Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Mol Med ; 27(1): 39, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33858325

ABSTRACT

OBJECTIVES: Targeted inhibition of inflammatory response can reduce diabetic cerebral ischemia-reperfusion (I/R) injure. Pyroptosis is characterized by caspase-1 dependence and the release of a large number of pro-inflammatory factors. LncRNA-Fendrr is associated with a variety of diseases, but Fendrr has not been studied in diabetic cerebral I/R. NLR-family CARD-containing protein 4 (NLRC4) regulate the pyroptosis of microglia cells. This study was designed to investigate whether Fendrr is involved in the effects of diabetic cerebral I/R injury. METHODS: The diabetic brain I/R model in mice was constructed. Mouse microglia cell line BV-2 cells were exposed to high glucose followed by hypoxia/reoxygenation (H/R). Fendrr and some pyroptosis-associated proteins were detected by qRT-PCR, western blot or ELISA. HE staining was used to detect pathological changes. Microglia pyroptosis was detected by TUNEL staining. RNA pull-down and RNA Immunoprecipitation were used to detect binding of Fendrr to HERC2 (E3 ubiquitin ligase), and CO-IP detected binding of HERC2 to NLRC4. The ubiquitination of NLRC4 was detected by ubiquitination experiments. RESULTS: Fendrr was significantly increased in the diabetic cerebral I/R model, and NLRC4 inflammatory complex and pyroptosis mediated inflammatory factors were increased. NLRC4 and inflammatory cytokines associated with pyroptosis were decreased in the high glucose-treated hypoxia/reoxygenation (H/R)-induced microglia after Fendrr knockdown. Fendrr bound to HERC2 protein, and HERC2 bound to NLRC4. Meanwhile, Fendrr could inhibit the ubiquitination of NLRC4, HERC2 promoted the ubiquitination of NLRC4 protein. Moreover, the effect of Fendrr overexpression in the diabetic cerebral I/R model of microglia can be reversed by HERC2 overexpression. CONCLUSION: Fendrr can protect against the ubiquitination and degradation of NLRC4 protein through E3 ubiquitin ligase HERC2, thereby accelerating the pyroptosis of microglia.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Diabetes Mellitus/genetics , Guanine Nucleotide Exchange Factors/metabolism , Infarction, Middle Cerebral Artery/genetics , Microglia/metabolism , RNA, Long Noncoding , Reperfusion Injury/genetics , Animals , Brain/metabolism , Cell Line , Diabetes Mellitus/metabolism , Infarction, Middle Cerebral Artery/metabolism , Inflammation/genetics , Male , Mice, Inbred C57BL , Pyroptosis , RNA, Long Noncoding/genetics , Reperfusion Injury/metabolism , Ubiquitination
2.
World J Clin Cases ; 7(5): 663-667, 2019 Mar 06.
Article in English | MEDLINE | ID: mdl-30863767

ABSTRACT

BACKGROUND: Empty sella syndrome is a condition in which the pituitary gland shrinks or flattens. Patients with empty sella syndrome often present with headache, hypertension, obesity, visual disturbances, cerebrospinal fluid (CSF) rhinorrhoea, or endocrine dysfunction. Herein, we report a rare case of empty sella syndrome discovered after the patient experienced postoperative hypotension and respiratory failure. CASE SUMMARY: A 60-year-old man was admitted for further workup of left shoulder pain. He was assessed by the orthopaedics team and booked for internal fixation of the left clavicle. General anaesthesia with a nerve block was administered. His blood pressure continued to decrease post-operation. Endocrine tests were performed, with the results supporting a diagnosis of hypopituitarism with hypocortisolism and hypothyroidism. Brain magnetic resonance imaging demonstrated that the sella was enlarged and filled with CSF, confirming a diagnosis of empty sella syndrome. The patient was started on endocrine replacement therapy. The patient regained consciousness and spontaneous breath finally. CONCLUSION: This case highlights the importance of considering pituitary hormone insufficiency in the context of respiratory and hemodynamic failure during the perioperative period.

