1
|
Kast KA, Sidelnik SA, Nejad SH, Suzuki J. Management of alcohol withdrawal syndromes in general hospital settings. BMJ 2025; 388:e080461. [PMID: 39778965 DOI: 10.1136/bmj-2024-080461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The covid-19 pandemic was associated with an unprecedented increase in alcohol consumption and associated morbidity, including hospitalizations for alcohol withdrawal. Clinicians based in hospitals must be ready to identify, assess, risk-stratify, and treat alcohol withdrawal with evidence based interventions. In this clinically focused review, we outline the epidemiology, pathophysiology, clinical manifestations, screening, assessment, and treatment of alcohol withdrawal in the general hospital population. We review and summarize studies addressing the drug treatment of alcohol withdrawal syndromes in inpatient populations, with a focus on the use of benzodiazepine drugs, phenobarbital, antiseizure drugs, and α-2 adrenergic drugs. Emerging areas of interest include the use of novel alcohol biomarkers, risk stratification instruments, alternative symptom severity scales, severe withdrawal syndromes resistant to benzodiazepine drugs, and treatment protocol variations-including non-symptom-triggered and benzodiazepine-sparing protocols. We identify key areas for research including identification of populations who will benefit from non-benzodiazepine strategies, more individualized risk stratification approaches to guide treatment, and greater inclusion of gender and racial and ethnic minorities in future studies.
Collapse
Affiliation(s)
- Kristopher A Kast
- Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - S Alex Sidelnik
- New York University Langone Health, New York University Grossman School of Medicine, New York, NY, USA
| | - Shamim H Nejad
- Addiction Medicine Consultation Services, Psychiatry Consultation Services, Valley Medical Center, Renton, WA, USA
| | - Joji Suzuki
- Division of Addiction Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Zheng T, Pei Z, Huang X. Progress and Frontiers of Research on Dexmedetomidine in Perioperative Medicine: A Bibliometric Analysis. Drug Des Devel Ther 2024; 18:6017-6029. [PMID: 39687681 PMCID: PMC11648549 DOI: 10.2147/dddt.s471602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Background Dexmedetomidine has received increasing attention for its sedative, analgesic, anxiolytic, anti-inflammatory and anti-stress effects in perioperative medicine. Numerous studies have been carried out to explore its influence on perioperative patients. Objective This study aimed to identify the most influential literature, trends and hotspots in dexmedetomidine research by bibliometric analysis. Methods Articles and reviews related to dexmedetomidine in perioperative medicine were collected from Web of Science Core Collection. VOSviewer and Citespace software were used for bibliometric analysis and data visualization. Results A total of 1652 suitable publications were extracted from the database for analysis, including 1,497 articles and 155 reviews. The number of publications in the field of dexmedetomidine research has increased markedly since 2013, with China being the major contributor, followed by United States. BMC Anesthesiology published the highest number of papers on this topic. Anesthesiology ranked first in terms of average citations per paper and co-citation journal. Ji Fuhai was the most prolific author, and Ma Daqing was the most cited authors. The main hotspots during this period were "elderly patients", "postoperative cognitive dysfunction", "injury" and "risk factors". Conclusion This study presents an overview of the development related to dexmedetomidine in perioperative medicine using bibliometric analysis. Dexmedetomidine research is thriving and expanding rapidly around the world. The effect of dexmedetomidine on cognitive function has been the latest research hotspot. To advance research in this field, more rigorous and scientific multi-center studies should be designed and further cooperation and academic exchange should be strengthened.
Collapse
Affiliation(s)
- Teng Zheng
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Zhi Pei
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaojing Huang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Pain Medicine, Shanghai Geriatric Medical Center, Minhang District, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Irving C, Durrmeyer X, Decobert F, Dassieu G, Ben Guirat A, Gouyon B, Tauzin M. Use of dexmedetomidine during mechanical ventilation in extremely preterm and extremely low birth weight neonates receiving morphine: A single-center retrospective study. PAEDIATRIC & NEONATAL PAIN 2024; 6:194-202. [PMID: 39677027 PMCID: PMC11645972 DOI: 10.1002/pne2.12130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/03/2024] [Accepted: 07/11/2024] [Indexed: 12/17/2024]
Abstract
Analgesia and sedation are often provided during mechanical ventilation in extremely preterm neonates. Opioids and benzodiazepines are the most frequently used agents but can have adverse effects. Dexmedetomidine, an alpha-2 agonist, might be interesting to spare opioid and benzodiazepine use. The objective of this study was to describe a cohort of mechanically ventilated extremely, preterm infants treated with morphine with or without dexmedetomidine. This was a retrospective, observational, single-center study in the neonatal intensive care unit of Creteil. We included preterm neonates born before 28 weeks of gestation and/or weighting less than 1000 g hospitalized between July 2017 and June 2020, on mechanical ventilation for at least 72 h and who received morphine with or without dexmedetomidine as a second- or third-line treatment. We described morphine and midazolam exposure, respiratory, and digestive outcomes for patients who received dexmedetomidine and those who did not. Twenty nine preterm infants received morphine and dexmedetomidine, and 44 received morphine without dexmedetomidine. Dexmedetomidine was used in patients of 25.7 [25.1-26.7] weeks, 680 [600-750] g and significantly more often in patients with vascular complications during pregnancy (p = 0.008), intrauterine growth restriction (p = 0.01) and in patients who received higher cumulative doses of morphine (p = 0.01). Morphine and midazolam doses tended to decrease after the introduction of dexmedetomidine. Dexmedetomidine was never discontinued because of side effects. In this study, dexmedetomidine, used as a second or third-line treatment during mechanical ventilation, was associated with a decrease in morphine and midazolam doses after introduction. Dexmedetomidine was used in a specific population of extremely preterm infants, with severe respiratory disease, who required prolonged mechanical ventilation and high morphine doses. This study highlights the need for pharmacokinetic/pharmacodynamic studies in this population, followed by randomized controlled trials and studies on the long-term effects of dexmedetomidine to determine its place in analgosedation of ventilated preterm infants.
Collapse
Affiliation(s)
- Camille Irving
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
| | - Xavier Durrmeyer
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
- Université Paris Est Créteil, Faculté de Santé de Créteil, IMRB, GRC CARMASCréteilFrance
| | - Fabrice Decobert
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
| | - Gilles Dassieu
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
| | - Aroua Ben Guirat
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
| | - Béatrice Gouyon
- Centre d'Etudes Périnatales de L'Océan Indien (CEPOI, EA7388)Université de La RéunionSaint PierreFrance
| | - Manon Tauzin
- Réanimation néonataleCentre Hospitalier Intercommunal de CréteilCréteilFrance
| |
Collapse
|
4
|
Vetter C, Meyer ER, Seidel K, Bervini D, Huber M, Krejci V. Co-administration of dexmedetomidine with total intravenous anaesthesia in carotid endarterectomy reduces requirements for propofol and improves haemodynamic stability: A single-centre, prospective, randomised controlled trial. Eur J Anaesthesiol 2024:00003643-990000000-00243. [PMID: 39529482 DOI: 10.1097/eja.0000000000002099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Total intravenous anaesthesia guided by electroencephalography and neurophysiological monitoring may be used for carotid endarterectomy. Reduction of brain metabolic demand during cross-clamping of the internal carotid artery with propofol titrated to burst suppression requires effect-site concentrations that may delay emergence and interfere with intraoperative neurophysiological monitoring. OBJECTIVE To test the hypothesis that dexmedetomidine decreases the effect-site concentration of propofol required for burst-suppression in patients undergoing carotid endarterectomy. DESIGN Randomised controlled trial. PARTICIPANTS Patients undergoing carotid endarterectomy. SETTING University Hospital of Berne, Switzerland, from October 2018 to September 2024. INTERVENTIONS Patients were randomised into a control (n = 23) and a dexmedetomidine groups (n = 22). Total intravenous anaesthesia was administered to both groups. Patients in the dexmedetomidine group received an intravenous bolus of dexmedetomidine (0.4 μg kg-1 over 10 min) before induction, followed by a continuous intravenous infusion (0.4 μg kg-1 h-1). The effect-site concentrations of propofol were titrated against frontal electroencephalography parameters. Burst suppression was induced with propofol during cross-clamping of the internal carotid artery. OUTCOME MEASURES The primary outcome was the effect-site concentration of propofol required for burst-suppression. The secondary outcomes were the requirement for vasoactive substances, neurophysiological monitoring parameters, and postoperative delirium. RESULTS The effect-site concentration of propofol required for burst suppression was 4.0 μg ml-1 [3.50 to 4.90] (median [interquartile range]) in the dexmedetomidine group compared with 6.0 μg ml-1 [5.5 to 7.3] in the control group (P < 0.001). Less norepinephrine was required in the dexmedetomidine group (total 454 μg [246 to 818] compared with 1000 μg [444 to 1326] (P = 0.015) in the control group). Dexmedetomidine did not affect intraoperative neurophysiological monitoring. CONCLUSION Co-administration of dexmedetomidine to total intravenous anaesthesia for carotid endarterectomy decreased the effect-site concentrations of propofol required for burst suppression by 33%. The propofol-sparing effect and peripheral alpha-agonism of dexmedetomidine may explain the reduced requirement for vasopressors. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT04662177.
