Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 96, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627764

RESUMEN

BACKGROUND: Gasdermin D (GSDMD)-mediated pyroptotic cell death is implicated in the pathogenesis of cognitive deficits in sepsis-associated encephalopathy (SAE), yet the underlying mechanisms remain largely unclear. Dynamin-related protein 1 (Drp1) facilitates mitochondrial fission and ensures quality control to maintain cellular homeostasis during infection. This study aimed to investigate the potential role of the GSDMD/Drp1 signaling pathway in cognitive impairments in a mouse model of SAE. METHODS: C57BL/6 male mice were subjected to cecal ligation and puncture (CLP) to establish an animal model of SAE. In the interventional study, mice were treated with the GSDMD inhibitor necrosulfonamide (NSA) or the Drp1 inhibitor mitochondrial division inhibitor-1 (Mdivi-1). Surviving mice underwent behavioral tests, and hippocampal tissues were harvested for histological analysis and biochemical assays at corresponding time points. Haematoxylin-eosin staining and TUNEL assays were used to evaluate neuronal damage. Golgi staining was used to detect synaptic dendritic spine density. Additionally, transmission electron microscopy was performed to assess mitochondrial and synaptic morphology in the hippocampus. Local field potential recordings were conducted to detect network oscillations in the hippocampus. RESULTS: CLP induced the activation of GSDMD, an upregulation of Drp1, leading to associated mitochondrial impairment, neuroinflammation, as well as neuronal and synaptic damage. Consequently, these effects resulted in a reduction in neural oscillations in the hippocampus and significant learning and memory deficits in the mice. Notably, treatment with NSA or Mdivi-1 effectively prevented these GSDMD-mediated abnormalities. CONCLUSIONS: Our data indicate that the GSDMD/Drp1 signaling pathway is involved in cognitive deficits in a mouse model of SAE. Inhibiting GSDMD or Drp1 emerges as a potential therapeutic strategy to alleviate the observed synaptic damages and network oscillations abnormalities in the hippocampus of SAE mice.


Asunto(s)
Disfunción Cognitiva , Encefalopatía Asociada a la Sepsis , Sepsis , Animales , Masculino , Ratones , Disfunción Cognitiva/metabolismo , Dinaminas/metabolismo , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Sepsis/patología , Encefalopatía Asociada a la Sepsis/metabolismo , Transducción de Señal
2.
Macromol Rapid Commun ; 44(17): e2300170, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37243910

RESUMEN

Herein, a novel tough luminescent hydrogel with Europium is fabricated using a facile copolymerization process by introducing 2,2':6',2-terpyridine (TPy) into a dual physical cross-linked hydrogel. The obtained P(NAGA-co-MAAc)/Eu/TPy (x) (x refers to the feed ratio of NAGA to MAAc) hydrogels not only show outstanding mechanical performances (fracture strength, ≈2.5 MPa), but also give a special ability of rapid detection to low concentrations of zinc ions. Attractively, the theoretical limits of detection (LOD) of the hydrogel sensors are calculated as 1.6 µm, which is acceptable within the WHO limit. Furthermore, the continuous change in fluorescence of P(NAGA-co-MAAc)/Eu/TPy (10) strips upon contact with Zn2+ can be clearly observed by the naked eyes with the aid of a portable UV lamp, resulting in semi-quantitative naked-eyes detection through a standard colorimetric card. Moreover, by identifying the RGB value of the hydrogel sensor, it can also realize quantitative analysis. Therefore, excellence in sensing, simplicity in structure, and convenience in using make P(NAGA-co-MAAc)/Eu/TPy (10) hydrogel as a superior fluorescent chemosensor of Zn2+ ions.