3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 44(4): 451-7, 2015 07.
Article in Chinese | MEDLINE | ID: mdl-26555425

ABSTRACT

Cerebral blood flow autoregulation is physiologically protective mechanism to maintain the stability of cerebral blood flow. Once autoregulation is impaired, the cerebral blood flow fluctuates with blood pressure, leading to the risk of brain ischemia or cerebral hyperemia. Multiple research results indicate that cerebral blood flow can be monitored indirectly and continuously with transcranial Doppler, near infrared spectroscopy or ICP. The correlation coefficient calculated by the surrogate for cerebral blood flow and blood pressure is used to judge cerebral blood flow autoregulation. When the correlation coefficient is close to 1, cerebral blood flow will be passively fluctuated by blood pressure, indicating autoregulation is impaired. When the coefficient is less than 0, cerebral blood flow will not be changed with blood pressure, indicating autoregulaiton is intact. The status of autoregualtion is closely associated with mortality or poor neurological outcomes in patients with cardiac surgery underwent cardiopulmonary bypass, liver transplantation patients or patients with deep trendelenburg position for long time or beach chair position. Continuous monitoring of cerebral blood flow autoregulation can identify the lower or the upper limit of autoregulation, and provide information to individualize the perioperative management of blood pressure.


Subject(s)
Cerebrovascular Circulation , Monitoring, Intraoperative , Blood Pressure , Cardiopulmonary Bypass , Homeostasis , Humans , Liver Transplantation , Spectroscopy, Near-Infrared
4.
Neurochem Res ; 39(1): 216-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24317635

ABSTRACT

Stroke results in inflammation, brain edema, and neuronal death. However, effective neuroprotectants are not available. Recent studies have shown that high mobility group box-1 (HMGB1), a proinflammatory cytokine, contributes to ischemic brain injury. Aquaporin 4 (AQP4), a water channel protein, is considered to play a pivotal role in ischemia-induced brain edema. More recently, studies have shown that pannexin 1 channels are involved in cerebral ischemic injury and the cellular inflammatory response. Here, we examined whether the pannexin 1 channel inhibitor probenecid could reduce focal ischemic brain injury by inhibiting cerebral inflammation and edema. Transient focal ischemia was induced in C57BL/6J mice by middle cerebral artery occlusion (MCAO) for 1 h. Infarct volume, neurological score and cerebral water content were evaluated 48 h after MCAO. Immunostaining, western blot analysis and ELISA were used to assess the effects of probenecid on the cellular inflammatory response, HMGB1 release and AQP4 expression. Administration of probenecid reduced infarct size, decreased cerebral water content, inhibited neuronal death, and reduced inflammation in the brain 48 h after stroke. In addition, HMGB1 release from neurons was significantly diminished and serum HMGB1 levels were substantially reduced following probenecid treatment. Moreover, AQP4 protein expression was downregulated in the cortical penumbra following post-stroke treatment with probenecid. These results suggest that probenecid, a powerful pannexin 1 channel inhibitor, protects against ischemic brain injury by inhibiting cerebral inflammation and edema.


Subject(s)
Brain Edema/prevention & control , Brain Injuries/prevention & control , HMGB1 Protein/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Probenecid/therapeutic use , Animals , Aquaporin 4/biosynthesis , Astrocytes/drug effects , Cerebral Infarction/pathology , Down-Regulation , HMGB1 Protein/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL
5.
Chin Med J (Engl) ; 125(22): 3977-80, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23158128