Collapse
Affiliation(s)
- Christian Vetter
- From the Department of Anaesthesiology and Pain Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland (CV, ERM, MH, VK), and Department of Neurosurgery, Inselspital, University Hospital, University of Bern, Bern, Switzerland (KS, DB)
| | | | | | | | | | | |
Collapse
|
5
|
Ma H, Perez J, Bertsch J, Albanese ML, Korn EG, Mueller A, Houle T, Burris BJ, McCain J, Wang J. Intraoperative Non-Opiate Anesthesia for Patients Undergoing Arthroscopic Temporomandibular Joint Surgery: A Randomized Controlled Trial. Drug Des Devel Ther 2024; 18:4915-4925. [PMID: 39525049 PMCID: PMC11546274 DOI: 10.2147/dddt.s486134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Background Pain intensity after temporomandibular joint (TMJ) surgery is often underestimated, and inadequate pain control may relate to poor recovery quality, increased opioid consumption, and longer hospital stay. This trial aims to evaluate whether non-opiate anesthesia provides a promising option of pain management for patients undergoing TMJ surgery. Methods Sixty patients receiving TMJ surgery were randomly assigned to either the control group or the non-opiate group. Non-opiate anesthesia used lidocaine, dexmedetomidine, and ketamine infusion therapy for pain management. The primary outcome was the highest documented pain score while in the post-anesthesia care unit (PACU). Secondary outcomes included perioperative opioid consumption, utilization, dosage, and timing of rescue analgesia in the PACU, incidence of postoperative nausea and vomiting in the PACU and at home, pain satisfaction levels, occurrence of opioid-related adverse effects, duration of PACU and hospital stays, and total consumption of oxycodone-acetaminophen tablets at 24 and 48 hours post-surgery. Results Patients were predominantly female (88.3%) and had a median age of 37.5 [IQR 26.0, 52.5] years. There were no significant differences observed in the highest documented pain scores (mean difference [MD] -0.36 points, 95% CI: -1.84, 1.12, p = 0.63), postoperative oxycodone-acetaminophen consumption (MD 6.68 mg, 95% CI: -2.48, 15.84, p = 0.15), pain satisfaction (odds ratio [OR] 0.81, 95% CI: 0.23, 2.81, p = 0.74), time to PACU discharge (hazard ratio [HR] 1.24, 95% CI: 0.67, 2.30, p = 0.49) or time to hospital discharge (HR 1.48, 95% CI: 0.80, 2.75, p = 0.21) between the two groups. Similarly, no significant difference was observed in time to rescue analgesia, calculated in minutes from the end of surgery (HR 1.69, 95% CI: 0.79, 3.61, p = 0.18). Conclusion Non-opiate anesthesia for pain management shows a similar postoperative analgesia effect, compared to opioid-based anesthesia, in patients undergoing arthroscopic TMJ surgery.
Collapse
Affiliation(s)
- He Ma
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, People’s Republic of China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Juan Perez
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Julia Bertsch
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marissa L Albanese
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth G Korn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ariel Mueller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy Houle
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Briana June Burris
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph McCain
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jingping Wang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Scheckenbach V, Fideler F. Optimizing Pediatric Sedation: Evaluating Remimazolam and Dexmedetomidine for Safety and Efficacy in Clinical Practice. Paediatr Drugs 2024:10.1007/s40272-024-00659-1. [PMID: 39485634 DOI: 10.1007/s40272-024-00659-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2024] [Indexed: 11/03/2024]
Abstract
Daily, children undergo countless investigations and interventions, which require sedation and immobilization to ensure safety and accuracy. This remains associated with a persistent risk of sedation-induced life-threatening events as children are particularly vulnerable to adverse medical events and complications. Consequently, there is an urgent need to increase the safety of pediatric sedation and anesthesia. An ideal approach involves the use of drugs with fewer intrinsic side effects. In this context, on the basis of their pharmacokinetic properties, remimazolam (RMZ) and dexmedetomidine (DEX) were evaluated for their suitability as ideal sedatives. RMZ and DEX, both of which are currently available in pediatric medicine, have shown great promise in initial publications. To date, only very limited data concerning RMZ in small children are available. RMZ is a novel, ultrashort-acting benzodiazepine that is metabolized by tissue esterase, largely independent of organ function. It has a context-sensitive half-life of approximately 10 min, with minimal accumulation even with prolonged use. Its effects can be completely reversed with flumazenil. DEX, an isomer of medetomidine, is a potent α2-receptor-agonist with multiple indications in anesthesia and intensive care medicine. It has coanalgesic potential, allows for 'arousal sedations' and has a low profile for cardiorespiratory side effects. DEX is metabolized in the liver and is predominantly excreted renally. Both drugs show potential in the prevention and treatment of delirium, with DEX having additional neuroprotective effects. DEX and RMZ possess several properties of an optimal sedative, including clinically insignificant main metabolites and a broad dosage range, indicating their potential to reduce the incidence of sedation-related life-threatening events in children. However, further clinical research is necessary to better evaluate their potential risks.
Collapse
Affiliation(s)
- Vera Scheckenbach
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Eberhard Karls University Tübingen, Hoppe-Seyler Str. 3, 72076, Tübingen, Germany.
| | - Frank Fideler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Eberhard Karls University Tübingen, Hoppe-Seyler Str. 3, 72076, Tübingen, Germany
| |
Collapse
|
7
|
Tao Z, Li P, Zhao X. Progress on the Mechanisms and Neuroprotective Benefits of Dexmedetomidine in Brain Diseases. Brain Behav 2024; 14:e70116. [PMID: 39482839 PMCID: PMC11527817 DOI: 10.1002/brb3.70116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION Dexmedetomidine, a highly specific α2 agonist, has been extensively utilized in clinical sedation and surgical anesthesia since its introduction in 2000 due to its excellent sympatholytic, sedative, and analgesic effects. This review aimed to identify new approaches for the treatment of patients with brain disorders by thoroughly describing the mechanism of action of dexmedetomidine and examining its neuroprotective effects from the standpoints of basic and clinical research. METHODS The PubMed and Web of Science databases were searched using the keywords dexmedetomidine and related brain diseases, although relevant articles from the last decade were included for detailed summarization and analysis. RESULTS Dexmedetomidine has shown strong neuroprotective effects, such as protection of the blood-brain barrier, decreased neuronal death, maintained hemodynamic stability, and reduced postoperative agitation and cognitive dysfunction. Furthermore, dexmedetomidine has been shown to exert various neuroprotective effects, including anti-inflammatory and antioxidative stress effects, modulation of autophagy, and reduction of apoptosis in cerebral diseases. CONCLUSIONS Dexmedetomidine acts as a neuroprotective agent against brain diseases during all phases of treatment. However, clinical trials with larger sample sizes are required to optimize dosage and dosing strategies.
Collapse
Affiliation(s)
- Zhenxing Tao
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Pengpeng Li
- Wuxi Medical SchoolJiangnan UniversityWuxiChina
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
| | - Xudong Zhao
- Department of NeurosurgeryJiangnan University Medical CenterWuxiChina
- Wuxi Neurosurgical InstituteWuxiChina
| |
Collapse
|
8
|
Feng H, Zhang Z, Lyu W, Kong X, Li J, Zhou H, Wei P. The Effects of Appropriate Perioperative Exercise on Perioperative Neurocognitive Disorders: a Narrative Review. Mol Neurobiol 2024; 61:4663-4676. [PMID: 38110646 PMCID: PMC11236851 DOI: 10.1007/s12035-023-03864-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Perioperative neurocognitive disorders (PNDs) are now considered the most common neurological complication in older adult patients undergoing surgical procedures. A significant increase exists in the incidence of post-operative disability and mortality in patients with PNDs. However, no specific treatment is still available for PNDs. Recent studies have shown that exercise may improve cognitive dysfunction-related disorders, including PNDs. Neuroinflammation is a key mechanism underlying exercise-induced neuroprotection in PNDs; others include the regulation of gut microbiota and mitochondrial and synaptic function. Maintaining optimal skeletal muscle mass through preoperative exercise is important to prevent the occurrence of PNDs. This review summarizes current clinical and preclinical evidence and proposes potential molecular mechanisms by which perioperative exercise improves PNDs, providing a new direction for exploring exercise-mediated neuroprotective effects on PNDs. In addition, it intends to provide new strategies for the prevention and treatment of PNDs.
Collapse
Affiliation(s)
- Hao Feng
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Zheng Zhang
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Haipeng Zhou
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China.
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China.
| |
Collapse
|
9
|
Gavet M, Junot S. Anesthetic management of a dog undergoing unilateral adrenalectomy for phaeochromocytoma excision using a partial intravenous anesthetic protocol. Open Vet J 2024; 14:1483-1490. [PMID: 39055755 PMCID: PMC11268903 DOI: 10.5455/ovj.2024.v14.i6.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Background The anesthetic management of adrenalectomies for phaeochromocytoma excision, a catecholamine-secreting tumor, is challenging due to the potential for fatal complications following severe hemodynamic variations, including hypertensive crisis following tumor manipulation or sympathetic stimulation, but also severe hypotension and volume depletion post resection. Case Description An 11 kg, 15-year-old male neutered Jack Russel Terrier, with mitral valve disease stage B2, was referred for adrenalectomy for phaeochromocytoma resection. The patient was administered per os prazosin 0.11 mg/kg twice a day and amlodipine 0.125 mg/kg once a day for preoperative stabilization. On the day of surgery, the dog received maropitant 1 mg/kg intravenously (IV) and was premedicated with 0.2 mg/kg methadone IV. Anesthesia was induced with alfaxalone 1 mg/kg IV and midazolam 0.2 mg/kg IV and maintained with partial intravenous anesthesia using sevoflurane in 70% oxygen and constant rate infusions of dexmedetomidine 0.5 μg/kg/hour and maropitant 100 μg/kg/hour. After induction of anesthesia, the dog was mechanically ventilated, and a transversus abdominal plane block was performed with ropivacaine 0.2%. The dog remained remarkably stable with a single, self-limiting, hypertension episode recorded intraoperatively. Postoperative rescue analgesia consisted of methadone and ketamine. The dog was discharged 48 hours after surgery, but persistent hypertension was reported at suture removal. Conclusion The use of a low-dose dexmedetomidine CRI, a maropitant CRI, and a transversus abdominal plane block provided stable perioperative hemodynamic conditions for phaeochromocytoma excision in a dog.
