Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Neurotherapeutics ; 21(1): e00309, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38241164

ABSTRACT

The primary sensory neurons of the dorsal root ganglia (DRG) are subject to transcriptional alterations following peripheral nerve injury. These alterations are believed to play a pivotal role in the genesis of neuropathic pain. Alternative RNA splicing is a process that generates multiple transcript variants from a single gene, significantly contributing to the complexity of the transcriptome. However, little is known about the functional significance and control of alternative RNA splicing in injured DRG after spinal nerve ligation (SNL). In our study, we conducted a comprehensive transcriptome profiling and bioinformatic analysis to approach and identified a neuron-specific isoform of an RNA splicing regulator, RNA-binding Fox1 (Rbfox1, also known as A2BP1), as a crucial regulator of alternative RNA splicing in injured DRG after SNL. Notably, Rbfox1 expression is markedly reduced in injured DRG following peripheral nerve injury. Restoring this reduction effectively mitigates nociceptive hypersensitivity. Conversely, mimicking the downregulation of Rbfox1 expression generates neuropathic pain symptoms. Mechanistically, we uncovered that Rbfox1 may be a key factor influencing alternative RNA splicing of neuron-glial related cell adhesion molecule (NrCAM), a key neuronal cell adhesion molecule. In injured DRG after SNL, the downregulation of Rbfox1amplifies the insertion of exon 10 in Nrcam transcripts, leading to an increase in long Nrcam variants (L-Nrcam) and a corresponding decrease in short Nrcam variants (S-Nrcam) within injured DRG. In summary, our study supports the essential role of Rbfox1 in neuropathic pain within DRG, probably via the regulation of Nrcam splicing. These findings suggest that Rbfox1 could be a potential target for neuropathic pain therapy.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Humans , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Alternative Splicing , Neuralgia/genetics , Neuralgia/metabolism , Cell Adhesion Molecules/metabolism , Sensory Receptor Cells/metabolism , Ganglia, Spinal/metabolism
2.
Int Immunopharmacol ; 124(Pt B): 110991, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37774485

ABSTRACT

BACKGROUND: Thalamic pain frequently occurs after stroke and is a challenging clinical issue. However, the mechanisms underlying thalamic pain remain unclear. Neuroinflammation is a key determining factor in the occurrence and maintenance of hemorrhage-induced thalamic pain. Pioglitazone is an agonist of peroxisome proliferator-activated receptor gamma (PPARγ) and shows anti-inflammatory effects in multiple diseases. The present work focused on exploring whether PPARγ is related to hemorrhage-induced thalamic pain. METHODS: Immunostaining was conducted to analyze the cellular localization of PPARγ and co-localization was evaluated with NeuN, ionized calcium-binding adapter molecular 1 (IBA1), and glia fibrillary acidic protein (GFAP). Western blot analyses were used to evaluate MyD88, pNF-κB/NF-κB, pSTAT6/STAT6, IL-1ß, TNF-α, iNOS, Arg-1, IL-4, IL-6, and IL-10 expression. Behavioral tests in mice were conducted to evaluate continuous pain hypersensitivity. RESULTS: We found that pioglitazone appeared to mitigate the contralateral hemorrhage-induced thalamic pain while inhibiting inflammatory responses. Additionally, Pioglitazone induced phosphorylation of STAT6 and suppressed the phosphorylation NF-κB in our model of thalamic pain. These effects could be partially reversed with the PPARγ antagonist GW9662. CONCLUSION: The PPARγ agonist pioglitazone can mitigate mechanical allodynia by suppressing the NF-κB inflammasome while activating the STAT6 signal pathway, which are well-known to be associated with inflammation.


Subject(s)
PPAR gamma , Thiazolidinediones , Mice , Animals , Pioglitazone/therapeutic use , PPAR gamma/metabolism , Thiazolidinediones/therapeutic use , Thiazolidinediones/pharmacology , NF-kappa B/metabolism , Neuroinflammatory Diseases , PPAR-gamma Agonists , Hemorrhage , Pain/drug therapy
3.
Br J Anaesth ; 131(2): 253-265, 2023 08.
Article in English | MEDLINE | ID: mdl-37474241

ABSTRACT

BACKGROUND: Delirium is a common and disturbing postoperative complication that might be ameliorated by propofol-based anaesthesia. We therefore tested the primary hypothesis that there is less delirium after propofol-based than after sevoflurane-based anaesthesia within 7 days of major cancer surgery. METHODS: This multicentre randomised trial was conducted in 14 tertiary care hospitals in China. Patients aged 65-90 yr undergoing major cancer surgery were randomised to either propofol-based anaesthesia or to sevoflurane-based anaesthesia. The primary endpoint was the incidence of delirium within 7 postoperative days. RESULTS: A total of 1228 subjects were enrolled and randomised, with 1195 subjects included in the modified intention-to-treat analysis (mean age 71 yr; 422 [35%] women); one subject died before delirium assessment. Delirium occurred in 8.4% (50/597) of subjects given propofol-based anaesthesia vs 12.4% (74/597) of subjects given sevoflurane-based anaesthesia (relative risk 0.68 [95% confidence interval {CI}: 0.48-0.95]; P=0.023; adjusted relative risk 0.59 [95% CI: 0.39-0.90]; P=0.014). Delirium reduction mainly occurred on the first day after surgery, with a prevalence of 5.4% (32/597) with propofol anaesthesia vs 10.7% (64/597) with sevoflurane anaesthesia (relative risk 0.50 [95% CI: 0.33-0.75]; P=0.001). Secondary endpoints, including ICU admission, postoperative duration of hospitalisation, major complications within 30 days, cognitive function at 30 days and 3 yr, and safety outcomes, did not differ significantly between groups. CONCLUSIONS: Delirium was a third less common after propofol than sevoflurane anaesthesia in older patients having major cancer surgery. Clinicians might therefore reasonably select propofol-based anaesthesia in patients at high risk of postoperative delirium. CLINICAL TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR-IPR-15006209) and ClinicalTrials.gov (NCT02662257).


