Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Sleep Med ; 121: 287-294, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39038404

ABSTRACT

BACKGROUND: Postoperative sleep disturbance (PSD) is prevalent in perioperative patients,and has significant impact on postoperative recovery and prognosis. The aim of this study was to investigate the effect of desflurane maintenance on postoperative sleep quality, in order to optimize patients' perioperative sleep management. METHOD: A total of 118 patients undergoing elective breast surgery were randomized to receive either desflurane-based volatile anesthesia (desflurane group) or propofol-based total intravenous anesthesia (propofol group) for anesthesia maintenance. The primary outcome was the quality of sleep, which was assessed by the Pittsburgh Sleep Quality Index (PSQI) on 3 days after operation (POD3). Secondary outcomes were PSQI on postoperative day 7 (POD7) and 30 (POD30), and postoperative anxiety, depression, and pain score, as well as objective sleep parameters including total sleep time (TST), WASO (Wakefulness after sleep onset), REM (Rapid eye movement) and NREM (Non-rapid Eye Movement) measured by Fitbit Charge 2TM during the initial 3 postoperative days. RESULTS: The global PSQI scores on POD3 in the desflurane group was non-inferior to that in the propofol group [mean (SD) 8.47 (3.46) vs. 7.65 (3.16); mean difference (95 % CI) 0.82 (-0.43, 2.07); p < 0.001 for non-inferiority]. There were no significant differences in PSQI scores on POD3 and POD7. In addition, the score of anxiety, depression, and pain on the 3rd, 7th, and 30th day after surgery have no significant differences between the propofol and the desflurane group, respectively. The postoperative NREM was higher in the desflurane group than that in the propofol group. CONCLUSION: The effects of desflurane-based volatile anesthesia maintenance on postoperative sleep quality is not inferior to that of propofol-based total intravenous anesthesia, and these two drugs may have different effects on the sleep structure. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04805775.


Subject(s)
Anesthetics, Inhalation , Desflurane , Propofol , Sleep Quality , Humans , Desflurane/administration & dosage , Female , Propofol/administration & dosage , Middle Aged , Adult , Elective Surgical Procedures , Breast/surgery , Anesthetics, Intravenous , Postoperative Complications/prevention & control , Postoperative Period
2.
Sleep Breath ; 28(3): 1365-1372, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38499834

ABSTRACT

BACKGROUND AND PURPOSE: Bispectral Index (BIS) and University of Michigan Sedation Scale (UMSS) were two commonly used methods of monitoring the sedation depth, but their correlation was not clear. The purpose of this study is to ascertain if BIS correlates with UMSS in determining the sedation level during pediatric drug-induced sleep endoscopy (DISE). METHODS: One-hundred children, aged 36-143 months, with ASA I~II grade, were enrolled. They were subject to general anesthesia for an elective adenotonsillectomy. Two drug regimens were used. After UMSS ≥ 3, the sites of airway obstructions were located by checking the supraglottic airway structures with a fibrous laryngoscope. UMSS scores, BIS values, electromyography (EMG), and signal quality indices (SQIs) were recorded at the pre-medication and pre-DISE baseline (T0), 5 min subsequent to medication administration but prior to DISE initiation (T1), 1 min after DISE was initiated (T2), 1 min after DISE was completed (T3), 1 min subsequent to tracheal intubation (T4), 1 min following extubation (T5), and 30 min past extubation (T6). RESULTS: There were strong correlations between BIS monitor readings and UMSS scores for total and two regimens. Kappa values revealed moderate agreement between BIS and UMSS for total and two regimens. The agreement rates were 67.47% for the total, 61.43% for Regimen 1, and 73.42% for Regimen 2, respectively. CONCLUSION: BIS correlates with UMSS in determining the sedation level during pediatric DISE for two regimens. BIS might serve as an appropriate indicator of sedation intensity when UMSS could not be used.