Collapse
Affiliation(s)
- Morgane Gavet
- Service d’Anesthésie, Université de Lyon, Marcy l’Etoile, France
| | - Stéphane Junot
- Service d’Anesthésie, Université de Lyon, Marcy l’Etoile, France
| |
Collapse
|
10
|
Yuan HX, Zhang LN, Li G, Qiao L. Brain protective effect of dexmedetomidine vs propofol for sedation during prolonged mechanical ventilation in non-brain injured patients. World J Psychiatry 2024; 14:370-379. [PMID: 38617978 PMCID: PMC11008391 DOI: 10.5498/wjp.v14.i3.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Dexmedetomidine and propofol are two sedatives used for long-term sedation. It remains unclear whether dexmedetomidine provides superior cerebral protection for patients undergoing long-term mechanical ventilation. AIM To compare the neuroprotective effects of dexmedetomidine and propofol for sedation during prolonged mechanical ventilation in patients without brain injury. METHODS Patients who underwent mechanical ventilation for > 72 h were randomly assigned to receive sedation with dexmedetomidine or propofol. The Richmond Agitation and Sedation Scale (RASS) was used to evaluate sedation effects, with a target range of -3 to 0. The primary outcomes were serum levels of S100-β and neuron-specific enolase (NSE) every 24 h. The secondary outcomes were remifentanil dosage, the proportion of patients requiring rescue sedation, and the time and frequency of RASS scores within the target range. RESULTS A total of 52 and 63 patients were allocated to the dexmedetomidine group and propofol group, respectively. Baseline data were comparable between groups. No significant differences were identified between groups within the median duration of study drug infusion [52.0 (IQR: 36.0-73.5) h vs 53.0 (IQR: 37.0-72.0) h, P = 0.958], the median dose of remifentanil [4.5 (IQR: 4.0-5.0) μg/kg/h vs 4.6 (IQR: 4.0-5.0) μg/kg/h, P = 0.395], the median percentage of time in the target RASS range without rescue sedation [85.6% (IQR: 65.8%-96.6%) vs 86.7% (IQR: 72.3%-95.3), P = 0.592], and the median frequency within the target RASS range without rescue sedation [72.2% (60.8%-91.7%) vs 73.3% (60.0%-100.0%), P = 0.880]. The proportion of patients in the dexmedetomidine group who required rescue sedation was higher than in the propofol group with statistical significance (69.2% vs 50.8%, P = 0.045). Serum S100-β and NSE levels in the propofol group were higher than in the dexmedetomidine group with statistical significance during the first six and five days of mechanical ventilation, respectively (all P < 0.05). CONCLUSION Dexmedetomidine demonstrated stronger protective effects on the brain compared to propofol for long-term mechanical ventilation in patients without brain injury.
Collapse
Affiliation(s)
- Hong-Xun Yuan
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Li-Na Zhang
- Central Operating Room, The Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing 100020, China
| | - Gang Li
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| | - Li Qiao
- Intensive Care Unit, Peking University International Hospital, Beijing 102206, China
| |
Collapse
|
11
|
Fu Y, Jin Z. Effects of Dexmedetomidine on Cognitive Function, Oxidative Stress and Brain Protection in Patients Undergoing Craniocerebral Surgery. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:19-27. [PMID: 38454897 PMCID: PMC10926013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
BACKGROUND The protective mechanism of dexmedetomidine on the brains of patients undergoing craniocerebral surgery remains unclear. The aim of this study was to examine the impact of dexmedetomidine on cognitive function, oxidative stress, and brain protection in such patients. METHODS Fifty-four patients who underwent craniocerebral surgery at our hospital from January 2020 to June 2023 were retrospectively selected as study subjects. They were divided into two groups: the control group (n = 27) and the study group (n = 27), based on different auxiliary anesthesia protocols. Patients in the study group received dexmedetomidine before anesthesia induction, using a midline intravenous pump to assist anesthesia, while the control group received an equivalent amount of normal saline. The remaining anesthesia induction and maintenance protocols were consistent for both groups. Cognitive function was assessed using the Mini Mental State Examination (MMSE) before and 1 day after surgery for both groups. Oxidative stress indicators, including malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) levels in the serum of both groups, were measured using enzyme-linked immunosorbent assay (ELISA). Additionally, changes in postoperative brain injury indicators, namely neuron-specific enolase (NSE) and central nervous system-specific protein (S100β), were detected and compared in the serum of both groups. Concurrently, postoperative adverse reactions were recorded for both groups. RESULTS The MMSE scale scores of both groups of patients 24 hours after surgery were significantly lower than those before surgery. However, the MMSE scale scores of the study group patients were notably higher than those in the control group, with a statistically significant difference (p < 0.05). One hour after surgery, the serum levels of MDA, GSH-Px, and SOD in both groups of patients were significantly elevated compared to pre-surgery levels. Yet, the study group exhibited significantly lower levels of MDA, GSH-Px, and SOD in comparison to the control group, and these differences were statistically significant (p < 0.05). The serum levels of NSE and S100β in both groups were markedly higher than preoperative levels 24 hours after surgery. However, the study group demonstrated significantly lower levels of serum NSE and S100β compared to the control group, with a statistically significant difference (p < 0.05). The incidence of postoperative complications in the study group was 7.41% (2/27), indicating a decreasing trend compared to 18.52% (5/27) in the control group. However, this difference did not reach statistical significance (χ2 = 1.477, p = 0.224). CONCLUSION Dexmedetomidine-assisted anesthesia in craniocerebral surgery can effectively enhance postoperative cognitive function, mitigate oxidative stress, and facilitate overall postoperative recovery for patients. The intervention exhibits a favorable safety profile with no reported serious adverse reactions, establishing it as a relatively safe and reliable approach.
Collapse
Affiliation(s)
- Yan Fu
- Department of Anesthesiology, The First People’s Hospital of Daishan, 316200 Zhoushan, Zhejiang, China
| | - Zhu Jin
- Department of Anesthesiology, Sahzu International Medical Center, 311215 Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
He J, Zhang X, Li C, Fu B, Huang Y, Li H. Dexmedetomidine nasal administration improves perioperative sleep quality and neurocognitive deficits in elderly patients undergoing general anesthesia. BMC Anesthesiol 2024; 24:42. [PMID: 38291398 PMCID: PMC10826024 DOI: 10.1186/s12871-024-02417-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/14/2024] [Indexed: 02/01/2024] Open
Abstract
OBJECTIVE To investigate the improvement of perioperative sleep quality and neurocognitive impairment in elderly patients under general anesthesia by nasal administration of dexmedetomidine. METHODS One hundred and twenty patients admitted to our hospital for various laparoscopic elective gynecological surgeries lasting more than 1 h under general anesthesia from July 2021 to March 2023 were selected. All subjects were divided into 3 groups according to the random number table method. From 21:00 to 21:30 every night from one day before to 5 days after surgery, group A was given alprazolam 0.4 mg orally; group B was given dexmedetomidine 1.5ug/kg nasal drip; group C was given saline nasal drip. All subjects were observed for general information, sleep quality, postoperative cognitive function, anxiety status, sleep quality, adverse effects and complication occurrence. RESULTS The difference in general information between the three groups was not statistically significant, P > 0.05; the sleep quality scores of the three groups on admission were not statistically significant, P > 0.05. At the Preoperative 1d, postoperative 1d, 3d and 5d, the RCSQ scores of the subjects in group A and group B were higher than those in groups C, and with the postoperative RCSQ scores of subjects in group B were higher as the time increased; the assessment of anxiety status in the three groups 1d before surgery was not statistically significant, P > 0.05. The cognitive function scores of subjects in the three groups were not statistically significant in the preoperative 1d, P > 0.05. The postoperative 1d (24.63 ± 2.23), 3d (25.83 ± 2.53), and 5d (26.15 ± 2.01) scores of the subjects in group B were higher than those in groups A and C (P < 0.05), and the subjects in group B had better recovery of postoperative cognitive function with increasing time; the occurrence of postoperative delirium (POD) in group B (12.5%) were lower on postoperative 5d than those in groups A (37.5%) and C (32.5%) (P < 0.05). There was no statistical significance in the evaluation of anxiety state of the three groups on the first day before operation (P > 0.05). The scores in group B were lower than those in group C on the postoperative 1d, 3d, 5 d (P < 0.05). The overall incidence of adverse reactions and complications in subjects in group B was 17.5% significantly lower than that in groups A and C (P < 0.05). CONCLUSION Dexmedetomidine can effectively improve the sleep disorder of elderly general anesthesia patients, reduce the damage to their neurocognitive function and the occurrence of POD, effectively reduce the anxiety of patients and the occurrence of adverse reactions and complications, and has better sedative, improve postoperative cognitive function and anti-anxiety effects, with a high drug safety, worthy of clinical application and promotion.
