1
|
Seymour KA, Strain M, Ashley-Koch A, Muehlbauer MJ, Ilkayeva OR, George TK, Hill D, Ellison M, Ito S, Lagoo-Deenadayalan S, Plichta JK, Purves JT, Thacker JKM, Nalley J, Kirk AD, Hwang ES, Bain JR. Acute perioperative alterations in metabolism: A pilot study using mass spectrometry-based metabolomics. Surgery 2025; 180:109055. [PMID: 39740607 PMCID: PMC11936720 DOI: 10.1016/j.surg.2024.109055] [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: 06/12/2024] [Revised: 10/15/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVE To characterize early physiologic stresses imposed by surgery by applying metabolomic analyses to deeply phenotype pre- and postoperative plasma and urine of patients undergoing elective surgical procedures. BACKGROUND Patients experience perioperative stress through depletion of metabolic fuels. Bowel stasis or injury might allow more microbiome-derived uremic toxins to enter the blood, while the liver and kidney are simultaneously clearing analgesic and anesthetic drugs. Metabolomics provides a broad-scale snapshot of small-molecule chemicals generated in vital energetic and detoxification pathways, enabling a mechanistic understanding of surgical stressors. METHODS We performed metabolomic analysis of paired preoperative and early-recovery plasma (n = 34) and urine (n = 35) from patients who underwent elective surgeries, spanning cardiovascular, gastrointestinal, hernia, oncologic, and urologic procedures. Mass spectrometry-based metabolomics analyses were performed together with the analysis of select metabolites and macromolecules via conventional clinical assays. RESULTS Fuel stress during elective surgery manifested in changes across all major metabolic pathways, encompassing lipolysis, glycolysis-Krebs cycle, ketogenesis, and glycogenolysis. A common signature of enhanced amino acid oxidation and urea-cycle activity emerged, which was especially pronounced in patients given citrulline boluses before visceral procedures. Excretion of amino acid-derived catabolite toxins increased during surgery, notably those derived from gut microbes, as did an extract of disposable surgical plasticware, bis(2-ethylhexyl)phthalate. CONCLUSION Elective surgery imposes broad-scale early and measurable metabolic changes. The use of citrulline-enriched preoperative carbohydrate drinks needs further study to limit metabolic burden. Attention to perioperative nutrition and intraoperative control of gut-microbial toxins might reduce metabolic derangements and lead to better postoperative outcomes.
Collapse
Affiliation(s)
- Keri A Seymour
- Department of Surgery, Duke University School of Medicine, Durham, NC.
| | - Madison Strain
- University Program in Genetics and Genomics, Duke University School of Medicine, Durham, NC; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Allison Ashley-Koch
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC; Department of Medicine (Nephrology), Duke University School of Medicine, Durham, NC; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC; Department of Medicine (Endocrinology), Duke University School of Medicine, Durham, NC
| | - Tabitha K George
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Demitrius Hill
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Mark Ellison
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Satoru Ito
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | | | | | - J Todd Purves
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Julie K M Thacker
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Justin Nalley
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - E Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - James R Bain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC; Department of Medicine (Endocrinology), Duke University School of Medicine, Durham, NC
| |
Collapse
|
2
|
Liu Y, Liang H, Sun Y, Liu W, Ye L, He W, Wang H. Effect of perioperative dexmedetomidine on recovery of postoperative gastrointestinal function in patients with general anesthesia: a systematic review and meta-analysis. BMC Anesthesiol 2024; 24:479. [PMID: 39732663 DOI: 10.1186/s12871-024-02868-0] [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: 02/19/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND There is controversy surrounding the influence of dexmedetomidine on the recovery of postoperative gastrointestinal dysfunction in patients under general anesthesia. The main purpose of this meta-analysis is to evaluate the effect of dexmedetomidine administration during the perioperative period on the recovery of gastrointestinal function in patients under general anesthesia. METHODS A systematic review and meta-analysis with trial sequential analysis was performed to identify randomized controlled trials comparing dexmedetomidine administration with placebo for the recovery of gastrointestinal function. The primary outcomes were gastrointestinal function; first oral feeding time; incidences of postoperative nausea and vomiting, postoperative nausea, and postoperative vomiting; time to first bowel sound; time to first flatus; and time to first defecation. The secondary outcome was the length of hospital stay. RESULTS A total of 20 studies comparing 2,470 participants were included in this meta-analysis. Perioperative dexmedetomidine administration did not result in a significant reduction in the time to first oral feeding (MD= -7.91, 95% CI = - 16.45 to 0.62, P = 0.07). However, dexmedetomidine administration was associated with a decreased incidence of postoperative nausea and vomiting (RR = 0.72, 95% CI = 0.58 to 0.88, P = 0.001), time to first flatus (MD= -6.73, 95% CI= -10.31 to -3.15, P = 0.0002), and time to first defecation (MD= -12.01, 95% CI = -22.40 to -1.61, P = 0.02). CONCLUSIONS Perioperative dexmedetomidine administration can promote the recovery of gastrointestinal function and reduce the length of hospital stay after abdominal surgery. The optimal dose and timing of dexmedetomidine and the influence on non-abdominal surgery need further investigation. TRIAL REGISTRATION The study protocol was registered in the PROSPERO database (registration number: CRD42023443708) on July 9, 2023.
Collapse
Affiliation(s)
- Yanping Liu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan City, 25000, China
| | - Hongbin Liang
- Department of Anesthesiology, The First People's Hospital of Foshan, Lingnan Road 81#, Foshan City, 528000, China
| | - Yuanyuan Sun
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan City, 25000, China
| | - Weihua Liu
- Shandong First Medical University & Shandong Academy of Medical Science, Jinan City, 25000, China
| | - Li Ye
- Department of Anesthesiology, The First People's Hospital of Foshan, Lingnan Road 81#, Foshan City, 528000, China
| | - Wanyou He
- Department of Anesthesiology, The First People's Hospital of Foshan, Lingnan Road 81#, Foshan City, 528000, China.