Subject(s)
Conscious Sedation , Endoscopy , Tonsillectomy , Humans , Male , Female , Child , Child, Preschool , Adenoidectomy , Hypnotics and Sedatives/administration & dosage , Consciousness Monitors , Anesthesia, General , Electromyography
3.
J Cell Mol Med ; 27(23): 3706-3716, 2023 12.
Article in English | MEDLINE | ID: mdl-37950418

ABSTRACT

Excessive productions of inflammatory cytokines and free radicals are involved in spinal cord injury (SCI). Fibroblast growth factor 5 (FGF5) is associated with inflammatory response and oxidative damage, and we herein intend to determine its function in SCI. Lentivirus was instilled to overexpress or knockdown FGF5 expression in mice. Compound C or H89 2HCl were used to suppress AMP-activated protein kinase (AMPK) or protein kinase A (PKA), respectively. FGF5 level was significantly decreased during SCI. FGF5 overexpression mitigated, while FGF5 silence further facilitated inflammatory response, oxidative damage and SCI. Mechanically, FGF5 activated AMPK to attenuate SCI in a cAMP/PKA-dependent manner, while inhibiting AMPK or PKA with pharmacological methods significantly abolished the neuroprotective effects of FGF5 against SCI. More importantly, serum FGF5 level was decreased in SCI patients, and elevated serum FGF5 level often indicate better prognosis. Our study identifies FGF5 as an effective therapeutic and prognostic target for SCI.


Subject(s)
AMP-Activated Protein Kinases , Fibroblast Growth Factor 5 , Oxidative Stress , Spinal Cord Injuries , Animals , Humans , Mice , AMP-Activated Protein Kinases/metabolism , Fibroblast Growth Factor 5/genetics , Fibroblast Growth Factor 5/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Mice, Knockout , Male , Female , Adult , Middle Aged
4.
Biochem Pharmacol ; 212: 115554, 2023 06.
Article in English | MEDLINE | ID: mdl-37080437

ABSTRACT

BACKGROUND: Melanoma has become more common, and its therapeutic management has remained challenging in recent decades. The purpose of our study is to explore new prognostic therapeutic markers of melanoma and to find new therapeutic methods and therapeutic targets of novel drugs, which have great significance. METHOD: First, the arachidonate 5-lipoxygenase (ALOX5) gene associated with both autophagy and ferroptosis was identified by R version 4.2.0. We used human melanoma and para-cancer tissues, human melanoma cell lines, and melanoma-bearing mouse tissues. We used qRT-PCR, Western blotting, immunohistochemistry, immunofluorescence staining, CCK-8, iron ion assay, GSH assay, and MDA assay. In vivo, the ferroptosis activation and antitumor effects of recombinant human ALOX5 protein were evaluated using a xenograft model. RESULT: We report that the downregulation of ALOX5 in melanoma is positively correlated with the prognosis of patients and is an independent prognostic factor. Elevated ALOX5 contributes to autophagy and ferroptosis in vitro and in vivo. At the same time, inhibition of autophagy can reduce ferroptosis enhanced by ALOX5, and autophagy and ALOX5 have a synergistic effect. The results of the mechanistic study showed that the increase in ALOX5 could activate the AMPK/mTOR pathway and inhibit GPX4 expression, promoting the occurrence of autophagy-dependent ferroptosis, while the decrease in p-AMPK/AMPK inhibited the occurrence of ferroptosis. CONCLUSION: ALOX5 deficiency was resistant to autophagy and ferroptosis by inhibiting the AMPK/mTOR pathway. Therefore, it can provide new targets and methods for melanoma drug development.


Subject(s)
Ferroptosis , Melanoma , Humans , Mice , Animals , AMP-Activated Protein Kinases/metabolism , Signal Transduction , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Cell Line, Tumor , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Autophagy
5.
Neurosci Bull ; 38(12): 1491-1507, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36282466

ABSTRACT

Preoperative sleep loss can amplify post-operative mechanical hyperalgesia. However, the underlying mechanisms are still largely unknown. In the current study, rats were randomly allocated to a control group and an acute sleep deprivation (ASD) group which experienced 6 h ASD before surgery. Then the variations in cerebral function and activity were investigated with multi-modal techniques, such as nuclear magnetic resonance, functional magnetic resonance imaging, c-Fos immunofluorescence, and electrophysiology. The results indicated that ASD induced hyperalgesia, and the metabolic kinetics were remarkably decreased in the striatum and midbrain. The functional connectivity (FC) between the nucleus accumbens (NAc, a subregion of the ventral striatum) and the ventrolateral periaqueductal gray (vLPAG) was significantly reduced, and the c-Fos expression in the NAc and the vLPAG was suppressed. Furthermore, the electrophysiological recordings demonstrated that both the neuronal activity in the NAc and the vLPAG, and the coherence of the NAc-vLPAG were suppressed in both resting and task states. This study showed that neuronal activity in the NAc and the vLPAG were weakened and the FC between the NAc and the vLPAG was also suppressed in rats with ASD-induced hyperalgesia. This study highlights the importance of preoperative sleep management for surgical patients.


