Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Medicine (Baltimore) ; 102(39): e35226, 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37773828

ABSTRACT

RATIONALE: Cardiac arrest (CA) caused by trigeminocardiac reflex (TCR) after endoscopic nasal surgery is rare. Hence, when a patient suffers from TCR induced CA in the recovery room, most doctors may not be able to find the cause in a short time, and standard cardiopulmonary resuscitation and resuscitation measures may not be effective. Providing circulatory assistance through venous-arterial extracorporeal membrane oxygenation (VA-ECMO) can help healthcare providers gain time to identify the etiology and initiate symptom-specific treatment. PATIENT CONCERNS: We report a rare case of CA after endoscopic nasal surgery treated with VA-ECMO. DIAGNOSES: We excluded myocardial infarction, pulmonary embolism, allergies, hypoxia, and electrolyte abnormalities based on the relevant examination results. Following a multidisciplinary consultation, clinical manifestation and a review of previous literature, we reasoned that the CA was due to TCR. INTERVENTIONS: VA-ECMO was established to resuscitate the patient successfully during effective cardiopulmonary resuscitation. OUTCOMES: ECMO was successfully evacuated a period of 190 minutes of therapy. The patient was discharged home on day 8. LESSONS: TCR is notable during endoscopic nasal surgery. Our case indicates that CA in operating room is worth prolonged CCPR. The ideal time for ECPR implementation should not be limited within 20 minutes after CCPR.


Subject(s)
Cardiopulmonary Resuscitation , Extracorporeal Membrane Oxygenation , Heart Arrest , Nasal Surgical Procedures , Reflex, Trigeminocardiac , Humans , Extracorporeal Membrane Oxygenation/methods , Heart Arrest/etiology , Heart Arrest/therapy , Cardiopulmonary Resuscitation/methods , Nasal Surgical Procedures/adverse effects , Receptors, Antigen, T-Cell , Retrospective Studies
2.
BMC Anesthesiol ; 22(1): 51, 2022 02 19.
Article in English | MEDLINE | ID: mdl-35183122

ABSTRACT

BACKGROUND: Dexmedetomidine (DEX) has a pharmacological profile that should allow rapid recovery and prevent undesirable outcomes such as pulmonary complications. METHODS: This large retrospective study compared the beneficial effects of perioperative infusion of DEX with propofol on the postoperative outcome after coronary artery bypass graft surgery. We reviewed patients' medical notes at Luoyang Central Hospital from 1st January 2012 to 31st December 2019. All continuous variables, if normally distributed, were presented as mean ± SD; Otherwise, the non-normally distributed data and categorical data were presented as median (25-75 IQR) or number (percentage). The Mann-Whitney U test and Chi-square test were used to evaluate the difference of variables between the DEX and propofol groups. Multivariate logistic regression analysis was performed on the main related and differential factors in the perioperative period. RESULTS: A total of 1388 patients were included in the study; of those, 557 patients received propofol infusion, and 831 patients received dexmedetomidine. DEX significantly reduced postoperative pulmonary complications compared with propofol, 7.82% vs 13.29%; P < 0.01, respectively. When compared with propofol, DEX significantly shortened the duration of mechanical lung ventilation, 18 (13,25) hours vs 21 (16,37) hours; P < 0.001, the length of stay in the intensive care unit, 51 (42,90) vs 59 (46,94.5) hours; P = 0.001 and hospital stay, 20 (17,24) vs 22 (17,28) days; P < 0.001, respectively. The incidences of postoperative wound dehiscence and infection were significantly reduced with DEX compared with propofol groups, 2.53% vs 6.64%; P < 0.001, respectively. Interestingly, patients receiving DEX had significantly shorter surgical time compared to propofol; 275 (240,310) vs 280 (250,320) minutes respectively (P = 0.005) and less estimated blood loss (P = 0.001). CONCLUSION: Perioperative infusion of dexmedetomidine improved the desirable outcomes in patients who had coronary artery bypass graft surgery compared with propofol.