Subject(s)
Anesthetics, Inhalation , Emergence Delirium , Neoplasms , Propofol , Humans , Female , Aged , Male , Propofol/adverse effects , Sevoflurane/adverse effects , Anesthetics, Inhalation/adverse effects , Follow-Up Studies , Anesthesia, General/adverse effects , Emergence Delirium/chemically induced , Neoplasms/surgery
4.
Br J Anaesth ; 131(2): 266-275, 2023 08.
Article in English | MEDLINE | ID: mdl-37474242

ABSTRACT

BACKGROUND: Experimental evidence indicates that i.v. anaesthesia might reduce cancer recurrence compared with volatile anaesthesia, but clinical information is observational only. We therefore tested the primary hypothesis that propofol-based anaesthesia improves survival over 3 or more years after potentially curative major cancer surgery. METHODS: This was a long-term follow-up of a multicentre randomised trial in 14 tertiary hospitals in China. We enrolled 1228 patients aged 65-90 yr who were scheduled for major cancer surgery. They were randomised to either propofol-based i.v. anaesthesia or to sevoflurane-based inhalational anaesthesia. The primary endpoint was overall survival after surgery. Secondary endpoints included recurrence-free and event-free survival. RESULTS: Amongst subjects randomised, 1195 (mean age 72 yr; 773 [65%] male) were included in the modified intention-to-treat analysis. At the end of follow-up (median 43 months), there were 188 deaths amongst 598 patients (31%) assigned to propofol-based anaesthesia compared with 175 deaths amongst 597 patients (29%) assigned to sevoflurane-based anaesthesia; adjusted hazard ratio 1.02; 95% confidence interval (CI): 0.83-1.26; P=0.834. Recurrence-free survival was 223/598 (37%) in patients given propofol anaesthesia vs 206/597 (35%) given sevoflurane anaesthesia; adjusted hazard ratio 1.07; 95% CI: 0.89-1.30; P=0.465. Event-free survival was 294/598 (49%) in patients given propofol anaesthesia vs 274/597 (46%) given sevoflurane anaesthesia; adjusted hazard ratio 1.09; 95% CI 0.93 to 1.29; P=0.298. CONCLUSIONS: Long-term survival after major cancer surgery was similar with i.v. and volatile anaesthesia. Propofol-based iv. anaesthesia should not be used for cancer surgery with the expectation that it will improve overall or cancer-specific survival. CLINICAL TRIAL REGISTRATIONS: ChiCTR-IPR-15006209; NCT02660411.


Subject(s)
Neoplasms , Propofol , Sevoflurane , Propofol/adverse effects , Sevoflurane/adverse effects , Neoplasms/surgery , Humans , Male , Female , Aged , Follow-Up Studies , Anesthetics, Intravenous , Anesthesia, Inhalation , Cancer Survivors
5.
PLoS One ; 18(6): e0286590, 2023.
Article in English | MEDLINE | ID: mdl-37267303

ABSTRACT

BACKGROUND: Esketamine has higher potency, stronger receptor affinity, a stronger analgesic effect, a higher in vivo clearance rate, and a lower incidence of adverse reactions when compared to ketamine. However, there have been few ketamine studies to assess patient-centered, overall recovery outcomes from the perspective of patients with colorectal cancer. METHODS: This was a prospective, randomized controlled trial. Ninety-two patients undergoing laparoscopic radical resection of colorectal cancer were randomly assigned to either the esketamine (K group) or non-eskatamine (C group) group. After anesthesia induction, a loading dose of 0.25 mg/kg was administered, followed by continuous infusion at a rate of 0.12 mg.kg-1.h-1 until closure of surgical incisions in the K group. In the C group, an equivalent volume of normal saline was infused. The primary outcome was quality of recovery at 24 h after surgery, as measured by the Quality of Recovery-15 (QoR-15) scale. The QoR-15 was evaluated at three timepoints: before (Tbefore), 24 h (T24h) and 72 h (T72h) after surgery. MAIN RESULTS: A total of 88 patients completed this study. The total QoR-15 scores in K group (n = 45) were higher than in the C group (n = 43) at 24 h: 112.33 ± 8.79 vs. 103.93 ± 9.03 (P = 0.000) and at 72 h: 118.73 ± 7.82 vs. 114.79 ± 7.98 (P = 0.022). However, the differences between the two groups only had clinical significance at 24 h after surgery. Among the five dimensions of the QoR-15, physical comfort (P = 0.003), emotional state (P = 0.000), and physical independence (P = 0.000) were significantly higher at 24 h in the K group, and physical comfort (P = 0.048) was higher at 72 h in the K group. CONCLUSIONS: This study found that intraoperative intravenous low-dose esketamine could improve the early postoperative quality of recovery in patients undergoing laparoscopic radical resection of colorectal cancer from the perspective of patients.