Subject(s)
Hyperalgesia , Sleep Deprivation , Rats , Animals , Hyperalgesia/metabolism , Sleep Deprivation/complications , Sleep Deprivation/diagnostic imaging , Sleep Deprivation/metabolism , Rats, Sprague-Dawley , Periaqueductal Gray/metabolism , Periaqueductal Gray/pathology , Proto-Oncogene Proteins c-fos/metabolism , Pain, Postoperative/metabolism , Pain, Postoperative/pathology
6.
J Oncol ; 2022: 4000617, 2022.
Article in English | MEDLINE | ID: mdl-36204178

ABSTRACT

Ferroptosis is characterized by lipid peroxidation and iron accumulation, closely associated with clear cell renal cell carcinoma (ccRCC). It is of great significance for prognostic prediction and treatment of ccRCC to find biomarkers related to ferroptosis. We conducted several bioinformatic analyses using the transcriptome data and clinical information derived from online databases. Firstly, we identified the differentially expressed target genes in ccRCC. Then, t test and COX analysis were used to determine whether it was an independent prognostic factor combined with clinical information. String and gene set enrichment analysis (GSEA) were used to predict its function. Finally, we used ccRCC cells: 769-P and KAKI-1 in vitro to verify the regulation of target genes on cell proliferation apoptosis, iron metabolism, and GSH metabolism, which were used to judge the effect of target genes on ferroptosis. The study showed that MT1G is downregulated in ccRCC tissues compared with normal renal tissues. However, the ccRCC patients with higher expression relatively had higher malignancy and advanced stages. MT1G is an independent adverse factor for the prognosis of ccRCC. The protein interaction network analysis and GSEA showed that MT1G was closely related to GSH metabolism-related proteins (GSR) and lipid oxidation-related proteins (PLA2G2A). Samples with high expression of MT1G were enriched in "glutathione metabolism," "oxidative phosphorylation," and "proteasome," whose function was involved in GSH metabolism and lipid peroxidation. The term associated with the occurrence and development of tumors included "P53 signaling pathway." Furthermore, in vitro experiments showed that MT1G partially blocked ferroptosis induced by erastin and sorafenib-induced ccRCC cell lines (769-P and CAKI-1). The mechanism may be that MT1G affects ferroptosis by regulating GSH consumption in ccRCC cells. MT1G may be a negative regulator of ferroptosis in ccRCC cells and a biomarker of poor prognosis.

7.
Neuroscience ; 470: 88-99, 2021 08 21.
Article in English | MEDLINE | ID: mdl-34242730

ABSTRACT

It is well known that the central nervous system (CNS) is a complex neuronal network and its function depends on the balance between excitatory and inhibitory neurons. Disruption of the excitatory/inhibitory (E/I) balance is the main cause for the majority of the CNS diseases. In this review, we will discuss roles of the inhibitory system in the CNS diseases. The GABAergic system as the main inhibitory system, is essential for the appropriate functioning of the CNS, especially as it is engaged in the formation of learning and memory. Many researchers have reported that the GABAergic system is involved in regulating synaptic plasticity, cognition and long-term potentiation. Some clinical manifestations (such as cognitive dysfunctions, attention deficits, etc.) have also been shown to emerge after abnormalities in the GABAergic system accompanied with concomitant diseases, that include Alzheimer's disease (AD), Parkinson's disease (PD), Autism spectrum disorder (ASD), Schizophrenia, etc. The GABAergic system consists of GABA, GABA transporters, GABAergic receptors and GABAergic neurons. Changes in any of these components may contribute to the dysfunctions of the CNS. In this review, we will synthesize studies which demonstrate how the GABAergic system participates in the pathogenesis of the CNS disorders, which may provide a new idea that might be used to treat the CNS diseases.