Collapse
Affiliation(s)
- Jiang He
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Xinning Zhang
- Department of Gynaecology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Cuicui Li
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Baojun Fu
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yizhou Huang
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Heng Li
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China.
| |
Collapse
|
13
|
Hamedani SG, Pourmasoumi M, Askari G, Bagherniya M, Sathyapalan T, Sahebkar A. An Investigation into the Effects of Chemical, Pharmaceutical, and Herbal Compounds on Neuroglobin: A Literature Review. Curr Med Chem 2024; 31:2944-2954. [PMID: 37069714 DOI: 10.2174/0929867330666230413093409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 04/19/2023]
Abstract
Neuroglobin (Ngb) is an oxygen-binding globin protein that is mainly expressed in the neurons of the central and peripheral nervous system. However, moderate levels of Ngb have also been detected in non-neural tissues. Ngb and Ngb modulating factors have been increasingly studied over the last decade due to their neuroprotective role in neurological disorders and hypoxia. Studies have shown that a number of chemicals, pharmaceuticals, and herbal compounds can modulate the expression of Ngb at different dose levels, indicating a protective role against neurodegenerative diseases. Iron chelators, hormones, antidiabetic drugs, anticoagulants, antidepressants, plant derivatives and short-chain fatty acids are among these compounds. Therefore, this study aimed to review the literature focused on the possible effects and mechanisms of chemical, pharmaceutical, and herbal compounds on Ngbs.
Collapse
Affiliation(s)
- Sahar Golpour Hamedani
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Makan Pourmasoumi
- Gastrointestinal & Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Yuan B, Huang X, Wen J, Peng M. Dexmedetomidine Pretreatment Confers Myocardial Protection and Reduces Mechanical Ventilation Duration for Patients Undergoing Cardiac Valve Replacement under Cardiopulmonary Bypass. Ann Thorac Cardiovasc Surg 2024; 30:23-00210. [PMID: 38684422 PMCID: PMC11082494 DOI: 10.5761/atcs.oa.23-00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
PURPOSE The study aims to assess the effects of dexmedetomidine (Dex) pretreatment on patients during cardiac valve replacement under cardiopulmonary bypass. METHODS For patients in the Dex group (n = 52), 0.5 μg/kg Dex was given before anesthesia induction, followed by 0.5 μg/kg/h pumping injection before aortic occlusion. For patients in the control group (n = 52), 0.125 ml/kg normal saline was given instead of Dex. RESULTS The patients in the Dex group had longer time to first dose of rescue propofol than the control group (P = 0.003). The Dex group required less total dosage of propofol than the control group (P = 0.0001). The levels of cardiac troponin I (cTnI), creatine kinase isoenzyme MB (CK-MB), malondialdehyde (MDA), and tumor necrosis factor-α (TNF-α) were lower in the Dex group than the control group at T4, 8 h after the operation (T5), and 24 h after the operation (T6) (P <0.01). The Dex group required less time for mechanical ventilation than the control group (P = 0.003). CONCLUSION The study suggests that 0.50 µg/kg Dex pretreatment could reduce propofol use and the duration of mechanical ventilation, and confer myocardial protection without increased adverse events during cardiac valve replacement.
Collapse
Affiliation(s)
- Binglin Yuan
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Xiqiang Huang
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Junlin Wen
- Department of Anesthesiology, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Mengzhe Peng
- Pharmacy Intravenous Admixture Services, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
15
|
Liu L, Luo Z, Mai Y, Lu Y, Sun Z, Chen J, Zeng T, Chen L, Liu Z, Yang H, Xu Q, Lan L, Tang C. Dexmedetomidine relieves inflammatory pain by enhancing GABAergic synaptic activity in pyramidal neurons of the anterior cingulate cortex. Neuropharmacology 2023; 240:109710. [PMID: 37683885 DOI: 10.1016/j.neuropharm.2023.109710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Pyramidal neuron (Pyn) hyperactivity in the anterior cingulate cortex (ACC) is involved in the modulation of pain. Previous studies indicate that the activation of α2 adrenoceptors (α2-ARs) by dexmedetomidine (DEX) is a safe and effective means of alleviating multiple types of pain. Here, we showed that systemically administered DEX can ameliorate the inflammatory pain induced by hindpaw injection of formalin (FA) and further examined the molecular and synaptic mechanisms of this DEX-elicited antinociceptive effect. We found that FA caused an increase in c-Fos expression in contralateral layer 2/3 (L2/3) ACC, and that intra-ACC infusion of DEX could also relieve phase 2 inflammatory pain behavior. DEX elicited an increase in the amplitude and frequency of miniature inhibitory post-synaptic currents (mIPSCs) and evoked IPSC amplitude, as well as a reduction in the hyperexcitability and both paired-pulse and excitation/inhibition ratios in contralateral L2/3 ACC Pyns of FA mice. These electrophysiological effects were associated with the upregulation of GABA A receptor (GABAAR) subunits. The interaction of phosphorylated Akt (p-Akt) with GABAAR subunits increased in the ACC following administration of DEX. These results suggest that DEX treatment reduces hyperactivity and enhances GABAergic inhibitory synaptic transmission in ACC Pyns, which produces analgesic effects by increasing GABAAR levels and activating the Akt signaling pathway.
Collapse
Affiliation(s)
- Ling Liu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhihao Luo
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuanying Mai
- Department of Nursing, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yi Lu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, 510130, China
| | - Zhaoxia Sun
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jianfeng Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Tianyu Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Lei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zihao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hanyu Yang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qin Xu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Lan Lan
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Chunzhi Tang
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
16
|
Bargnes V, Oliver B, Wang E, Greenspan S, Jin Z, Yeung I, Bergese S. Taming Postoperative Delirium with Dexmedetomidine: A Review of the Therapeutic Agent's Neuroprotective Effects following Surgery. Pharmaceuticals (Basel) 2023; 16:1453. [PMID: 37895924 PMCID: PMC10610260 DOI: 10.3390/ph16101453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Postoperative delirium (POD) represents a perioperative neurocognitive disorder that has dreaded ramifications on a patient's recovery from surgery. Dexmedetomidine displays multiple mechanisms of neuroprotection to assist in preventing POD as a part of a comprehensive anesthetic care plan. This review will cover dexmedetomidine's pharmacological overlap with the current etiological theories behind POD along with pre-clinical and clinical studies on POD prevention with dexmedetomidine. While the body of evidence surrounding the use of dexmedetomidine for POD prevention still requires further development, promising evidence exists for the use of dexmedetomidine in select dosing and circumstances to enhance recovery from surgery.
Collapse
Affiliation(s)
- Vincent Bargnes
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Brian Oliver
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Emily Wang
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Seth Greenspan
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Isaac Yeung
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| |
Collapse
|
17
|
Bagheri J, Fallahnezhad S, Alipour N, Babaloo H, Tahmasebi F, Kheradmand H, Sazegar G, Haghir H. Maternal diabetes decreases the expression of α2-adrenergic and M2 muscarinic receptors in the visual cortex of male rat neonates. J Chem Neuroanat 2023; 132:102326. [PMID: 37619956 DOI: 10.1016/j.jchemneu.2023.102326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
AIMS This study investigates the impact of maternal diabetes on the expression of α2-adrenergic and M2 muscarinic receptors in the primary visual cortex of male offspring born to diabetic rats. MAIN METHODS In adult female rats, a single dose of intraperitoneal streptozotocin (STZ) was used to induce diabetes (Diabetic group). Diabetes was controlled with insulin in the Insulin-treated group. Female rats in the control group received normal saline instead of STZ. Male newborns were euthanized at P0, P7, and P14, and the expression of α2-adrenergic and M2 muscarinic receptors in the primary visual cortex was determined using immunohistochemistry (IHC). KEY FINDINGS The study showed that α2-adrenergic and M2 muscarinic receptors were significantly suppressed in all layers of the primary visual cortex of male neonates born to diabetic rats at P0, P7, and P14 compared to the control group. The highest expression was for the Con group at P14 and the lowest one was in the Dia group at P0 for both receptors. The insulin treatment in diabetic mothers modulated the expression of these receptors to normal levels in their newborns. SIGNIFICANCE The results demonstrate maternal diabetes decreases the expression of α2-adrenergic and M2 muscarinic receptors in the primary visual cortex of male offspring born to diabetic rats. Insulin treatment can offset these effects of diabetes.
Collapse
Affiliation(s)
- Javad Bagheri
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Somaye Fallahnezhad
- Nervous System Stem Cell Research Center, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasim Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamideh Babaloo
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Kheradmand
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Sazegar
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Haghir
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Huang Q, Zhou R, Hao X, Zhang W, Chen G, Zhu T. Circulating biomarkers in perioperative management of cancer patients. PRECISION CLINICAL MEDICINE 2023; 6:pbad018. [PMID: 37954451 PMCID: PMC10634636 DOI: 10.1093/pcmedi/pbad018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 11/14/2023] Open
Abstract
Owing to the advances in surgical technology, most solid tumours can be controlled by surgical excision. The priority should be tumour control, while some routine perioperative management might influence cancer progression in an unnoticed way. Moreover, it is increasingly recognized that effective perioperative management should include techniques to improve postoperative outcomes. These influences are elucidated by the different functions of circulating biomarkers in cancer patients. Here, circulating biomarkers with two types of clinical functions were reviewed: (i) circulating biomarkers for cancer progression monitoring, for instance, those related to cancer cell malignancy, tumour microenvironment formation, and early metastasis, and (ii) circulating biomarkers with relevance to postoperative outcomes, including systemic inflammation, immunosuppression, cognitive dysfunction, and pain management. This review aimed to provide new perspectives for the perioperative management of patients with cancer and highlight the potential clinical translation value of circulating biomarkers in improving outcomes.