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Lingnan Road 81#, Foshan City, 528000, China.
| |
Collapse
|
3
|
Xie D, Wang F, Wen W, Li H. Postoperative analgesic effect of intravenous coinjection of lidocaine and dexmedetomidine in gynaecological surgery: a systematic review and meta-analysis. BMJ Open 2024; 14:e091904. [PMID: 39663167 PMCID: PMC11647388 DOI: 10.1136/bmjopen-2024-091904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
OBJECTIVES Hysterectomy or myomectomy is a common gynaecological procedure that results in moderate to severe acute postoperative pain, which can cause many adverse effects. This study aimed to compare the postoperative analgesic efficacy, opioid consumption, quality of postoperative recovery (QOR) and adverse reactions of intravenous coinjection of lidocaine and dexmedetomidine versus lidocaine or dexmedetomidine alone in gynaecological surgery. DESIGN Systematic review and meta-analysis was performed. DATA SOURCES The PubMed, Embase, Cochrane Library and Web of Science databases were used to access the articles. Electronic databases were searched for eligible studies published before 1 May 2024. ELIGIBILITY CRITERIA All randomised controlled trials (RCTs) were included in the final analysis in which the intraoperative intervention group received intravenous coinjection of lidocaine and dexmedetomidine, and the control group received intravenous injection of lidocaine or dexmedetomidine alone in gynaecologic procedures. DATA EXTRACTION AND SYNTHESIS Study retrieval, literature screening, data extraction and risk of bias assessment were performed independently by two reviewers. The quality of included studies was assessed by the Cochrane Collaboration Risk of Bias (ROB V.2.0). Data were expressed as standardised mean difference, weighted mean difference or relative risk with 95% CI. Review Manager V.5.4 was used for data analysis. RESULTS A total of five RCTs were included, involving 672 patients, of which 224 patients received coinjection of lidocaine and dexmedetomidine. The results revealed that coinjection of lidocaine and dexmedetomidine was superior to individual lidocaine in the visual analogue scale (VAS) scores at 1 hour (MD=-0.90, 95% CI (-1.11 to -0.69), p<0.001), 2 hours (MD=-0.99, 95% CI (-1.19 to -0.80), p<0.001), 4 hours (MD=-1.20, 95% CI (-1.75 to -0.66), p<0.001), 6 hours (MD=-1.09, 95% CI (-1.48 to -0.70), p<0.001), 8 hours (MD=-1.22, 95% CI (-1.61 to -0.83), p<0.001) and 12 hours (MD=-0.76, 95% CI (-1.35 to -0.17), p=0.o1) after surgery. Compared with the dexmedetomidine group, the lidocaine+dexmedetomidine group had low VAS scores at 1 hour (MD=-0.60, 95% CI (-0.83 to -0.37), p<0.001), 2 hours (MD=-0.70, 95% CI (-0.87 to -0.53), p<0.001), 6 hours (MD=-0.79, 95% CI (-0.98 to -0.59), p<0.001), 8 hours (MD=-0.77, 95% CI (-1.25 to -0.28), p=0.002) and 12 hours (MD=-0.56, 95% CI (-1.00 to -0.11), p=0.01) after surgery. Coinjection of lidocaine and dexmedetomidine resulted in significantly lower postoperative opioid consumption, postoperative nausea and vomiting and bradycardia than lidocaine alone (all p<0.05). Compared with the dexmedetomidine group, the lidocaine+dexmedetomidine group shortened the time to intestinal transit resumption (p=0.003). Coinjection of lidocaine and dexmedetomidine reduced intraoperative opioid consumption and increased QOR scores compared with lidocaine and dexmedetomidine alone (all p<0.05). CONCLUSION Lidocaine combined with dexmedetomidine had superior analgesic efficacy and safety. However, due to the limitation in the number of available studies, more large-scale, prospective RCTs are needed for further investigation.PROSPERO registration numberCRD42023384018.
Collapse
Affiliation(s)
- Dan Xie
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Fangjun Wang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Wen Wen
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongqiong Li
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
4
|
Luo X, Hou HJ, Chen PS, Chang XL, Li Y, An LX, Liu FK, Xue FS. Addition of Dexmedetomidine to the Anesthesia Regimen Attenuates Pain and Improves Early Recovery After Esophageal Endoscopic Submucosal Dissection: A Randomized Controlled Trial. Drug Des Devel Ther 2024; 18:4551-4562. [PMID: 39411154 PMCID: PMC11476426 DOI: 10.2147/dddt.s475749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Objective Postoperative pain is a common yet often underestimated complication following esophageal endoscopic submucosal dissection (ESD), with limited strategies for effective management. This prospective, double-blind, randomized controlled trial assessed the effects of adding dexmedetomidine (DEX) to the anesthesia regimen on postoperative pain and early recovery in patients undergoing esophageal ESD. Methods In total, 60 patients scheduled for elective esophageal ESD under general anesthesia were randomly assigned to the DEX or control group. The DEX group received an intravenous loading dose of DEX at 1 μg/kg for 10 min, followed by a continuous intravenous infusion of 0.6 µg/kg/h, which was stopped 30 min before the end of the procedure. The control group received normal saline as a placebo. The study's primary outcome was the incidence of moderate-to-severe postoperative pain. Secondary outcomes included postoperative pain scores, hemodynamic parameters, the occurrence of postoperative nausea and vomiting (PONV), patient satisfaction, and lengths of stay in the post-anesthesia care unit (PACU) and hospital. Results The incidence of moderate-to-severe postoperative pain in the DEX group was significantly lower than that in the control group (absolute difference: -33.4%; OR: 0.250; 95% CI: 0.085-0.731, P = 0.01). Pain scores at 1 h postoperatively (0.5[2.0] vs 3.0[1.3], P = 0.003) were significantly lower in the DEX group. Additionally, morphine dosage in the PACU (0[0] vs 1.0[2.0] P = 0.004) was significantly reduced in the DEX group compared with the control group. In the DEX group, the incidence and severity of PONV were significantly decreased and the length of PACU stay was shorter than in the control group (P < 0.01). However, the rates of intraoperative hypotension, tachycardia, and bradycardia were similar between the two groups. Patient satisfaction and length of hospital stay were also comparable. Conclusion Adding DEX to the anesthesia regimen for esophageal ESD significantly attenuates postoperative pain and improves early recovery outcomes.