Subject(s)
Alzheimer Disease , Autism Spectrum Disorder , Cognitive Dysfunction , Central Nervous System , GABAergic Neurons , Humans
8.
Front Neurosci ; 15: 593654, 2021.
Article in English | MEDLINE | ID: mdl-33867911

ABSTRACT

It has been well recognized that exposure to chronic stress could increase pain responding and exacerbate pain symptoms, resulting in stress-induced hyperalgesia. However, the mechanisms underlying stress-induced hyperalgesia are not yet fully elucidated. To this end, we observed that restraint as a stressful event exacerbated mechanical and thermal hyperalgesia, accompanied with up-regulation of nitric oxide (NO) (P < 0.001), GTP cyclohydrolase 1 (GCH1) (GCH1 mRNA: P = 0.001; GCH1 protein: P = 0.001), and tetrahydrobiopterin (BH4) concentration (plasma BH4: P < 0.001; spinal BH4: P < 0.001) on Day 7 in restraint stress (RS) rats. Intrathecal injection of N ω-nitro-L-arginine methyl ester (L-NAME), a non-specific NO synthase inhibitor, or N-([3-(aminomethyl)phenyl]methyl) ethanimidamide, a special inhibitor of inducible NO synthase (iNOS), for seven consecutive days attenuated stress-induced hyperalgesia and decreased the production of NO (P < 0.001). Interestingly, 7-nitro indazole, a special inhibitor of neuronal NO synthase, alleviated stress-induced hyperalgesia but did not affect spinal NO synthesis. Furthermore, intrathecal injection of BH4 not only aggravated stress-induced hyperalgesia but also up-regulated the expression of spinal iNOS (iNOS mRNA: P = 0.015; iNOS protein: P < 0.001) and NO production (P < 0.001). These findings suggest that hyperalgesia induced by RS is associated with the modulation of the GCH1-BH4 system and constitutively expressed spinal iNOS. Thus, the GCH1-BH4-iNOS signaling pathway may be a new novel therapeutic target for pain relief in the spinal cord.

9.
Pain ; 161(8): 1824-1836, 2020 08.
Article in English | MEDLINE | ID: mdl-32701842

ABSTRACT

Cognitive impairment is one of the most common complications associated with chronic pain. Almost 20% of chronic pain patients suffer from cognitive impairment, which may substantially influence their quality of life. Levels of major excitatory neurotransmitters in the central nervous system and alterations in the glutamatergic system may influence cognitive function and the pain sensory pathway. In this study, we adopted the spared nerve injury model to establish the progress of chronic pain and investigated the mechanism underlying the cognitive aspect related to it. At behavioral level, using the novel-object recognition test, mechanical hypersensitivity was observed in peripheral nerve-injured rats because they exhibited recognition deficits. We showed a dramatic decrease in hippocampal glutamate concentration using nuclear magnetic resonance and reduced glutamatergic synaptic transmission using whole-cell recordings. These were associated with deficient hippocampal long-term potentiation induced by high-frequency stimulation of the Schaffer collateral afferent. Ultra-high-performance liquid chromatography revealed lower levels of D-serine in the hippocampus of the spared nerve injury rats and that D-serine treatment could restore synaptic plasticity and cognitive dysfunction. The reduction of excitatory synapses was also increased by administering D-serine. These findings suggest that chronic pain has a critical effect on synaptic plasticity linked to cognitive function and may built up a new target for the development of cognitive impairment under chronic pain conditions.


Subject(s)
Neuralgia , Quality of Life , Animals , Hippocampus , Long-Term Potentiation , Neuronal Plasticity , Rats , Synapses
10.
Gene ; 758: 144973, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-32707303

ABSTRACT

Renal ischemia-reperfusion (rI/R) is a risk factor for acute lung injury (ALI). Alveolar macrophages (AMs) activation mediated by rI/R-induced ALI is one of the pathogeneses associated with the development of ALI. In rI/R, α2-adrenergic receptor agonists have been indicated to be effective in decreasing urea nitrogen concentrations. In this study, we explored the underlying pathogenesis of the clinically obtainable α2-adrenergic receptor agonist dexmedetomidine (DEX) in protecting against rI/R -mediated AMs activation. We incubated AMs with the serum of sham and rI/R rats in the presence or absence of various concentrations of DEX. We used an enzyme-linked immunosorbent assay to detect the secretion levels of GSH, LDH, IL-18, IL-1ß, and HMGB1 in the culture supernatant. We employed real-time polymerase chain reaction to assess the expression of NOX-4 mRNA, and western blotting to observe the protein levels of NOX-4, the NLRP3 inflammasome, AMPK, and eNOS. In addition, we used immunofluorescence to analyze ROS and MMP activity. Incubation of AMs with DEX suppressed rI/R-mediated cellular LDH production and ROS release. DEX also abolished the rI/R-mediated decrease in the activity of GSH and increased the levels of the rI/R-related NADPH oxidase protein NOX-4. Furthermore, DEX reduced the amelioration of the mitochondrial potential induced by rI/R. Our study showed that DEX inhibits rI/R-mediated levels of the NLRP3 inflammasome proteins ASC, NLRP3, HMGB1 and p20, and ameliorates rI/R-mediated AMPK signaling inactivation. Therefore, DEX reduces the levels of two mediators that are activated by the NLRP3 inflammasome: IL-18 and IL-1ß. Finally, our study established that DEX mitigates the rI/R-mediated decrease in eNOS, demonstrating its protective functions against AMs activation. In conclusion, our study demonstrated that the protective action of DEX in AMs is induced through amelioration of HMGB1-NLRP3 inflammasome-AMPK signaling. Our results suggest that the anesthetic reagent DEX exerts beneficial effects to ameliorate rI/R-induced ALI.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Lung Injury/prevention & control , Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , HMGB1 Protein/metabolism , Ischemia/pathology , Macrophages, Alveolar/pathology , NADPH Oxidase 4/biosynthesis , NADPH Oxidase 4/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction
11.
CNS Neurosci Ther ; 26(9): 913-924, 2020 09.
Article in English | MEDLINE | ID: mdl-32488976