Asunto(s)
Hidrogeles , Luminiscencia , Hidrogeles/química , Iones/química , Fluorescencia , Zinc/química
3.
Neuroreport ; 34(4): 220-231, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36719835

RESUMEN

OBJECTIVES: This study was aimed at evaluating the effects of dexpramipexole (DPX) - a mitochondrial protectant that sustains mitochondrial function and energy production - on cognitive function in a mouse model of sepsis-associated encephalopathy (SAE) induced by peripheral administration of lipopolysaccharide (LPS) and examining the potential mechanisms. METHODS: C57BL/6 male mice were randomized into one of four treatment protocols: Con+Sal, Con+DPX, LPS+Sal or LPS+DPX. The mice were intraperitoneally (i.p.) injected with LPS or equivalent volumes of normal saline once daily for 3 consecutive days. To evaluate the protective effects of DPX, we administered DPX or normal saline i.p. to the mice once daily for 6 consecutive days. Six mice in each group were decapitated on day 7, and each brain was rapidly removed and separated into two halves for biochemical and histochemical analysis. The remaining surviving mice in each group were subjected to behavioral tests from days 7 to 10. RESULTS: Peripheral administration of LPS to mice led to learning and memory deficits in behavioral tests, which were associated with mitochondrial impairment and ATP depletion in the hippocampus. Repeated DPX treatment protected the mitochondria against LPS-induced morphological and functional impairment; inhibited the activation of the Nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome-caspase-1-dependent pyroptosis pathway and cytochrome c (Cyt-c)-caspase-3-dependent apoptosis pathway; and attenuated LPS-induced neuroinflammation and cell death in the hippocampus in SAE mice. CONCLUSIONS: Mitochondria-mediated pyroptosis and apoptosis are involved in the pathogenesis of cognitive deficits in a mouse model of SAE and DPX protects mitochondria and suppresses the mitochondria-medicated pyroptosis and apoptosis pathways, and ameliorates LPS-induced neuroinflammation and cognitive deficits. This study provides theoretical evidence supporting DPX for the treatment of SAE.


Asunto(s)
Encefalopatía Asociada a la Sepsis , Masculino , Ratones , Animales , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Piroptosis , Pramipexol , Lipopolisacáridos/toxicidad , Enfermedades Neuroinflamatorias , Solución Salina/metabolismo , Solución Salina/farmacología , Ratones Endogámicos C57BL , Apoptosis , Cognición , Mitocondrias/metabolismo
4.
Int Immunopharmacol ; 82: 106317, 2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32087497

RESUMEN

Perioperative neurocognitive disorders (PND) are characterized by deficits in cognitive functions in the elderly following anesthesia and surgery. Effective clinical interventions for preventing this disease are limited. Growing evidence demonstrates that activation of NOD-like receptor protein3 (NLRP3) inflammasome is involved in neurodegenerative diseases. We therefore hypothesized that activation of NLRP3 inflammasome is linked to neuroinflammation and the subsequent cognitive impairments that occurred in an animal model of PND. In this study, 18-month-old C57BL/6 mice were subjected to an exploratory laparotomy under isoflurane anesthesia to mimic clinical human abdominal surgery. For interventional studies, mice received NLRP3 specific inhibitor MCC950 (10 mg/kg) or the vehicle only intraperitoneally. Behavioral studies were performed at 6 and 7 d after surgery using open field and fear conditioning tests, respectively. Interleukin-1ß (IL-1ß), interleukin-18 (IL-18), tumor necrosis factor-α (TNF-α), ionized calcium-binding adaptor molecule-1 (IBA1) positive cells, glial fibrillary acidic protein (GFAP) positive cells, NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), and cleaved caspase-1 were measured at 3 days post-surgery. Brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95) were measured at 7 days post-surgery. Our data indicates that surgery-induced cognitive impairments were associated with significant increases in IL-1ß, IL-18, TNF-α, NLRP3, ASC, cleaved caspase-1, IBA1-positive cells and GFAP-positive cells, and decreases in BDNF and PSD95 expression in the hippocampus. Notably, administration with MCC950 attenuated inflammatory changes and rescued surgery-induced cognitive impairments. Our study suggests that surgery induces neuroinflammation and cognitive deficits that are partly attributed to the activation of NLRP3 inflammasome in the hippocampus of aged mice.