Collapse
Affiliation(s)
- Xin Luo
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hai-Jun Hou
- Department of Pain Medicine, Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Pei-Shan Chen
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xin-Lu Chang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Li-Xin An
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Fu-Kun Liu
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Fu-Shan Xue
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, People’s Republic of China
| |
Collapse
|
5
|
Sun JJ, Wang H, Tang LL, Jiang H, Liu XS. Effect of intraoperative dexmedetomidine on recovery of gastrointestinal function after caesarean section undergoing spinal and epidural anesthesia: A randomized, double blind, placebo-controlled clinical trial. Eur J Obstet Gynecol Reprod Biol 2024; 297:30-35. [PMID: 38574697 DOI: 10.1016/j.ejogrb.2024.03.038] [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: 10/07/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
OBJECTIVE Gastrointestinal dysfunction after cesarean section negatively affects postoperative recovery. Dexmedetomidine has been shown to improve postoperative gastrointestinal function in patients undergoing lumbar spinal fusion surgery and laparoscopic gastrectomy, but its role in cesarean section has not been fully elucidated. The study aimed to investigate the effect of dexmedetomidine on gastrointestinal function after cesarean section. STUDY DESIGN 220 pregnant women who underwent elective cesarean section were randomized into group D and group S. Group D patients received a loading dose of 0.5 μg/kg of dexmedetomidine for 10 mins followed by a maintenance dose of 0.5 μg/kg/h intravenously immediately after the umbilical cord was cut intraoperatively, whereas the other group (group S) received an equivalent quantity of normal saline as loading and maintenance dose IV by infusion pump. The primary outcome was time to first flatus after surgery (hours). Secondary outcomes included time to first feces and first bowel sounds (hours), incidence rates of postoperative gastrointestinal complications, and the length of postoperative hospital stay (days). RESULTS Modified intention-to-treat analysis showed that patients in Group D had a significantly shorter time to first flatus (21 [16 to 28.25] vs. 25 [18 to 32.25] h; P = 0.014), time to first feces (45.5 [35.75 to 55.25] vs. 53 [40 to 60] h; P = 0.019), and time to first bowel sounds (P = 0.010), a lower incidence of abdominal distension (21[20.6 %] vs. 36[34.3 %], P = 0.027), shorter length of postoperative hospital stay (P = 0.010) compared to patients in Group S. CONCLUSION Intraoperative dexmedetomidine infusion reduces the time to first flatus, the incidence of abdominal distension, and shortens the length of hospital stay, promoting gastrointestinal function after cesarean section.
Collapse
Affiliation(s)
- Jing-Jing Sun
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li-Li Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Xue-Sheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
6
|
Lai YC, Wang WT, Hung KC, Chen JY, Wu JY, Chang YJ, Lin CM, Chen IW. Impact of intravenous dexmedetomidine on postoperative gastrointestinal function recovery: an updated meta-analysis. Int J Surg 2024; 110:1744-1754. [PMID: 38085848 PMCID: PMC10942148 DOI: 10.1097/js9.0000000000000988] [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: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 03/16/2024]
Abstract
BACKGROUND Postoperative ileus (POI) is a complication that may occur after abdominal or nonabdominal surgery. Intravenous dexmedetomidine (Dex) has been reported to accelerate postoperative gastrointestinal function recovery; however, updated evidence is required to confirm its robustness. METHODS To identify randomized controlled trials examining the effects of perioperative intravenous Dex on gastrointestinal function recovery in patients undergoing noncardiac surgery, databases including MEDLINE, EMBASE, Google Scholar, and Cochrane Library were searched on August 2023. The primary outcome was time to first flatus. Secondary outcomes included time to oral intake and defecation as well as postoperative pain scores, postoperative nausea/vomiting (PONV), risk of hemodynamic instability, and length of hospital stay (LOS). To confirm its robustness, subgroup analyses and trial sequential analysis were performed. RESULTS The meta-analysis of 22 randomized controlled trials with 2566 patients showed that Dex significantly reduced the time to flatus [mean difference (MD):-7.19 h, P <0.00001), time to oral intake (MD: -6.44 h, P =0.001), time to defecation (MD:-13.84 h, P =0.008), LOS (MD:-1.08 days, P <0.0001), and PONV risk (risk ratio: 0.61, P <0.00001) without differences in hemodynamic stability and pain severity compared with the control group. Trial sequential analysis supported sufficient evidence favoring Dex for accelerating bowel function. Subgroup analyses confirmed the positive impact of Dex on the time to flatus across different surgical categories and sexes. However, this benefit has not been observed in studies conducted in regions outside China. CONCLUSIONS Perioperative intravenous Dex may enhance postoperative gastrointestinal function recovery and reduce LOS, thereby validating its use in patients for whom postoperative ileus is a significant concern.