ABSTRACT

AIMS: Perioperative neurocognitive disorders (PND) are associated with cognitive impairment in the preoperative or postoperative period, and neuroinflammation is thought to be the most important mechanisms especially during the postoperative period. The GABAergic system is easily disrupted by neuroinflammation. This study investigated the impact of the GABAergic system on PND after anesthesia and surgery. METHODS: An animal model of laparotomy with inhalation anesthesia in 16-month-old mice was addressed. Effects of the GABAergic system were assessed using biochemical analysis. Pharmacological blocking of α5GABAA Rs or P38 mitogen-activated protein kinase (MAPK) were applied to investigate the effects of the GABAergic system. RESULTS: After laparotomy, the hippocampus-dependent memory and long-term potentiation were impaired, the levels of IL-6, IL-1ß and TNF-α up-regulated in the hippocampus, the concentration of GABA decreased, and the protein levels of the surface α5GABAA Rs up-regulated. Pharmacological blocking of α5GABAA Rs with L655,708 alleviated laparotomy induced cognitive deficits. Further studies found that the P38 MAPK signaling pathway was involved and pharmacological blocking with SB203,580 alleviated memory dysfunctions. CONCLUSIONS: Anesthesia and surgery caused neuroinflammation in the hippocampus, which consequently disrupted the GABAergic system, increased the expressions of surface α5GABAA Rs especially through the P38 MAPK signaling pathway, and eventually led to hippocampus-dependent memory dysfunctions.


Subject(s)
Anesthesia/adverse effects , GABAergic Neurons/metabolism , Laparotomy/adverse effects , Postoperative Cognitive Complications/metabolism , Receptors, GABA-A/metabolism , Animals , Female , GABAergic Neurons/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Imidazoles/pharmacology , Mice , Mice, Inbred C57BL , Postoperative Cognitive Complications/etiology , Pyridines/pharmacology
12.
Neuroscience ; 388: 357-366, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30086367

ABSTRACT

Peripheral immune activation could cause neuroinflammation, leading to a series of central nervous system (CNS) disorders, such as spatial learning and memory dysfunction. However, its pathogenic mechanism and therapeutic strategies are not yet determined. The present study aimed to investigate the therapeutic effects of sulforaphane (SFN) on lipopolysaccharide (LPS)-induced spatial learning and memory dysfunction, and tried to elucidate its relationship with the role of hippocampal brain-derived neurotrophic factor (BDNF)-mammalian target of rapamycin (mTOR) signaling pathway. Intraperitoneal injection of LPS for consecutive 7 days to mice caused abnormal behaviors in Morris water maze test (MWMT), while systemic administration of SFN notably reversed the abnormal behaviors. In addition, hippocampal levels of inflammatory cytokines, synaptic proteins, BDNF-tropomyosin receptor kinase B (TrkB) and mTOR signaling pathways were altered in the processes of LPS-induced cognitive dysfunction and SFN's therapeutic effects. Furthermore, we found that ANA-12 (a TrkB inhibitor) or rapamycin (a mTOR inhibitor) could block the beneficial effects of SFN on LPS-induced cognitive dysfunction, and that hippocampal levels of synaptic proteins, BDNF-TrkB and mTOR signaling pathways were also notably changed. In conclusion, the results of the present study suggest that SFN could elicit improving effects on LPS-induced spatial learning and memory dysfunction, which is likely related to the regulation of hippocampal BDNF-mTOR signaling pathway.