ABSTRACT

BACKGROUND: Epidural lidocaine can be used when regional anesthesia needs to be established quickly, but the effect of co-administering epidural fentanyl on the minimum local analgesic concentration (MLAC) of lidocaine is not known. We compared the MLAC of epidural lidocaine in combination with different doses of fentanyl for epidural anesthesia in adults. METHODS: One hundred and twenty patients requiring epidural analgesia were randomly allocated to receive 20 ml of one of four solutions: lidocaine, or lidocaine plus fentanyl 1 µg/ml, 2 µg/ml, or 3 µg/ml. The first patient in each group was administered 1% lidocaine weight by volume; subsequent patients received a concentration determined by the response of the previous patient to a higher or lower concentration according to up and down sequential allocation in 0.1% increments. Efficacy was assessed using a visual analog pain scale, and accepted if this was = 10 mm on a 100 mm scale within 30 minutes. The extent of motor block and of nausea and vomiting were recorded at 30 minutes after administration of the epidural solution and two hours after surgery, respectively. RESULTS: The MLAC of lidocaine in those receiving lidocaine alone was 0.785% (95%CI 0.738 - 0.864). A significant dose-dependent reduction was observed with the addition of fentanyl: the MLAC of lidocaine with fentanyl at 2 µg/ml was 0.596% (95%CI 0.537 - 0.660) and 0.387% with fentanyl at 3 µg/ml (95%CI 0.329 - 0.446, P < 0.001). CONCLUSION: Epidural fentanyl significantly reduces the dose of lidocaine required for effective epidural analgesia in adults without causing adverse side effects.


Subject(s)
Analgesia, Epidural/methods , Fentanyl/administration & dosage , Fentanyl/therapeutic use , Lidocaine/administration & dosage , Lidocaine/therapeutic use , Adult , Drug Interactions , Female , Fistula/surgery , Hemorrhoidectomy , Humans , Male , Middle Aged , Urinary Bladder Neoplasms/surgery
6.
Chin Med J (Engl) ; 123(14): 1888-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20819573

ABSTRACT

BACKGROUND: Astrocyte swelling is an important consequence of hepatic encephalopathy, and aquaporin-4 has been reported to play a vital role in this swelling. Ammonia causes astrocyte swelling and is also known to modulate aquaporin-4 expression in the astrocyte foot processes. The purpose of this study was to explore the mechanism of ammonia-induced aquaporin-4 expression, which has been suggested to involve the p38 mitogen-activated protein kinase pathway. METHODS: We exposed cultured astrocytes to ammonium chloride, an in vitro model of hepatic encephalopathy. The purity of cultured astrocytes was evaluated by fluorescent glial fibrillary acidic protein labeling; cell morphology was assessed by light microscopy; the expression of aquaporin-4, phospho-p38, and p38 were detected by Western blotting analysis. Statistical analysis was performed by one-way factorial analysis of variance, and the relationship between variables was calculated by linear regression using SPSS version 13.0 program for Windows (SPSS, Chicago, IL, USA). RESULTS: The purity of cultured astrocytes was (96.6 +/- 1.4)%. Astrocytes swelled significantly when exposed to 5 mmol/L ammonium chloride for 24 hours as compared to non-exposed astrocytes. Co-treatment with 10 micromol/L SB203580 (an inhibitor of p38) attenuated the degree of ammonium chloride induced astrocyte swelling. Western blotting analysis revealed that the expression levels of phospho-p38 and aquaporin-4 in ammonium chloride treated cells were significantly increased relative to the control group (P < 0.001); SB203580 co-treatment inhibited the increased expression of phospho-p38 and aquaporin-4 relative to the ammonium chloride treated group (P = 0.002 and P = 0.015 respectively). The phosphorylation of p38 and upregulation of aquaporin-4 were highly correlated (r = 0.909). There were no significant differences in total p38 expression among the groups (P = 0.341). CONCLUSIONS: Ammonium chloride induced upregulation of aquaporin-4 in astrocytes is regulated by the p38 mitogen-activated protein kinase pathway. Inhibiting p38 activation prevented ammonium chloride induced aquaporin-4 protein upregulation.