Collapse
Affiliation(s)
- Qiyuan Huang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruihao Zhou
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuechao Hao
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weiyi Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guo Chen
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Zhu
- Department of Anaesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
19
|
Al-Mahrouqi T, Al Alawi M, Freire RC. Dexmedetomidine in the Treatment of Depression: An Up-to-date Narrative Review. Clin Pract Epidemiol Ment Health 2023; 19:e174501792307240. [PMID: 37916205 PMCID: PMC10507216 DOI: 10.2174/17450179-v19-230823-2023-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 11/03/2023]
Abstract
Depressive disorders (DD) are common, and their prevalence is expected to rise over the next decade. Depressive disorders are linked to significant morbidity and mortality. The clinical conundrum of depressive disorders lies in the heterogeneity of their phenomenology and etiology. Further, the currently available antidepressants have several limitations, including a delayed onset of action, limited efficacy, and an unfavorable side effect profile. In this review, Dexmedetomidine (DEX), a highly selective and potent α2-adrenergic receptor (α2-AR) agonist, is proposed as a potentially novel antidepressant with multiple mechanisms of action targeting various depression pathophysiological processes. These mechanisms include modulation of the noradrenergic system, regulation of neuroinflammation and oxidative stress, influence on the Brain-Derived Neurotrophic Factor (BDNF) levels, and modulation of neurotransmitter systems, such as glutamate. The review begins with an introduction before moving on to a discussion of DEX's pharmacological features. The pathophysiological and phenomenological targets of DD are also explored, along with the review of the existing preclinical and clinical evidence for DEX's putative anti-depressant effects. Finally, the review ends by presenting the pertinent conclusions and future directions.
Collapse
Affiliation(s)
- Tamadhir Al-Mahrouqi
- Department of Behavioural Medicine, Sultan Qaboos University Hospital, Muscat, Oman
- Psychiatry Residency Training Program, Oman Medical Speciality Board, Muscat, Oman
| | - Mohammed Al Alawi
- Department of Behavioural Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| | - Rafael C. Freire
- Department of Psychiatry and Centre for Neuroscience Studies, Queens University, Kingston, Canada
- Laboratory of Panic and Respiration, Institute of Psychiatry, Federal University of Rio de, Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Seong H, Jeong D, Kim EH, Yoon KS, Na D, Yoon SZ, Cho JE. MicroRNA-323-5p Involved in Dexmedetomidine Preconditioning Impart Neuroprotection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1518. [PMID: 37763638 PMCID: PMC10532972 DOI: 10.3390/medicina59091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives: Cerebral ischemia is one of the major preoperative complications. Dexmedetomidine is a well-known sedative-hypnotic agent that has potential organ-protective effects. We examine the miRNAs associated with preconditioning effects of dexmedetomidine in cerebral ischemia. Materials and Methods: Transient infarcts were induced in mice via reperfusion after temporary occlusion of one side of the middle cerebral artery. A subset of these mice was exposed to dexmedetomidine prior to cerebral infarction and miRNA profiling of the whole brain was performed. We administered dexmedetomidine and miRNA-323-5p mimic/inhibitor to oxygen-glucose deprivation/reoxygenation astrocytes. Additionally, we administered miR-323-5p mimic and inhibitor to mice via intracerebroventricular injection 2 h prior to induction of middle cerebral artery occlusion. Results: The infarct volume was significantly lower in the dexmedetomidine-preconditioned mice. Analysis of brain samples revealed an increased expression of five miRNAs and decreased expression of three miRNAs in the dexmedetomidine-pretreated group. The viability of cells significantly increased and expression of miR-323-5p was attenuated in the dexmedetomidine-treated oxygen-glucose deprivation/reoxygenation groups. Transfection with anti-miR-323-5p contributed to increased astrocyte viability. When miRNA-323-5p was injected intraventricularly, infarct volume was significantly reduced when preconditioned with the miR-323-5p inhibitor compared with mimic and negative control. Conclusions: Dexmedetomidine has a protective effect against transient neuronal ischemia-reperfusion injury and eight specific miRNAs were profiled. Also, miRNA-323-5p downregulation has a cell protective effect under ischemic conditions both in vivo and in vitro. Our findings suggest the potential of the miR-323-5p inhibitor as a therapeutic agent against cerebral infarction.
Collapse
Affiliation(s)
- Hyunyoung Seong
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Daun Jeong
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Eung Hwi Kim
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Seob Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Donghyun Na
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Zhoo Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jang Eun Cho
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
21
|
Horan R, Sortica da Costa C, Nambyiah P. The persistent effects of anaesthesia on the brain. BJA Educ 2023; 23:304-311. [PMID: 37465234 PMCID: PMC10350555 DOI: 10.1016/j.bjae.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 04/06/2023] [Indexed: 07/20/2023] Open
Affiliation(s)
- R. Horan
- Great Ormond Street Hospital, London, UK
| | | | | |
Collapse
|
22
|
Li J, Zhang H, Deng B, Wang X, Liang P, Xu S, Jing Z, Xiao Z, Sun L, Gao C, Wang J, Sun X. Dexmedetomidine Improves Anxiety-like Behaviors in Sleep-Deprived Mice by Inhibiting the p38/MSK1/NFκB Pathway and Reducing Inflammation and Oxidative Stress. Brain Sci 2023; 13:1058. [PMID: 37508990 PMCID: PMC10377202 DOI: 10.3390/brainsci13071058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Sleep deprivation (SD) triggers a range of neuroinflammatory responses. Dexmedetomidine can improve sleep deprivation-induced anxiety by reducing neuroinflammatory response but the mechanism is unclear; (2) Methods: The sleep deprivation model was established by using an interference rod device. An open field test and an elevated plus maze test were used to detect the emotional behavior of mice. Mouse cortical tissues were subjected to RNA sequence (RNA-seq) analysis. Western blotting and immunofluorescence were used to detect the expression of p38/p-p38, MSK1/p-MSK1, and NFκBp65/p- NFκBp65. Inflammatory cytokines were detected using enzyme-linked immunosorbent assay (ELISA); (3) Results: SD triggered anxiety-like behaviors in mice and was closely associated with inflammatory responses and the MAPK pathway (as demonstrated by transcriptome analysis). SD led to increased expression levels of p-p38, p-MSK1, and p-NFκB. P38 inhibitor SB203580 was used to confirm the important role of the p38/MSK1/NFκB pathway in SD-induced neuroinflammation. Dexmedetomidine (Dex) effectively improves emotional behavior in sleep-deprived mice by attenuating SD-induced inflammatory responses and oxidative stress in the cerebral cortex, mainly by inhibiting the activation of the p38/MSK1/NFκB pathway; (4) Conclusions: Dex inhibits the activation of the p38/MSK1/NFκB pathway, thus attenuating SD-induced inflammatory responses and oxidative stress in the cerebral cortex of mice.
Collapse
Affiliation(s)
- Jiangjing Li
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Heming Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Bin Deng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710065, China
| | - Xin Wang
- Department of Otolaryngology Head and Neck Surgery, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Peng Liang
- Department of Rehabilitative Physioltherapy, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Shenglong Xu
- Department of Radiation Medical Protection, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, The Fourth Military Medical University, Xi'an 710068, China
| | - Ziwei Jing
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Zhibin Xiao
- Department of Anesthesiology, The 986th Air Force Hospital, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Li Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Changjun Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| | - Jin Wang
- Department of Radiation Medical Protection, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, The Fourth Military Medical University, Xi'an 710068, China
| | - Xude Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an 710038, China
| |
Collapse
|
23
|
Chauhan R, Singh N, Reddy A, Singh A. The role of dexmedetomidine in post cranioplasty refractory status epilepticus. Anaesth Rep 2023; 11:e12246. [PMID: 37736063 PMCID: PMC10511152 DOI: 10.1002/anr3.12246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
Cranioplasty, specifically a repair of the skull defect resulting from a previous decompressive craniectomy, is a relatively simple procedure associated with a minimal rate of complications. Even though seizures are seen in up to 30% of the patients postoperatively, status epilepticus is not commonly described. Cerebral oedema, ischaemia and neuro-inflammation have been reported as putative causes of seizures in this population. Here, we report a case of refractory status epilepticus unresponsive to standard anti-epileptic and anaesthetic agents. The use of dexmedetomidine helped terminate the episode and led to a favourable outcome. Most of the standard anti-epileptic and anaesthetic agents act through potentiation of GABAergic transmission or sodium channel blockade and postsynaptic adrenoceptor activation by dexmedetomidine may help potentiate their effect. Further studies are needed to investigate its anticonvulsant effect on post-traumatic brain injury and elaborate on optimal dosage.