Subject(s)
Inflammation/drug therapy , Isothiocyanates/pharmacology , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Nootropic Agents/pharmacology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Hippocampus/immunology , Inflammation/complications , Inflammation/psychology , Learning Disabilities/etiology , Learning Disabilities/immunology , Lipopolysaccharides , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/immunology , Mice, Inbred C57BL , Signal Transduction/drug effects , Spatial Memory/drug effects , Spatial Memory/physiology , Sulfoxides , TOR Serine-Threonine Kinases/metabolism
13.
CNS Neurosci Ther ; 24(10): 917-929, 2018 10.
Article in English | MEDLINE | ID: mdl-29488342

ABSTRACT

AIM: Multifactors contribute to the development of postoperative cognitive dysfunction (POCD), of which the most important mechanism is neuroinflammation. Prostaglandin E2 (PGE2) is a key neuroinflammatory molecule and could modulate hippocampal synaptic transmission and plasticity. This study was designed to investigate whether PGE2 and its receptors signaling pathway were involved in the pathophysiology of POCD. METHODS: Sixteen-month old male C57BL/6J mice were exposed to laparotomy. Cognitive function was evaluated by fear conditioning test. The levels of PGE2 and its 4 distinct receptors (EP1-4) were assessed by biochemical analysis. Pharmacological or genetic methods were further applied to investigate the role of the specific PGE2 receptors. RESULTS: Here, we found that the transcription and translation level of the EP3 receptor in hippocampus increased remarkably, but not EP1, EP2, or EP4. Immunofluorescence results showed EP3 positive cells in the hippocampal CA1 region were mainly neurons. Furthermore, pharmacological blocking or genetic suppression of EP3 could alleviate surgery-induced hippocampus-dependent memory deficits and rescued the expression of plasticity-related proteins, including cAMP response element-binding protein (CREB), activity-regulated cytoskeletal-associated protein (Arc), and brain-derived neurotrophic factor (BDNF) in hippocampus. CONCLUSION: This study showed that PGE2-EP3 signaling pathway was involved in the progression of POCD and identified EP3 receptor as a promising treatment target.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/pathology , Dinoprostone/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Laparotomy/adverse effects , Signal Detection, Psychological/physiology , AIDS-Related Complex/genetics , AIDS-Related Complex/metabolism , Aging , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Conditioning, Psychological , Exploratory Behavior , Fear , Male , Mice , Mice, Inbred C57BL , Postoperative Complications/pathology , Postoperative Complications/physiopathology , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Transduction, Genetic
14.
J Pain ; 18(8): 933-946, 2017 08.
Article in English | MEDLINE | ID: mdl-28323246

ABSTRACT

Cancer-induced bone pain (CIBP) remains a major challenge in advanced cancer patients because of our lack of understanding of its mechanisms. Previous studies have shown the vital role of γ-aminobutyric acid B receptors (GABABRs) in regulating nociception and various neuropathic pain models have shown diminished activity of GABABRs. However, the role of spinal GABABRs in CIBP remains largely unknown. In this study, we investigated the specific cellular mechanisms of GABABRs in the development and maintenance of CIBP in rats. Our behavioral results show that acute as well as chronic intrathecal treatment with baclofen, a GABABR agonist, significantly attenuated CIBP-induced mechanical allodynia and ambulatory pain. The expression levels of GABABRs were significantly decreased in a time-dependent manner and colocalized mostly with neurons and a minority with astrocytes and microglia. Chronic treatment with baclofen restored the expression of GABABRs and markedly inhibited the activation of cyclic adenosine monophosphate (cAMP)-dependent protein kinase and the cAMP-response element-binding protein signaling pathway. PERSPECTIVE: Our findings provide, to our knowledge, the first evidence that downregulation of GABABRs contribute to the development and maintenance of CIBP and restored diminished GABABRs attenuate CIBP-induced pain behaviors at least partially by inhibiting the protein kinase/cAMP-response element-binding protein signaling pathway. Therefore, spinal GABABR may become a potential therapeutic target for the management of CIBP.