Subject(s)
Colorectal Neoplasms , Ketamine , Laparoscopy , Humans , Ketamine/therapeutic use , Prospective Studies , Laparoscopy/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/surgery , Colorectal Neoplasms/etiology , Pain, Postoperative/drug therapy , Double-Blind Method
6.
JAMA Surg ; 157(10): 888-895, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35947398

ABSTRACT

Importance: Older patients may benefit from the hemodynamic stability of etomidate for general anesthesia. However, it remains uncertain whether the potential for adrenocortical suppression with etomidate may increase morbidity. Objective: To test the primary hypothesis that etomidate vs propofol for anesthesia does not increase in-hospital morbidity after abdominal surgery in older patients. Design, Setting, and Participants: This multicenter, parallel-group, noninferiority randomized clinical trial (Etomidate vs Propofol for In-hospital Complications [EPIC]) was conducted between August 15, 2017, and November 20, 2020, at 22 tertiary hospitals in China. Participants were aged 65 to 80 years and were scheduled for elective abdominal surgery. Patients and outcome assessors were blinded to group allocation. Data analysis followed a modified intention-to-treat principle. Interventions: Patients were randomized 1:1 to receive either etomidate or propofol for general anesthesia by target-controlled infusion. Main Outcomes and Measures: Primary outcome was a composite of major in-hospital postoperative complications (with a noninferiority margin of 3%). Secondary outcomes included intraoperative hemodynamic measurements; postoperative adrenocortical hormone levels; self-reported postoperative pain, nausea, and vomiting; and mortality at postoperative months 6 and 12. Results: A total of 1944 participants were randomized, of whom 1917 (98.6%) completed the trial. Patients were randomized to the etomidate group (n = 967; mean [SD] age, 70.3 [4.0] years; 578 men [59.8%]) or propofol group (n = 950; mean [SD] age, 70.6 [4.2] years; 533 men [56.1%]). The primary end point occurred in 90 of 967 patients (9.3%) in the etomidate group and 83 of 950 patients (8.7%) in the propofol group, which met the noninferiority criterion (risk difference [RD], 0.6%; 95% CI, -1.6% to 2.7%; P = .66). In the etomidate group, mean (SD) cortisol levels were lower at the end of surgery (4.8 [2.7] µg/dL vs 6.1 [3.4] µg/dL; P < .001), and mean (SD) aldosterone levels were lower at the end of surgery (0.13 [0.05] ng/dL vs 0.15 [0.07] ng/dL; P = .02) and on postoperative day 1 (0.14 [0.04] ng/dL vs 0.16 [0.06] ng/dL; P = .001) compared with the propofol group. No difference in mortality was observed between the etomidate and propofol groups at postoperative month 6 (2.2% vs 3.0%; RD, -0.8%; 95% CI, -2.2% to 0.7%) and 12 (3.3% vs 3.9%; RD, -0.6%; 95% CI, -2.3% to 1.0%). More patients had pneumonia in the etomidate group than in the propofol group (2.0% vs 0.3%; RD, 1.7%; 95% CI, 0.7% to 2.8%; P = .001). Results were consistent in the per-protocol population. Conclusions and Relevance: Results of this trial showed that, compared with propofol, etomidate anesthesia did not increase overall major in-hospital morbidity after abdominal surgery in older patients, although it induced transient adrenocortical suppression. Trial Registration: ClinicalTrials.gov Identifier: NCT02910206.


Subject(s)
Etomidate , Propofol , Aged , Aldosterone , Anesthesia, General , Anesthesia, Intravenous , Anesthetics, Intravenous/adverse effects , Hospitals , Humans , Hydrocortisone , Male , Postoperative Complications/etiology , Propofol/adverse effects
7.
Mol Neurobiol ; 59(4): 2629-2641, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35141864

ABSTRACT

Nerve trauma-induced toll-like receptor 7 (TLR7) expression level increases in primary sensory neurons in injured dorsal root ganglion (DRG) avails to neuropathic pain, but the reason is still unknown. In the current study, we showed that unilateral lumbar 4 (L4) spinal nerve ligation (SNL) upregulated CCAAT/enhancer-binding protein-ß (C/EBPß) expression in ipsilateral L4 DRG. Preventing this elevation attenuated the SNL-induced upregulation of TLR7 in the ipsilateral L4 DRG and inhibited cold/thermal hyperalgesia and mechanical allodynia. In injected DRG, mimicking nerve trauma-induced C/EBPß upregulation increased TLR7 levels, augmented responses to cold/thermal/mechanical stimuli, and caused ipsilateral spontaneous pain with no SNL. Mechanistically, SNL upregulated binding of increased C/EBPß to Tlr7 promoter in ipsilateral L4 DRG. Accorded that C/EBPß could trigger the activation of Tlr7 promoter and co-expressed with Tlr7 mRNA in individual DRG neurons, our findings strongly suggest the role of C/EBPß in nerve trauma-mediated TLR7 upregulation in injured primary sensory neurons.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Toll-Like Receptor 7 , Trauma, Nervous System , Animals , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Trauma, Nervous System/metabolism , Up-Regulation
8.
Ann Palliat Med ; 10(6): 6768-6778, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34237977