Collapse
Affiliation(s)
- R. Chauhan
- Department of Anaesthesia and Intensive CarePostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - N. Singh
- Department of AnaesthesiologySanjay Gandhi Postgraduate Institute of Medical Sciences and ResearchLucknowIndia
| | - A. Reddy
- Department of Anaesthesia and Intensive CarePostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - A. Singh
- Department of NeurosurgeryPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| |
Collapse
|
24
|
Tsivitis A, Wang A, Murphy J, Khan A, Jin Z, Moore R, Tateosian V, Bergese S. Anesthesia, the developing brain, and dexmedetomidine for neuroprotection. Front Neurol 2023; 14:1150135. [PMID: 37351266 PMCID: PMC10282145 DOI: 10.3389/fneur.2023.1150135] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/18/2023] [Indexed: 06/24/2023] Open
Abstract
Anesthesia-induced neurotoxicity is a set of unfavorable adverse effects on central or peripheral nervous systems associated with administration of anesthesia. Several animal model studies from the early 2000's, from rodents to non-human primates, have shown that general anesthetics cause neuroapoptosis and impairment in neurodevelopment. It has been difficult to translate this evidence to clinical practice. However, some studies suggest lasting behavioral effects in humans due to early anesthesia exposure. Dexmedetomidine is a sedative and analgesic with agonist activities on the alpha-2 (ɑ2) adrenoceptors as well as imidazoline type 2 (I2) receptors, allowing it to affect intracellular signaling and modulate cellular processes. In addition to being easily delivered, distributed, and eliminated from the body, dexmedetomidine stands out for its ability to offer neuroprotection against apoptosis, ischemia, and inflammation while preserving neuroplasticity, as demonstrated through many animal studies. This property puts dexmedetomidine in the unique position as an anesthetic that may circumvent the neurotoxicity potentially associated with anesthesia.
Collapse
Affiliation(s)
- Alexandra Tsivitis
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Ashley Wang
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Jasper Murphy
- Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, NY, United States
| | - Ayesha Khan
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Robert Moore
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Vahe Tateosian
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, New York, NY, United States
| |
Collapse
|
25
|
Avci O, Taskiran AS, Gundogdu O. Dexmedetomidine, an α 2 agonist, increases the morphine analgesic effect and decreases morphine tolerance development by suppressing oxidative stress and TNF/IL-1 signalling pathway in rats. REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2023; 70:327-340. [PMID: 37286034 DOI: 10.1016/j.redare.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/18/2022] [Indexed: 06/09/2023]
Abstract
BACKGROUND The aim of the present study is to examine the possible effect de dexmedetomidine on the development of morphine tolerance in rats including nociception, morphine analgesia, apoptosis, oxidative stress, and tumour necrosis factor (TNF)/ interleukin-1 (IL-1) pathways. MATERIALS AND METHODS In this study, 36 Wistar Albino (225-245 g) rats were used. Animals were divided into 6 groups: saline (S), 20 mcg/kg dexmedetomidine (D), 5 mg/kg morphine (M), M + D, morphine tolerance (MT), and MT + D. The analgesic effect was measured with hot plate and tail-flick analgesia tests. After the analgesia tests, the dorsal root ganglia (DRG) tissues were excised. Oxidative stress parameters [total antioxidant status (TAS), total oxidant status (TOS)], TNF, IL-1 and apoptosis enzymes (Caspase-3, Caspase-9), were measured in DRG tissues. RESULTS Dexmedetomidine showed an antinociceptive effect when given alone (p < 0.05 to p < 0.001). In addition, dexmedetomidine increased the analgesic effect of morphine (p < 0.001), and also decreased the tolerance to morphine at a significant level (p < 0.01 to p < 0.001). Moreover, it decreased oxidative stress (p < 0.001) and TNF/IL-1 levels when given as an additional drug of single-dose morphine and morphine tolerance group (p < 0.001). Furthermore, dexmedetomidine decreased Caspase-3 and Caspase-9 levels after tolerance development (p < 0.001). CONCLUSION Dexmedetomidine has antinociceptive properties, and it increases the analgesic effect of morphine and also prevents tolerance development. These effects probably occur by the modulation of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- O Avci
- Facultad de Medicina, Universidad Sivas Cumhuriyet, Departamento de Anestesiología y Reanimación, Sivas, Turkey
| | - A S Taskiran
- Facultad de Medicina, Universidad Sivas Cumhuriyet, Departamento de Fisiología, Sivas, Turkey
| | - O Gundogdu
- Facultad de Medicina, Universidad Sivas Cumhuriyet, Departamento de Anestesiología y Reanimación, Sivas, Turkey.
| |
Collapse
|
26
|
Li J, Wang K, Liu M, He J, Zhang H, Liu H. Dexmedetomidine alleviates cerebral ischemia-reperfusion injury via inhibiting autophagy through PI3K/Akt/mTOR pathway. J Mol Histol 2023:10.1007/s10735-023-10120-1. [PMID: 37186301 DOI: 10.1007/s10735-023-10120-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 03/20/2023] [Indexed: 05/17/2023]
Abstract
Dexmedetomidine has been shown to protect against cerebral ischemia-reperfusion injury (CIRI). Nevertheless, the precise mechanism is obscure. In order to explore the effect of dexmedetomidine pre-conditioning on autophagy against CIRI in rats, middle cerebral artery occlusion (MCAO) was conducted to establish cerebral ischemia-reperfusion (I/R) model in male SD rats with 2 h ischemia and 24 h reperfusion. Dexmedetomidine was delivered to rats at 10, 50 and 100 µg/kg doses respectively, and LY294002, a PI3K/Akt/mTOR pathway inhibitor, was administered at 10 mg/kg intraperitoneally 30 min before MCAO. Neurological deficit score was assessed and cerebral infarct size was detected by TTC staining. Morris water maze (MWM) was performed to estimate spatial learning and memory ability. Furthermore, to detect activity of PI3K/Akt/mTOR pathway and autophagy, p-Akt, p-mTOR, Beclin-1 and LC3 were measured by western blot. Our findings revealed that 50 and 100 µg/kg of dexmedetomidine pretreatment could improve the neurological deficit score and reduce cerebral infarct size after CIRI, while these effects were markedly suppressed by LY294002. In MWM test, dexmedetomidine was confirmed to shorten escape latency and increase times across platform after CIRI. Nevertheless, LY294002 pretreatment eliminated the improvement of dexmedetomidine on spatial learning and memory ability. Furthermore, dexmedetomidine pretreatment reduced ratios of Beclin-1 and LC3II/LC3I and elevated p-Akt/Akt and p-mTOR/mTOR after CIRI. However, above effects of dexmedetomidine were partly reversed by LY294002. Overall, dexmedetomidine pretreatment exerted neuroprotection against CIRI in rats by attenuating autophagy via the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Jianli Li
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China.
| | - Keyan Wang
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Meinv Liu
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Jinhua He
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Huanhuan Zhang
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Huan Liu
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, 050051, China
| |
Collapse
|
27
|
Katoh K. Effects of Electrical Stimulation of the Cell: Wound Healing, Cell Proliferation, Apoptosis, and Signal Transduction. Med Sci (Basel) 2023; 11:medsci11010011. [PMID: 36810478 PMCID: PMC9944882 DOI: 10.3390/medsci11010011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/10/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
Electrical stimulation of the cell can have a number of different effects depending on the type of cell being stimulated. In general, electrical stimulation can cause the cell to become more active, increase its metabolism, and change its gene expression. For example, if the electrical stimulation is of low intensity and short duration, it may simply cause the cell to depolarize. However, if the electrical stimulation is of high intensity or long duration, it may cause the cell to become hyperpolarized. The electrical stimulation of cells is a process by which an electrical current is applied to cells in order to change their function or behavior. This process can be used to treat various medical conditions and has been shown to be effective in a number of studies. In this perspective, the effects of electrical stimulation on the cell are summarized.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
28
|
Gatica S, Aravena C, Prado Y, Aravena D, Echeverría C, Santibanez JF, Riedel CA, Stehberg J, Simon F. Appraisal of the Neuroprotective Effect of Dexmedetomidine: A Meta-Analysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:163-181. [PMID: 37093427 DOI: 10.1007/978-3-031-26163-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Dexmedetomidine is an adrenergic receptor agonist that has been regarded as neuroprotective in several studies without an objective measure to it. Thus, the aim of this meta-analysis was to analyze and quantify the current evidence for the neuroprotective effects of dexmedetomidine in animals. The search was performed by querying the National Library of Medicine. Studies were included based on their language, significancy of their results, and complete availability of data on animal characteristics and interventions. Risk of bias was assessed using SYRCLE's risk of bias tool and certainty was assessed using the ARRIVE Guidelines 2.0. Synthesis was performed by calculating pooled standardized mean difference and presented in forest plots and tables. The number of eligible records included per outcome is the following: 22 for IL-1β, 13 for IL-6, 19 for apoptosis, 7 for oxidative stress, 7 for Escape Latency, and 4 for Platform Crossings. At the cellular level, dexmedetomidine was found protective against production of IL-1β (standardized mean difference (SMD) = - 4.3 [- 4.8; - 3.7]) and IL-6 (SMD = - 5.6 [- 6.7; - 4.6]), apoptosis (measured through TUNEL, SMD = - 6.0 [- 6.8; - 4.6]), and oxidative stress (measured as MDA production, SMD = - 2.0 [- 2.4; - 1.4]) exclusively in the central nervous system. At the organism level, dexmedetomidine improved behavioral outcomes measuring escape latency (SMD = - 2.4 [- 3.3; - 1.6]) and number of platform crossings (SMD = 9.1 [- 6.8; - 11.5]). No eligible study had high risk of bias and certainty was satisfactory for reproducibility in all cases. This meta-analysis highlights the complexity of adrenergic stimulation and sheds light into the mechanisms potentiated by dexmedetomidine, which could be exploited for improving current neuroprotective formulations.