Subject(s)
Bone Neoplasms/complications , Cancer Pain/etiology , Cancer Pain/pathology , Carcinoma/complications , Receptors, GABA-B/metabolism , Spinal Cord/metabolism , Animals , Baclofen/pharmacology , CREB-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Female , GABA-B Receptor Agonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glial Fibrillary Acidic Protein/metabolism , Pain Measurement , Pain Threshold/physiology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Time Factors
15.
Brain Behav Immun ; 60: 161-173, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27742579

ABSTRACT

Major histocompatibility class II (MHC II)-specific activation of CD4+ T helper cells generates specific and persistent adaptive immunity against tumors. Emerging evidence demonstrates that MHC II is also involved in basic pain perception; however, little is known regarding its role in the development of cancer-induced bone pain (CIBP). In this study, we demonstrate that MHC II expression was markedly induced on the spinal microglia of CIBP rats in response to STAT1 phosphorylation. Mechanical allodynia was ameliorated by either pharmacological or genetic inhibition of MHC II upregulation, which was also attenuated by the inhibition of pSTAT1 and pERK but was deteriorated by intrathecal injection of IFNγ. Furthermore, inhibition of ERK signaling decreased the phosphorylation of STAT1, as well as the production of MHC II in vivo and in vitro. These findings suggest that STAT1 contributes to bone cancer pain as a downstream mediator of ERK signaling by regulating MHC II expression in spinal microglia.


Subject(s)
Bone Neoplasms/metabolism , Microglia/metabolism , STAT1 Transcription Factor/metabolism , Spinal Cord/metabolism , Animals , Cancer Pain/metabolism , Female , Hyperalgesia/metabolism , Injections, Spinal/methods , MAP Kinase Signaling System/physiology , Rats, Sprague-Dawley
16.
J Neurochem ; 141(4): 553-564, 2017 05.
Article in English | MEDLINE | ID: mdl-27926984

ABSTRACT

Morphine is viewed as one of the classical treatments for intractable pain, but its role is limited by side effects, including analgesic tolerance. A few chemokines have been reported to be engaged in the mechanisms of morphine tolerance. However, the exact roles of CXC chemokine 11 (CXCL11) in chronic morphine tolerance remain unknown. In this study, Walker 256 mammary gland carcinoma cells were inoculated into the tibia of rats to provoke cancer-induced bone pain. Then, morphine was intrathecally administered twice daily for seven consecutive days to induce drug tolerance. We found that the level of CXCL11 in lumbar spinal cord was increased during the development of morphine tolerance in cancer-induced bone pain rats. Meanwhile, CXCL11 was co-localized with markers of astrocytes and neurons in the spinal cord. Inhibition of CXCL11 by neutralizing antibodies could remarkably attenuate the degree of morphine tolerance and decrease the activation of astrocytes. Moreover, blocking astrocyte activation by d, l-Fluorocitric acid could distinctly alleviate morphine tolerance and reduce the expression of CXCL11. Finally, morphine stimulation could induce the release of CXCL11 by cultured astrocytes and neurons in vitro. In summary, our results provide evidence that spinal CXCL11 plays a powerful modulatory role in the development of morphine tolerance through cross-talking between astrocytes and neurons. Read the Review series "Pain".


Subject(s)
Analgesics, Opioid/therapeutic use , Bone Neoplasms/complications , Chemokine CXCL11/genetics , Chemokine CXCL11/physiology , Morphine/therapeutic use , Pain/drug therapy , Pain/etiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal , Carcinoma 256, Walker , Drug Tolerance , Female , Injections, Spinal , Male , Neoplasm Transplantation , Pain/psychology , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism
17.
Acta Pharmacol Sin ; 37(6): 753-62, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27157092

ABSTRACT

AIM: To investigate the mechanisms underlying the anti-nociceptive effect of minocycline on bone cancer pain (BCP) in rats. METHODS: A rat model of BCP was established by inoculating Walker 256 mammary carcinoma cells into tibial medullary canal. Two weeks later, the rats were injected with minocycline (50, 100 µg, intrathecally; or 40, 80 mg/kg, ip) twice daily for 3 consecutive days. Mechanical paw withdrawal threshold (PWT) was used to assess pain behavior. After the rats were euthanized, spinal cords were harvested for immunoblotting analyses. The effects of minocycline on NF-κB activation were also examined in primary rat astrocytes stimulated with IL-1ß in vitro. RESULTS: BCP rats had marked bone destruction, and showed mechanical tactile allodynia on d 7 and d 14 after the operation. Intrathecal injection of minocycline (100 µg) or intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced mechanical tactile allodynia. Furthermore, intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced upregulation of GFAP (astrocyte marker) and PSD95 in spinal cord. Moreover, intraperitoneal injection of minocycline (80 mg/kg) reversed BCP-induced upregulation of NF-κB, p-IKKα and IκBα in spinal cord. In IL-1ß-stimulated primary rat astrocytes, pretreatment with minocycline (75, 100 µmol/L) significantly inhibited the translocation of NF-κB to nucleus. CONCLUSION: Minocycline effectively alleviates BCP by inhibiting the NF-κB signaling pathway in spinal astrocytes.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Astrocytes/drug effects , Bone Neoplasms/complications , Cancer Pain/drug therapy , Minocycline/therapeutic use , NF-kappa B/immunology , Spinal Cord/drug effects , Analgesics/therapeutic use , Animals , Astrocytes/immunology , Astrocytes/pathology , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Cancer Pain/complications , Cancer Pain/immunology , Cancer Pain/pathology , Cell Line, Tumor , Female , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/immunology , Hyperalgesia/pathology , Rats, Wistar , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/pathology
18.
J Neurochem ; 134(3): 590-600, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25919859