ABSTRACT

BACKGROUND: Cerebral ischemic reperfusion injury (CI/RI) is a common cerebrovascular disease with high morbidity and disability that threatens human health. This study was conducted to explore the effects of dexmedetomidine (Dex) on the c-Jun N-terminal kinase (JNK) pathway in CI/RI, and to provide a theoretical basis for the recovery of brain function after cerebral ischemia. METHODS: Sprague Dawley (SD) rats (n=24) were randomly divided into Sham, Sham + Dex, Sham + yohimbine (Yoh) + Dex, Sham + SP600125, ischemic reperfusion (I/R), I/R + Dex, I/R + Yoh + Dex, and I/R + SP600125 groups, and a focal cerebral ischemia reperfusion rat model was established by linear thrombus. The neurological deficit score and infarct volume were measured. Wet/dry weight ratios were used to measure brain water content, and cerebral infarct volume was determined by 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. The cellular distribution of p-JNK and cleaved caspase-3 were examined using immunofluorescent staining (IF) and the total JNK and p-JNK were determined by Western blotting (WB). RESULTS: Compared with the Sham group, the rats in I/R, I/R + Dex, I/R + Yoh + Dex, and I/R + SP600125 groups developed hemiparesis of the left forelimb at different levels with a higher neurological deficit score, brain water content, infarct volume, and markedly upregulated expression of cleaved caspase-3, p-JNK (P<0.05). Compared with the I/R group, the neurological deficit score, brain water content, infarct volume, and expression of cleaved caspase-3, p-JNK were markedly decreased in I/R + Dex, I/R + Yoh + Dex, and I/R + SP600125 groups (P<0.05), and compared with the I/R + Dex group, the neurological deficit score, brain water content, infarct volume, and expression of cleaved caspase-3, p-JNK were markedly increased in the I/R + Yoh + Dex group (P<0.05). Double immunofluorescence staining showed there was a strong colocalization between p-JNK and the astroglial marker GFAP. CONCLUSIONS: The JNK signaling pathway is involved in CI/RI. Inhibition of the JNK pathway blocked caspase-3 activation which can decrease CI/RI. Dex can alleviate cerebral CI/RI in rats by increasing α2-adrenergic receptor and blocking JNK phosphorylation and activation of caspase-3.


Subject(s)
Brain Ischemia , Dexmedetomidine , Reperfusion Injury , Animals , Brain Ischemia/drug therapy , Dexmedetomidine/therapeutic use , MAP Kinase Signaling System , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy
9.
Colloids Surf B Biointerfaces ; 202: 111681, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33714187

ABSTRACT

Despite the potential of immunotherapy in various solid tumors, the efficiency of immunotherapy is limited by little tumor-infiltrating lymphocytes (TILs) and abundant immunosuppressive M2-type tumor-associated macrophages (M2-TAMs) in the tumor microenvironment (TME). Herein, we design a versatile photo-immunotherapy nanoparticle (termed as HA-AuNR/M-M2pep NP) to conquer above challenges. The HA-AuNR/M-M2pep NP is composed of hyaluronic acid modified gold nanorod (HA-AuNR) surface-modified with matrix metalloproteinase-2 (MMP2)-responsive M2pep fusion peptides (M-M2pep). Upon tumor site, the fabricated HA-AuNR/M-M2pep NP releases M2pep through the cleavage of MMP2-sensitive peptide to selectively deplete M2-TAMs and improve immunoactivity of TME. Meanwhile, HA-AuNR could target to tumor cells and realize precise tumor photothermal therapy (PTT) under near infrared light irradiation, which further triggers immunogenic cell death (ICD) of tumor cells and elicits antitumor immunity. In vivo antitumor studies reveal that HA-AuNR/M-M2pep NPs-mediated PTT and M2-TAMs depletion recruit TILs, activate effector T lymphocytes, secrete antitumor cytokines (e.g. IFN-γ, TNF-α), and effectively inhibit the growth of tumor. Collectively, HA-AuNR/M-M2pep NP-mediated photo-immunotherapy based on dual targeted delivery and bio-responsive drug release holds tremendous promise to enhance antitumor efficacy.


Subject(s)
Nanoparticles , Neoplasms , Humans , Immunotherapy , Matrix Metalloproteinase 2 , Neoplasms/therapy , Tumor Microenvironment
10.
Front Cell Dev Biol ; 8: 814, 2020.
Article in English | MEDLINE | ID: mdl-33015035