5.
J Cell Biochem ; 120(9): 15045-15056, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31016764

RESUMEN

BACKGROUND: This study investigated the activation of mitogen-activated protein kinases in the spinal dorsal horn to explore the mechanisms underlying morphine-induced acute and chronic hyperalgesia in mice. METHODS: Male adult mice were given a single subcutaneous injection (SC) of morphine (1 µg/kg) or twice-daily administration of morphine (10 mg/kg/day) for 8 days. Thermal hyperalgesia and mechanical allodynia were assessed using the radiant heat and von Frey filament test. Levels of phospho (p)-extracellular signal-regulated kinases (p-ERK), p-c-Jun N-terminal kinase (p-JNK), p-p38, p-PKCγ, N-methyl-d-aspartate receptor (NMDAr), and c-Fos protein in the spinal dorsal horn were examined by Western blot assays. RESULTS: A single ultra-low dose or repeated administration of morphine induced hyperalgesia in mice and caused a significant increase in the levels of p-ERK and p-JNK, but not p-p38, in the spinal dorsal horn. The level of c-Fos protein was significantly elevated following administration of morphine. The protein levels of p-PKCγ and NMDAr subunits (NR2B and NR2A) were also altered. Pretreatment with the NMDAr antagonist MK-801 or the protein kinase C (PKC) inhibitor calphostin C (CC) suppressed the morphine-induced increase in p-ERK, p-JNK, and c-Fos. Administration of MK-801 and CC also relieved morphine-induced hyperalgesia. CONCLUSION: These findings suggest that activation of the spinal ERK and JNK signaling pathways contribute to morphine-induced acute and chronic hyperalgesia in mice.


Asunto(s)
Analgésicos Opioides/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hiperalgesia/inducido químicamente , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Morfina/farmacología , Asta Dorsal de la Médula Espinal/metabolismo , Animales , Activación Enzimática , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Mol Pain ; 14: 1744806918761238, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29424271

RESUMEN

Background Several studies have shown that scorpion venom peptide BmK AGAP has an analgesic activity. Our previous study also demonstrated that intraplantar injection of BmK AGAP ameliorates formalin-induced spontaneous nociceptive behavior. However, the effect of intrathecal injection of BmK AGAP on nociceptive processing is poorly understood. Methods We investigated the effects of intrathecal injection of BmK AGAP on spinal nociceptive processing induced by chronic constrictive injury or formalin. Thermal hyperalgesia and mechanical allodynia were measured using radiant heat and the von Frey filaments test. Formalin-induced spontaneous nociceptive behavior was also investigated. C-Fos expression was assessed by immunohistochemistry. Phosphorylated mitogen-activated protein kinase (p-MAPK) expression was monitored by Western blot assay. Results Intrathecal injection of BmK AGAP reduced chronic constrictive injury-induced neuropathic pain behavior and pain from formalin-induced inflammation, accompanied by decreased expression of spinal p-MAPKs and c-Fos protein. The results of combining low doses of different MAPK inhibitor (U0126, SP600125, or SB203580; 0.1 µg for each inhibitor) with a low dose of BmK AGAP (0.2 µg) suggested that BmK AGAP could potentiate the effects of MAPK inhibitors on inflammation-associated pain. Conclusion Our results demonstrate that intrathecal injection of BmK AGAP produces a sensory-specific analgesic effect via a p-MAPK-dependent mechanism.


Asunto(s)
Analgésicos/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Venenos de Escorpión/uso terapéutico , Sensación , Médula Espinal/enzimología , Analgésicos/farmacología , Animales , Constricción , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Formaldehído , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/patología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/patología , Inyecciones Espinales , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Neuralgia/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-fos/metabolismo , Venenos de Escorpión/administración & dosificación , Venenos de Escorpión/farmacología , Sensación/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
7.
ScientificWorldJournal ; 2014: 654183, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386611

