Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neuroinflammation ; 21(1): 96, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627764

ABSTRACT

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.


Subject(s)
Cognitive Dysfunction , Sepsis-Associated Encephalopathy , Sepsis , Animals , Male , Mice , Cognitive Dysfunction/metabolism , Dynamins/metabolism , Hippocampus/metabolism , Mice, Inbred C57BL , Sepsis/pathology , Sepsis-Associated Encephalopathy/metabolism , Signal Transduction
2.
J Cell Biochem ; 120(9): 15045-15056, 2019 09.
Article in English | MEDLINE | ID: mdl-31016764

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/chemically induced , JNK Mitogen-Activated Protein Kinases/metabolism , Morphine/pharmacology , Spinal Cord Dorsal Horn/metabolism , Animals , Enzyme Activation , MAP Kinase Signaling System/physiology , Male , Mice , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Mol Pain ; 14: 1744806918761238, 2018.
Article in English | MEDLINE | ID: mdl-29424271

ABSTRACT

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.


Subject(s)
Analgesics/therapeutic use , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Scorpion Venoms/therapeutic use , Sensation , Spinal Cord/enzymology , Analgesics/pharmacology , Animals , Constriction , Disease Models, Animal , Down-Regulation/drug effects , Formaldehyde , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Injections, Spinal , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Neuralgia/complications , Neuralgia/drug therapy , Neuralgia/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-fos/metabolism , Scorpion Venoms/administration & dosage , Scorpion Venoms/pharmacology , Sensation/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology
4.
Anesthesiology ; 112(5): 1234-49, 2010 May.
Article in English | MEDLINE | ID: mdl-20395829

ABSTRACT

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.


Subject(s)
Ephrin-B1/administration & dosage , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Pain/enzymology , Receptors, Eph Family/physiology , Animals , Butadienes/administration & dosage , Ephrin-B1/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/enzymology , Hyperalgesia/pathology , Inflammation Mediators/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Inflammation Mediators/toxicity , Injections, Spinal , MAP Kinase Signaling System/drug effects , Male , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nitriles/administration & dosage , Pain/chemically induced , Pain/pathology , Pain Measurement/methods
5.
Pain ; 139(3): 617-631, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18706764

ABSTRACT

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.


Subject(s)
Ephrin-B1/physiology , Hyperalgesia/physiopathology , MAP Kinase Signaling System , Receptors, Eph Family/physiology , Animals , Anthracenes/pharmacology , Butadienes/pharmacology , Dizocilpine Maleate/pharmacology , Ephrin-B1/pharmacology , Foot , Formaldehyde/toxicity , Hot Temperature/adverse effects , Hyperalgesia/etiology , Injections, Spinal , MAP Kinase Signaling System/drug effects , Male , Mice , Nitriles/pharmacology , Physical Stimulation/adverse effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Receptors, Eph Family/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Spinal Cord/chemistry , Spinal Cord/physiopathology , Touch , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...