Collapse
Affiliation(s)
- Yi-Chen Lai
- Department of Anesthesiology, Chi Mei Medical Center, Tainan city, Taiwan
| | - Wei-Ting Wang
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Kuo-Chuan Hung
- Department of Anesthesiology, Chi Mei Medical Center, Tainan city, Taiwan
| | - Jen-Yin Chen
- Department of Anesthesiology, Chi Mei Medical Center, Tainan city, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ying-Jen Chang
- Department of Anesthesiology, Chi Mei Medical Center, Tainan city, Taiwan
| | - Chien-Ming Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan city, Taiwan
| | - I-Wen Chen
- Department of Anesthesiology, Chi Mei Medical Center, Liouying, Tainan city, Taiwan
| |
Collapse
|
7
|
Srinivasan SS, Dosso J, Huang HW, Selsing G, Alshareef A, Kuosmanen J, Ishida K, Jenkins J, Madani WAM, Hayward A, Traverso G. An ingestible self-propelling device for intestinal reanimation. Sci Robot 2024; 9:eadh8170. [PMID: 38416855 DOI: 10.1126/scirobotics.adh8170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 01/31/2024] [Indexed: 03/01/2024]
Abstract
Postoperative ileus (POI) is the leading cause of prolonged hospital stay after abdominal surgery and is characterized by a functional paralysis of the digestive tract, leading to symptoms such as constipation, vomiting, and functional obstruction. Current treatments are mainly supportive and inefficacious and yield acute side effects. Although electrical stimulation studies have demonstrated encouraging pacing and entraining of the intestinal slow waves, no devices exist today to enable targeted intestinal reanimation. Here, we developed an ingestible self-propelling device for intestinal reanimation (INSPIRE) capable of restoring peristalsis through luminal electrical stimulation. Optimizing mechanical, material, and electrical design parameters, we validated optimal deployment, intestinal electrical luminal contact, self-propelling capability, safety, and degradation of the device in ex vivo and in vivo swine models. We compared the INSPIRE's effect on motility in models of normal and depressed motility and chemically induced ileus. Intestinal contraction improved by 44% in anesthetized animals and up to 140% in chemically induced ileus cases. In addition, passage time decreased from, on average, 8.6 days in controls to 2.5 days with the INSPIRE device, demonstrating significant improvement in motility. Luminal electrical stimulation of the intestine via the INSPIRE efficaciously restored peristaltic activity. This noninvasive option offers a promising solution for the treatment of ileus and other motility disorders.
Collapse
Affiliation(s)
- Shriya S Srinivasan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Julien Dosso
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hen-Wei Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - George Selsing
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Amro Alshareef
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Johannes Kuosmanen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Keiko Ishida
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua Jenkins
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wiam Abdalla Mohammed Madani
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alison Hayward
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
8
|
Lv G, Zhang T, Wang L, Fu X, Wang Y, Yao H, Fang H, Xia X, Yang J, Wang B, Zhang Z, Jin X, Kang Y, Cheng Y, Wu Q. Prediction of prokinetic agents in critically ill patients with feeding intolerance: a prospective observational clinical study. Front Nutr 2023; 10:1244517. [PMID: 37964927 PMCID: PMC10641452 DOI: 10.3389/fnut.2023.1244517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Background Prokinetic agents are currently considered the first-line therapy to improve gastric emptying when feeding intolerance occurred in critically ill adults. In this study, we developed a technique to assess the feasibility of predicting prokinetic agent efficacy in critically ill patients. Methods The first images of each patient were obtained after EFI had occurred but before the first dose of prokinetic agents was administered and additional images were obtained every morning until the seventh day. The gastric antrum echodensity was recorded based on grayscale values (50th percentile, ED50; 85th percentile, ED85; mean, EDmean) and daily energy and protein intake was collected as the judgment for effective and ineffective group. A receiver operating characteristic curve was analyzed to distinguish the thresholds between the two groups and thus determine the ability of the gastric antrum echodensity to predict the efficacy of prokinetic agents. Results In total, 83 patients were analyzed. Patients in the ineffective group had a higher ED50 (58.13 ± 14.48 vs. 49.88 ± 13.78, p < 0.001, difference 95% CI: 5.68, 10.82), ED85 (74.81 ± 16.41 vs. 65.70 ± 16.05, p < 0.001, difference 95% CI:6.16, 12.05), and EDmean (60.18 ± 14.31 vs. 51.76 ± 14.08, p < 0.001, difference 95% CI: 5.85, 11.00) than those in the effective group. Patients in the effective group more easily reached the target energy 16.21 ± 7.98 kcal/kg vs. 9.17 ± 6.43 kcal/kg (p < 0.001), 0.72 ± 0.38 g/kg vs. 0.42 ± 0.31 g/kg (p < 0.001) than in the ineffective group intake by day. Conclusion The gastric antrum echodensity might serve as a tool for judging the efficacy of prokinetic agents, helping clinicians to decide whether to use prokinetic agents or place a post-pyloric tube when feeding intolerance occurs in critically ill patients.Clinical trial registration:http://www.chictr.org.cn/addproject2.aspx, ChiCTR2200058373. Registered 7 April 2022.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Qin Wu
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Dexmedetomidine Has Differential Effects on the Contractility of Equine Jejunal Smooth Muscle Layers In Vitro. Animals (Basel) 2023; 13:ani13061021. [PMID: 36978562 PMCID: PMC10044630 DOI: 10.3390/ani13061021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
α2 agonists are frequently used in horses with colic, even though they have been shown to inhibit gastrointestinal motility. The aim of this study was to determine the effect of dexmedetomidine on small intestinal in vitro contractility during different phases of ischaemia. Experimental segmental jejunal ischaemia was induced in 12 horses under general anaesthesia, and intestinal samples were taken pre-ischaemia and following ischaemia and reperfusion. Spontaneous and electrically evoked contractile activity of the circular and longitudinal smooth muscles were determined in each sample with and without the addition of dexmedetomidine. During a second experiment, tetrodotoxin was added to determine if the effect was neurogenic. We found that the circular smooth muscle (CSM) contractility was not affected by ischaemia, whereas the longitudinal smooth muscle (LSM) showed an increase in both spontaneous and induced contractile activity. The addition of dexmedetomidine caused a decrease in the spontaneous contractile activity of CSM, but an increase in that of LSM, which was not mediated by the enteric nervous system. During ischaemia, dexmedetomidine also mildly increased the electrically induced contractile activity in LSM. These results may indicate a stimulatory effect of dexmedetomidine on small intestinal contractility. However, the influence of dexmedetomidine administration on intestinal motility in vivo needs to be further investigated.