ABSTRACT

Bone cancer pain (BCP) is one of the most common and severe complications in patients suffering from primary bone cancer or metastatic bone cancer such as breast, prostate, or lung, which profoundly compromises their quality of life. Emerging lines of evidence indicate that central sensitization is required for the development and maintenance of BCP. However, the underlying mechanisms are largely unknown. In this study, we investigated the role of PI3Kγ/Akt in the central sensitization in rats with tumor cell implantation in the tibia, a widely used model of BCP. Our results showed that PI3Kγ and its downstream target pAkt were up-regulated in a time-dependent manner and distributed predominately in the superficial layers of the spinal dorsal horn neurons, astrocytes and a minority of microglia, and were colocalized with non-peptidergic, calcitonin gene-related peptide-peptidergic, and A-type neurons in dorsal root ganglion ipsilateral to tumor cell inoculation in rats. Inhibition of spinal PI3Kγ suppressed BCP-associated behaviors and the up-regulation of pAkt in the spinal cord and dorsal root ganglion. This study suggests that PI3Kγ/Akt signal pathway mediates BCP in rats. Central sensitization is required for the development and maintenance of bone cancer pain (BCP). In this study, we reported that PI3Kγ/Akt mediated the function of ephrinBs/EphBs in the central sensitization under BCP condition, and inhibition of spinal PI3Kγ suppressed BCP-associated behaviors. Our results suggest that inhibition of PI3Kγ/Akt may be a new target for the treatment of BCP.


Subject(s)
Bone Neoplasms/metabolism , Central Nervous System Sensitization/physiology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Pain/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Bone Neoplasms/complications , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Immunohistochemistry , Pain/etiology , Rats , Rats, Wistar , Spinal Cord/metabolism
19.
Exp Neurol ; 263: 39-49, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281485

ABSTRACT

Previously, we showed that activation of the spinal CXCL9, 10/CXCR3 pathway mediated bone cancer pain (BCP) in rats. However, the cellular mechanism involved is poorly understood. Here, we found that the activated CXCR3 was co-localized with either neurons, microglia, and astrocytes in the spinal cord, or non-peptidergic-, peptidergic-, and A-type neurons in the dorsal root ganglion. The inoculation of Walker-256 mammary gland carcinoma cells into the rat's tibia induced a time-dependent phosphorylation of Akt and extracellular signal-regulated kinase (ERK1/2) in the spinal cord, and CXCR3 was necessary for the phosphorylation of Akt and ERK 1/2. Meanwhile, CXCR3 was co-localized with either pAkt or pERK1/2. Blockage of either Akt or ERK1/2 prevented or reversed the mechanical allodynia in BCP rats. Furthermore, there was cross-activation between PI3K/Akt and Raf/MEK/ERK pathway under the BCP condition. Our results demonstrated that the activation of spinal chemokine receptor CXCR3 mediated BCP through Akt and ERK 1/2 kinase, and also indicated a crosstalk between PI3K/Akt and Raf/MEK/ERK signaling pathways under the BCP condition.


Subject(s)
Bone Neoplasms/metabolism , MAP Kinase Signaling System/physiology , Pain/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR3/metabolism , Animals , Blotting, Western , Bone Neoplasms/complications , Disease Models, Animal , Female , Hyperalgesia/metabolism , Immunohistochemistry , Pain/etiology , Rats , Rats, Wistar , Receptor Cross-Talk/physiology
SELECTION OF CITATIONS
SEARCH DETAIL