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) were analyzed for surface sediments and a sediment core from the Yellow River-dominated margin. The concentration of 16 USEPA priority PAHs in surface sediments ranged from 5.6 to 175.4 ng g(-1) dry weight sediment (dws) with a mean of 49.1 ng g(-1) dws. From 1930 to 2011, the distribution of PAHs (37.2 to 210.6 ng g(-1) dws) was consistent with the socioeconomic development of China. The PAHs' concentration peaked in 1964 and 1986, corresponding to the rapid economic growth in China (1958-1965) and the initiation of the "Reform and Open" policy in 1978, respectively. The applications of molecular diagnostic ratios and principal component analysis suggest that PAHs are predominantly produced by the coal and biomass combustion, whereas the contribution of petroleum combustions slightly increased after the 1970s, synchronous with an increasing usage of oil and gas in China.


Asunto(s)
Monitoreo del Ambiente , Sedimentos Geológicos/química , Hidrocarburos Policíclicos Aromáticos/aislamiento & purificación , Contaminantes Químicos del Agua/aislamiento & purificación , China , Humanos , Petróleo/toxicidad , Hidrocarburos Policíclicos Aromáticos/química , Análisis de Componente Principal , Ríos/química , Contaminantes Químicos del Agua/química
8.
PLoS One ; 8(11): e78239, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244296

RESUMEN

In the present study, we investigated the anti-nociceptive effect and the underlying mechanism of the analgesic-antitumor peptide (AGAP), a neurotoxin from the scorpion Buthus martensii Karsch. AGAP in doses of 0.2, 1 and 5 µg was injected intraplantarly (i.pl.) before formalin injection 10 min at the same site. The suppression by intraplantar injection of AGAP on formalin-induced spontaneous nociceptive behaviors was investigated. The results show that AGAP could dose-dependently inhibit formalin-induced two-phase spontaneous flinching response. To investigate the mechanism of action of treatment with AGAP in inflammatory pain, the expressions of peripheral and spinal phosphorylated mitogen-activated protein kinases (phospho-MAPKs) including p-p38, p-ERK and p-JNK were examined. We found that formalin increased the expressions of peripheral and spinal MAPKs, which were prevented by pre-intraplantar injection of AGAP in inflammation pain model in mice. AGAP could also decrease the expression of spinal Fos induced by formalin. Furthermore, combinations the lower doses of the inhibitors of MAPKs (U0126, SP600125, or SB203580 0.1 µg) with the lower dose of AGAP (0.2 µg), the results suggested that AGAP could potentiate the effects of the inhibitors of MAPKs on the inflammatory pain. The present results indicate that pre-intraplantar injection of AGAP prevents the inflammatory pain induced by formalin through a MAPKs-mediated mechanism in mice.


Asunto(s)
Analgésicos/farmacología , Antineoplásicos/farmacología , Proteínas de Artrópodos/farmacología , Desinfectantes/efectos adversos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Formaldehído/efectos adversos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neurotoxinas/farmacología , Dolor/prevención & control , Péptidos/farmacología , Venenos de Escorpión/farmacología , Escorpiones/química , Analgésicos/química , Animales , Antineoplásicos/química , Proteínas de Artrópodos/química , Desinfectantes/farmacología , Relación Dosis-Respuesta a Droga , Formaldehído/farmacología , Inflamación/inducido químicamente , Inflamación/patología , Inflamación/prevención & control , Masculino , Ratones , Neurotoxinas/química , Dolor/inducido químicamente , Dolor/patología , Péptidos/química , Venenos de Escorpión/química
9.
Anesthesiology ; 112(5): 1234-49, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20395829