Collapse
|
10
|
Sui X, Wang Y, Jin M, Li K, Jiang G, Song A, He Z, Yin C, Zhao J, Wang L, Han F. The effects of dexmedetomidine for patient-controlled analgesia on postoperative sleep quality and gastrointestinal motility function after surgery: A prospective, randomized, double-blind, and controlled trial. Front Pharmacol 2022; 13:990358. [PMID: 36299885 PMCID: PMC9588938 DOI: 10.3389/fphar.2022.990358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Postoperative poor sleep quality and decreased gastrointestinal motility function are common clinical problems. This study investigated the effects of dexmedetomidine (DEX) combined with sufentanil for patient-controlled analgesia (PCA) on postoperative sleep quality and gastrointestinal motility function after surgery in patients with colorectal cancer. Methods: Patients undergoing colorectal cancer surgery were randomly divided into three groups, DEX 0, 200, or 400 μg, each combined with sufentanil 150 μg for PCA immediately after surgery. The primary outcome was sleep quality in the first 7 days after surgery based on the Athens Insomnia Scale (AIS) score. The secondary outcome was postoperative gastrointestinal motility recovery evaluated by the time of first flatus, first feces and first diet. Postoperative pain intensity, side effects and the length of postoperative hospital stay were also compared among groups. The study was registered with the Chinese Clinical Trial Registry (https://www.chictr.org.cn/enIndex.aspx, ChiCTR2000032601). Results: Ultimately, 210 cases were included. Sleep quality was better in the DEX 200 μg group and DEX 400 μg group than in the DEX 0 μg group. Overall, in the DEX 200 μg group and DEX 400 μg group, the AIS score (p < 0.05) and the incidence of sleep disturbance (7.3%, 4.5% vs. 19.6%, p < 0.001) were lower than those in the DEX 0 μg group in the first 7 days after surgery. There were no significant differences in postoperative gastrointestinal motility among the three groups in the total surgical categories (p > 0.05). In the laparoscopic surgery patients of each group, the time of postoperative first flatus (p = 0.02) and first feces (p = 0.01) was significantly longer in the DEX 400 μg group than in the DEX 0 μg group. There were no differences in postoperative pain intensity, side effects or length of postoperative hospital stay (p > 0.05). Conclusion: The continuous infusion of DEX (200 or 400 μg) for PCA significantly improved postoperative sleep quality after colorectal cancer surgery. DEX (200 μg) was better at improving postoperative sleep quality without affecting gastrointestinal motility function than DEX (400 μg) in patients who underwent laparoscopic colorectal cancer surgery.
Collapse
Affiliation(s)
- Xin Sui
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yue Wang
- Department of Pain Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Mingxin Jin
- Department of Anesthesiology, Cancer Hospital Chinese Academy of Medical Science, Shenzhen Center, Shenzhen, China
| | - Kun Li
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Ge Jiang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Ailing Song
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Zhaoyi He
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chengke Yin
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jingshun Zhao
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Liping Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Fei Han
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
- *Correspondence: Fei Han,
| |
Collapse
|
11
|
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
|
12
|
Abass M, Ibrahim H, Salci H, Hamed MA. Evaluation of the effect of different sedative doses of dexmedetomidine on the intestinal motility in clinically healthy donkeys (Equus asinus). BMC Vet Res 2022; 18:274. [PMID: 35836159 PMCID: PMC9281064 DOI: 10.1186/s12917-022-03376-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
AIM Gastrointestinal effects of different doses of dexmedetomidine in donkeys are still unidentified. The current study aimed to evaluate the impact of different doses of dexmedetomidine on the motility of selected parts of the gastrointestinal tracts in donkeys using transabdominal ultrasonography. MATERIALS AND METHODS An experimental crossover study was conducted on 30 healthy donkeys of both sexes (15 males and 15 females; 160 ± 60 kg). With a two-week washout period, each donkey received an injection of either a normal saline solution or three different doses of dexmedetomidine (3, 5, and 7 μg/kg, respectively). All medications were administered intravenously in equal volumes. The contractility of selected intestinal segments (duodenum, jejunum, left colon, right colon, and cecum) was measured 3 min before administration (zero time) and at 15, 30, 45, 60, 90, and 120 minutes after administration. RESULTS Small and large intestinal motility was within the normal ranges before IV injection of normal isotonic saline or dexmedetomidine at a dose of 3, 5, and 7 μg/kg. Two Way Repeated Measures ANOVA output of the data displayed a statistically significant the between time and treatments for the contractility of each of the duodenum (P = 0.0029), jejunum (P = 0.0033), left colon (P = 0.0073), right colon (P = 0.0035), and cecum (P = 0.0026), implying that the impact of treatment on the gastric motility varied among different time points. The simple main effect analysis revealed that the IV dexmedetomidine at 3, 5, and 7 μg/kg doses significantly inhibited (P ≤ 0.01) the bowel contractility compared to the administration of isotonic saline. CONCLUSION Dose-dependent inhibitory effect of dexmedetomidine on intestinal motility was reported in donkeys following intravenous administration. This inhibitory effect on intestinal motility should be considered in clinical practice.
