persistent toxicological effects in generations F1 and F2 following low/mild doses of MeHg and/or VitA exposure during dams' (F0) gestation and breastfeeding. Toxicological effects observed in F2 included chronic DNA damage, bone marrow toxicity, altered microglial content, reduced neuronal signal, and diminished male longevity. Sex-specific patterns were also observed. Co-exposure to MeHg and VitA showed both synergistic and antagonistic effects. Additionally, the study demonstrated that MeHg and VitA affected histone methylation and caused consistent effects in F2. While MeHg exposure has been associated with transgenerational inheritance effects in other organisms, this study provides the first evidence of transgenerational inheritance of MeHg and VitA-induced toxicological effects in rodents. Although the exact mechanism is not yet fully understood, these findings suggest that MeHg and VitA may perpetuate their impacts across generations. The study highlights the need for remedial policies and interventions to mitigate the potential health problems faced by future generations exposed to MeHg or VitA. Further research is warranted to investigate the transgenerational effects beyond F2 and determine the matrilineal or patrilineal inheritance patterns.


Asunto(s)
Mercurio , Compuestos de Metilmercurio , Humanos , Preescolar , Ratas , Animales , Embarazo , Femenino , Masculino , Compuestos de Metilmercurio/toxicidad , Ratas Wistar , Vitamina A , Metilación
5.
Brain Behav Immun Health ; 14: 100253, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34589762

RESUMEN

HSP70 is one of the main molecular chaperones involved in the cellular stress response. Besides its chaperone action, HSP70 also modulates the immune response. Increased susceptibility to toxic insults in intra- and extracellular environments has been associated with insufficient amounts of inducible HSP70 in adult neurons. On the other hand, exogenous HSP70 administration has demonstrated neuroprotective effects in experimental models of age-related disorders. In this regard, this study investigated the effects of exogenous HSP70 in an animal model of dopaminergic denervation of the nigrostriatal axis. After unilateral intrastriatal injection with 6-hydroxydopamine (6-OHDA), the animals received purified recombinant HSP70 through intranasal administration (2 µg/rat/day) for 15 days. Our results indicate a neuroprotective effect of intranasal HSP70 against dopaminergic denervation induced by 6-OHDA. Exogenous HSP70 improved motor impairment and reduced the loss of dopaminergic neurons caused by 6-OHDA. Moreover, HSP70 modulated neuroinflammatory response in the substantia nigra, an important event in Parkinson's disease pathogenesis. Specifically, HSP70 treatment reduced microglial activation and astrogliosis induced by 6-OHDA, as well as IL-1ß mRNA expression in this region. Also, recombinant HSP70 increased the protein content of HSP70 in the substantia nigra of rats that received 6-OHDA. These data suggest the neuroprotection of HSP70 against dopaminergic neurons damage after cellular stress. Finally, our results indicate that HSP70 neuroprotective action against 6-OHDA toxicity is related to inflammatory response modulation.

6.
Mol Neurobiol ; 56(5): 3079-3089, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30094805

RESUMEN

The receptor for advanced glycation endproducts (RAGE) is a transmembrane, immunoglobulin-like receptor that interacts with a broad repertoire of extracellular ligands. RAGE belongs to a family of cell adhesion molecules and is considered a key receptor in the inflammation axis and a potential contributor to the neurodegeneration. The present study aimed to investigate the content and cell localization of RAGE in the brain of Wistar rats subjected to systemic inflammation induced by a single dose of lipopolysaccharide (LPS, 5 mg/kg, i.p.). Fifteen days after LPS administration, the content of RAGE was analyzed in the prefrontal cortex (PFC), hippocampus (HIPP), cerebellum (CB), and substantia nigra (SN) were investigated. RAGE levels increased in all structures, except HIPP; however, immunohistochemistry analysis demonstrated that the cell site of RAGE expression changed from blood vessel-like structures to neuronal cells in all brain areas. Besides, the highest level of RAGE expression was found in SN. Immunofluorescence analysis in SN confirmed that RAGE expression was mainly co-localized in endothelial cells (RAGE/PECAM-1 co-staining) in untreated animals, while LPS-treated animals had RAGE expression predominantly in dopaminergic neurons (RAGE/TH co-staining). Decreased TH levels, as well as increased pro-inflammatory markers (TNF-α, IL-1ß, Iba-1, GFAP, and phosphorylated ERK1/2) in SN, occurred concomitantly to RAGE stimulation in the same site. These results suggest a role for RAGE in the establishment of a neuroinflammation-neurodegeneration axis that develops as a long-term response to systemic inflammation by LPS.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Células Endoteliales/metabolismo , Inflamación/metabolismo , Neuronas/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Biomarcadores/metabolismo , Neuronas Dopaminérgicas/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Masculino , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas Wistar , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
7.
Neurochem Int ; 126: 27-35, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30849398