Subject(s)
Ammonium Chloride/pharmacology , Aquaporin 4/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Aquaporin 4/genetics , Blotting, Western , Cells, Cultured , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Anesth Analg ; 109(5): 1493-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19843787

ABSTRACT

BACKGROUND: Aquaporin 4 (AQP4) plays a key role in maintaining water balance in the central nervous system, and its dysfunction may lead to brain edema. Previous studies have suggested that propofol may be involved in neuroprotection by preventing brain edema. In this study, we examined the effects of propofol on edema and assessed its neuroprotective actions in an oxygen and glucose deprivation (OGD) model of cultured rat astrocytes. We assessed the effects of propofol on AQP4 expression and the possible role of the protein kinase C (PKC) pathway on this effect. METHODS: Neocortical astrocytes were exposed to OGD in an anaerobic chamber. After 6 h of OGD exposure, astrocytes were subsequently subjected to 24 h of reoxygenation. Propofol was added during the OGD phase of the model. Cell morphology was assessed by light microscopy. Astrocyte viability was assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide absorbency (optical density value) and the percentage of lactate dehydrogenase released by injured astrocytes. AQP4 expression was evaluated with Western blot analysis. To investigate the possible mechanism of propofol's effects on AQP4 expression, cultured astrocytes were pretreated for 24 h with the PKC activator, 12-O-tetradecanoylphorbol 13-acetate, before the propofol treatment/OGD 6 h/reoxygenation 24 h. RESULTS: We found by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide testing that astrocyte viability began to decrease after about 4 h of OGD exposure and decreased to 60% after 6 h of OGD. When 6 h of OGD was followed by 24 h of reoxygenation, cell viability was further decreased. AQP4 expression was attenuated after 6 h of OGD exposure but was reversed and exceeded baseline levels after 24 h of reoxygenation. Propofol dose-dependently reduced cell death assessed by lactate dehydrogenase test (P < 0.05), and 10 muM propofol significantly down-regulated AQP4 expression in astrocytes after 6 h of OGD followed by 24 h of reoxygenation (P < 0.01). Prolonged (24 h) pretreatment with the phorbol ester, 12-O-tetradecanoylphorbol 13-acetate before OGD significantly reversed the effect of propofol on AQP4 expression (P < 0.01). CONCLUSION: Propofol, administered during OGD, provided neuroprotective effects and down-regulated AQP4 expression in the OGD/reoxygenation model of cultured rat astrocytes. Activation of the PKC pathway may block the effects of propofol.


Subject(s)
Aquaporin 4/drug effects , Astrocytes/drug effects , Brain Diseases/prevention & control , Neocortex/drug effects , Neuroprotective Agents/pharmacology , Propofol/pharmacology , Protein Kinase C/metabolism , Reperfusion Injury/prevention & control , Animals , Animals, Newborn , Aquaporin 4/metabolism , Astrocytes/enzymology , Astrocytes/pathology , Brain Diseases/enzymology , Brain Diseases/pathology , Brain Edema/enzymology , Brain Edema/prevention & control , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Enzyme Activators/pharmacology , Glucose/deficiency , L-Lactate Dehydrogenase/metabolism , Neocortex/enzymology , Neocortex/pathology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
8.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 38(4): 392-8, 2009 Jul.
Article in Chinese | MEDLINE | ID: mdl-19693978

ABSTRACT

OBJECTIVE: To investigate effect of tramadol on c-fos expression in spinal cord dorsal horn and serum IL-6 levels induced by plantar incision in rats. METHODS: The Brennan pain model was induced by incision on the planter surface of left hind paw in rats. Forty-eight rats were randomly divided into six groups: Sham group (Group C), control group (Group I,pretreatment with saline 5 ml), three tramadol pretreatment groups (Group T1, T10 and T20,pretreated with 1 mg/kg, 10 mg/kg and 20 mg/kg tramadol, respectively) and one tramadol treatment group (Group PT10, treated with tramadol 10 mg/kg immediately after operation). Pain behavior was assessed by withdrawal threshold to von Frey filament stimulation intensity, response latency of the hind paw to radiant thermal and a cumulative pain score 2 h after incision. Fos-positive neurons in spinal cord were identified by the immunohistochemical technique. Serum IL-6 levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: WithdrawIal threshold to von Frey filament stimulation intensity and response latency of the hind paw to radiant thermal in Group I were significantly lower than those in Group C (P<0.01). Cumulative pain score in Group I was significantly higher than that in Group C (P<0.01). In Groups of T10 and T20, withdrawal threshold to von Frey filament stimulation intensity and response latency of the hind paw to radiant thermal were significantly higher than those in Group I (P<0.01), cumulative pain score was significantly lower than that in Group I in a dose-dependent manner (P<0.01), and were also those in Group PT10. The greatest density of Fos-positive neurons was located in lamine I-II in Group I. Serum IL-6 levels were significantly elevated in Group I. Pretreatment with tramadol showed a dose-depended inhibitory effect on c-fos expression and serum IL-6 production,but not in Group T1. Administration of tramadol postoperatively also suppressed the c-fos expression and serum IL-6 production as showed in PT10 but were weaker than those in Group T10. CONCLUSION: Pretreatment with tramadol can produce dose-dependent inhibitory effect on c-fos expression in spinal cord dorsal horn and then suppress the inflammatory response to the trauma.


Subject(s)
Interleukin-6/blood , Pain, Postoperative/metabolism , Posterior Horn Cells/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Tramadol/pharmacology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Male , Pain Threshold/drug effects , Pain, Postoperative/drug therapy , Posterior Horn Cells/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Tramadol/therapeutic use
9.
Anesth Analg ; 107(6): 2009-16, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020152

ABSTRACT

BACKGROUND: Cerebral edema is a major threat for stroke victims. Most studies have focused on the neuroprotective activities of propofol, addressing infarct volume rather than cerebral edema. Aquaporin-4 (AQP4) plays an important role in maintaining brain water homeostasis under various neurological insults. We explored the effect of propofol pretreatment on cerebral edema in a rat model of brain ischemia reperfusion and assessed the involvement of AQP4. METHODS: To induce brain ischemia reperfusion, we introduced a silicone-coated monofilament nylon suture into the origin of the middle cerebral artery, withdrawing it after 90 min. Treatment groups (n = 32), received propofol (0.1 mL x kg(-1) x min(-1)) infusion for 30 min before occlusion; the vehicle group (n = 32) and the sham-operated group (n = 28), which received the intralipid vehicle at the same time and rate. To assess cerebral infarct volume, we used 2, 3, 5-triphenyl-tetrazolium chloride staining; wet-dry weight ratio was the basis for cerebral edema estimation, and we used immunohistochemistry and Western blot to detect AQP4 expression. RESULTS: The wet-dry weight ratio decreased from 86.89% +/- 0.71% in the vehicle group (n = 6) to 72.42% +/- 0.74% in the propofol group (n = 6), corresponding to an average decrease of 16%. In parallel and based on immunohistochemical semi-quantification, the propofol group exhibited remarkable attenuation of AQP4 over-expression in the ischemic border zone compared with the vehicle group: 1.28 +/- 0.03 vs 1.40 +/- 0.05, n = 7, respectively; P < 0.05. Values derived from Western blot quantification were similarly decreased in the propofol group compared to the vehicle group: 20.85% +/- 4.18% vs 31.67% +/- 3.23%, n = 4, respectively; P < 0.05. However, infarct volume and neurologic deficit in postischemic rats in the propofol group were not statistically different from values in the vehicle group. CONCLUSIONS: We conclude that prestroke treatment with propofol reduces postischemic cerebral edema in rats, possibly through inhibiting AQP4 over-expression in the boundary zone of ischemia.


Subject(s)
Anesthetics, Intravenous/therapeutic use , Aquaporin 4/genetics , Brain Edema/drug therapy , Ischemic Attack, Transient/complications , Propofol/therapeutic use , Reperfusion Injury/drug therapy , Animals , Aquaporin 4/analysis , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...