Subject(s)
Coronary Artery Bypass/methods , Dexmedetomidine/pharmacology , Hypnotics and Sedatives/pharmacology , Postoperative Complications/epidemiology , Propofol/pharmacology , Aged , China/epidemiology , Cohort Studies , Female , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , Respiration, Artificial/statistics & numerical data , Retrospective Studies
3.
Int J Neurosci ; 131(9): 843-853, 2021 Sep.
Article in English | MEDLINE | ID: mdl-32345086

ABSTRACT

AIM OF THE STUDY: Hypoxic-ischemic encephalopathy (HIE) is a major cause of newborn brain injury. Apoptosis and necroptosis are two forms of cell death which may occur in HIE but reported data are yet limited. This study investigates the expression of receptor interacting protein kinase (RIPK) 1 and 3, and caspase3, the key modulators of necroptosis and apoptosis, respectively, in a model of HIE to determine whether both forms of cell death occur in the corresponding brain regions. MATERIALS AND METHODS: Postneonatal day 7 Sprague-Dawley rats were subjected to right carotid artery ligation followed by hypoxia or subjected to skin incision under surgical anesthesia without ligation and hypoxia. Neuroglioma (H4) cell was cultured and subjected to 24 h hypoxic insults. Necrostatin-1, a RIPK1 inhibitor, was administered in both in vivo and in vitro settings before insult. RESULTS: After hypoxic-ischemic insults, both RIPK1 and RIPK3 expression were significantly increased in the region of hippocampal dentate gyrus in the injurious hemisphere. However, cleaved caspase3 was significantly increased in the hippocampal cornu ammonis 1 region in the injurious hemisphere. After hypoxic insults, RIPK1 and RIPK3 expression was also found in H4 cells. In addition, it was identified that the increased RIPK1 and RIPK3 can be inhibited by necrostatin-1 in both in vivo and in vitro. CONCLUSIONS: These data indicated that apoptosis and necroptosis occur in different brain regions of hippocampus in a model of HIE which may suggest that strategies to prevent each form of neuronal death is valuable to be developed.


Subject(s)
Apoptosis , Asphyxia/metabolism , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Necroptosis , Animals , Asphyxia/pathology , Cell Line, Tumor , Disease Models, Animal , Hippocampus/pathology , Humans , Hypoxia-Ischemia, Brain/pathology , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
4.
J Anesth ; 28(5): 740-58, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24610035

ABSTRACT

Inhalational anesthetics-induced organoprotection has received much research interest and has been consistently demonstrated in different models of organ damage, in particular, ischemia-reperfusion injury, which features prominently in the perioperative period and in cardiovascular events. The cellular mechanisms accountable for effective organoprotection over heart, brain, kidneys, and other vital organs have been elucidated in turn in the past two decades, including receptor stimulations, second-messenger signal relay and amplification, end-effector activation, and transcriptional modification. This review summarizes the signaling pathways and the molecular participants in inhalational anesthetics-mediated organ protection published in the current literature, comparing and contrasting the 'preconditioning' and 'postconditioning' phenomena, and the similarities and differences in mechanisms between organs. The salubrious effects of inhalational anesthetics on vital organs, if reproducible in human subjects in clinical settings, would be of exceptional clinical importance, but clinical studies with better design and execution are prerequisites for valid conclusions to be made. Xenon as the emerging inhalational anesthetic, and its organoprotective efficacy, mechanism, and relative advantages over other anesthetics, are also discussed.


Subject(s)
Anesthetics, Inhalation/pharmacology , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Animals , Brain/drug effects , Heart/drug effects , Humans , Perioperative Period
5.
Ann Surg ; 259(6): 1235-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24263322