ABSTRACT

Despite the great increase in human lifespan with improved medical care, the physiological and pathological changes such as memory and cognitive disorders and associated anxiety and depression are major concern with aging. Molecular mechanisms underlying these changes are little known. The present study examined the differentially expressed genes (DEGs) and the genes with differentially expressed isoforms in three brain regions, anterior cingulate cortex (ACC), amygdala and hippocampus, throughout the lifespan of mice. Compared to 2-month old mice, both 12- and 24-month old mice displayed memory and cognitive impairments in the Morris water maze, Y-maze, and novel object recognition tests and depression- and anxiety-like behaviors in the tail suspension, forced swimming, open field, and elevated plus maze tests. RNA sequencing analysis identified 634 and 1078 DEGs in ACC, 453 and 1015 DEGs in the amygdala and 884 and 1054 DEGs in hippocampus in the 12- and 24-month old mice, respectively. Similarly, many genes with differentially expressed isoforms were also identified in these three brain regions in the 12- and 24-month old mice. Further functional analysis revealed that many DEGs and the genes with differentially expressed isoforms in the ACC and amygdala were mapped to depression- and anxiety-related genes, respectively and that a lot of DEGs and the genes with differentially expressed isoforms in hippocampus were mapped to cognitive dysfunction-related genes from both 12- and 24-month old mice. All of these mapped DEGs and the genes with differentially expressed isoforms were closely related to neuroinflammation. Our findings indicate that these neuroinflammation-related DEGs and the genes with differentially expressed isoforms are likely new targets in the management of memory/cognitive impairment and emotional disorders during the aging.

11.
Oncol Rep ; 44(6): 2429-2442, 2020 12.
Article in English | MEDLINE | ID: mdl-33125091

ABSTRACT

As a novel halogenated hydroxyl ether­inhaled general anesthetic, sevoflurane has been reported to affect the progression of diverse human cancers. In the present study, we aimed to explore the functions and underlying mechanisms of sevoflurane in colon cancer. MTT assay, flow cytometric analysis and Transwell assay were conducted to evaluate cell viability, apoptosis and invasion, respectively. Western blot analysis was performed to determine the protein level of sphingosine­1­phosphate phosphatase 1 (SGPP1). The morphology and size of exosomes were analyzed by TEM and NTA. The levels of circular RNA 3­hydroxy­3­methylglutaryl­CoA synthase 1 (circ­HMGCS1), microRNA (miR)­34a­5p and SGPP1 mRNA were examined by RT­qPCR. Dual­luciferase reporter and RNA RIP assays were utilized to explore the interaction between miR­34a­5p and circ­HMGCS1 or SGPP1. A murine xenograft model was established to investigate the effect of circ­HMGCS1 in vivo. As a result, it was determined that sevoflurane suppressed cell viability and invasion and induced apoptosis in colon cancer in a dose­dependent way. Exosomal circ­HMGCS1 was increased in the serums and cells of colon cancer patients. Circ­HMGCS1 was downregulated by sevoflurane treatment in colon cancer cells and circ­HMGCS1 overexpression could restore the effect of sevoflurane on colon cancer cell development. miR­34a­5p was a target of circ­HMGCS1 and miR­34a­5p inhibition reversed the effect of circ­HMGCS1 silencing on colon cancer cell progression. Moreover, circ­HMGCS1 knockdown suppressed SGPP1 expression via sponging miR­34a­5p. Knockdown of circ­HMGCS1 blocked tumor growth in vivo. In conclusion, sevoflurane inhibited colon cancer progression by modulating the exosome­transmitted circ­HMGCS1/miR­34a­5p/SGPP1 axis.


Subject(s)
Colonic Neoplasms/drug therapy , Membrane Proteins/genetics , MicroRNAs/metabolism , Phosphoric Monoester Hydrolases/genetics , RNA, Circular/metabolism , Sevoflurane/pharmacology , Aged , Animals , Apoptosis/drug effects , Apoptosis/genetics , Case-Control Studies , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Colon/pathology , Colonic Neoplasms/blood , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Progression , Exosomes/drug effects , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Healthy Volunteers , Humans , Intestinal Mucosa/pathology , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , RNA, Circular/genetics , Sevoflurane/therapeutic use , Xenograft Model Antitumor Assays
12.
Am J Transl Res ; 12(8): 4337-4346, 2020.
Article in English | MEDLINE | ID: mdl-32913509

ABSTRACT

AIMS: The present study investigated the function and mechanism of lncRNA Gm43050 in sevoflurane-induced abnormal cognition. METHODS: Primary hippocampal neurons were used to establish the model of abnormal cognitive disorder. Overexpression and knockdown experiments were performed to analyze cell death rates, proliferation, apoptosis and the inflammatory response. The dual-luciferase reporter assay was used to analyze the potential binding targets of lncRNA Gm43050. Rescue experiments were used to assess the downstream targets of Gm43050. RESULTS: We found that lncRNA Gm43050 was in the cytoplasm. Overexpression of lncRNA Gm43050 had no impact on proliferation but significantly reduced the cell death rates and apoptosis. The inflammation markers IL-6, IL-1ß, IL-8 and TNF-α were manifestly downregulated in the overexpression group. Opposite effects were detected in the lncRNA Gm43050 knockdown group. Bioinformatics analysis showed that miR-640 may be the potential target of Gm43050. Additionally, we found that ZFP91 was the downstream target of miR-640. CONCLUSION: We provided comprehensive data of the function and mechanism of lncRNA Gm43050 in abnormal cognition. Our study showed that lncRNA Gm43050 exerted its important role via the regulation of miR-640 and ZFP91.