RESUMEN

Carvacrol (CARV) presents valuable biological properties such as anti-inflammatory and antioxidant activities. However, pharmacological uses of CARV are largely limited due to disadvantages related to solubility, bioavailability, preparation and storage processes. The complexation of monoterpenes with ß-cyclodextrin (ß-CD) increases their stability, solubility and oral bioavailability. Here, the protective effect of oral treatment with CARV/ß-CD complex (25 µg/kg/day) against dopaminergic (DA) denervation induced by unilateral intranigral injection of 6-hydroxydopamine (6-OHDA - 10 µg per rat) was analyzed, in order to evaluate a putative application in the development of neuroprotective therapies for Parkinson's disease (PD). Pretreatment with CARV/ß-CD for 15 days prevented the loss of DA neurons induced by 6-OHDA in adult Wistar rats. This effect may occur through CARV anti-inflammatory and antioxidant properties, as the pretreatment with CARV/ß-CD inhibited the release of IL-1ß and TNF-α; besides, CARV prevented the increase of mitochondrial superoxide production induced by 6-OHDA in cultured SH-SY5Y cells. Importantly, hepatotoxicity or alterations in blood cell profile were not observed with oral administration of CARV/ß-CD. Therefore, this study showed a potential pharmacological application of CARV/ß-CD in PD using a non-invasive route of drug delivery, i.e., oral administration.


Asunto(s)
Cimenos/administración & dosificación , Desnervación/efectos adversos , Neuronas Dopaminérgicas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Oxidopamina/toxicidad , beta-Ciclodextrinas/administración & dosificación , Administración Oral , Animales , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Combinación de Medicamentos , Masculino , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
8.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 317-328, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30529222

RESUMEN

Retinoic acid (RA) promotes differentiation in multiple neurogenic cell types by promoting gene reprogramming through retinoid receptors and also by inducing cytosolic signaling events. The nuclear RXR receptors are one of the main mediators of RA cellular effects, classically by joining the direct receptors of RA, the nuclear RAR receptors, in RAR/RXR dimers which act as transcription factors. Distinct RXR genes lead to RXRα, RXRß and RXRγ subtypes, but their specific roles in neuronal differentiation remain unclear. We firstly investigated both RXRs and RARs expression profiles during RA-mediated neuronal differentiation of human neuroblastoma cell line SH-SY5Y, and found varying levels of retinoid receptors transcript and protein contents along the process. In order to understand the roles of the expression of distinct RXR subtypes to RA signal transduction, we performed siRNA-mediated silencing of RXRα and RXRß during the first stages of SH-SY5Y differentiation. Our results showed that RXRα is required for RA-induced neuronal differentiation of SH-SY5Y cells, since its silencing compromised cell cycle arrest and prevented the upregulation of neuronal markers and the adoption of neuronal morphology. Besides, silencing of RXRα affected the phosphorylation of ERK1/2. By contrast, silencing of RXRß improved neurite extension and led to increased expression of tau and synaptophysin, suggesting that RXRß may negatively regulate neuronal parameters related to neurite outgrowth and function. Our results indicate distinct functions for RXR subtypes during RA-dependent neuronal differentiation and reveal new perspectives for studying such receptors as clinical targets in therapies aiming at restoring neuronal function.


Asunto(s)
Neuritas/metabolismo , Receptor alfa X Retinoide/fisiología , Receptor beta X Retinoide/fisiología , Animales , Puntos de Control del Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/metabolismo , Receptores de Ácido Retinoico/fisiología , Receptor alfa X Retinoide/metabolismo , Receptor beta X Retinoide/metabolismo , Receptores X Retinoide , Transducción de Señal/efectos de los fármacos , Activación Transcripcional , Tretinoina/metabolismo , Tretinoina/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA