1
|
Warner BK, Munhall CC, Nguyen SA, Schlosser RJ, Guldan GJ, Meyer TA. Dexmedetomidine and surgical field visibility in nasal surgery: A systematic review and meta-analysis. J Perioper Pract 2025; 35:112-126. [PMID: 38831613 DOI: 10.1177/17504589241252107] [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] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Nasal and sinus surgery, especially using endoscopy, relies upon adequate haemostasis to be safe and effective. Often other haemostatic methods, such as cautery are not viable, and other methods must be employed. This study examines the effectiveness of dexmedetomidine in controlled hypotension and for surgical field visibility in endoscopic sinus surgery and other nasal surgeries. REVIEW METHODS A literature search was conducted in PubMed, Scopus, CINAHL and Central for randomised controlled trials using dexmedetomidine for controlled hypotension in adult patients undergoing endoscopic sinus surgery or other nasal surgery. Meta-analysis of mean differences and single means were performed. RESULTS Of 935 identified studies, 31 met the inclusion criteria. A statistically significant difference in Fromme-Boezaart surgical field visibility scores was found comparing dexmedetomidine to placebo (p < 0.00001) and propofol (p < 0.0001), but not other agents. A significant difference in intraoperative blood loss volume was found compared with placebo (51.5mL, p < 0.00001) and propofol (13.6mL, p < 0.0001), but not other agents. CONCLUSION Dexmedetomidine demonstrated significantly improved surgical field visibility and blood loss volume compared with placebo and propofol, but not other agents. Dexmedetomidine is viable and useful for controlled hypotension in nasal surgery. Choice of controlled hypotension agent should follow patient and procedure-specific considerations.
Collapse
Affiliation(s)
- Brendon K Warner
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - C Cooper Munhall
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Shaun A Nguyen
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Rodney J Schlosser
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - George J Guldan
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ted A Meyer
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
2
|
Tiainen SM, Heinonen H, Koskinen A, Mäkelä S, Laitio R, Löyttyniemi E, Mäkelä K, Saari TI, Uusalo P. Premedication with intranasal dexmedetomidine in patients undergoing total knee arthroplasty under spinal anaesthesia (TKADEX)-a prospective, double-blinded, randomised controlled trial. BJA OPEN 2025; 13:100382. [PMID: 40114985 PMCID: PMC11923756 DOI: 10.1016/j.bjao.2025.100382] [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: 09/24/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025]
Abstract
Background Previous studies have shown that perioperative use of adjuvants, such as the alpha-2 agonist dexmedetomidine, may reduce postoperative pain and opioid requirements. However, information about optimal dosing is lacking. We investigated if premedication with intranasal dexmedetomidine compared with placebo reduces postoperative pain in patients undergoing total knee arthroplasty under spinal anaesthesia. Methods This single-centre, double-blind, two-arm study compared premedication with intranasal dexmedetomidine (single 1 μg kg-1 dose) to intranasal saline in 101 consecutive elective patients undergoing total knee arthroplasty under spinal anaesthesia. The primary outcome was postoperative pain measured with the numerical rating scale during the first 24 h. Secondary outcomes were postoperative opioid requirement, perioperative haemodynamic variables, requirement of additional intraoperative sedation, incidence of postoperative nausea and vomiting, and patient satisfaction at 30 days after surgery. Results Patients in the dexmedetomidine group had lower numerical rating scale scores [median (interquartile range) 2.0 (0.0-3.0)] at 3 h when compared with the control group [3.0 (2.0-4.0)] (P=0.037). Cumulative 24 h opioid requirements (in morphine equivalents) did not differ between dexmedetomidine [45 mg (30-68 mg)] and control groups [53 mg (38-88 mg)] (P=0.334). More patients in the dexmedetomidine group were satisfied with pain management in the ward (P=0.0013). The groups did not differ in the incidence of postoperative nausea and vomiting (P=0.310) or haemodynamic adverse events (P>0.27 for all). Conclusions Our results indicate that intranasal dexmedetomidine may reduce postoperative pain and the requirement for additional sedation and increase short-term patient satisfaction in patients undergoing total knee arthroplasty. Clinical trial registration ClinicalTrials.gov (NCT04859283).
Collapse
Affiliation(s)
- Suvi-Maria Tiainen
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anaesthesiology and Intensive Care, Satasairaala Central Hospital, Satakunta Hospital District, Pori, Finland
| | - Heta Heinonen
- Medical Faculty, University of Turku, Turku, Finland
| | - Atte Koskinen
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
| | - Sanna Mäkelä
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
| | - Ruut Laitio
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Keijo Mäkelä
- Department of Orthopaedics and Traumatology, Turku University Hospital, and University of Turku, Turku, Finland
| | - Teijo I Saari
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
| | - Panu Uusalo
- Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
3
|
Kilcoyne I, Nieto J, Nottle BF, Flynn H, Knych HK. Morphine synovial fluid concentrations after intravenous regional limb perfusion in horses during standing sedation. Equine Vet J 2025; 57:502-512. [PMID: 38887833 DOI: 10.1111/evj.14114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Addition of morphine to the perfusate while performing intravenous regional limb perfusion (IVRLP) may be helpful in treating painful infectious orthopaedic conditions of the distal limb. OBJECTIVES The main objective of this study was to determine synovial morphine concentrations following IVRLP with morphine alone or in combination with amikacin. STUDY DESIGN Randomised cross-over in vivo experiment. METHODS Six horses underwent IVRLP with 0.1 mg/kg morphine sulphate diluted to 60 mL using 0.9% NaCl (M group) or combined with 2 g amikacin and 0.9% NaCl (MA group) with a 2-week washout period between treatments. Synovial fluid was collected from the radiocarpal joint (RCJ) at 10, 20, 30, 120, 240, 480, 720 and 1440 min after IVRLP. The tourniquet was removed after the 30-min sample was collected. Synovial concentrations of morphine and major metabolites were measured using liquid chromatography-tandem mass spectrometry. Amikacin concentrations were quantified by a fluorescence polarisation immunoassay. RESULTS Measurable concentrations of morphine were apparent in the RCJ of all horses. Median CMAX of morphine in the M group was 4753.1 (2115.7-14 934.5) ng/mL and 4477 (3434.3-7363) ng/mL in the MA group (p = 0.5). Median CMAX of synovial amikacin was 322.6 (157.5-1371.6 μg/mL). MAIN LIMITATIONS Limitations include small sample size. Investigators were not blinded to the treatments and a third treatment group where amikacin alone was administered via IVRLP to the study population was not included. CONCLUSIONS IVRLP using morphine is a feasible technique and synovial morphine concentrations were measurable following IVRLP and were not affected when used concurrently with amikacin. Administration of morphine via IVRLP may be beneficial as an analgesic technique for orthopaedic conditions of the distal limb while limiting potential serious systemic side-effects.
Collapse
Affiliation(s)
- Isabelle Kilcoyne
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Jorge Nieto
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Bridget F Nottle
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Harriet Flynn
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Heather K Knych
- K.L. Maddy Equine Analytical Chemistry Laboratory, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
4
|
Peterson CD, Larson CM, Bruce DJ, Clements BM, Pflepsen KR, Akgün E, Kitto KF, Lunzer MM, Fairbanks CA, Portoghese PS, Wilcox GL. Peripherally mediated opioid combination therapy in mouse and pig. THE JOURNAL OF PAIN 2025; 27:104735. [PMID: 39542192 PMCID: PMC11807752 DOI: 10.1016/j.jpain.2024.104735] [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: 05/29/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
The concomitant epidemics of chronic pain and opioid misuse in the United States have led to a call for novel analgesics with limited abuse potential. Previously, we have shown that co-delivery of a novel combination targeting both μ- and δ-opioid receptors in the peripheral and central nervous systems can produce synergistic analgesia. Loperamide, a peripherally restricted μ-opioid agonist, and oxymorphindole, a δ-opioid receptor partial agonist, synergize in multiple mouse models of hyperalgesia. We predicted this effect would generalize across species and therefore assessed this combination for analgesic synergy in a mouse model of post-incisional hypersensitivity. In mice, oxymorphindole and loperamide displayed significant analgesic synergy. Similar synergy was observed with N-benzyl-oxymorphindole and loperamide. In cross-bred pigs, we compared the analgesic effects of either morphine alone or the combination of oxymorphindole and loperamide or the combination of N-benzyl-oxymorphindole and loperamide. Both combinations showed increased potency as compared to morphine sulfate and effectively reduced hypersensitivity following injury without side effects. From these data we conclude that the combination of oxymorphindole and loperamide or the combination of N-benzyl-oxymorphindole and loperamide reverse incisional hyperalgesia, likely by acting in the periphery, in a large animal model without adverse effects on respiration or heart rate. PERSPECTIVE: This article presents novel opioid combinations, the μ-opioid agonist loperamide with a δ-opioid agonist, either oxymorphindole (OMI) or N-benzyl-oxymorphindole (BOMI), that relieve pain in mice and pigs without adverse side effects. These therapies could help clinicians manage pain in patients while reducing overall opioid burden and limiting side effects.
Collapse
Affiliation(s)
- C D Peterson
- Department of Pharmaceutics, University of Minnesota College of Pharmacy, USA; Department of Neuroscience, University of Minnesota Medical School, USA; Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, USA.
| | - C M Larson
- Comparative and Molecular Biosciences, University of Minnesota College of Veterinary Medicine, USA
| | - D J Bruce
- Department of Neuroscience, University of Minnesota Medical School, USA; Department of Pharmacology, University of Minnesota Medical School, USA
| | - B M Clements
- Department of Pharmaceutics, University of Minnesota College of Pharmacy, USA
| | - K R Pflepsen
- Department of Pharmaceutics, University of Minnesota College of Pharmacy, USA
| | - E Akgün
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, USA
| | - K F Kitto
- Department of Neuroscience, University of Minnesota Medical School, USA
| | - M M Lunzer
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, USA
| | - C A Fairbanks
- Department of Pharmaceutics, University of Minnesota College of Pharmacy, USA; Department of Neuroscience, University of Minnesota Medical School, USA; Department of Pharmacology, University of Minnesota Medical School, USA
| | - P S Portoghese
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, USA
| | - G L Wilcox
- Department of Neuroscience, University of Minnesota Medical School, USA; Department of Pharmacology, University of Minnesota Medical School, USA; Department of Dermatology, University of Minnesota Medical School, USA
| |
Collapse
|
5
|
Poonia A, Tiwari DK, Saharan S, Arora N, Kumar G, Kumar T. Echocardiographic indices probe during dexmedetomidine/midazolam - ketamine anesthesia in dogs undergoing ovariohysterectomy. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:591-597. [PMID: 39807392 PMCID: PMC11725295 DOI: 10.30466/vrf.2024.2019897.4130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/07/2024] [Indexed: 01/16/2025]
Abstract
Cardiovascular and respiratory alterations during anesthesia are of major concern in canines. Thus, it is essential to understand the potential depressant effects of anesthetic drugs on cardio-vascular system; so that, anesthetic procedures are conducted in the best possible way. The objective of the study was to assess and compare the echocardiographic indices during dex-medetomidine and midazolam anesthesia in dogs undergoing elective ovariohysterectomy. Twenty-eight female dogs brought to the department for elective ovariohysterectomy were randomly divided into two groups comprising of 14 each. Sedation was achieved with dex-medetomidine and GMID. Physiological parameters and echocardiographic indices were evaluated before drug administration (T0), after 10 min of sedation (T1), after induction (T2) and at the end of surgery (T3) in both groups. Heart rate was significantly higher at T1, T2 and T3; while, rectal temperature was significantly lower at T3 in GMID compared to the GDEX. There was a significant decrease in stroke volume and cardiac output values at T1 and then, a significant increase at T2; whereas, there was a non-significant decrease at T3 in both groups. Ejection fraction and fractional shortening values decreased significantly at T1, increased significantly at T2 and then, decreased significantly at the end of surgery (T3). Dexmedetomidine-ketamine and midazolam-ketamine combinations provide better hemodynamic and respiratory stability in the dogs undergoing elective ovariohysterectomy. Systolic functions were minimally altered with GMID compared to GDEX. Thus, GMID is more cardio stable compared to GDEX.
Collapse
Affiliation(s)
- Anju Poonia
- Department of Veterinary Surgery and Radiology, College of Veterinary Sciences, Guru Angad Dev University of Veterinary and Animal Sciences, Ludhiana, India;
| | - Deepak Kumar Tiwari
- Department of Veterinary Surgery and Radiology, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India;
| | - Sandeep Saharan
- Department of Veterinary Clinical Complex, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India;
| | - Neeraj Arora
- Department of Veterinary Surgery and Radiology, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India;
| | - Gaurav Kumar
- Department of Veterinary Surgery and Radiology, College of Veterinary Sciences, Guru Angad Dev University of Veterinary and Animal Sciences, Rampura Phul, India.
| | - Tarun Kumar
- Department of Veterinary Clinical Complex, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India;
| |
Collapse
|
6
|
Wagner MA, Smith EML, Ayyash N, Holden JE. Prazosin as an Adjuvant to Increase Effectiveness of Duloxetine in a Rat Model of Oxaliplatin-Induced Peripheral Neuropathy. Semin Oncol Nurs 2024; 40:151686. [PMID: 38897856 DOI: 10.1016/j.soncn.2024.151686] [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: 04/20/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Duloxetine, the only American Society of Clinical Oncology (ASCO) treatment recommended for chemotherapy-induced peripheral neuropathy (CIPN) in cancer survivors, is not effective for 40% of survivors. This study examined the ability of a duloxetine-prazosin combination to prevent the development of allodynia and hyperalgesia in a rat model of oxaliplatin-induced peripheral neuropathy (OPIN). METHODS Female (n = 24) and male (n = 41) rats were started on duloxetine (15 mg), prazosin (2 mg), or a duloxetine-prazosin combination one week prior to administration of the chemotherapy drug, oxaliplatin, and continued the duloxetine-prazosin combination for 32 days. Behavioral testing for mechanical allodynia and mechanical hyperalgesia was done with selected von Frey filaments over the course of the study. RESULTS Overall percent paw withdrawal for rats that received the duloxetine-prazosin combination was significantly lower in female (p < .001 for both conditions) and male (p = .029 for allodynia; p < .001 for hyperalgesia) than those that received water. No significant posttreatment differences were found for allodynia or hyperalgesia between rats treated with duloxetine and rats that received the duloxetine-prazosin combination in either sex. CONCLUSIONS These finding provide preliminary evidence that a duloxetine-prazosin combination can prevent the posttreatment development of allodynia and hyperalgesia in both male and female rats; however, the results suggest that the duloxetine-prazosin combination is no more efficacious than duloxetine alone in preventing chronic OIPN. IMPLICATIONS FOR NURSING PRACTICE The profession of nursing is built on clinical practice supported by scientific research. The current study addressed the clinical practice problem of prevention and management of painful OIPN, which is a priority area in oncology nursing.
Collapse
Affiliation(s)
- Monica A Wagner
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, OH.
| | | | - Naji Ayyash
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, OH
| | | |
Collapse
|
7
|
Sun H, Zhong Y, Wang M, Niu S, Yang R, Tian Y, Li B. Postoperative Dexmedetomidine Infusion and Chronic Postsurgical Pain in Thoracoscopic Pulmonary Nodule Surgery: A Retrospective Study with Propensity-Score-Matched Analysis. Pain Ther 2024; 13:865-881. [PMID: 38805167 PMCID: PMC11254894 DOI: 10.1007/s40122-024-00611-8] [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: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION Patients frequently suffer from debilitating chronic postsurgical pain (CPSP) subsequent to thoracoscopic surgery. The impact of postoperative dexmedetomidine infusion on CPSP remains elusive. This study aimed to scrutinize the effect of dexmedetomidine on both 1-year incidence of CPSP and the quality of recovery after thoracoscopic pulmonary nodule surgery. METHODS This retrospective analysis encompassed clinical and follow-up data from 1148 patients undergoing thoracoscopic pulmonary nodule surgery at our institution between September 2021 and August 2022. Depending on whether dexmedetomidine was infused intravenously or not on the first night after surgery, patients were stratified into the dexmedetomidine group or the control group, with propensity score matching applied to harmonize baseline characteristics. Comparative analysis sought to delineate distinctions of CPSP and recovery quality 1 year after surgery. RESULTS Following propensity score matching, a cohort of 258 patients in each group underwent analysis. Comparisons after matching revealed no statistically significant disparities in 1-year CPSP incidence [76/258 (29.5%) versus 78/258 (30.2%), P = 0.847], moderate-to-severe pain occurrence [17/76 (22.4%) versus 22/78 (28.2%), P = 0.405], neuropathic pain occurrence [11/76 (14.5%) versus 11/78 (14.1%), P = 0.948], and postoperative recovery quality assessed by 12-Item Short Form Health Survey (SF-12) score (113.1 [107.2, 116.0] versus 113.0 [107.4, 116.0], P = 0.328). Multivariate logistic regression analysis encompassing the entire cohort identified being female [odds ratio (OR) 2.10, 95% confidence interval (CI) 1.59-2.79, P < 0.001) and postoperative rescue analgesia (OR 1.47, 95% CI 1.09-1.96, P = 0.010) as risk factors for CPSP, while intraoperative fentanyl dosage (OR 0.92, 95% CI 0.87-0.98, P = 0.006) emerged as a protective factor. CONCLUSION The prolonged administration of dexmedetomidine did not yield discernible amelioration in either 1-year CPSP or the recovery quality after thoracoscopic surgery. Noteworthy risk factors for CPSP encompassed female sex, postoperative rescue analgesia, and diminished fentanyl dosage intraoperatively.
Collapse
Affiliation(s)
- Hang Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210000, China
| | - Yiwei Zhong
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210000, China
| | - Min Wang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, 321 Zhongshan Road, Nanjing, 210000, China
| | - Shujie Niu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210000, China
| | - Rusong Yang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210000, China
| | - Yali Tian
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210000, China.
| | - Bingbing Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210000, China.
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210000, China.
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Xuzhou Medical University, 321 Zhongshan Road, Nanjing, 210000, China.
| |
Collapse
|
8
|
Kästner SB, Amon T, Tünsmeyer J, Noll M, Söbbeler FJ, Laakso S, Saloranta L, Huhtinen M. Anaesthetic-sparing effect of the anxiolytic drug tasipimidine in Beagle dogs. Vet Anaesth Analg 2024; 51:244-252. [PMID: 38555213 DOI: 10.1016/j.vaa.2024.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/03/2023] [Accepted: 02/05/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE To evaluate the effect of oral tasipimidine on dog handling, ease of catheter placement and propofol and isoflurane requirements for anaesthesia. STUDY DESIGN Placebo-controlled, randomized, blinded, experimental trial. ANIMALS A group of seven adult Beagle dogs weighing (mean ± standard deviation) 13.1 ± 2.7 kg with a mean age of 18.6 ± 1 months. METHODS The dogs underwent four treatments before induction of anaesthesia with propofol. PP: placebo orally (PO) 60 minutes before induction of anaesthesia followed by placebo (NaCl 0.9%) intravenously (IV). TP: tasipimidine 30 μg kg-1 (PO) 60 minutes before induction of anaesthesia followed by placebo (NaCl 0.9%) IV. TMP: tasipimidine 30 μg kg-1 PO 60 minutes before induction of anaesthesia followed by methadone 0.2 mg kg-1 IV. TMPD: tasipimidine 30 μg kg-1 PO 60 minutes before induction of anaesthesia followed by methadone 0.2 mg kg-1 and dexmedetomidine 1 μg kg-1 IV followed by a dexmedetomidine constant rate infusion of 1 μg kg-1 hour-1. Sedation, response to catheter placement, intubation quality, time to loss of consciousness, time to intubation, required dose of propofol and minimum alveolar isoflurane concentration preventing motor movement (MACNM) were determined. A mixed-model analysis or the Friedman and Mann-Whitney test were used; p-value < 0.05. RESULTS Response to catheter placement did not differ between treatments. Tasipimidine alone reduced the propofol dose by 30%. Addition of methadone or methadone and dexmedetomidine reduced the propofol dose by 48% and 50%, respectively. Isoflurane MACNM was reduced by 19% in tasipimidine-medicated dogs, whereas in combination with methadone or methadone and dexmedetomidine, isoflurane MACNM was reduced by 35%. CONCLUSIONS AND CLINICAL RELEVANCE An anxiolytic dose of tasipimidine induced mild signs of sedation in dogs and reduced propofol and isoflurane requirements to induce and maintain anaesthesia, which needs to be considered in an anaesthetic plan.
Collapse
Affiliation(s)
- Sabine Br Kästner
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany.
| | - Thomas Amon
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Julia Tünsmeyer
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Mike Noll
- Evidensia Small Animal Clinic, Norderstedt, Germany
| | - Franz-Josef Söbbeler
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | | | | |
Collapse
|
9
|
Acosta-Mares P, Violante-Soria V, Browne T, Cruz SL. Xylazine potentiates the lethal but not the rewarding effects of fentanyl in mice. Drug Alcohol Depend 2023; 253:110993. [PMID: 37883846 DOI: 10.1016/j.drugalcdep.2023.110993] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Fentanyl is commonly laced with xylazine. People who use this combination report heightened effects, but it also increases death risk. Although no medication has been approved to counteract overdoses produced by fentanyl and xylazine, naloxone is frequently used. This paper studies the preclinical rewarding and lethal effects of fentanyl combined with xylazine and the efficacy of yohimbine or naloxone to prevent death. METHODS Male Swiss Webster mice were treated with (in mg/kg, i.p.) xylazine (0.3, 1, 3, or 5.6), fentanyl (0.01, 0.3, or 0.1), or 1 xylazine plus 0.01 (non-effective) or 0.1 (effective) fentanyl doses during the conditioned-place preference (CPP) test. In addition, independent groups received (in mg/kg, i.p.): xylazine (31.6, 60, 74.2, or 100), fentanyl (3.1 or 10), or both substances at two doses: 31.6 xylazine + 3.1 fentanyl, or 60 xylazine + 10 fentanyl to analyze lethal effects. We determined whether yohimbine or naloxone (each medication tested at 10 or 30mg/kg) could prevent the lethality produced by fentanyl/xylazine combinations. Female mice were also tested in key experiments. RESULTS Xylazine neither induced CPP nor altered fentanyl's rewarding effects. In contrast, lethality was potentiated when fentanyl was combined with xylazine. Naloxone, but not yohimbine, effectively prevented the lethality of the fentanyl/xylazine combinations. CONCLUSIONS At the doses tested, xylazine does not increase the rewarding effect of fentanyl on the CPP in male mice but potentiates the risk of fatal overdose in male and female mice. A high naloxone dose prevents death induced by coadministration of fentanyl and xylazine in both sexes.
Collapse
Affiliation(s)
- Palmira Acosta-Mares
- Department of Pharmacobiology, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Valeria Violante-Soria
- Department of Pharmacobiology, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Thom Browne
- Colombo Plan Secretariat, Drug Advisory Program, Colombo, Sri Lanka
| | - Silvia L Cruz
- Department of Pharmacobiology, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| |
Collapse
|
10
|
Nelson TS, Santos DFS, Prasoon P, Gralinski M, Allen HN, Taylor BK. Endogenous μ-opioid-Neuropeptide Y Y1 receptor synergy silences chronic postoperative pain in mice. PNAS NEXUS 2023; 2:pgad261. [PMID: 37649580 PMCID: PMC10465188 DOI: 10.1093/pnasnexus/pgad261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/09/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023]
Abstract
Tissue injury creates a delicate balance between latent pain sensitization (LS) and compensatory endogenous analgesia. Inhibitory G-protein-coupled receptor (GPCR) interactions that oppose LS, including μ-opioid receptor (MOR) or neuropeptide Y Y1 receptor (Y1R) activity, persist in the spinal cord dorsal horn (DH) for months, even after the resolution of normal pain thresholds. Here, we demonstrate that following recovery from surgical incision, a potent endogenous analgesic synergy between MOR and Y1R activity persists within DH interneurons to reduce the intensity and duration of latent postoperative hypersensitivity and ongoing pain. Failure of such endogenous GPCR signaling to maintain LS in remission may underlie the transition from acute to chronic pain states.
Collapse
Affiliation(s)
- Tyler S Nelson
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Diogo F S Santos
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Pranav Prasoon
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Margaret Gralinski
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Heather N Allen
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Bradley K Taylor
- Department of Anesthesiology and Perioperative Medicine, Center for Neuroscience, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Porras-García E, Mas-Nieto M, Delgado-García JM, Domínguez-Del-Toro E. Noradrenergic projections regulate the acquisition of classically conditioned eyelid responses in wild-type and are impaired in kreisler mice. Sci Rep 2023; 13:11458. [PMID: 37454229 PMCID: PMC10349844 DOI: 10.1038/s41598-023-38278-4] [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: 03/14/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
During embryonic development, heterozygous mutant kreisler mice undergo ectopic expression of the Hoxa3 gene in the rostral hindbrain, affecting the opioid and noradrenergic systems. In this model, we have investigated behavioral and cognitive processes in their adulthood. We confirmed that pontine and locus coeruleus neuronal projections are impaired, by using startle and pain tests and by analyzing immunohistochemical localization of tyrosine hydroxylase. Our results showed that, even if kreisler mice are able to generate eyelid reflex responses, there are differences with wild-types in the first component of the response (R1), modulated by the noradrenergic system. The acquisition of conditioned motor responses is impaired in kreisler mice when using the trace but not the delay paradigm, suggesting a functional impairment in the hippocampus, subsequently confirmed by reduced quantification of alpha2a receptor mRNA expression in this area but not in the cerebellum. Moreover, we demonstrate the involvement of adrenergic projection in eyelid classical conditioning, as clonidine prevents the appearance of eyelid conditioned responses in wild-type mice. In addition, hippocampal motor learning ability was restored in kreisler mice by administration of adrenergic antagonist drugs, and a synergistic effect was observed following simultaneous administration of idazoxan and naloxone.
Collapse
Affiliation(s)
- Elena Porras-García
- Division of Neurosciences, University Pablo de Olavide, Ctra. de Utrera, Km. 1, 41013, Sevilla, Spain
| | - Magdalena Mas-Nieto
- Division of Neurosciences, University Pablo de Olavide, Ctra. de Utrera, Km. 1, 41013, Sevilla, Spain
| | - José María Delgado-García
- Division of Neurosciences, University Pablo de Olavide, Ctra. de Utrera, Km. 1, 41013, Sevilla, Spain
| | | |
Collapse
|
12
|
D'Onofrio G, Izzi A, Manuali A, Bisceglia G, Tancredi A, Marchello V, Recchia A, Tonti MP, Icolaro N, Fazzari E, Carotenuto V, De Bonis C, Savarese L, Gorgoglione LP, Del Gaudio A. Anesthetic Management for Awake Craniotomy Applied to Neurosurgery. Brain Sci 2023; 13:1031. [PMID: 37508963 PMCID: PMC10377309 DOI: 10.3390/brainsci13071031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Our anesthetic technique proposed for awake craniotomy is the monitored anesthesia care (MAC) technique, with the patient in sedation throughout the intervention. Our protocol involves analgo-sedation through the administration of dexmedetomidine and remifentanil in a continuous intravenous infusion, allowing the patient to be sedated and in comfort, but contactable and spontaneously breathing. Pre-surgery, the patient is pre-medicated with intramuscular clonidine (2 µg/kg); it acts both as an anxiolytic and as an adjuvant in pain management and improves hemodynamic stability. In the operating setting, dexmedetomidine in infusion and remifentanil in target controlled infusion (TCI) for effect are started. The purpose of the association is to exploit the pharmacodynamics of dexmedetomidine which guarantees the control of respiratory drive, and the pharmacokinetics of remifentanil characterized by insensitivity to the drug. Post-operative management: at the end of the surgical procedure, the infusion of drugs was suspended. Wake-up craniotomy is associated with reduced hospital costs compared to craniotomy performed in general anesthesia, mainly due to reduced costs in the operating room and shorter hospital stays. Greater patient satisfaction and the benefits of avoiding hospital stay have led to the evolution of outpatient intracranial neurosurgery.
Collapse
Affiliation(s)
- Grazia D'Onofrio
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Antonio Izzi
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Aldo Manuali
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Giuliano Bisceglia
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Angelo Tancredi
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Vincenzo Marchello
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Andreaserena Recchia
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Maria Pia Tonti
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Nadia Icolaro
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Elena Fazzari
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Vincenzo Carotenuto
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Costanzo De Bonis
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Luciano Savarese
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Leonardo Pio Gorgoglione
- Complex Unit of Neurosurgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Alfredo Del Gaudio
- Complex Unit of Anaesthesia-2, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| |
Collapse
|
13
|
Beier SL, de Lima MPA, de Sousa FG, Silva RA, Fagundes N, Bovi MF, Tôrres RCS. Comparison of two sedation protocols for diagnostic radiography in dogs with hip dysplasia. Vet Anim Sci 2023; 20:100294. [PMID: 37180767 PMCID: PMC10172832 DOI: 10.1016/j.vas.2023.100294] [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] [Indexed: 05/16/2023] Open
Abstract
Hip dysplasia is an alteration with a high incidence in large dogs. The aim of the study was to compare the association of xylazine or dexmedetomidine with fentanyl for radiography with joint distractor for the diagnosis of hip dysplasia. Fifteen healthy dogs, German Shepherd and Belgian Shepherd, were randomly submitted to treatments 0.2 mg/kg xylazine + 2.5 µg/kg fentanyl (XF) or 2 µg/kg dexmedetomidine + 2.5 µg/kg fentanyl (DF), intravenously. HR, f, SAP, MAP, DAP and TR were evaluated at intervals of 5 min before and after the administration of treatments; pH, PaCO2, PaO2, BE, HCO3-, SaO2, Na+, K+ and Hb at 5 and 15 min after treatment administration; and the quality of sedation at intervals of 5 min after administration of treatments. Latency, duration, and recovery times were also compared. The HR values showed a significant reduction in both groups, as well as pH, PaCO2, PaO2 and SaO2. Latency, duration and recovery times, and quality of sedation did not differ statistically between groups. Xylazine and fentanyl or dexmedetomidine and fentanyl combinations provide adequate sedation and analgesia for performing diagnostic radiographic procedures for hip dysplasia. However, oxygen supplementation is recommended to increase protocol safety.
Collapse
Affiliation(s)
| | | | - Felipe Gaia de Sousa
- Corresponding author at: Department of Veterinary Clinic and Surgery, Veterinary School, Federal University of Minas Gerais 6627 Antônio Carlos Av, Pampulha, Belo Horizonte 31270-901, Minas Gerais, Brazil.
| | | | | | | | | |
Collapse
|
14
|
Mukhopadhyay S, Gupta S, Wilkerson JL, Sharma A, McMahon LR, McCurdy CR. Receptor Selectivity and Therapeutic Potential of Kratom in Substance Use Disorders. CURRENT ADDICTION REPORTS 2023. [DOI: 10.1007/s40429-023-00472-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
15
|
Donaldson AC, Fuller A, Meyer LCR, Buss PE. Chemical immobilisation of lions: weighing up drug effectiveness versus clinical effects. J S Afr Vet Assoc 2023; 94:23-32. [PMID: 37358315 DOI: 10.36303/jsava.544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Selection of an effective drug combination to immobilise African lions (Panthera leo) requires balancing immobilisation effectiveness with potential side effects. We compared the immobilisation effectiveness and changes to physiological variables induced by three drug combinations used for free-ranging African lions. The lions (12 animals per drug combination) were immobilised with tiletamine-zolazepam-medetomidine (TZM), ketamine-medetomidine (KM) or ketamine-butorphanol-medetomidine (KBM). Induction, immobilisation, and recovery were timed, evaluated using a scoring system, and physiological variables were monitored. The drugs used for immobilisation were antagonised with atipamezole and naltrexone. The quality of induction was rated as excellent for all drug combinations and induction times (mean ± SD) did not differ between the groups (10.54 ± 2.67 min for TZM, 10.49 ± 2.63 min for KM, and 11.11 ± 2.91 min for KBM). Immobilisation depth was similar over the immobilisation period in the TZM and KBM groups, and initially light, progressing to deeper in lions administered KM. Heart rate, respiratory rate and peripheral arterial haemoglobin saturation with oxygen were within the expected range for healthy, awake lions in all groups. All lions were severely hypertensive and hyperthermic throughout the immobilisation. Following antagonism of immobilising drugs, lions immobilised with KM and KBM recovered to walking sooner than those immobilised with TZM, at 15.29 ± 10.68 min, 10.88 ± 4.29 min and 29.73 ± 14.46 min, respectively. Only one lion in the KBM group exhibited ataxia during recovery compared to five and four lions in the TZM and KM groups, respectively. All three drug combinations provided smooth inductions and effective immobilisations but resulted in hypertension. KBM had an advantage of allowing for shorter, less ataxic recoveries.
Collapse
Affiliation(s)
- A C Donaldson
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, South Africa and Centre for Veterinary Wildlife Research, Faculty of Veterinary Science, University of Pretoria, South Africa and Center for Zoo and Wild Animal Health, Copenhagen Zoo, Denmark and Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - A Fuller
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, South Africa and Centre for Veterinary Wildlife Research, Faculty of Veterinary Science, University of Pretoria, South Africa and Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - L C R Meyer
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, South Africa and Centre for Veterinary Wildlife Research, Faculty of Veterinary Science, University of Pretoria, South Africa and Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| | - P E Buss
- Centre for Veterinary Wildlife Research, Faculty of Veterinary Science, University of Pretoria, South Africa and Veterinary Wildlife Services, South African National Parks, Kruger National Park, South Africa and Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, South Africa
| |
Collapse
|
16
|
Johnston KJ, Huckins LM. Chronic Pain and Psychiatric Conditions. Complex Psychiatry 2023; 9:24-43. [PMID: 37034825 PMCID: PMC10080192 DOI: 10.1159/000527041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Chronic pain is a common condition with high socioeconomic and public health burden. A wide range of psychiatric conditions are often comorbid with chronic pain and chronic pain conditions, negatively impacting successful treatment of either condition. The psychiatric condition receiving most attention in the past with regard to chronic pain comorbidity has been major depressive disorder, despite the fact that many other psychiatric conditions also demonstrate epidemiological and genetic overlap with chronic pain. Further understanding potential mechanisms involved in psychiatric and chronic pain comorbidity could lead to new treatment strategies both for each type of disorder in isolation and in scenarios of comorbidity. Methods This article provides an overview of relationships between DSM-5 psychiatric diagnoses and chronic pain, with particular focus on PTSD, ADHD, and BPD, disorders which are less commonly studied in conjunction with chronic pain. We also discuss potential mechanisms that may drive comorbidity, and present new findings on the genetic overlap of chronic pain and ADHD, and chronic pain and BPD using linkage disequilibrium score regression analyses. Results Almost all psychiatric conditions listed in the DSM-5 are associated with increased rates of chronic pain. ADHD and BPD are significantly genetically correlated with chronic pain. Psychiatric conditions aside from major depression are often under-researched with respect to their relationship with chronic pain. Conclusion Further understanding relationships between psychiatric conditions other than major depression (such as ADHD, BPD, and PTSD as exemplified here) and chronic pain can positively impact understanding of these disorders, and treatment of both psychiatric conditions and chronic pain.
Collapse
Affiliation(s)
- Keira J.A. Johnston
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
| | - Laura M. Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Farkas DJ, Foss JD, Ward SJ, Rawls SM. Kratom alkaloid mitragynine: Inhibition of chemotherapy-induced peripheral neuropathy in mice is dependent on sex and active adrenergic and opioid receptors. IBRO Neurosci Rep 2022; 13:198-206. [PMID: 36093282 PMCID: PMC9459671 DOI: 10.1016/j.ibneur.2022.08.007] [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: 06/02/2022] [Accepted: 08/26/2022] [Indexed: 11/06/2022] Open
Abstract
Mitragynine (MG) is an alkaloid found in Mitragyna speciosa (kratom) that is used as an herbal remedy for pain relief and opioid withdrawal. MG acts at μ-opioid and α-adrenergic receptors in vitro, but the physiological relevance of this activity in the context of neuropathic pain remains unknown. The purpose of the present study was to characterize the effects of MG in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN), and to investigate the potential impact of sex on MG's therapeutic efficacy. Inhibition of oxaliplatin-induced mechanical hypersensitivity was measured following intraperitoneal administration of MG. Both male and female C57BL/6J mice were used to characterize potential sex-differences in MG's therapeutic efficacy. Pharmacological mechanisms of MG were characterized through pretreatment with the opioid and adrenergic antagonists naltrexone, prazosin, yohimbine, and propranolol (1, 2.5, 5 mg/kg). Oxaliplatin produced significant mechanical allodynia of equal magnitude in both male and females, which was dose-dependently attenuated by repeated MG exposure. MG was more potent in males vs females, and the highest dose of MG (10 mg/kg) exhibited greater anti-allodynic efficacy in males. Mechanistically, activity at µ-opioid, α1- and α2-adrenergic receptors, but not β-adrenergic receptors contributed to the effects of MG against oxaliplatin-induced mechanical hypersensitivity. Repeated MG exposure significantly attenuated oxaliplatin-induced mechanical hypersensitivity with greater potency and efficacy in males, which has crucial implications in the context of individualized pain management. The opioid and adrenergic components of MG indicate that it shares pharmacological properties with clinical neuropathic pain treatments.
Collapse
Affiliation(s)
- Daniel J. Farkas
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Corresponding author.
| | - Jeffery D. Foss
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Sara Jane Ward
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Scott M. Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
18
|
Wu Y, Cai Z, Liu L, Wang J, Li Y, Kang Y, An N. Impact of intravenous dexmedetomidine on gastrointestinal function recovery after laparoscopic hysteromyomectomy: a randomized clinical trial. Sci Rep 2022; 12:14640. [PMID: 36030343 PMCID: PMC9420113 DOI: 10.1038/s41598-022-18729-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/18/2022] [Indexed: 01/14/2023] Open
Abstract
Postoperative intestinal ileus is common after laparoscopic surgery, the incidence of those after hysterectomy was 9.2%. Anesthesia is one of the independent risk factors of postoperative ileus. Dexmedetomidine has been widely used in perioperative anesthesia and previous reports suggested that intraoperative dexmedetomidine may be associated with the improvement of gastrointestinal function recovery after abdominal surgery. We hypothesized that dexmedetomidine could improve gastrointestinal function recovery after laparoscopic hysteromyomectomy. Participants in elective laparoscopic hysteromyomectomy were enrolled with a single dose of 0.5 μg kg−1 dexmedetomidine or the same volume of placebo intravenously administered for 15 min, followed by continuous pumping of 0.2 μg kg−1 h−1 of corresponding drugs until 30 min before the end of surgery. The primary outcome was the time to first flatus. Secondary outcomes were the time to first oral feeding and the first defecation, the occurrence of flatulence, pain score and postoperative nausea and vomiting until 48 h after the surgery. Eventually, 106 participants (54 in dexmedetomidine group and 52 in placebo group) were included for final analysis. The time to first flatus (SD, 25.83 [4.18] vs 27.67 [3.77], P = 0.019), oral feeding time (SD, 27.29 [4.40] vs 28.92 [3.82], P = 0.044), the time to first defecation (SD, 59.82 [10.49] vs 63.89 [7.71], P = 0.025), abdominal distension (n%, 12 (22.2) vs 21 (40.4), P = 0.044), PONV at 24 h (n%, 10 (18.5) vs 19 (36.5), P = 0.037), NRS 6 h (3.15(0.68) vs 3.46 (0.87), P = 0.043) and NRS 12 h (3.43 (0.88) vs 3.85 (0.85), P = 0.014) of dexmedetomidine group were significantly shorter than those of the placebo group. Intraoperative dexmedetomidine reduced the time to first flatus, first oral feeding, and first defecation. These results suggested that this treatment may be a feasible strategy for improving postoperative gastrointestinal function recovery in patients undergoing laparoscopic hysteromyomectomy.
Collapse
Affiliation(s)
- Yu Wu
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, 050082, China.
| | - Zenghua Cai
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, 050082, China
| | - Lishuang Liu
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital, Shijiazhuang, 050082, China
| | - Jinbao Wang
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, 050082, China
| | - Yanli Li
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, 050082, China
| | - Yuling Kang
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, 050082, China
| | - Ni An
- Department of Anesthesiology and Pain, Troop 32295 of the Chinese People's Liberation Army, Liaoyang, China
| |
Collapse
|
19
|
Root-Bernstein R. Biased, Bitopic, Opioid-Adrenergic Tethered Compounds May Improve Specificity, Lower Dosage and Enhance Agonist or Antagonist Function with Reduced Risk of Tolerance and Addiction. Pharmaceuticals (Basel) 2022; 15:214. [PMID: 35215326 PMCID: PMC8876737 DOI: 10.3390/ph15020214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 01/03/2023] Open
Abstract
This paper proposes the design of combination opioid-adrenergic tethered compounds to enhance efficacy and specificity, lower dosage, increase duration of activity, decrease side effects, and reduce risk of developing tolerance and/or addiction. Combinations of adrenergic and opioid drugs are sometimes used to improve analgesia, decrease opioid doses required to achieve analgesia, and to prolong the duration of analgesia. Recent mechanistic research suggests that these enhanced functions result from an allosteric adrenergic binding site on opioid receptors and, conversely, an allosteric opioid binding site on adrenergic receptors. Dual occupancy of the receptors maintains the receptors in their high affinity, most active states; drops the concentration of ligand required for full activity; and prevents downregulation and internalization of the receptors, thus inhibiting tolerance to the drugs. Activation of both opioid and adrenergic receptors also enhances heterodimerization of the receptors, additionally improving each drug's efficacy. Tethering adrenergic drugs to opioids could produce new drug candidates with highly desirable features. Constraints-such as the locations of the opioid binding sites on adrenergic receptors and adrenergic binding sites on opioid receptors, length of tethers that must govern the design of such novel compounds, and types of tethers-are described and examples of possible structures provided.
Collapse
|
20
|
Degrandmaison J, Rochon-Haché S, Parent JL, Gendron L. Knock-In Mouse Models to Investigate the Functions of Opioid Receptors in vivo. Front Cell Neurosci 2022; 16:807549. [PMID: 35173584 PMCID: PMC8841419 DOI: 10.3389/fncel.2022.807549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/28/2022] Open
Abstract
Due to their low expression levels, complex multi-pass transmembrane structure, and the current lack of highly specific antibodies, the assessment of endogenous G protein-coupled receptors (GPCRs) remains challenging. While most of the research regarding their functions was performed in heterologous systems overexpressing the receptor, recent advances in genetic engineering methods have allowed the generation of several unique mouse models. These animals proved to be useful to investigate numerous aspects underlying the physiological functions of GPCRs, including their endogenous expression, distribution, interactome, and trafficking processes. Given their significant pharmacological importance and central roles in the nervous system, opioid peptide receptors (OPr) are often referred to as prototypical receptors for the study of GPCR regulatory mechanisms. Although only a few GPCR knock-in mouse lines have thus far been generated, OPr are strikingly well represented with over 20 different knock-in models, more than half of which were developed within the last 5 years. In this review, we describe the arsenal of OPr (mu-, delta-, and kappa-opioid), as well as the opioid-related nociceptin/orphanin FQ (NOP) receptor knock-in mouse models that have been generated over the past years. We further highlight the invaluable contribution of such models to our understanding of the in vivo mechanisms underlying the regulation of OPr, which could be conceivably transposed to any other GPCR, as well as the limitations, future perspectives, and possibilities enabled by such tools.
Collapse
Affiliation(s)
- Jade Degrandmaison
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Samuel Rochon-Haché
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Jean-Luc Parent
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Jean-Luc Parent,
| | - Louis Gendron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
- *Correspondence: Louis Gendron,
| |
Collapse
|
21
|
Gao X, Zhao T, Xu G, Ren C, Liu G, Du K. The Efficacy and Safety of Ultrasound-Guided, Bi-Level, Erector Spinae Plane Block With Different Doses of Dexmedetomidine for Patients Undergoing Video-Assisted Thoracic Surgery: A Randomized Controlled Trial. Front Med (Lausanne) 2021; 8:577885. [PMID: 34901039 PMCID: PMC8655682 DOI: 10.3389/fmed.2021.577885] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/27/2021] [Indexed: 01/26/2023] Open
Abstract
Background: The anesthetic characteristics of ultrasound-guided bi-level erector spinae plane block (ESPB) plus dexmedetomidine (Dex) remain unclear. We compared the efficacy and safety of ultrasound-guided bi-level ESPB plus different doses of Dex in patients undergoing video-assisted thoracic surgery (VATS). Methods: One-hundred eight patients undergoing VATS were randomized into three groups: R group (n = 38, 15 ml of 0.375% ropivacaine with 0.1 mg/kg dexamethasone), RD1 group (n = 38, 15 ml of 0.375% ropivacaine plus 0.5 μg/kg DEX with 0.1 mg/kg dexamethasone) and RD2 group (n = 38, 15 ml of 0.375% ropivacaine plus 1.0 μg/kg DEX with 0.1 mg/kg dexamethasone). The primary outcome was the pain 12 h after surgery. Secondary outcomes included the Prince Henry Hospital Pain Score; hemodynamics; consumption of sufentanil; anesthetized dermatomal distribution; recovery time; rescue analgesia; satisfaction scores of patients and surgeon; quick recovery index; adverse effects; the prevalence of chronic pain and quality of recovery. Results: The visual analog scale (VAS) and the Prince Henry pain score were significantly lower in both the RD1 and RD2 groups during the first 24 h after surgery (P
< 0.05). Both VAS with coughing and the Prince Henry pain score were significantly lower in the RD2 group than in the RD1 group 8–24 h after surgery (P < 0.05). Both heart rate and mean arterial pressure were significantly different from T2 to T6 in the RD1 and RD2 groups (P < 0.05). The receipt of remifentanil, propofol, Dex, and recovery time was significantly reduced in the RD2 group (P < 0.05). The requirement for sufentanil during the 8–72 h after surgery, less rescue medication, and total press times were significantly lower in the RD2 group (P < 0.05). The time to the first dose of rescue ketorolac was significantly longer in the RD2 group (P < 0.05). Further, anal exhaust, removal of chest tubes, and ambulation were significantly shorter in the RD2 group (P < 0.05). The incidence of tachycardia, post-operative nausea and vomiting, and chronic pain was significantly reduced in the RD2 group, while the QoR-40 score was significantly higher in the RD2 group (P < 0.05). Conclusions: Pre-operative bi-level, single-injection ESPB plus 1 μg/kg DEX provided superior pain relief and long-term post-operative recovery for patients undergoing VATS. Clinical Trial Registration:http://www.chictr.org.cn/searchproj.aspx.
Collapse
Affiliation(s)
- Xiujuan Gao
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Tonghang Zhao
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Guangjun Xu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Chunguang Ren
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Guoying Liu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Ke Du
- Department of Thoracic Surgery, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
22
|
Mondardini MC, Daverio M, Caramelli F, Conti G, Zaggia C, Lazzarini R, Muscheri L, Azzolina D, Gregori D, Sperotto F, Amigoni A. Dexmedetomidine for prevention of opioid/benzodiazepine withdrawal syndrome in pediatric intensive care unit: Interim analysis of a randomized controlled trial. Pharmacotherapy 2021; 42:145-153. [PMID: 34882826 DOI: 10.1002/phar.2654] [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: 09/19/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 11/09/2022]
Abstract
STUDY OBJECTIVE Withdrawal syndrome (WS) may be a critical drawback of opioid/benzodiazepine weaning in children. The most effective intervention to reduce WS prevalence is yet to be determined. Dexmedetomidine (DEX) was estimated to be effective in reducing WS-related symptoms, but no randomized trial has been conducted to prove its efficacy so far. We aimed to evaluate the efficacy and safety of DEX in reducing the occurrence of WS. DESIGN AND SETTING This was an adaptive randomized double-blind placebo-controlled trial conducted at three Italian Pediatric Intensive Care Units (PICUs). PATIENTS It included children admitted to PICU, undergoing at least five days of opioids/benzodiazepines continuous infusion, and ready to start the analgosedation weaning. INTERVENTION Twenty-four hours before the start of weaning, an infusion of DEX/placebo was started. WS symptoms were monitored using the Withdrawal-Assessment-Tool-version-1 (WAT-1). In case of WS symptoms (WAT-1 ≥ 3) an opioid/benzodiazepine bolus was given and the DEX/placebo infusion-rate was increased. MEASUREMENTS The primary outcome measure was the prevalence of WS. Secondary outcomes were the trend of WAT-1 over time, number of rescue doses, length of weaning and PICU-stay, and onset of adverse events (AEs). MAIN RESULTS Forty-five patients were enrolled, of whom 5 dropped-out and 40 entered the interim analysis. There were no significant baseline differences between groups. WS prevalence did not significantly differ between groups (77.8% DEX vs 90.9% placebo, p = 0.381). By generalized linear mixed modeling, the WAT-1 trend showed a significant increase per unit of time in the DEX arm (estimate 0.27, CI 0.07-0.47, p = 0.009) compared to placebo. Most frequent AEs were hemodynamic, and all of them happened in the DEX arm. CONCLUSIONS A continuous infusion of DEX, started 24 h before the analgosedation weaning and increased based on WS signs, was not able to significantly modify the prevalence of WS in children who received at least five days of opioids/benzodiazepines treatment compared to placebo.
Collapse
Affiliation(s)
- Maria Cristina Mondardini
- Pediatric Anesthesia and Intensive Care Unit, Department of Woman's and Child's Health, University Hospital of Bologna IRCCS S. Orsola Polyclinic, Bologna, Italy
| | - Marco Daverio
- Pediatric Intensive Care Unit, Department of Woman's and Child's Health, University-Hospital, Padua, Italy
| | - Fabio Caramelli
- Pediatric Anesthesia and Intensive Care Unit, Department of Woman's and Child's Health, University Hospital of Bologna IRCCS S. Orsola Polyclinic, Bologna, Italy
| | - Giorgio Conti
- Pediatric Intensive Care Unit and Pediatric Trauma Center, Department of Anesthesia and Intensive Care, Catholic University of Rome, A Gemelli Polyclinic, Rome, Italy
| | - Cristina Zaggia
- Pediatric Intensive Care Unit, Department of Woman's and Child's Health, University-Hospital, Padua, Italy
| | - Rossella Lazzarini
- Pediatric Anesthesia and Intensive Care Unit, Department of Woman's and Child's Health, University Hospital of Bologna IRCCS S. Orsola Polyclinic, Bologna, Italy
| | - Lidia Muscheri
- Pediatric Intensive Care Unit and Pediatric Trauma Center, Department of Anesthesia and Intensive Care, Catholic University of Rome, A Gemelli Polyclinic, Rome, Italy
| | - Danila Azzolina
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University-Hospital of Padua, Padua, Italy
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University-Hospital of Padua, Padua, Italy
| | - Francesca Sperotto
- Pediatric Intensive Care Unit, Department of Woman's and Child's Health, University-Hospital, Padua, Italy.,Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Angela Amigoni
- Pediatric Intensive Care Unit, Department of Woman's and Child's Health, University-Hospital, Padua, Italy
| |
Collapse
|
23
|
Comparative study of dezocine, pentazocine and tapentadol on antinociception and physical dependence. Life Sci 2021; 285:119996. [PMID: 34597607 DOI: 10.1016/j.lfs.2021.119996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/20/2022]
Abstract
AIMS Dezocine and pentazocine, widely prescribed in China for postoperative pain, were initially considered as mixed agonist/antagonist targeting μ-opioid receptors (MORs) and κ-opioid receptors (KORs). However, dezocine has been revealed to alleviate chronic neuropathic pain through MOR activation and norepinephrine reuptake inhibition (NRI). This study investigated dezocine- and pentazocine-induced antinociception and physical dependence development, compared to the typical MOR-NRI opioid tapentadol. MAIN METHODS Calcium mobilization assay was conducted to assess the potency of the drugs while hot-plate test was performed to compare the antinociception. Physical dependence development was compared with morphine. KEY FINDINGS Treatment with dezocine, pentazocine and tapentadol stimulated calcium mobilization in HEK293 cells stably expressed MORs but not KORs, whereas dezocine and pentazocine inhibited KOR activities. Subcutaneously injected dezocine-, tapentadol- and pentazocine-induced antinociception dose-dependently, in hot-plate test. Intrathecally injected MOR antagonist CTAP, norepinephrine depletor 6-OHDA and α2-adrenoceptor (α2-AR) antagonist yohimbine partially antagonized dezocine, pentazocine and tapentadol antinociception. Whereas specific KOR antagonist GNTI did not alter their antinociception, the putative inverse KOR agonist nor-BNI reduced dezocine and pentazocine antinociception. Moreover, combined CTAP and 6-OHDA or yohimbine blocked dezocine and tapentadol antinociception but displayed the same partial inhibition on pentazocine antinociception as CTAP alone. Furthermore, compared to morphine and pentazocine, long-term treatment with dezocine and tapentadol produced much less physical dependence-related withdrawal signs, which were restored by spinal 6-OHDA or yohimbine treatment. SIGNIFICANCE Our findings illustrated that dezocine and tapentadol, but not pentazocine, exert remarkable antinociception in nociceptive pain with less abuse liability via dual mechanisms of MOR activation and NRI.
Collapse
|
24
|
Conventional and advanced echocardiographic assessment of systolic function in dogs sedated with dexmedetomidine or acepromazine. Res Vet Sci 2021; 141:129-137. [PMID: 34740044 DOI: 10.1016/j.rvsc.2021.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/25/2021] [Accepted: 09/26/2021] [Indexed: 01/16/2023]
Abstract
Dexmedetomidine and acepromazine, sedatives commonly used in dogs have opposite vascular effects, resulting in afterload increase and decrease, respectively. This could variably affect systolic myocardial function. Previous echocardiographic studies assessing the cardiovascular effects of these drugs used conventional echocardiography, while advanced techniques such as speckle tracking echocardiography (STE) and tissue Doppler imaging (TDI), which are known to provide a more accurate assessment of systolic function, have been rarely used for this aim. Moreover, in the few studies using advanced techniques, the drugs where combined with opioids. Therefore, the main objective of this prospective study was to assess systolic myocardial function by conventional and advanced echocardiography (STE and TDI), in dogs sedated exclusively with dexmedetomidine or acepromazine not combined with other drugs. Twenty healthy dogs were randomly divided into two groups, Group A (acepromazine, 20 μg/kg IM), and Group D (dexmedetomidine, 5 μg/kg IM), cardiovascular parameters were assessed before sedation (T0), and thirty minutes afterwards (T1). Systolic arterial pressure and heart rate decreased in both groups at T1 as compared to T0. Only one conventional echocardiographic raw variable (left ventricular internal dimension in systole) and three out of five advanced echocardiographic variables (radial TDI systolic velocities at the epicardial region of the left ventricular free wall, longitudinal TDI systolic velocities of the septal mitral valve annulus and the STE-derived left ventricular global strain), were affected in Group D. A systolic impairment was observed in Group D and better estimated by advanced echocardiography. In Group A, only the end diastolic voume index (conventional echocardiography) was decreased. Both protocols seem to induce echocardiographic changes more likely secondary to their vascular action.
Collapse
|
25
|
Elvir-Lazo OL, White PF, Cruz Eng H, Yumul F, Chua R, Yumul R. Impact of chronic medications in the perioperative period -anesthetic implications (Part II). Postgrad Med 2021; 133:920-938. [PMID: 34551658 DOI: 10.1080/00325481.2021.1982298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Background: This review article discusses the pharmacodynamic effects of the most commonly used chronic medications by patients undergoing elective surgical procedures, namely cardiovascular drugs (e.g., beta blockers, alpha-2 agonist, calcium channel blockers, ACE inhibitors, diuretics, etc.), lipid-lowering drugs, gastrointestinal medications (H2-blockers, proton pump inhibitors), pulmonary medications (inhaled β-agonists, anticholinergics,), antibiotics (tetracyclines, clindamycin and macrolide, linezolid.), opioids and non-opioids analgesics (NSAIDs, COX-2 inhibitors, acetaminophen), gabapentanoids, erectile dysfunction (ED) drugs, psychotropic drugs (tricyclic antidepressants [TCAs], monoamine oxidase inhibitors [MAOI], selective serotonin reuptake inhibitors [SSRIs], serotonin norepinephrine reuptake inhibitors [SNRIs], and cannabinol-containing drugs). In addition, the potential adverse drug-interactions between these chronic medications and commonly used anesthetic drugs during the perioperative period will be reviewed. Finally, recommendations regarding the management of chronic medications during the preoperative period will be provided.Materials and Methods: An online search was conducted from January 2000 through February 2021 with the Medline database through PubMed and Google Scholar using the following search terms/keywords: "chronic medications in the perioperative period", and "chronic medications and anesthetic implications." In addition, we searched for anesthetic side effects associated with the major drug groups.Results and Conclusions: An understanding of the pharmacodynamic effects of most used chronic medications is important to avoid untoward outcomes in the perioperative period. These drug interactions may result in altered efficacy and toxicity of the anesthetic medications administered during surgery. These drug-drug interactions can also affect the morbidity, mortality, recovery time of surgical patients and acute relapse of chronic illnesses which could lead to last minute cancellation of surgical procedures. Part II of this two-part review article focuses on the reported interactions between most commonly taken chronic medications by surgical patients and anesthetic and analgesic drugs, as well as recommendations regarding the handling these chronic medications during the perioperative period.
Collapse
Affiliation(s)
| | - Paul F White
- Department of Anesthesiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,The White Mountain Institute, The Sea Ranch, CA, USA
| | - Hillenn Cruz Eng
- Adena Health System, department of anesthesiology, Chillicothe, OH, US
| | - Firuz Yumul
- Department of family medicine, Skagit Regional Health, Family Medicine, Arlington, WA, USA
| | - Raissa Chua
- Department of Internal Medicine, Huntington Hospital, Prasadena, CA, USA
| | - Roya Yumul
- Department of Anesthesiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,David Geffen School of Medicine-UCLA, Charles R, Drew University of Medicine and Science, Los Angeles, CA, USA
| |
Collapse
|
26
|
5-HT 1A Serotonergic, α-Adrenergic and Opioidergic Receptors Mediate the Analgesic Efficacy of Vortioxetine in Mice. Molecules 2021; 26:molecules26113242. [PMID: 34071269 PMCID: PMC8199248 DOI: 10.3390/molecules26113242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 01/09/2023] Open
Abstract
Vortioxetine is a multimodal antidepressant drug that affects several brain neurochemicals and has the potential to induce various pharmacological effects on the central nervous system. Therefore, we investigated the centrally mediated analgesic efficacy of this drug and the mechanisms underlying this effect. Analgesic activity of vortioxetine (5, 10 and 20 mg/kg, p.o.) was examined by tail-clip, tail-immersion and hot-plate tests. Motor performance of animals was evaluated using Rota-rod device. Time course measurements (30-180 min) showed that vortioxetine (10 and 20 mg/kg) administrations significantly increased the response latency, percent maximum possible effect and area under the curve values in all of the nociceptive tests. These data pointed out the analgesic effect of vortioxetine on central pathways carrying acute thermal and mechanical nociceptive stimuli. Vortioxetine did not alter the motor coordination of mice indicating that the analgesic activity of this drug was specific. In mechanistic studies, pre-treatments with p-chlorophenylalanine (serotonin-synthesis inhibitor), NAN-190 (serotonin 5-HT1A receptor antagonist), α-methyl-para-tyrosine (catecholamine-synthesis inhibitor), phentolamine (non-selective α-adrenoceptor blocker), and naloxone (non-selective opioid receptor blocker) antagonised the vortioxetine-induced analgesia. Obtained findings indicated that vortioxetine-induced analgesia is mediated by 5-HT1A serotonergic, α-adrenergic and opioidergic receptors, and contributions of central serotonergic and catecholaminergic neurotransmissions are critical for this effect.
Collapse
|
27
|
Bessière B, Iris F, Milet A, Beopoulos A, Billoet C, Farjot G. A new mechanistic approach for the treatment of chronic neuropathic pain with nitrous oxide integrated from a systems biology narrative review. Med Gas Res 2021; 11:34-41. [PMID: 33642336 PMCID: PMC8103977 DOI: 10.4103/2045-9912.310058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022] Open
Abstract
The limitations of the currently available treatments for chronic neuropathic pain highlight the need for safer and more effective alternatives. The authors carried out a focused review using a systems biology approach to integrate the complex mechanisms of nociception and neuropathic pain, and to decipher the effects of nitrous oxide (N2O) on those pathways, beyond the known effect of N2O on N-methyl-D-aspartate receptors. This review identified a number of potential mechanisms by which N2O could impact the processes involved in peripheral and central sensitization. In the ascending pathway, the effects of N2O include activating TWIK-related K+ channel 1 potassium channels on first-order neurons, blocking voltage-dependent calcium channels to attenuate neuronal excitability, attenuating postsynaptic glutamatergic receptor activation, and possibly blocking voltage-dependent sodium channels. In the descending pathway, N2O induces the release of endogenous opioid ligands and stimulates norepinephrine release. In addition, N2O may mediate epigenetic changes by inhibiting methionine synthase, a key enzyme involved in DNA and RNA methylation. This could explain why this short-acting analgesic has shown long-lasting anti-pain sensitization effects in animal models of chronic pain. These new hypotheses support the rationale for investigating N2O, either alone or in combination with other analgesics, for the management of chronic neuropathic pain.
Collapse
Affiliation(s)
- Baptiste Bessière
- Air Liquide Santé International, Paris Innovation Campus, Jouy-en-Josas, France
| | | | - Aude Milet
- Air Liquide Santé International, Paris Innovation Campus, Jouy-en-Josas, France
| | | | - Catherine Billoet
- Air Liquide Santé International, Paris Innovation Campus, Jouy-en-Josas, France
| | - Géraldine Farjot
- Air Liquide Santé International, Paris Innovation Campus, Jouy-en-Josas, France
| |
Collapse
|
28
|
Grubb T, Sager J, Gaynor JS, Montgomery E, Parker JA, Shafford H, Tearney C. 2020 AAHA Anesthesia and Monitoring Guidelines for Dogs and Cats. J Am Anim Hosp Assoc 2020; 56:59-82. [PMID: 32078360 DOI: 10.5326/jaaha-ms-7055] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Risk for complications and even death is inherent to anesthesia. However, the use of guidelines, checklists, and training can decrease the risk of anesthesia-related adverse events. These tools should be used not only during the time the patient is unconscious but also before and after this phase. The framework for safe anesthesia delivered as a continuum of care from home to hospital and back to home is presented in these guidelines. The critical importance of client communication and staff training have been highlighted. The role of perioperative analgesia, anxiolytics, and proper handling of fractious/fearful/aggressive patients as components of anesthetic safety are stressed. Anesthesia equipment selection and care is detailed. The objective of these guidelines is to make the anesthesia period as safe as possible for dogs and cats while providing a practical framework for delivering anesthesia care. To meet this goal, tables, algorithms, figures, and "tip" boxes with critical information are included in the manuscript and an in-depth online resource center is available at aaha.org/anesthesia.
Collapse
Affiliation(s)
- Tamara Grubb
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - Jennifer Sager
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - James S Gaynor
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - Elizabeth Montgomery
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - Judith A Parker
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - Heidi Shafford
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| | - Caitlin Tearney
- From Washington State University College of Veterinary Medicine, Pullman, Washington (T.G.); University of Florida Veterinary Hospitals, Gainesville, Florida (J.S.); Peak Performance Veterinary Group, Breckenridge, Colorado (J.S.G.); University of California, Davis, School of Veterinary Medicine, Davis, California (E.M.); Pima Pet Clinic, Tucson, Arizona (J.A.P.); Veterinary Anesthesia Specialists, Clackamas, Oregon (H.S.); and Wheat Ridge Animal Hospital, Wheat Ridge, Colorado (C.T.)
| |
Collapse
|
29
|
Root-Bernstein R, Churchill B, Turke M. Glutathione and Glutathione-Like Sequences of Opioid and Aminergic Receptors Bind Ascorbic Acid, Adrenergic and Opioid Drugs Mediating Antioxidant Function: Relevance for Anesthesia and Abuse. Int J Mol Sci 2020; 21:E6230. [PMID: 32872204 PMCID: PMC7504417 DOI: 10.3390/ijms21176230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Opioids and their antagonists alter vitamin C metabolism. Morphine binds to glutathione (l-γ-glutamyl-l-cysteinyl-glycine), an intracellular ascorbic acid recycling molecule with a wide range of additional activities. The morphine metabolite morphinone reacts with glutathione to form a covalent adduct that is then excreted in urine. Morphine also binds to adrenergic and histaminergic receptors in their extracellular loop regions, enhancing aminergic agonist activity. The first and second extracellular loops of adrenergic and histaminergic receptors are, like glutathione, characterized by the presence of cysteines and/or methionines, and recycle ascorbic acid with similar efficiency. Conversely, adrenergic drugs bind to extracellular loops of opioid receptors, enhancing their activity. These observations suggest functional interactions among opioids and amines, their receptors, and glutathione. We therefore explored the relative binding affinities of ascorbic acid, dehydroascorbic acid, opioid and adrenergic compounds, as well as various control compounds, to glutathione and glutathione-like peptides derived from the extracellular loop regions of the human beta 2-adrenergic, dopamine D1, histamine H1, and mu opioid receptors, as well as controls. Some cysteine-containing peptides derived from these receptors do bind ascorbic acid and/or dehydroascorbic acid and the same peptides generally bind opioid compounds. Glutathione binds not only morphine but also naloxone, methadone, and methionine enkephalin. Some adrenergic drugs also bind to glutathione and glutathione-like receptor regions. These sets of interactions provide a novel basis for understanding some ways that adrenergic, opioid and antioxidant systems interact during anesthesia and drug abuse and may have utility for understanding drug interactions.
Collapse
Affiliation(s)
- Robert Root-Bernstein
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (B.C.); (M.T.)
| | - Beth Churchill
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (B.C.); (M.T.)
| | - Miah Turke
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (B.C.); (M.T.)
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
30
|
Transcriptional profile of spinal dynorphin-lineage interneurons in the developing mouse. Pain 2020; 160:2380-2397. [PMID: 31166300 DOI: 10.1097/j.pain.0000000000001636] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mounting evidence suggests that the spinal dorsal horn (SDH) contains multiple subpopulations of inhibitory interneurons that play distinct roles in somatosensory processing, as exemplified by the importance of spinal dynorphin-expressing neurons for the suppression of mechanical pain and chemical itch. Although it is clear that GABAergic transmission in the SDH undergoes significant alterations during early postnatal development, little is known about the maturation of discrete inhibitory "microcircuits" within the region. As a result, the goal of this study was to elucidate the gene expression profile of spinal dynorphin (pDyn)-lineage neurons throughout life. We isolated nuclear RNA specifically from pDyn-lineage SDH interneurons at postnatal days 7, 21, and 80 using the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) technique, followed by RNA-seq analysis. Over 650 genes were ≥2-fold enriched in adult pDyn nuclei compared with non-pDyn spinal cord nuclei, including targets with known relevance to pain such as galanin (Gal), prepronociceptin (Pnoc), and nitric oxide synthase 1 (Nos1). In addition, the gene encoding a membrane-bound guanylate cyclase, Gucy2d, was identified as a novel and highly selective marker of the pDyn population within the SDH. Differential gene expression analysis comparing pDyn nuclei across the 3 ages revealed sets of genes that were significantly upregulated (such as Cartpt, encoding cocaine- and amphetamine-regulated transcript peptide) or downregulated (including Npbwr1, encoding the receptor for neuropeptides B/W) during postnatal development. Collectively, these results provide new insight into the potential molecular mechanisms underlying the known age-dependent changes in spinal nociceptive processing and pain sensitivity.
Collapse
|
31
|
Li X, Liu Y, Zhao J, Xiang Z, Ren C, Qiao K. The Safety and Efficacy of Ultrasound-Guided Serratus Anterior Plane Block (SAPB) Combined with Dexmedetomidine for Patients Undergoing Video-Assisted Thoracic Surgery (VATS): A Randomized Controlled Trial. J Pain Res 2020; 13:1785-1795. [PMID: 32801843 PMCID: PMC7381820 DOI: 10.2147/jpr.s258443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/06/2020] [Indexed: 12/31/2022] Open
Abstract
Background Although video-assisted thoracic surgery (VATS) can significantly reduce postoperative pain, the incidence is as high as 30–50%. The purpose of this study was to explore the safety and efficacy of ultrasound-guided serratus anterior plane block (SAPB) combined with dexmedetomidine (Dex) for patients undergoing VATS. Methods Eighty patients were randomized into two groups (20 mL 0.5% ropivacaine plus 0.5 µg/kg or 1 µg/kg Dex). Primary outcome was the visual analog scale of pain while coughing (VASc) score at 24 h after surgery. Secondary outcomes included hemodynamics, sufentanil consumption, number of patients needing rescue analgesia, time to first rescue analgesic, total dose of rescue analgesic, satisfaction scores of patients and surgeons, time of chest tube removal, length of hospital stay, adverse effects, the prevalence of chronic pain and quality of life. Results Compared with D1 group, visual analog scale of pain at rest (VASr) was significantly lower during the first 24 h after surgery, while VASc was significantly lower during the first 48 h after surgery (P<0.05). Mean arterial pressure was significantly decreased from T2 to T8, and heart rate was significantly decreased from T2 to T7 in the D2 group (P<0.05). Consumption of sevoflurane, remifentanil, DEX and the recovery time were significantly reduced in the D2 group (P <0.05). Consumption of sufentanil 8–72 h after surgery was significantly lower in the D2 group (P<0.05). Additionally, the number of patients who required rescue analgesia, the time to the first dose of rescue analgesia, and the total dose of rescue analgesia was significantly lower in the D2 group (P<0.05). Conclusion The results of this study show that 1 µg/kg DEX is a beneficial adjuvant to ropivacaine for single-injection SAPB in VATS patients while stable hemodynamics were maintained.
Collapse
Affiliation(s)
- Xiang Li
- Department of Anesthesiology, The First People's Hospital of Tianmen, Tianmen, Hubei, People's Republic of China
| | - Yanchao Liu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Jing Zhao
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Zhixiong Xiang
- Department of Anesthesiology, The First People's Hospital of Tianmen, Tianmen, Hubei, People's Republic of China
| | - Chunguang Ren
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Kekun Qiao
- Department of Anesthesiology, The First People's Hospital of Tianmen, Tianmen, Hubei, People's Republic of China
| |
Collapse
|
32
|
Akashi N, Murahata Y, Kishida H, Hikasa Y, Azuma K, Imagawa T. Effects of constant rate infusions of dexmedetomidine, remifentanil and their combination on minimum alveolar concentration of sevoflurane in dogs. Vet Anaesth Analg 2020; 47:490-498. [PMID: 32471632 DOI: 10.1016/j.vaa.2020.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the effects of constant rate infusions (CRIs) of dexmedetomidine and remifentanil alone and their combination on minimum alveolar concentration (MAC) of sevoflurane in dogs. STUDY DESIGN Randomized crossover experimental study. ANIMALS A total of six (three males, three females) healthy, adult neutered Beagle dogs weighing 12.6 ± 1.4 kg. METHODS Anesthesia was induced with sevoflurane in oxygen until endotracheal intubation was possible and anesthesia maintained with sevoflurane using positive-pressure ventilation. Each dog was anesthetized five times and was administered each of the following treatments: saline (1 mL kg-1 hour-1) or dexmedetomidine at 0.1, 0.5, 1.0 or 5.0 μg kg-1 loading dose intravenously over 10 minutes followed by CRI at 0.1, 0.5, 1.0 or 5.0 μg kg-1 hour-1, respectively. Following 60 minutes of CRI, sevoflurane MAC was determined in duplicate using an electrical stimulus (50 V, 50 Hz, 10 ms). Then, CRI of successively increasing doses of remifentanil (0.15, 0.60 and 2.40 μg kg-1 minute-1) was added to each treatment. MAC was also determined after 30 minutes equilibration at each remifentanil dose. Isobolographic analysis determined interaction from the predicted doses required for a 50% MAC reduction (ED50) with remifentanil, dexmedetomidine and remifentanil combined with dexmedetomidine, with the exception of dexmedetomidine 5.0 μg kg-1 hour-1, obtained using log-linear regression analysis. RESULTS The sevoflurane MAC decreased dose-dependently with increasing infusion rates of dexmedetomidine and remifentanil. Remifentanil ED50 values were lower when combined with dexmedetomidine than those obtained during saline-remifentanil. Synergistic interactions between dexmedetomidine and remifentanil for MAC reduction occurred with dexmedetomidine at 0.5 and 1.0 μg kg-1 hour-1. CONCLUSIONS AND CLINICAL RELEVANCE Combined CRIs of dexmedetomidine and remifentanil synergistically resulted in sevoflurane MAC reduction. The combination of dexmedetomidine and remifentanil effectively reduced the requirement of sevoflurane during anesthesia in dogs.
Collapse
Affiliation(s)
- Natsuki Akashi
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yusuke Murahata
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan.
| | - Hikaru Kishida
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yoshiaki Hikasa
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Kazuo Azuma
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Tomohiro Imagawa
- Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
33
|
Ahsan MZ, Khan FU, Zhao MJ, Wang YX. Synergistic interaction between butorphanol and dexmedetomidine in antinociception. Eur J Pharm Sci 2020; 149:105322. [PMID: 32289662 DOI: 10.1016/j.ejps.2020.105322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/21/2020] [Accepted: 03/22/2020] [Indexed: 01/14/2023]
Abstract
Opioid analgesics and the α2-adrenergic receptor (α2AR) agonists are found to produce synergistic antinociception when administered in combination. In this study interactions between butorphanol and dexmedetomidine were investigated in the thermal pain and autonomous locomotor activity. Butorphanol and dexmedetomidine were administered subcutaneously alone and in combination in a fixed-dose ratio (3:1) to assess the antinociceptive and sedative responses. Butorphanol produced antinociception in the hot-plate test via three major opioid receptor subtypes, i.e. MORs, KORs and DORs, while in the tail-immersion test the antinociception was produced by MORs and KORs, whereas dexmedetomidine exhibited antinociception by α2ARs in both tests. They exhibited dose- and time-dependent antinociception and inhibition of locomotor activity when administered alone, while their combination displayed enhanced therapeutic effects. Isobolographic analysis revealed that combined butorphanol and dexmedetomidine produced synergistic interactions in the hot-plate, tail-immersion and locomotor activity tests. Furthermore, the analgesic synergy was also approved to be modulated by MORs, KORs, DORs and α2ARs. Hence we concluded from this study that combined butorphanol and dexmedetomidine produced synergistic antinociception that may be helpful in facilitating clinical management of acute nociceptive pain.
Collapse
Affiliation(s)
- Muhammad Zaeem Ahsan
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240 China.
| | - Farhan Ullah Khan
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240 China.
| | - Meng-Jing Zhao
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240 China.
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240 China.
| |
Collapse
|
34
|
Di Bella C, Skouropoulou D, Stabile M, Muresan C, Grasso S, Lacitignola L, Valentini L, Crovace A, Staffieri F. Respiratory and hemodynamic effects of 2 protocols of low-dose infusion of dexmedetomidine in dogs under isoflurane anesthesia. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2020; 84:96-107. [PMID: 32255904 PMCID: PMC7088514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/02/2019] [Indexed: 06/11/2023]
Abstract
The aim of this study was to evaluate the respiratory and hemodynamic effects of a low-dose dexmedetomidine infusion [1 μg/kg body weight (BW) per hour], with or without a loading dose (1 μg/kg BW), in dogs under isoflurane anesthesia. Thirty dogs were premedicated with methadone [0.3 mg/kg BW intramuscular (IM)], induced with propofol intravenous (IV) and maintained with isoflurane (1.3% to 1.4%) under mechanical ventilation. Animals were randomly assigned to 3 intravenous (IV) treatments (n = 10): 1 μg/kg BW dexmedetomidine, followed by 1 μg/kg BW per hour (group BI); or saline solution bolus, followed by either an infusion of 1 μg/kg BW per hour dexmedetomidine (group I) or saline solution (group C). The infusions were interrupted after 30 minutes. Respiratory system static compliance (Cstat) and respiratory system resistance (Rrs), partial pressure of oxygen/fractional inspired oxygen ratio (PaO2/FIO2), intrapulmonary shunt (Fshunt), and cardiac output (CO) were determined 5 minutes before the bolus (BASELINE), at the end of the bolus (BOLUS), and at 15 (T15), 30 (T30), and 45 minutes (T45) intervals. In group BI, Cstat and PaO2/FiO2 were higher at T15 and T30 than at BASELINE in the same group and than group C at the same times. In group I, the same parameters at T30 were higher than at BASELINE and than group C at the same time. In group BI, Rrs and Fshunt were lower than at BASELINE and than group C at the same time. In group I, the same parameters at T30 were lower than at BASELINE and those of group C at the same time. Cardiac output (CO) at T30 was higher in groups BI and I than in group C. The results of this study showed that low-dose dexmedetomidine infusion improves oxygenation and respiratory system mechanics and has a stabilizing hemodynamic effect in dogs anesthetized with isoflurane and mechanically ventilated.
Collapse
Affiliation(s)
- Caterina Di Bella
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Despoina Skouropoulou
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Marzia Stabile
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Cosmin Muresan
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Salvatore Grasso
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Luca Lacitignola
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Luisa Valentini
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| | - Francesco Staffieri
- Department of Emergency and Organ Transplantation, Section of Veterinary Clinics and Animal Production (Di Bella, Skouropoulou, Stabile, Lacitignola, Valentini, Crovace, Staffieri) and Section of Anesthesia and Intensive Care (Grasso), "Aldo Moro" University of Bari, Bari, Italy; Department of Surgery, University of Agriculture Sciences and Veterinary Medicine, Cluj-Napoca, Romania (Muresan)
| |
Collapse
|
35
|
Nayebzadeh N, Vazir B, Zendehdel M, Asghari A. Central Opioidergic and Adrenergic systems Mediates Food Intake via α1, α2 and β2 Receptors in Neonatal Layer-Type Chicken. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09810-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
36
|
Bruce DJ, Peterson CD, Kitto KF, Akgün E, Lazzaroni S, Portoghese PS, Fairbanks CA, Wilcox GL. Combination of a δ-opioid Receptor Agonist and Loperamide Produces Peripherally-mediated Analgesic Synergy in Mice. Anesthesiology 2020; 131:649-663. [PMID: 31343460 DOI: 10.1097/aln.0000000000002840] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The long-term use of opioids for analgesia carries significant risk for tolerance, addiction, and diversion. These adverse effects are largely mediated by μ-opioid receptors in the central nervous system. Based on the authors' previous observation that morphine and δ-opioid receptor agonists synergize in spinal cord in a protein kinase Cε-dependent manner, they predicted that this μ-opioid receptor-δ-opioid receptor synergy would take place in the central terminals of nociceptive afferent fibers and generalize to their peripheral terminals. Therefore, the authors hypothesized that loperamide, a highly efficacious μ-opioid receptor agonist that is excluded from the central nervous system, and oxymorphindole, a δ-opioid receptor agonist that was shown to synergize with morphine spinally, would synergistically reverse complete Freund's adjuvant-induced hyperalgesia. METHODS Using the Hargreaves assay for thermal nociception, the von Frey assay for mechanical nociception and the complete Freund's adjuvant-induced model of inflammatory pain, we tested the antinociceptive and antihyperalgesic effect of loperamide, oxymorphindole, or the loperamide-oxymorphindole combination. Animals (Institute for Cancer Research [ICR] CD1 strain mice; n = 511) received drug by systemic injection, intraplantar injection to the injured paw, or a transdermal solution on the injured paw. Dose-response curves for each route of administration and each nociceptive test were generated, and analgesic synergy was assessed by isobolographic analysis. RESULTS In naïve animals, the loperamide-oxymorphindole combination ED50 value was 10 times lower than the theoretical additive ED50 value whether given systemically or locally. In inflamed animals, the combination was 150 times more potent systemically, and 84 times more potent locally. All combinations showed statistically significant synergy when compared to the theoretical additive values, as verified by isobolographic analysis. The antihyperalgesia was ablated by a peripherally-restricted opioid antagonist. CONCLUSIONS From these data we conclude that the loperamide-oxymorphindole combination synergistically reverses complete Freund's adjuvant-induced inflammatory hyperalgesia. The authors also conclude that this interaction is mediated by opioid receptors located in the peripheral nervous system.
Collapse
Affiliation(s)
- Daniel J Bruce
- From the Departments of Pharmacology (D.J.B., C.A.F., G.L.W.) Neuroscience (C.D.P., K.F.K., S.L., C.A.F., G.L.W.) Pharmaceutics (C.A.F.) Dermatology (G.L.W.) Medicinal Chemistry (E.A., P.S.P.), University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Exploring functional consequences of GPCR oligomerization requires a different lens. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 169:181-211. [DOI: 10.1016/bs.pmbts.2019.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
Mazy A, Ghanem MA, Abd Elatif MSE, Basyoni YE. Spinal anesthesia for lengthy lower limb orthopedic surgeries: dexmedetomidine plus fentanyl versus dexmedetomidine. AIN-SHAMS JOURNAL OF ANESTHESIOLOGY 2019; 11:10. [DOI: 10.1186/s42077-019-0024-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/12/2019] [Indexed: 09/02/2023]
|
39
|
Abstract
Opioids are very potent and efficacious drugs, traditionally used for both acute and chronic pain conditions. However, the use of opioids is frequently associated with the occurrence of adverse effects or clinical problems. Other than adverse effects and dependence, the development of tolerance is a significant problem, as it requires increased opioid drug doses to achieve the same effect. Mechanisms of opioid tolerance include drug-induced adaptations or allostatic changes at the cellular, circuitry, and system levels. Dose escalation in long-term opioid therapy might cause opioid-induced hyperalgesia (OIH), which is a state of hypersensitivity to painful stimuli associated with opioid therapy, resulting in exacerbation of pain sensation rather than relief of pain. Various strategies may provide extra-opioid analgesia. There are drugs that may produce independent analgesic effects. A tailored treatment provided by skilled personnel, in accordance with the individual condition, is mandatory. Any treatment aimed at reducing opioid consumption may be indicated in these circumstances. Interventional techniques able to decrease the pain input may allow a decrease in the opioid dose, thus reverting the mechanisms producing tolerance of OIH. Intrathecal therapy with local anesthetics and a sympathetic block are the most common techniques utilized in these circumstances.
Collapse
Affiliation(s)
- Sebastiano Mercadante
- Main Regional Center of Supportive/Palliative Care, La Maddalena Cancer Center, Palermo, Italy.
- Palliative/Supportive Care and Rehabilitation, MD Anderson, Houston, TX, USA.
| | | | | |
Collapse
|
40
|
Creighton DW, Kumar AH, Grant SA. Perioperative Multimodal Pain Management: an Evidence-Based Update. CURRENT ANESTHESIOLOGY REPORTS 2019. [DOI: 10.1007/s40140-019-00340-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
41
|
Mercado-Reyes J, Almanza A, Segura-Chama P, Pellicer F, Mercado F. D2-like receptor agonist synergizes the μ-opioid agonist spinal antinociception in nociceptive, inflammatory and neuropathic models of pain in the rat. Eur J Pharmacol 2019; 853:56-64. [PMID: 30876975 DOI: 10.1016/j.ejphar.2019.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/25/2019] [Accepted: 03/12/2019] [Indexed: 11/19/2022]
Abstract
Opioids are potent analgesic drugs, but their use has been limited due to their side effects. Antinociceptive effects of D2-like receptor agonists such as quinpirole have been shown at the spinal cord level; however, their efficacy is not as high as that of opioids. Dopaminergic agonists are long-prescribed and well-tolerated drugs that have been useful to treat clinically and experimentally painful conditions. Because current pain treatments are not completely effective, the aim of this work was to determine if a D2-like receptor agonist improves the antinociceptive effects of a μ-opioid receptor agonist. Drugs were intrathecally administered in adult rats; mechanonociceptive and thermonociceptive tests were carried out. Intraplantar injection of complete Freund's adjuvant (CFA) and sciatic loose ligation (SLL) were used for inflammatory and neuropathic models of pain, respectively. In intact animals, D-Ala2, N-MePhe4, Gly-ol-enkephalin (DAMGO; a µ-opioid receptor agonist) increased the paw withdrawal latencies (PWL) in thermal and mechanical nociceptive tests in a dose-dependent manner. Quinpirole (D2-like receptor agonist) increased PWL only in mechanonociception. In the presence of quinpirole (1 nmol), the ED50 of the mechanical antinociceptive effect of DAMGO was significantly decreased (8-fold). Coadministration of 1 nmol quinpirole and 30 pmol DAMGO completely reversed hyperalgesia in the CFA model, whereas 100 pmol DAMGO plus 1 nmol quinpirole reversed the allodynia in the SLL model. This work offers evidence about a synergistic antinociceptive effect between opioidergic and dopaminergic drugs. This combination may relieve painful conditions resistant to conventional treatments, and it may reduce the adverse effects of chronic opioid administration.
Collapse
Affiliation(s)
- Jonathan Mercado-Reyes
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Angélica Almanza
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Pedro Segura-Chama
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico; Cátedras CONACyT - Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Pellicer
- Laboratorio de Neurofisiología Integrativa, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Mercado
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico.
| |
Collapse
|
42
|
Bahari Z, Meftahi GH. Spinal α 2 -adrenoceptors and neuropathic pain modulation; therapeutic target. Br J Pharmacol 2019; 176:2366-2381. [PMID: 30657594 DOI: 10.1111/bph.14580] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/23/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022] Open
Abstract
Neuropathic pain can arise from disease or damage to the nervous system. The most common symptoms of neuropathic pain include spontaneous pain, allodynia, and hyperalgesia. There is still limited knowledge about the factors that initiate and maintain neuropathic pain. However, ample evidence has proved the antinociceptive role of spinal α-adrenoceptors following nerve injury. It is well-documented that noradrenergic descending pathways from supraspinal loci exert an inhibitory influence on the spinal cord nociceptive neurons, mostly through the activation of spinal α2 -adrenoceptors. This, in turn, suppresses transmission of pain input and the hyperexcitability of spinal dorsal horn neurons. There is considerable evidence demonstrating that spinal application of α2 -adrenoceptor agonists leads to analgesic effects in animal models of neuropathic pain. Today, despite the recent rapid development of neuroscience and drug discovery, effective drugs with clear basic mechanisms have remained a mystery. Here, we give an overview of the cellular mechanisms through which brainstem adrenergic descending inhibitory processing can alter spinal pain transmission to the higher centres, and how these pathways change in neuropathic pain conditions focusing on the role of spinal α2 -adrenoceptors in the spinal dorsal horn. We then suggest that α2 -adrenoceptor agonist may be useful to treat neuropathic pain. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Zahra Bahari
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
43
|
Song F, Ye C, Qi F, Zhang P, Wang X, Lü Y, Fernandez-Escobar A, Zheng C, Li L. Effect of perioperative infusion of Dexmedetomidine combined with Sufentanil on quality of postoperative analgesia in patients undergoing laparoscopic nephrectomy: a CONSORT-prospective, randomized, controlled trial. BMC Anesthesiol 2018; 18:145. [PMID: 30342467 PMCID: PMC6195732 DOI: 10.1186/s12871-018-0608-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/05/2018] [Indexed: 01/27/2023] Open
Abstract
Background Postoperative pain is one of the most common symptoms after surgery, which brings physical discomfort to patients. In addition, it may cause a series of complications, and even affect the long-term quality of life. The purpose of this prospective, randomized, double-blinded, controlled trial is to investigate the efficacy and safety of dexmedetomidine combined with sufentanil to attenuate postoperative pain in patients after laparoscopic nephrectomy. Methods Ninety patients undergoing laparoscopic nephrectomy were randomized into three groups: the control (sufentanil 0.02 μg/kg/h, Group C), sufentanil plus low dose of dexmedetomidine (0.02 μg/kg/h each, Group D1), and sufentanil plus high dose of dexmedetomidine (0.04 μg/kg/h, Group D2). The patient-controlled analgesia was programmed to deliver a bolus dose of 0.5 ml, followed by an infusion of 2 ml/h and a lockout time of 10 min. The primary goal was to calculate the cumulative amount of self-administered sufentanil; the secondary goals were to estimate pain intensity using the numerical rating scale (NRS), level of sedation, the first bowel movement, concerning adverse effects as well as duration of postoperative hospital stay. Results The total consumption of sufentanil in group D1 and D2 were significantly lower than in group C during the first 8 h after surgery (P < 0.05), whereas there were no statistically significant differences (P > 0.05) between group D1 and D2. Compared with group C, the NRS scores at rest during first 8 h after surgery were significantly lower in group D1 (P < 0.05). The NRS scores, neither at rest nor with movement, show statistically significant differences between group D1 and D2 at each time point following surgery (P > 0.05). The time to first flatus was shorter in group D1 compared with the control group (P < 0.05). In addition, compared with group C, group D1 and D2 had a shorter time for first defecation (P < 0.05). Conclusions Dexmedetomidine combined with sufentanil showed better postoperative analgesia without adverse effects, as well as facilitated bowel movements for patients undergoing laparoscopic nephrectomy. Trial registration We registered this study in a Chinese Clinical Trial Registry (ChiCTR) centre on Dec 23 2015 and received the registration number: ChiCTR-IPR-15007628.
Collapse
Affiliation(s)
- Fuxi Song
- Department of Anaesthesiology, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Chunmiao Ye
- Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Feng Qi
- Department of Anaesthesiology, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, 250012, China
| | - Ping Zhang
- Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Xuexiang Wang
- The Institute for Translational Medicine, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Yanfeng Lü
- Department of Anoproctology, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China
| | - Alejandro Fernandez-Escobar
- Translational Research Program, University of Toronto, 27 King's College Circle, Toronto, ON, M5S 1A1, Canada
| | - Chao Zheng
- Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China.
| | - Liang Li
- Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan, 250033, China.
| |
Collapse
|
44
|
Bivalent ligand that activates mu opioid receptor and antagonizes mGluR5 receptor reduces neuropathic pain in mice. Pain 2018; 158:2431-2441. [PMID: 28891868 DOI: 10.1097/j.pain.0000000000001050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The mu opioid receptor (MOR) and metabotropic glutamate receptor 5 (mGluR5) are well-established pharmacological targets in the management of chronic pain. Both receptors are expressed in the spinal cord. MMG22, a bivalent ligand containing 2 pharmacophores separated by 22 atoms, which simultaneously activates MOR and antagonizes mGluR5, has been shown to produce potent reversal of tactile hypersensitivity in rodent models of lipopolysaccharide (LPS)-and bone cancer-induced chronic pain. This study assessed whether intrathecal MMG22 also is effective in reducing pain of neuropathic origin. Furthermore, we theorized that MMG22 should reduce hyperalgesia in nerve-injured mice in a manner consistent with a synergistic interaction between MOR and mGluR5. Several weeks after spared nerve injury, tactile hypersensitivity was reversed in mice by the intrathecal injection of MMG22 (0.01-10 nmol) but also by its shorter spacer analog, MMG10, with similar potency. The potencies of the bivalent ligands were 10- to 14-fold higher than those of the compounds upon which the bivalent structure was based, the MOR agonist oxymorphone and the mGluR5 antagonist MPEP. Coadministration of oxymorphone and MPEP demonstrated analgesic synergism, an interaction confirmed by isobolographic analysis. This study indicates that in the spared nerve injury-induced model of neuropathic pain, the 2 pharmacophores of the bivalent ligands MMG22 and MMG10 target MOR and mGluR5 as separate receptor monomers. The observed increase in the potency of MMG22 and MMG10, compared with oxymorphone and MPEP, may reflect the synergistic interaction of the 2 pharmacophores of the bivalent ligand acting at their respective separate receptor monomers.
Collapse
|
45
|
Adrenergic Agonists Bind to Adrenergic-Receptor-Like Regions of the Mu Opioid Receptor, Enhancing Morphine and Methionine-Enkephalin Binding: A New Approach to "Biased Opioids"? Int J Mol Sci 2018; 19:ijms19010272. [PMID: 29342106 PMCID: PMC5796218 DOI: 10.3390/ijms19010272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/10/2018] [Accepted: 01/13/2018] [Indexed: 11/16/2022] Open
Abstract
Extensive evidence demonstrates functional interactions between the adrenergic and opioid systems in a diversity of tissues and organs. While some effects are due to receptor and second messenger cross-talk, recent research has revealed an extracellular, allosteric opioid binding site on adrenergic receptors that enhances adrenergic activity and its duration. The present research addresses whether opioid receptors may have an equivalent extracellular, allosteric adrenergic binding site that has similar enhancing effects on opioid binding. Comparison of adrenergic and opioid receptor sequences revealed that these receptors share very significant regions of similarity, particularly in some of the extracellular and transmembrane regions associated with adrenergic binding in the adrenergic receptors. Five of these shared regions from the mu opioid receptor (muOPR) were synthesized as peptides and tested for binding to adrenergic, opioid and control compounds using ultraviolet spectroscopy. Adrenergic compounds bound to several of these muOPR peptides with low micromolar affinity while acetylcholine, histamine and various adrenergic antagonists did not. Similar studies were then conducted with purified, intact muOPR with similar results. Combinations of epinephrine with methionine enkephalin or morphine increased the binding of both by about half a log unit. These results suggest that muOPR may be allosterically enhanced by adrenergic agonists.
Collapse
|
46
|
Mizobe F, Wakuno A, Okada J, Otsuka T, Ishikawa Y, Kurimoto S. Clinical usefulness of intravenous constant rate infusion of fentanyl and medetomidine under sevoflurane anesthesia in Thoroughbred racehorses undergoing internal fixation surgery. J Equine Sci 2017; 28:143-147. [PMID: 29270071 PMCID: PMC5735311 DOI: 10.1294/jes.28.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/18/2017] [Indexed: 11/30/2022] Open
Abstract
A total of 20 racehorses with longitudinal fractures underwent internal fixation surgery under sevoflurane anesthesia combined with infusion of medetomidine (3 µg/kg/hr) alone (10 horses, group M) or medetomidine and
fentanyl (7 µg/kg/hr) (10 horses, group FM). In group FM, the end-tidal sevoflurane concentration during surgery was maintained significantly lower than in group M (2.8–2.9% for group M vs. 2.2–2.6% for group FM,
P<0.01). The mean arterial blood pressure was maintained over 70 mmHg using dobutamine infusion (group M, 0.36–0.54 µg/kg/min; group FM, 0.27–0.65 µg/kg/min), and the recovery qualities were clinically acceptable in
both groups. In conclusion, co-administration of fentanyl and medetomidine by constant rate infusion may be a clinically useful intraoperative anesthetic adjunct for horses to reduce the requirement of sevoflurane when
they undergo orthopedic surgery.
Collapse
Affiliation(s)
- Fumiaki Mizobe
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| | - Ai Wakuno
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| | - Jun Okada
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| | - Tasuku Otsuka
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| | - Yuhiro Ishikawa
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| | - Shinjiro Kurimoto
- Racehorse Hospital, Ritto Training Center, Japan Racing Association, Shiga 520-3085, Japan
| |
Collapse
|
47
|
Effects of I 2 -imidazoline receptor (IR) alkylating BU99006 in the mouse brain: Upregulation of nischarin/I 1 -IR and μ-opioid receptor proteins and modulation of associated signalling pathways. Neurochem Int 2017; 108:169-176. [DOI: 10.1016/j.neuint.2017.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022]
|
48
|
Abstract
There has been great progress in the understanding of basic neurobiologic mechanisms of pain, but this body of knowledge has not yet translated into new and improved analgesics. Progress has been made regarding pain assessment in horses, but more work is needed until sensitive and accurate pain assessment tools are available for use in clinical practice. This review summarizes and updates the knowledge concerning the cornerstones of pain medicine (understand, assess, prevent, and treat). It highlights the importance of understanding pain mechanisms and expressions to enable a rational approach to pain assessment, prevention, and management in the equine patient.
Collapse
|
49
|
Vo L, Hood S, Drummond PD. Involvement of Opioid Receptors and α2-Adrenoceptors in Inhibitory Pain Modulation Processes: A Double-Blind Placebo-Controlled Crossover Study. THE JOURNAL OF PAIN 2016; 17:1164-1173. [DOI: 10.1016/j.jpain.2016.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/27/2016] [Accepted: 07/27/2016] [Indexed: 12/25/2022]
|
50
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2016; 68:631-700. [PMID: 27343248 PMCID: PMC4931872 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|