13.
Cell Cycle ; 19(19): 2538-2552, 2020 10.
Article in English | MEDLINE | ID: mdl-32857667

ABSTRACT

The involvement of propofol and circular RNAs (circRNAs) in lung cancer progression has been identified. However, the relationship between propofol and circRNAs as well as the underlying molecular mechanisms on lung cancer development remain unclear. Cell viability, migration and invasion were measured by cell counting kit-8 assay, 5-bromo-2-deoxyuridine (BrdU) and transwell assay. Glycolytic metabolism was calculated by measuring the glucose consumption, lactate production and extracellular acidification. Western blot was used to detect the protein of glucose transporter 1 (GLUT1), glycolysis enzymes, and forkhead box M1 (FOXM1). The expression of circRNA transcriptional adaptor 2A (circTADA2A), microRNA (miR)-455-3p and FOXM1 mRNA was detected by quantitative real-time polymerase chain reaction. The interaction between miR-455-3p and circTADA2A or FOXM1 was analyzed using the dual-luciferase reporter assay. Murine xenograft model was established to perform in vivo experiments. We found propofol treatment alleviated lung cancer cell proliferation, migration, invasion and aerobic glycolysis in vitro as well as inhibited tumor growth in vivo. Propofol decreased the level of circTADA2A and exerted anti-tumor effects by regulating circTADA2A. MiR-455-3p directly interacted with circTADA2A and FOXM1 in lung cancer cells, and circTADA2A could regulate FOXM1 expression by binding to miR-455-3p. Subsequently, rescue assay showed that propofol inhibited cell proliferation, migration, invasion and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer. Collectively, propofol suppressed cell carcinogenesis and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer, providing an effective clinical implication for propofol to prevent the development of lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Box Protein M1/metabolism , Glycolysis/drug effects , Lung Neoplasms/drug therapy , MicroRNAs/metabolism , Propofol/pharmacology , RNA, Circular/metabolism , A549 Cells , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Circular/genetics , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Cancer Med ; 9(19): 7218-7230, 2020 10.
Article in English | MEDLINE | ID: mdl-32767662

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. Herein, we explored the underlying mechanism by which Propofol inhibited the development of HCC. METHODS: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was carried out to detect the viability and proliferation. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were performed to detect the expression of long noncoding RNA (lncRNA) H19, microRNA-520a-3p (miR-520a-3p), LIM domain kinase 1 (LIMK1), metastasis-associated markers (Snail, Twist, Vimentin and E-cadherin) and exosome markers (CD9 and CD81). Transmission electron microscopy (TEM) was used to observe the morphology and structure of exosomes. The apoptosis and metastasis were measured by flow cytometry and transwell assays. StarBase software was utilized to predict the targets of H19 and miR-520a-3p. Dual-luciferase reporter assay was performed to confirm the interaction between miR-520a-3p and H19 or LIMK1. Nude mice bearing tumors were used to validate the role of exosomal H19. RESULTS: The high expression of exosomal H19 accelerated the proliferation and motility while hampering the apoptosis of HCC cells. MiR-520a-3p could bind with H19. Exosomal H19 exacerbated HCC through sponging miR-520a-3p. The 3' untranslated region (3'UTR) of LIMK1 could bind to miR-520a-3p. MiR-520a-3p mimic transfection reversed the inhibitory effect of high expression of exosomal LIMK1 on the apoptosis of HCC cells and the promoting effects on the proliferation and metastasis of HCC cells. The mRNA and protein levels of LIMK1 were regulated by H19/miR-520a-3p signaling. The high level of exosomal H19 promoted the growth of HCC tumors in vivo. CONCLUSION: Circulating H19 promoted the proliferation, migration and invasion and inhibited the apoptosis of HCC cells treated with Propofol through upregulating LIMK1 via sponging miR-520a-3p.


Subject(s)
Antineoplastic Agents/pharmacology , Exosomes/metabolism , Lim Kinases/metabolism , Liver Neoplasms/drug therapy , MicroRNAs/metabolism , Propofol/pharmacology , RNA, Long Noncoding/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Disease Progression , Exosomes/genetics , Exosomes/pathology , Gene Expression Regulation, Neoplastic , Humans , Lim Kinases/genetics , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , RNA, Long Noncoding/genetics , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Anesthesiology ; 133(2): 318-331, 2020 08.
Article in English | MEDLINE | ID: mdl-32667155