ABSTRACT

OBJECTIVE: Postoperative cognitive decline is emerging as a significant complication of surgery among older adults. Animal models indicate a central role of hippocampal inflammatory responses in the pathophysiology of postoperative cognitive decline. We hypothesized that atorvastatin, shown to exert neuroprotective potential in central nervous system (CNS) disorders, would attenuate neuroinflammation and improve cognitive function in mice after surgery and anesthesia. METHODS: C57BL6 adult mice were pretreated with atorvastatin (250 µg) or vehicle, orally, for 5 days before undergoing unilateral nephrectomy under isoflurane anesthesia. We evaluated behavioral parameters related to cognitive function (fear conditioning and Morris Water Maze) and determined systemic and hippocampal interleukin-1ß levels, postoperatively. Endothelial COX-2 expression, gross NF-κB and microglial (IBA1, CD68) activation, synaptic function (synapsin-1, PSD95, COX-2), heme oxygenase-1, and GSK3ß were also examined. RESULTS: Surgery induced a significant reduction in hippocampal-dependent fear response that was attenuated by treatment with atorvastatin, which also preserved spatial memory on day 7 after surgery. Atorvastatin evoked significant protection from hippocampal interleukin-1ß production, but not systemic interleukin-1ß production, accompanied by a marked reduction in hippocampal endothelial COX-2, NF-κB activation and decreased microglial reactivity. Surgery triggered an acute decline in synapsin-1, paralleled by an increase in postsynaptic COX-2 that was partially attenuated by atorvastatin. Furthermore, phosphorylation and inactivation of neuronal GSK3ß was significantly enhanced after atorvastatin treatment. CONCLUSIONS: These findings indicate that cognitive decline is very likely associated with synaptic pathology after systemic and central inflammation induced by peripheral surgery/isoflurane anesthesia and suggest that the anti-inflammatory and neuroprotective properties of atorvastatin provide a rationale for its use as a therapeutic strategy for postoperative cognitive decline.


Subject(s)
Cognition Disorders/drug therapy , Cognition/drug effects , Heptanoic Acids/administration & dosage , Memory/drug effects , Nephrectomy/adverse effects , Pyrroles/administration & dosage , Recovery of Function/drug effects , Administration, Oral , Animals , Atorvastatin , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Mice , Mice, Inbred C57BL , Postoperative Complications , Treatment Outcome
6.
Neurobiol Dis ; 45(2): 743-50, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075165

ABSTRACT

Anesthetic-induced neurodegeneration in the developing brain has been well documented. However, the experiments carried out so far do not include surgical conditions. This proof of concept study was designed to investigate the impact of nociceptive stimuli on anesthetic induced neuroapoptosis in the rat developing brain. Separate cohorts of 7-day-old Sprague-Dawley rat pups were randomly assigned to six groups: Naïve (room air); Anesthesia alone (70% nitrous oxide and 0.75% isoflurane for 6 h); Formalin injection alone (subcutaneous injection with 10 µL 5% formalin into the left hind paw); Anesthesia+formalin injection; Surgical incision (to the left hind paw) alone; Anesthesia+surgical incision. Apoptosis (Caspase-3) and neuronal activation (c-Fos) in the brain and spinal cord section, and cortical TNF-α and IL-1ß were measured with in situ immunostaining and western blot respectively. Cognition was tested using Trace Fear conditioning 40 days after the insult. Prolonged anesthesia caused widespread apoptosis in the central nervous system compared to naïve animals. Nociceptive stimulation with formalin (F) or surgical incision (S) increased the injury in the brain cortex (F: 60% or S: 40% increase) and spinal cord (F: 80% vs. S: 50% increase) respectively. Both nociceptive stimuli further augmented cognitive impairment induced by the anesthetics when assessed 40 days later. The activated pain pathway and the increased expression of the pro-inflammatory cytokine, IL-1ß, in the cortex may be responsible for the enhanced neuroapoptosis. Nociceptive stimulation and prolonged anesthesia produced significantly more apoptosis than prolonged anesthesia alone when administered to neonates during the synaptogenic period.