Collapse
Affiliation(s)
- Marwa Abass
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Hussam Ibrahim
- Department of Internal Medicine, Infectious and Fish Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Hakan Salci
- Department of Surgery, Faculty of Veterinary Medicine, Uludag Universitesi, Bursa, Turkey
| | - Mohamed A Hamed
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| |
Collapse
|
13
|
Grajecki D, Tacke F. [Gastrointestinal motility disorders in critically ill patients]. Dtsch Med Wochenschr 2022; 147:696-704. [PMID: 35636421 DOI: 10.1055/a-1664-1823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The gastrointestinal tract is one of the most complex organ systems of the human body. On the one hand, it forms the absorption surface for nutrients, but on the other hand it is also a barrier for toxins, food components and against up to 1014 commensal microorganisms. The complexity of the interplay between absorption, motility and immune functions of the gastrointestinal tract is particularly evident in critically ill patients. In this article, we review the latest updates on pathogenic relationships of motility disorders and diagnostic algorithms in intensive care patients. In addition to established therapies, new developments in the treatment of hypomotility are outlined.
Collapse
Affiliation(s)
- Donata Grajecki
- Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte
| | - Frank Tacke
- Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte
| |
Collapse
|
14
|
Alkaissi HR, Khudyakov A, Belligund P. Acute Colonic Pseudo-Obstruction Following the Use of Dexmedetomidine. Cureus 2021; 13:e19465. [PMID: 34912607 PMCID: PMC8665670 DOI: 10.7759/cureus.19465] [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] [Accepted: 11/10/2021] [Indexed: 12/03/2022] Open
Abstract
Dexmedetomidine is a preferred agent for light sedation with minimal adverse effects. We report a case of acute colonic pseudo-obstruction following dexmedetomidine use in a patient with alcohol withdrawal. He was treated with benzodiazepines first to control the withdrawal symptoms, then escalated to dexmedetomidine once delirium tremens ensued. Later on, the patient developed abdominal distension and vomiting. Imaging showed dilated bowel loops and absence of peristalsis on ultrasound. Decompression with the nasogastric (NG) tube was done, with high output from the NG tube. Dexmedetomidine infusion was used twice, and once it was stopped, the NG tube output was reduced, with the resumption of gastrointestinal motility and improvement of the abdominal distension. Recent similar reports of functional intestinal obstruction following alpha-2 (α2) agonist use necessitate further studies of intestinal motility following dexmedetomidine use and awareness of the possible side effect of dexmedetomidine on intestinal motility.
Collapse
Affiliation(s)
- Hussam R Alkaissi
- Internal Medicine, State University of New York Downstate Medical Center, New York City, USA
| | - Aleksandr Khudyakov
- Pulmonology and Critical Care, Veterans Affairs New York Harbor Health Care (VA NYHHS), Brooklyn, USA
| | - Pooja Belligund
- Pulmonology and Critical Care, Veterans Affairs New York Harbor Health Care (VA NYHHS), Brooklyn, USA
| |
Collapse
|
15
|
Lu Y, Fang PP, Yu YQ, Cheng XQ, Feng XM, Wong GTC, Maze M, Liu XS. Effect of Intraoperative Dexmedetomidine on Recovery of Gastrointestinal Function After Abdominal Surgery in Older Adults: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2128886. [PMID: 34648009 PMCID: PMC8517746 DOI: 10.1001/jamanetworkopen.2021.28886] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE Postoperative ileus is common after abdominal surgery, and small clinical studies have reported that intraoperative administration of dexmedetomidine may be associated with improvements in postoperative gastrointestinal function. However, findings have been inconsistent and study samples have been small. Further examination of the effects of intraoperative dexmedetomidine on postoperative gastrointestinal function is needed. OBJECTIVE To evaluate the effects of intraoperative intravenous dexmedetomidine vs placebo on postoperative gastrointestinal function among older patients undergoing abdominal surgery. DESIGN, SETTING, AND PARTICIPANTS This multicenter, double-blind, placebo-controlled randomized clinical trial was conducted at the First Affiliated Hospital of Anhui Medical University in Hefei, China (lead site), and 12 other tertiary hospitals in Anhui Province, China. A total of 808 participants aged 60 years or older who were scheduled to receive abdominal surgery with an expected surgical duration of 1 to 6 hours were enrolled. The study was conducted from August 21, 2018, to December 9, 2019. INTERVENTIONS Dexmedetomidine infusion (a loading dose of 0.5 μg/kg over 15 minutes followed by a maintenance dose of 0.2 μg/kg per hour) or placebo infusion (normal saline) during surgery. MAIN OUTCOMES AND MEASURES The primary outcome was time to first flatus. Secondary outcomes were postoperative gastrointestinal function measured by the I-FEED (intake, feeling nauseated, emesis, physical examination, and duration of symptoms) scoring system, time to first feces, time to first oral feeding, incidence of delirium, pain scores, sleep quality, postoperative nausea and vomiting, hospital costs, and hospital length of stay. RESULTS Among 808 patients enrolled, 404 were randomized to receive intraoperative dexmedetomidine, and 404 were randomized to receive placebo. In total, 133 patients (60 in the dexmedetomidine group and 73 in the placebo group) were excluded because of protocol deviations, and 675 patients (344 in the dexmedetomidine group and 331 in the placebo group; mean [SD] age, 70.2 [6.1] years; 445 men [65.9%]) were included in the per-protocol analysis. The dexmedetomidine group had a significantly shorter time to first flatus (median, 65 hours [IQR, 48-78 hours] vs 78 hours [62-93 hours], respectively; P < .001), time to first feces (median, 85 hours [IQR, 68-115 hours] vs 98 hours [IQR, 74-121 hours]; P = .001), and hospital length of stay (median, 13 days [IQR, 10-17 days] vs 15 days [IQR, 11-18 days]; P = .005) than the control group. Postoperative gastrointestinal function (as measured by the I-FEED score) and delirium incidence were similar in the dexmedetomidine and control groups (eg, 248 patients [72.1%] vs 254 patients [76.7%], respectively, had I-FEED scores indicating normal postoperative gastrointestinal function; 18 patients [5.2%] vs 12 patients [3.6%] had delirium on postoperative day 3). CONCLUSIONS AND RELEVANCE In this randomized clinical trial, the administration of intraoperative dexmedetomidine reduced the time to first flatus, time to first feces, and length of stay after abdominal surgery. These results suggest that this therapy may be a viable strategy to enhance postoperative recovery of gastrointestinal function among older adults. TRIAL REGISTRATION Chinese Clinical Trial Registry Identifier: ChiCTR1800017232.