ABSTRACT

BACKGROUND: Suboptimal tissue perfusion and oxygenation during surgery may be responsible for postoperative nausea and vomiting in some patients. This trial tested the hypothesis that muscular tissue oxygen saturation-guided intraoperative care reduces postoperative nausea and vomiting. METHODS: This multicenter, pragmatic, patient- and assessor-blinded randomized controlled (1:1 ratio) trial was conducted from September 2018 to June 2019 at six teaching hospitals in four different cities in China. Nonsmoking women, 18 to 65 yr old, and having elective laparoscopic surgery involving hysterectomy (n = 800) were randomly assigned to receive either intraoperative muscular tissue oxygen saturation-guided care or usual care. The goal was to maintain muscular tissue oxygen saturation, measured at flank and on forearm, greater than baseline or 70%, whichever was higher. The primary outcome was 24-h postoperative nausea and vomiting. Secondary outcomes included nausea severity, quality of recovery, and 30-day morbidity and mortality. RESULTS: Of the 800 randomized patients (median age, 50 yr [range, 27 to 65]), 799 were assessed for the primary outcome. The below-goal muscular tissue oxygen saturation area under the curve was significantly smaller in patients receiving muscular tissue oxygen saturation-guided care (n = 400) than in those receiving usual care (n = 399; flank, 50 vs. 140% · min, P < 0.001; forearm, 53 vs. 245% · min, P < 0.001). The incidences of 24-h postoperative nausea and vomiting were 32% (127 of 400) in the muscular tissue oxygen saturation-guided care group and 36% (142 of 399) in the usual care group, which were not significantly different (risk ratio, 0.89; 95% CI, 0.73 to 1.08; P = 0.251). There were no significant between-group differences for secondary outcomes. No harm was observed throughout the study. CONCLUSIONS: In a relatively young and healthy female patient population, personalized, goal-directed, muscular tissue oxygen saturation-guided intraoperative care is effective in treating decreased muscular tissue oxygen saturation but does not reduce the incidence of 24-h posthysterectomy nausea and vomiting.


Subject(s)
Hysterectomy/adverse effects , Intraoperative Care/methods , Muscle, Skeletal/metabolism , Oxygen Consumption/physiology , Postoperative Nausea and Vomiting/metabolism , Postoperative Nausea and Vomiting/prevention & control , Adult , Double-Blind Method , Female , Humans , Hysterectomy/trends , Intraoperative Care/trends , Middle Aged , Postoperative Nausea and Vomiting/diagnosis
17.
BMC Anesthesiol ; 20(1): 86, 2020 04 18.
Article in English | MEDLINE | ID: mdl-32305062

ABSTRACT

BACKGROUND: Postoperative pain is the most prominent concern among surgical patients. It has previously been reported that venous cannulation-induced pain (VCP) can be used to predict postoperative pain after laparoscopic cholecystectomy within 90 mins in the recovery room. Its potential in predicting postoperative pain in patients with patient-controlled intravenous analgesia (PCIA) is worth establishing. The purpose of this prospective observational study was to investigate the application of VCP in predicting postoperative pain in patients with PCIA during the first 24 h after laparoscopic nephrectomy. METHODS: One hundred twenty patients scheduled for laparoscopic nephrectomy were included in this study. A superficial vein on the back of the hand was cannulated with a standard-size peripheral venous catheter (1.1 × 3.2 mm) by a nurse in the preoperative areas. Then the nurse recorded the VAS score associated with this procedure estimated by patients, and dichotomized the patients into low response group (VAS scores < 2.0) or high response group (VAS scores ≥2.0). After general anesthesia and surgery, all the patients received the patient-controlled intravenous analgesia (PCIA) with sufentanil. The VAS scores at rest and on coughing at 2 h, 4 h, 8 h, 12 h, 24 h, the effective number of presses and the number of needed rescue analgesia within 24 h after surgery were recorded. RESULTS: Peripheral venous cannulation-induced pain score was significantly correlated with postoperative pain intensity at rest (rs = 0.64) and during coughing (rs = 0.65), effective times of pressing (rs = 0.59), additional consumption of sufentanil (rs = 0.58). Patients with venous cannulation-induced pain intensity ≥2.0 VAS units reported higher levels of postoperative pain intensity at rest (P < 0.0005) and during coughing (P < 0.0005), needed more effective times of pressing (P < 0.0005) and additional consumption of sufentanil (P < 0.0005), and also needed more rescue analgesia (P = 0.01) during the first 24 h. The odds of risk for moderate or severe postoperative pain (OR 3.5, 95% CI 1.3-9.3) was significantly higher in patients with venous cannulation-induced pain intensity ≥2.0 VAS units compared to those <2.0 VAS units. CONCLUSIONS: Preoperative assessment of pain induced by venous cannulation can be used to predict postoperative pain intensity in patients with PCIA during the first 24 h after laparoscopic nephrectomy. TRIAL REGISTRATION: We registered this study in a Chinese Clinical Trial Registry (ChiCTR) center on July 6 2019 and received the registration number: ChiCTR1900024352.


Subject(s)
Catheterization, Peripheral/adverse effects , Laparoscopy/methods , Nephrectomy/methods , Pain, Postoperative/epidemiology , Adult , Analgesia, Patient-Controlled/methods , Analgesics, Opioid/administration & dosage , Catheterization, Peripheral/methods , Female , Humans , Male , Middle Aged , Pain Measurement , Preoperative Period , Prospective Studies , Sufentanil/administration & dosage
18.
Eur J Clin Pharmacol ; 76(7): 903-912, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32274525