Collapse
Affiliation(s)
- Sebastian Gatica
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| | - Cristobal Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Yolanda Prado
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cesar Echeverría
- Laboratory of Molecular Biology, Nanomedicine and Genomics, Faculty of Medicine, University of Atacama, Copiapo, Chile
| | - Juan F Santibanez
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
- Integrative Center for Biology and Applied Chemistry (CIBQA), Bernardo O'Higgins University, Santiago, Chile
| | - Claudia A Riedel
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Jimmy Stehberg
- Laboratory of Neurobiology, Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
29
|
Aroosa S, Sattar A, Javeed A, Usman M, Hafeez MA, Ahmad M. Protective Effects of Dexmedetomidine Infusion on Genotoxic Potential of Isoflurane in Patients Undergoing Emergency Surgery. Int J Clin Pract 2023; 2023:7414655. [PMID: 36874382 PMCID: PMC9977554 DOI: 10.1155/2023/7414655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/13/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Isoflurane (ISO) has been extensively uses in general anesthesia and reported to cause deoxyribonucleic acid (DNA) damage in prolonged surgical procedures. Dexmedetomidine (DEX) is an adrenergic agonist and having antioxidant activity that may reduce the genotoxic potential (DNA damage) and oxidative stress induced by ISO in patients undergoing major neurosurgical procedures. Methods and Findings. Twenty-four patients of ASA (American Society of Anesthesiologists) classes I and II were randomly divided into two groups (n = 12). Group A patients received ISO, while group B patients received DEX infusion for maintenance of anesthesia. Venous blood samples were collected at different time intervals and used to evaluate the oxidative stress marker malondialdehyde (MDA) and endogenous antioxidants superoxide dismutases (SOD) and catalases (CAT). A single-cell gel electrophoresis (SCGE)-comet assay was used to investigate the genotoxic potential of ISO. CONCLUSION Increased level of antioxidants and decreased value of MDA and genetic damage index were seen in group B (P < 0.001) in a time-dependent manner. Genetic damage was highest at point T 2 (0.77 vs. 1.37), and continued to decrease till T 3 (0.42 vs. 1.19), with respect to negative controls or baseline values following DEX infusion. Significantly, higher level of MDA was recorded in serum of group A (P < 0.001) as compared to group B (1.60 ± 0.33 vs. 0.03 ± 0.001). Enzymatic activities of CAT and SOD were significantly higher in group B than group A (10.11 ± 2.18 vs. 5.71 ± 0.33), (1.04 ± 0.05 vs. 0.95 ± 0.01), respectively. It may play a contributing role in daily anesthesia practice and improve the toxic effects on patients as well as anesthesia personnel. Trial Registration. Ethical Committee of Post Graduate Medical Institute (PGMI), Lahore General Hospital approved the use of humans in this study vide human subject application number ANS-6466 dated February 04, 2019. Furthermore, as the clinical trials required registration from an appropriate registry approved by World Health Organization (WHO), this trail also retrospectively registered at Thai Clinical Trials Registry (an approved WHO registry for clinical trials registration) under reference ID TCTR20211230001 on December 30, 2021.
Collapse
Affiliation(s)
- Sadaf Aroosa
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Adeel Sattar
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Aqeel Javeed
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Usman
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Mian Abdul Hafeez
- Department of Parasitology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Mehmood Ahmad
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
- Department of Pharmacology, Riphah International University, Lahore, Pakistan
| |
Collapse
|
30
|
Brain Bioenergetics in Chronic Hypertension: Risk Factor for Acute Ischemic Stroke. Biochem Pharmacol 2022; 205:115260. [PMID: 36179931 DOI: 10.1016/j.bcp.2022.115260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022]
Abstract
Chronic hypertension is one of the key modifiable risk factors for acute ischemic stroke, also contributing to determine greater neurological deficits and worse functional outcome when an acute cerebrovascular event would occur. A tight relationship exists between cerebrovascular autoregulation, neuronal activity and brain bioenergetics. In chronic hypertension, progressive adaptations of these processes occur as an attempt to cope with the demanding necessity of brain functions, creating a new steady-state homeostatic condition. However, these adaptive modifications are insufficient to grant an adequate response to possible pathological perturbations of the established fragile hemodynamic and metabolic homeostasis. In this narrative review, we will discuss the main mechanisms by which alterations in brain bioenergetics and mitochondrial function in chronic hypertension could lead to increased risk of acute ischemic stroke, stressing the interconnections between hemodynamic factors (i.e. cerebral autoregulation and neurovascular coupling) and metabolic processes. Both experimental and clinical pieces of evidence will be discussed. Moreover, the potential role of mitochondrial dysfunction in determining, or at least sustaining, the pathogenesis and progression of chronic neurogenic hypertension will be considered. In the perspective of novel therapeutic strategies aiming at improving brain bioenergetics, we propose some determinant factors to consider in future studies focused on the cause-effect relationships between chronic hypertension and brain bioenergetic abnormalities (and vice versa), so to help translational research in this so-far unfilled gap.
Collapse
|
31
|
Chen A, Chen X, Deng J, Wei J, Qian H, Huang Y, Wu S, Gao F, Gong C, Liao Y, Zheng X. Dexmedetomidine alleviates olfactory cognitive dysfunction by promoting neurogenesis in the subventricular zone of hypoxic-ischemic neonatal rats. Front Pharmacol 2022; 13:983920. [PMID: 36059991 PMCID: PMC9437207 DOI: 10.3389/fphar.2022.983920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Hypoxic-ischemic brain damage (HIBD) is the main cause of neurological dysfunction in neonates. Olfactory cognitive function is important for feeding, the ability to detect hazardous situations and social relationships. However, only a few studies have investigated olfactory cognitive dysfunction in neonates with HIBD; furthermore, the specific mechanisms involved are yet to be elucidated. It has been reported that neurogenesis in the subventricular zone (SVZ) is linked to olfactory cognitive function. Recently, dexmedetomidine (DEX) has been shown to provide neuroprotection in neonates following HIBD. In the present study, we investigated whether DEX could improve olfactory cognitive dysfunction in neonatal rats following HIBD and attempted to determine the underlying mechanisms. Methods: We induced HIBD in rats using the Rice–Vannucci model, and DEX (25 μg/kg, i.p.) was administered immediately after the induction of HIBD. Next, we used triphenyl tetrazolium chloride (TTC) staining and the Zea-longa score to assess the success of modelling. The levels of BDNF, TNF-α, IL-1β and IL-6 were determined by western blotting. Immunofluorescence staining was used to detect microglial activation and microglial M1/M2 polarization as well as to evaluate the extent of neurogenesis in the SVZ. To evaluate the olfactory cognitive function, the rats in each group were raised until post-natal days 28–35; then, we performed the buried food test and the olfactory memory test. Results: Analysis showed that HIBD induced significant brain infarction, neurological deficits, and olfactory cognitive dysfunction. Furthermore, we found that DEX treatment significantly improved olfactory cognitive dysfunction in rat pups with HIBD. DEX treatment also increased the number of newly formed neuroblasts (BrdU/DCX) and neurons (BrdU/NeuN) in the SVZ by increasing the expression of BDNF in rat pups with HIBD. Furthermore, analysis showed that the neurogenic effects of DEX were possibly related to the inhibition of inflammation and the promotion of M1 to M2 conversion in the microglia. Conclusion: Based on the present findings, DEX treatment could improve olfactory cognitive dysfunction in neonatal rats with HIBD by promoting neurogenesis in the SVZ and enhancing the expression of BDNF in the microglia. It was possible associated that DEX inhibited neuroinflammation and promoted M1 to M2 conversion in the microglia.
Collapse
Affiliation(s)
- Andi Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiaohui Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianhui Deng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jianjie Wei
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Haitao Qian
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yongxin Huang
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Shuyan Wu
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Fei Gao
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Cansheng Gong
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yanling Liao
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiaochun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Key Laboratory of Critical care Medicine, Fujian Provincial Co-Constructed Laboratory of “Belt and Road”, Fujian Emergency Medical Center, Fuzhou, China
- *Correspondence: Xiaochun Zheng,
| |
Collapse
|
32
|
Dexmedetomidine prevents hemorrhagic brain injury by reducing damage induced by ferroptosis in mice. Neurosci Lett 2022; 788:136842. [PMID: 35995304 DOI: 10.1016/j.neulet.2022.136842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/20/2022] [Accepted: 08/14/2022] [Indexed: 11/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition with significant morbidity and mortality for which few effective treatments are clinically available. After ICH, iron overload within the perihaematomal region can induce lethal reactive oxygen species (ROS) production and lipid peroxidation, which contribute to secondary brain injury. An iron-dependent form of non-apoptotic cell death known as ferroptosis was recently identified. Ferroptosis plays an important role in ICH pathology. It is characterized by an accumulation of iron-induced lipid ROS, which leads to intracellular oxidative stress. Dexmedetomidine (DEX), an α2-adrenergic agonist, is widely used for anesthesia, pain control, and intensive care unit sedation. DEX has numerous beneficial activities, including anti-inflammatory, anti-oxidative, and anti-cell death activities. Here, we established a mouse model of ICH using collagenase VII and evaluated the effect of DEX in preventing ICH-induced brain injury. Our study showed that administering DEX reduced the damage induced by ferroptosis after ICH by regulating iron metabolism, amino acid metabolism and lipid peroxidation processes.