Subject(s)
Anesthetics, Inhalation/toxicity , Apoptosis/physiology , Brain/drug effects , Neurons/drug effects , Pain/physiopathology , Animals , Animals, Newborn , Apoptosis/drug effects , Behavior, Animal/drug effects , Blotting, Western , Brain/pathology , Cognition/drug effects , Formaldehyde/toxicity , Immunohistochemistry , Irritants/toxicity , Isoflurane/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neurons/pathology , Nitrous Oxide/toxicity , Pain/chemically induced , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/pathology
7.
J Alzheimers Dis ; 27(1): 1-10, 2011.
Article in English | MEDLINE | ID: mdl-21734347

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease commonly seen in the elderly and is characterized by progressive cognitive and physical decline. Current understanding of AD pathogenesis revolves around amyloid-ß peptide (Aß), a product of the sequential proteolytic cleavage of the transmembrane amyloid-ß protein precursor (AßPP) by ß- and γ-secretase, enzymes found predominantly in the cholesterol rich micro domains of the cell membrane. Several risk factors for AD are associated with cholesterol metabolism, including dyslipidaemia, coronary artery and cerebrovascular disease. Statins are widely prescribed for their cholesterol lowering ability and show a favorable side effect profile overall. By competitive inhibition of hydroxymethyl co-enzyme A-reductase, statins reduce the production of cholesterol and isoprenoid intermediates including geranylgeranyl and farnesyl pyrophosphate. These isoprenoids modify recently translated proteins such as small GTPase molecules that are essential in numerous cell-signaling pathways, including vesicular trafficking and inflammation. In experimental models of AD, statins reduce the production of Aß by disrupting secretase enzyme function and by reducing neuroinflammation. Furthermore, epidemiological studies suggest that statins may reduce the incidence of AD. Consequently, statins, secondary of their anti-hypercholesterolaemic, plieotropic and anti-inflammatory effects, are being investigated for a potential therapeutic role. This review will discuss evidence for the role of statins in the treatment and prevention of AD neurodegeneration.


Subject(s)
Alzheimer Disease/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Nerve Degeneration/prevention & control , Age Factors , Aged , Aged, 80 and over , Amyloid beta-Peptides , Cerebrovascular Disorders/drug therapy , Cholesterol/metabolism , Disease Progression , Humans
8.
Anesthesiology ; 113(2): 360-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20613483

ABSTRACT

BACKGROUND: Administration of certain general anesthetics to rodents during the synaptogenic phase of neurodevelopment produces neuronal injury. Preconditioning (pretreatment) can reduce tissue injury caused by a severe insult; the authors investigated whether pretreatment strategies can protect the developing brain from anesthetic-induced neurotoxicity. METHODS: Seven-day-old Sprague-Dawley rats were pretreated with one of the following: 70% xenon, 70% nitrous oxide, or 8% hypoxia for 2 h; 24 h later, rats were exposed to the neurotoxic combination of 70% nitrous oxide and 0.75% isoflurane for 6 h. Cortical and hippocampal neuroapoptosis was assessed using caspase-3 immunostaining. Separate cohorts were maintained for 40 days at which time cognitive function with trace fear conditioning was performed. In other pretreated cohorts, rat cortices were isolated for immunoblotting of caspase-3, Bcl-2, cytochrome C, P53, and mitogen-activated protein kinases. To obviate physiologic influences, organotypic hippocampal slices harvested from postnatal rat pups were cultured for 5 days and exposed to the same conditions as obtained for the in vivo studies, and caspase-3 immunostaining was again the measured outcome. RESULT: Xenon pretreatment prevented nitrous oxide- and isoflurane-induced neuroapoptosis (in vivo and in vitro) and cognitive deterioration (in vivo). Contrastingly, nitrous oxide- and isoflurane-induced neuroapoptosis was exacerbated by hypoxic pretreatment. Nitrous oxide pretreatment had no effect. Xenon pretreatment increased Bcl-2 expression and decreased both cytochrome C release and P53 expression; conversely, the opposite was evident after hypoxic pretreatment. CONCLUSIONS: Although xenon pretreatment protects against nitrous oxide- and isoflurane-induced neuroapoptosis, hypoxic pretreatment exacerbates anesthetic-induced neonatal neurodegeneration.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Apoptosis/drug effects , Brain/growth & development , Hypoxia/metabolism , Nitrous Oxide/toxicity , Xenon/administration & dosage , Animals , Animals, Newborn , Apoptosis/physiology , Brain/cytology , Brain/drug effects , Cohort Studies , Hypoxia/chemically induced , Hypoxia/pathology , Ischemic Preconditioning/methods , Nitrous Oxide/administration & dosage , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...