ABSTRACT

AIM AND BACKGROUND: Postoperative nausea and vomiting (PONV) remains a significant clinical problem for surgical patients. Amisulpride is a well-studied D2/D3 antagonist that has the potential to be used for preventing and treating PONV. Our aim was to assess the efficacy and safety of amisulpride for prevention and treatment of PONV through a systematic review and meta-analysis. METHOD: A systematic literature search was performed using MEDLINE, EMBASE, PUBMED, clinicaltrials.gov, and the Cochrane Central Register of Controlled Trials from their inception to Feb 15th, 2019. The efficacy outcome was the incidence of complete response, defined as no emesis and no rescue antiemetic use in a 24-h period after study drug administration. The safety outcomes were the adverse effects associated with amisulpride. RESULTS: Five studies comprising 3243 patients met inclusion critieria. Compared with placebo, amisulpride showed a significantly improved incidence of complete response [relative risk (RR): 1.30; 95% confidence interval (CI): 1.20-1.41; P < 0.00001, I2 = 0%] with firm evidence from the trial sequential analysis. Particularly, the amisulpride at 5 mg dose indicated a significant benefit than placebo [relative risk (RR): 1.28; 95% confidence interval (CI): 1.18-1.39; P < 0.00001, I2 = 4%]. The adverse event profile of amisulpride was generally similar to the placebo. CONCLUSION: Based on our findings, low-dose, intravenous amisulpride is safe and efficacious for the prevention and treatment of PONV compared to placebo. Further studies are needed to explore the optimal dose and timing. CLINICAL TRIAL REGISTRATION: PROSPERO: CRD42019121483.


Subject(s)
Amisulpride/therapeutic use , Dopamine Antagonists/therapeutic use , Postoperative Nausea and Vomiting/drug therapy , Humans , Randomized Controlled Trials as Topic
19.
Cell Cycle ; 19(9): 1000-1011, 2020 05.
Article in English | MEDLINE | ID: mdl-32212891

ABSTRACT

Although a previous study reported that propofol had a therapeutic effect in status epilepticus (SE), the mechanisms underlying the effect of propofol in SE remain unclear. The aim of this study was to explore the regulatory mechanisms underlying propofol-induced inhibition of SE.A rat SE model was established using the lithium-pilocarpine injection method. A qRT-PCR and Western blot were utilized to detect the expression of relative molecules. Cell apoptosis was evaluated by a flow cytometry assay. The interaction between miR-15a-5p and NR2B was assessed using a luciferase reporter assay.Propofol inhibited cell apoptosis and increased miR-15a-5p expression both in hippocampal tissues of SE rats and low Mg2+-induced hippocampal neurons. Propofol-induced attenuation of apoptosis of low Mg2+-induced hippocampal neurons was mediated by miR-15a-5p. miR-15a-5p targeted NR2B and negatively regulated its expression. Propofol downregulated NR2B expression, mediated by miR-15a-5p. In terms of the mechanism of action, propofol suppressed the apoptosis of Mg2+-induced hippocampal neurons through the miR-15a-5p/NR2B/ERK1/2 pathway. In vivo experiment suggested that propofol inhibited the apoptosis of hippocampal neurons in SE rats by upregulating miR-15a-5p.In terms of the molecular mechanism of propofol, it appears to inhibit apoptosis of hippocampal neurons in SE through the miR-15a-5p/NR2B/ERK1/2 pathway. The findings provide theoretical support for propofol treatment of SE.


Subject(s)
Anesthetics, Intravenous/administration & dosage , Apoptosis/drug effects , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , MicroRNAs/metabolism , Neurons/metabolism , Propofol/administration & dosage , Receptors, N-Methyl-D-Aspartate/metabolism , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Animals , Apoptosis/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Knockdown Techniques , Lithium Compounds/adverse effects , MAP Kinase Signaling System/genetics , MicroRNAs/genetics , Neurons/drug effects , Pilocarpine/adverse effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Status Epilepticus/chemically induced , Transfection , Up-Regulation/drug effects , Up-Regulation/genetics
20.
Onco Targets Ther ; 13: 361-370, 2020.
Article in English | MEDLINE | ID: mdl-32021281

ABSTRACT

BACKGROUND: Propofol has been identified to perform anti-tumor functions in glioma. However, the molecular mechanisms underlying propofol-induced prevention on migration and invasion of glioma cells remain unclear. METHODS: Cell proliferation, invasion and migration were measured by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide assay and transwell assay, respectively. The expression of microRNA (miR)-206 and Rho-associated coiled coil-containing protein kinase 1 (ROCK1) was detected by quantitative real-time polymerase chain reaction. Western blot was used to measure the activation of the PI3K/AKT pathway. The interaction between miR-206 and ROCK1 was analyzed using the dual-luciferase reporter assay, RNA immunoprecipitation assay, and pull-down assay. RESULTS: Propofol treatment inhibited the migration, invasion, and PI3K/AKT pathway activation in glioma cells. MiR-206 was decreased in glioma tissues and cells, while propofol exposure induced the upregulation of miR-206 in glioma cells. Besides that, we also found overexpressed miR-206 enhanced propofol-mediated inhibition on the migration, invasion, and PI3K/AKT pathway activation of glioma cells. Subsequently, ROCK1 was confirmed to be a target of miR-206. ROCK1 was elevated in glioma tissues and cells, but was reduced by propofol exposure in glioma cells. The rescue assay indicated that the miR-206/ROCK1 axis was involved in propofol-induced inhibition on the migration, invasion, and PI3K/AKT pathway activation in glioma cells. CONCLUSION: Propofol inhibited the migration and invasion of glioma cells by blocking the PI3K/AKT pathway through the miR-206/ROCK1 axis, suggesting an effective clinical implication for the anesthetic to prevent the metastasis of glioma.

SELECTION OF CITATIONS
SEARCH DETAIL
...