Collapse
|
33
|
Lovell S, Simon B, Boudreau EC, Mankin J, Jeffery N. Randomized clinical trial comparing outcomes after fentanyl or ketamine-dexmedetomidine analgesia in thoracolumbar spinal surgery in dogs. J Vet Intern Med 2022; 36:1742-1751. [PMID: 35962706 PMCID: PMC9511085 DOI: 10.1111/jvim.16514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
Background Opioids are widely used for perioperative pain control in dogs undergoing spinal surgery, but alternatives may be required because data suggest that opioids exacerbate inflammation in the injured spinal cord and veterinary access to opioids may become more restricted in the future. Objectives To compare recovery of ambulation and other functions between spinal cord‐injured dogs receiving peri‐operative fentanyl and those receiving a ketamine‐dexmedetomidine combination. Animals A total of 102 client‐owned dogs undergoing decompressive surgery for thoracolumbar intervertebral disc herniation. Methods Randomized clinical trial. Dogs were randomized 1:1 to fentanyl or a ketamine‐dexmedetomidine combination for intra and postoperative analgesia. Primary outcome was time to recovery of ambulation; secondary outcomes were the postoperative Colorado Acute Pain Scale, the short‐form Glasgow Composite Measure Pain Scale, time to recovery of voluntary urination and time to unassisted eating. Results No difference was found in time to recovery of ambulation between groups (adjusted sub‐hazard ratio, 0.83; 95% confidence interval [CI], 0.55‐1.24; P = .36) or in pain scores (Colorado: χ2 = 14.74; P = .32; Glasgow: χ2 = 6.61; P = .76). Differences in time to recovery of eating and urination were small but favored ketamine‐dexmedetomidine (adjusted odds ratios, 3.31; 95% CI, 1.53‐7.16; P = .002 and 2.43; 95% CI, 1.00‐5.96; P = .05, respectively). Conclusions and Clinical Importance There was no evidence that, at the doses used, fentanyl impaired ambulatory outcome after surgery for thoracolumbar intervertebral disc herniation in dogs. Pain control appeared similar between groups. Secondary outcomes suggested minor benefits associated with ketamine‐dexmedetomidine. The ketamine‐dexmedetomidine combination appears to be a reasonable alternative to peri‐operative opioids.
Collapse
Affiliation(s)
- Stephanie Lovell
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Bradley Simon
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Elizabeth C Boudreau
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Joseph Mankin
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Nicholas Jeffery
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
34
|
Huang Y, Deng Y, Zhang R, Meng M, Chen D. Comparing the Effect of Dexmedetomidine and Midazolam in Patients with Brain Injury. Brain Sci 2022; 12:brainsci12060752. [PMID: 35741637 PMCID: PMC9221420 DOI: 10.3390/brainsci12060752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Studies have shown that dexmedetomidine improves neurological function. Whether dexmedetomidine reduces mortality or improves quantitative electroencephalography (qEEG) among patients post-craniotomy remains unclear. METHODS This single-center randomized study was conducted prospectively from 1 January 2019 to 31 December 2020. Patients who were transferred to the ICU after craniotomy within 24 h were included. The analgesic was titrated to a Critical care Pain Observation Tool (CPOT) score ≤2, and the sedative was titrated to a Richmond Agitation-Sedation Scale (RASS) score ≤-3 for at least 24 h. The qEEG signals were collected by four electrodes (F3, T3, F4, and T4 according to the international 10/20 EEG electrode practice). The primary outcome was 28-day mortality and qEEG results on day 1 and day 3 after sedation. RESULTS One hundred and fifty-one patients were enrolled in this study, of whom 77 were in the dexmedetomidine group and 74 in the midazolam group. No significant difference was found between the two groups in mortality at 28 days (14.3% vs. 24.3%; p = 0.117) as well as in the theta/beta ratio (TBR), the delta/alpha ratio (DAR), and the (delta + theta)/(alpha + beta) ratio (DTABR) between the two groups on day 1 or day 3. However, both the TBR and the DTABR were significantly increased in the dexmedetomidine group. The DTABR in the midazolam group was significantly increased. The DAR was significantly increased on the right side in the dexmedetomidine group (20.4 (11.6-43.3) vs. 35.1 (16.7-65.0), p = 0.006) as well as on both sides in the midazolam group (Left: 19.5 (10.1-35.8) vs. 37.3 (19.3-75.7), p = 0.006; Right: 18.9 (10.1-52.3) vs. 39.8 (17.5-99.9), p = 0.002). CONCLUSION Compared with midazolam, dexmedetomidine did not lead to a lower 28-day mortality or better qEEG results in brain injury patients after a craniotomy.
Collapse
|
35
|
Burlacu CC, Neag MA, Mitre AO, Sirbu AC, Badulescu AV, Buzoianu AD. The Role of miRNAs in Dexmedetomidine's Neuroprotective Effects against Brain Disorders. Int J Mol Sci 2022; 23:5452. [PMID: 35628263 PMCID: PMC9141783 DOI: 10.3390/ijms23105452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
There are limited neuroprotective strategies for various central nervous system conditions in which fast and sustained management is essential. Neuroprotection-based therapeutics have become an intensively researched topic in the neuroscience field, with multiple novel promising agents, from natural products to mesenchymal stem cells, homing peptides, and nanoparticles-mediated agents, all aiming to significantly provide neuroprotection in experimental and clinical studies. Dexmedetomidine (DEX), an α2 agonist commonly used as an anesthetic adjuvant for sedation and as an opioid-sparing medication, stands out in this context due to its well-established neuroprotective effects. Emerging evidence from preclinical and clinical studies suggested that DEX could be used to protect against cerebral ischemia, traumatic brain injury (TBI), spinal cord injury, neurodegenerative diseases, and postoperative cognitive disorders. MicroRNAs (miRNAs) regulate gene expression at a post-transcriptional level, inhibiting the translation of mRNA into functional proteins. In vivo and in vitro studies deciphered brain-related miRNAs and dysregulated miRNA profiles after several brain disorders, including TBI, ischemic stroke, Alzheimer's disease, and multiple sclerosis, providing emerging new perspectives in neuroprotective therapy by modulating these miRNAs. Experimental studies revealed that some of the neuroprotective effects of DEX are mediated by various miRNAs, counteracting multiple mechanisms in several disease models, such as lipopolysaccharides induced neuroinflammation, β-amyloid induced dysfunction, brain ischemic-reperfusion injury, and anesthesia-induced neurotoxicity models. This review aims to outline the neuroprotective mechanisms of DEX in brain disorders by modulating miRNAs. We address the neuroprotective effects of DEX by targeting miRNAs in modulating ischemic brain injury, ameliorating the neurotoxicity of anesthetics, reducing postoperative cognitive dysfunction, and improving the effects of neurodegenerative diseases.
Collapse
Affiliation(s)
- Codrin-Constantin Burlacu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandru-Constantin Sirbu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Andrei-Vlad Badulescu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
36
|
Vu T, Smith JA. An Update on Postoperative Cognitive Dysfunction Following Cardiac Surgery. Front Psychiatry 2022; 13:884907. [PMID: 35782418 PMCID: PMC9240195 DOI: 10.3389/fpsyt.2022.884907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022] Open
Abstract
Postoperative cognitive dysfunction is extremely prevalent following cardiac surgery. The increasing patient age and comorbidity profile increases their susceptibility to cognitive impairment. The underlying pathophysiological mechanisms leading to cognitive impairment are not clearly elucidated. Using the contemporary literature (2015-present), this narrative review has three aims. Firstly, to provide an overview of postoperative cognitive impairment. Secondly, to analyse the predominant pathophysiological mechanisms leading to cognitive dysfunction following cardiac surgery such as inflammation, cerebral hypoperfusion, cerebral microemboli, glycaemic control and anaesthesia induced neurotoxicity. Lastly, to assess the current therapeutic strategies of interest to address these pathophysiological mechanisms, including the administration of dexamethasone, the prevention of prolonged cerebral desaturations and the monitoring of cerebral perfusion using near-infrared spectroscopy, surgical management strategies to reduce the neurological effects of microemboli, intraoperative glycaemic control strategies, the effect of volatile vs. intravenous anaesthesia, and the efficacy of dexmedetomidine.
Collapse
Affiliation(s)
- Tony Vu
- Department of Cardiothoracic Surgery, Monash Health, Melbourne, VIC, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Julian A Smith
- Department of Cardiothoracic Surgery, Monash Health, Melbourne, VIC, Australia.,Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Chen Y, Fan Z, Wu Q. Dexmedetomidine improves oxygen-glucose deprivation/reoxygenation (OGD/R) -induced neurological injury through regulating SNHG11/miR-324-3p/VEGFA axis. Bioengineered 2021; 12:4794-4804. [PMID: 34334080 PMCID: PMC8806500 DOI: 10.1080/21655979.2021.1957071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dexmedetomidine (Dex) has been reported to exhibit neuroprotective effects through various regulatory mechanisms. This study aims to investigate the role and molecular mechanism of SNHG11 in Dex-mediated neuroprotection. The ischemic stroke (IS) model was established in vivo by middle cerebral artery occlusion (MCAO) and in vitro by oxygen-glucose deprivation and reperfusion (OGD/R)-treated SH-SY5Y. SNHG11 was highly expressed after OGD/R, and Dex improved OGD/R-induced neurological injury. Additionally, Dex reversed the effects of SNHG11 on OGD/R-induced neurological injury. Furthermore, we found that SNHG11 upregulated vascular endothelial growth factor A (VEGFA) expression by targeting miR-324-3p. Through rescue assays, it was confirmed that SNHG11 regulated OGD/R-induced neurological injury through increasing VEGFA expression. At last, Dex was also discovered to improve neurological injury through regulating SNHG11 in the rat model. In conclusion, our work demonstrated that Dex improved OGD/R-induced neurological injury via SNHG11/miR-324-3p/VEGFA axis. These findings may offer a novel therapeutic strategy for IS treatment.
Collapse
Affiliation(s)
- Yujie Chen
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhiying Fan
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qingwei Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|