Collapse
Affiliation(s)
- Yao Lu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Pan-Pan Fang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yong-Qi Yu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin-Qi Cheng
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao-Mei Feng
- Department of Anesthesiology, University of Utah, Salt Lake City
| | | | - Mervyn Maze
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco
| | - Xue-Sheng Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Li Y, Wang Y, Chang H, Cheng B, Miao J, Li S, Hu H, Huang L, Wang Q. Inhibitory Effects of Dexmedetomidine and Propofol on Gastrointestinal Tract Motility Involving Impaired Enteric Glia Ca 2+ Response in Mice. Neurochem Res 2021; 46:1410-1422. [PMID: 33656693 DOI: 10.1007/s11064-021-03280-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/31/2022]
Abstract
Propofol and dexmedetomidine are popular used for sedation in ICU, however, inadequate attention has been paid to their effect on gastrointestinal tract (GIT) motility. Present study aimed to compare the effect of propofol and dexmedetomidine on GIT motility at parallel level of sedation and explore the possible mechanism. Male C57BL/6 mice (8-10 weeks) were randomly divided into control, propofol and dexmedetomidine group. After intraperitoneal injection of propofol or dexmedetomidine, comparable sedative level was confirmed by sedative score, physiological parameters and electroencephalogram (EEG). Different segments of GIT motility in vivo (gastric emptying, small intestine transit, distal colon bead expulsion, stool weight and number of fecal pellets, gastrointestinal transit and whole gut transit time) and colonic migrating motor complexes (CMMCs) pattern in vitro were evaluated. The Ca2+ response of primary enteric glia was examined under the treatment of propofol or dexmedetomidine. There is little difference in physiological parameters and composite permutation entropy index (CPEI) between administration of 50 mg/kg propofol and 40 μg/kg dexmedetomidine, indicated that parallel level of sedation was reached. Data showed that propofol and dexmedetomidine had significantly inhibitory effect on GIT motility while dexmedetomidine was stronger. Also, the amplitude (ΔF/F0) of Ca2+ response in primary enteric glia was attenuated after treated with the sedatives while the effect of dexmedetomidine was greater than propofol. These findings demonstrated that dexmedetomidine caused stronger inhibitory effects on GIT motility in sedative mice, which may involve impaired Ca2+ response in enteric glia. Hence, dexmedetomidine should be carefully applied especially for potential GIT dysmotility patient.
Collapse
Affiliation(s)
- Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yubo Wang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiwen Miao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liyu Huang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
17
|
Padar M, Starkopf J, Reintam Blaser A. Deepening of sedation with propofol has limited effect on intra-abdominal pressure - An interventional study in mechanically ventilated adult patients with intra-abdominal hypertension. J Crit Care 2021; 65:98-103. [PMID: 34118506 DOI: 10.1016/j.jcrc.2021.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/18/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE To evaluate the effect of deepening of sedation on intra-abdominal pressure (IAP). METHODS 37 adult mechanically ventilated ICU patients with intra-abdominal hypertension received a bolus dose and subsequent infusion of propofol (bolus: 1 mg/kg, infusion: 3 mg/kg/h). IAP, mean arterial pressure (MAP), abdominal perfusion pressure (APP), depth of sedation according to Richmond Agitation-Sedation Scale (RASS), respiratory parameters, and vasopressor dose were assessed after bolus and at 15, 30 and 60 min of infusion of propofol. RESULTS Median IAP at baseline was 15 (13-16) mm Hg. During the intervention, median IAP decreased by 1 mm Hg at all time points. In 24% of patients IAP decreased by ≥3 mm Hg. Compared to baseline, MAP and APP were reduced at all time points. Deepening of sedation per RASS was achieved in 70% of patients at all time points. No changes in respiratory tidal volumes nor plateau pressures were observed. Vasopressor therapy with noradrenaline was started or increased in 43% of patients, whereas the increase in patients already receiving noradrenaline prior to the intervention was not significant. CONCLUSIONS Deepening of sedation with propofol results in a small decrease in IAP and greater simultaneous decrease in MAP and APP, requiring increased vasopressor support in some cases.
Collapse
Affiliation(s)
- Martin Padar
- Department of Anaesthesiology and Intensive Care, Tartu University Hospital, L. Puusepa 8, 51014 Tartu, Estonia; Department of Anaesthesiology and Intensive Care, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia.