RESUMEN

BACKGROUND: Our previous studies have demonstrated that EphBs receptors and ephrinBs ligands were involved in modulation of spinal nociceptive information. However, the downstream mechanisms that control this process are not well understood. The aim of this study was to further investigate whether mitogen-activated protein kinases (MAPKs), as the downstream effectors, participate in modulation of spinal nociceptive information related to ephrinBs/EphBs. METHODS: Thermal hyperalgesia and mechanical allodynia were measured using radiant heat and von Frey filaments test. Immunofluorescence staining was used to detect the expression of p-MAPKs and of p-MAPKs/neuronal nuclei, or p-MAPKs/glial fibrillary acidic protein double label. C-Fos expression was determined by immunohistochemistry. The expression of p-MAPKs was also determined by Western blot assay. RESULTS: Intrathecal injection of ephrinB1-Fc produced a dose- and time-dependent thermal and mechanical hyperalgesia, accompanied by the increase of spinal p-MAPKs and c-Fos expression. Immunofluorescence staining revealed that p-MAPKs colocalized with the neuronal marker (neuronal nuclei) and the astrocyte marker (glial fibrillary acidic protein). Inhibition of MAPKs prevented and reversed pain behaviors and the increase of spinal c-Fos expression induced by intrathecal injection of ephrinB1-Fc. Inhibition of EphBs receptors by intrathecal injection of EphB1-Fc reduced formalin-induced inflammation and chronic constrictive injury-induced neuropathic pain behaviors accompanied by decreased expression of spinal p-MAPKs and c-Fos protein. Furthermore, pretreatment with MK-801, an N-methyl-d-aspartate receptor antagonist, prevented behavioral hyperalgesia and activation of spinal MAPKs induced by intrathecal injection of ephrinB1-Fc. CONCLUSIONS: These results demonstrated that activation of MAPKs contributed to modulation of spinal nociceptive information related to ephrinBs/EphBs.


Asunto(s)
Efrina-B1/administración & dosificación , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Dolor/enzimología , Receptores de la Familia Eph/fisiología , Animales , Butadienos/administración & dosificación , Efrina-B1/toxicidad , Hiperalgesia/inducido químicamente , Hiperalgesia/enzimología , Hiperalgesia/patología , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , Mediadores de Inflamación/toxicidad , Inyecciones Espinales , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nitrilos/administración & dosificación , Dolor/inducido químicamente , Dolor/patología , Dimensión del Dolor/métodos
10.
Pain ; 139(3): 617-631, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18706764

RESUMEN

EphBs receptors and ephrinBs ligands are present in the adult brain and peripheral tissue and play a critical role in modulating multiple aspects of physiology and pathophysiology. Ours and other studies have demonstrated that spinal ephrinBs/EphBs signaling was involved in the modulation of nociceptive information and central sensitization. However, the role of ephrinBs/EphBs signaling in peripheral sensitization is poorly understood. This study shows that intraplantar (i.pl.) injection of ephrinB1-Fc produces a dose- and time-dependent thermal and mechanical hyperalgesia and the increase of spinal Fos protein expression in mice, which can be partially prevented by pre-treatment with EphB1-Fc. EphrinB1-Fc-induced hyperalgesia is accompanied with the NMDA receptor-mediated increase of expression in peripheral and spinal phosphorylated mitogen-activated protein kinases (phospho-MAPKs) including p-p38, pERK and pJNK, and also is prevented or reversed by the inhibition of peripheral and spinal MAPKs. Furthermore, in formalin inflammation pain model, pre-inhibition of EphBs receptors by the injection of EphB1-Fc reduces pain behavior, which is accompanied by the decreased expression of peripheral p-p38, pERK and pJNK. These data provide evidence that ephrinBs may act as a prominent contributor to peripheral sensitization, and demonstrate that activation of peripheral ephrinBs/EphBs system induces hyperalgesia through a MAPKs-mediated mechanism.


Asunto(s)
Efrina-B1/fisiología , Hiperalgesia/fisiopatología , Sistema de Señalización de MAP Quinasas , Receptores de la Familia Eph/fisiología , Animales , Antracenos/farmacología , Butadienos/farmacología , Maleato de Dizocilpina/farmacología , Efrina-B1/farmacología , Pie , Formaldehído/toxicidad , Calor/efectos adversos , Hiperalgesia/etiología , Inyecciones Espinales , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Nitrilos/farmacología , Estimulación Física/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fos/análisis , Receptores de la Familia Eph/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Médula Espinal/química , Médula Espinal/fisiopatología , Tacto , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...