| | - Joel Starkopf
- Department of Anaesthesiology and Intensive Care, Tartu University Hospital, L. Puusepa 8, 51014 Tartu, Estonia; Department of Anaesthesiology and Intensive Care, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia
| | - Annika Reintam Blaser
- Department of Anaesthesiology and Intensive Care, University of Tartu, L. Puusepa 8, 51014 Tartu, Estonia; Department of Intensive Care Medicine, Lucerne Cantonal Hospital, 6000 Lucerne, Switzerland
| |
Collapse
|
18
|
Bell CM, Procter LD, Parli SE. Asystole Following Concomitant Intravenous Administration of Neostigmine and Dexmedetomidine in a Patient With Acute Colonic Pseudo-Obstruction. J Pharm Pract 2021; 35:650-653. [PMID: 33739166 DOI: 10.1177/08971900211001278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Acute colonic pseudo-obstruction (ACPO) is a condition characterized by acute dilation of the large bowel without evidence of mechanical obstruction that occurs in a variety of hospitalized patients with many predisposing factors. Management includes supportive care and limitation of offending medications with mainstays of treatment of neostigmine administration and colonic decompression. We report the case of a critically ill patient with ACPO who experienced bradycardia and a brief episode of asystole when receiving concomitant dexmedetomidine and neostigmine infusions but who later remained hemodynamically stable when receiving propofol and neostigmine infusions. The bradycardia and associated hemodynamic instability experienced while on dexmedetomidine and neostigmine infusions were rapidly corrected with atropine and cessation of offending agents. Because ACPO is encountered frequently and the use of dexmedetomidine as a sedative agent in the ICU is increasing, practitioners should be aware of the additive risk of bradycardia and potential for asystole with the combination of neostigmine and dexmedetomidine. Electronic drug interaction databases should be updated and drug information sources should include a drug-drug interaction between dexmedetomidine and neostigmine to reduce the likelihood of concomitant administration.
Collapse
Affiliation(s)
- Carolyn Magee Bell
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, SC, USA
| | - Levi D Procter
- Department of General Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Sara E Parli
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, KY, USA.,Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
| |
Collapse
|
19
|
Zhao W, Hu Y, Chen H, Wang X, Wang L, Wang Y, Wu X, Han F. The Effect and Optimal Dosage of Dexmedetomidine Plus Sufentanil for Postoperative Analgesia in Elderly Patients With Postoperative Delirium and Early Postoperative Cognitive Dysfunction: A Single-Center, Prospective, Randomized, Double-Blind, Controlled Trial. Front Neurosci 2020; 14:549516. [PMID: 33192244 PMCID: PMC7645155 DOI: 10.3389/fnins.2020.549516] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
Background Postoperative delirium (POD) and postoperative cognitive dysfunction (POCD) are common complications after major surgery among elderly patients. Dexmedetomidine (DEX) is less frequently explored for its effects in patients with postoperative neurocognitive disorders. This study investigated the effect and optimal dosage of DEX for patient-controlled analgesia (PCA) on POD and early POCD after major surgery among elderly patients. Methods Patients in four groups received continuous infusion of DEX 0, 100, 200, and 400 μg with sufentanil 150 μg for PCA immediately after surgery. POD and POCD were assessed on postoperative days 1, 2, 3, and 7 by using the Confusion Assessment Method (CAM) and Mini-Mental State Examination (MMSE) scales. Furthermore, the incidence of POD and POCD of all the four groups in postoperative 7 days classified by high risk factors (age, education, surgical site, and surgical category), sedation level, postoperative pain intensity, and side effects were assessed. Results The overall incidence rates of POD and early POCD 7 days after surgery were lower in the DEX 200 μg 400 μg groups than in the DEX 0 μg and 100 μg groups (P < 0.05). Compared with DEX 200 μg, DEX 400 μg reduced early POCD in patients who underwent open surgery (P < 0.05). There were no intergroup differences in the postoperative sedation level, pain intensity, and side effects. Conclusion The continuous infusion of DEX 200 μg or DEX 400 μg in PCA significantly decreased the incidence of POD and early POCD after major surgery without increasing any side effects. Compared with DEX 200 μg, DEX 400 μg was preferred for reducing early POCD in patients who underwent open surgery.
Collapse
Affiliation(s)
- Wenshuai Zhao
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanan Hu
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hui Chen
- Department of Anesthesiology, Heilongjiang Provincial Corps Hospital, Chinese People's Armed Police Forces, Harbin, China
| | - Xifan Wang
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Liping Wang
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Wang
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaohong Wu
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fei Han
- Department of Anesthesiology, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
Chang H, Li S, Li Y, Hu H, Cheng B, Miao J, Gao H, Ma H, Gao Y, Wang Q. Effect of sedation with dexmedetomidine or propofol on gastrointestinal motility in lipopolysaccharide-induced endotoxemic mice. BMC Anesthesiol 2020; 20:227. [PMID: 32894042 PMCID: PMC7487735 DOI: 10.1186/s12871-020-01146-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Background Sepsis often accompanies gastrointestinal motility disorder that contributes to the development of sepsis in turn. Propofol and dexmedetomidine, as widely used sedatives in patients with sepsis, are likely to depress gastrointestinal peristalsis. We queried whether propofol or dexmedetomidine, at sedative doses, aggravated sepsis-induced ileus. Methods Sedative/Anesthetic Scores and vital signs of lipopolysaccharide (LPS)-induced endotoxemic mice were measured during sedation with propofol or dexmedetomidine. Endotoxemic mice were divided into 10% fat emulsion, propofol, saline, and dexmedetomidine group. The gastric emptying, small intestinal transit, tests of colonic motility, gastrointestinal transit and whole gut transit were evaluated at 15 mins and 24 h after intraperitoneal injection of sedatives/vehicles respectively. Results 40 mg·kg− 1propofol and 80 μg·kg− 1 dexmedetomidine induced a similar depth of sedation with comparable vital signs except that dexmedetomidine strikingly decreased heart rate in endotoxemic mice. Dexmedetomidine markedly inhibited gastric emptying (P = 0.006), small intestinal transit (P = 0.006), colonic transit (P = 0.0006), gastrointestinal transit (P = 0.0001) and the whole gut transit (P = 0.034) compared with the vehicle, whereas propofol showed no depression on all parts of gastrointestinal motility 15 mins after administration. The inhibitive effects of dexmedetomidine in these tests vanished 24 h after the administration. Conclusions Deep sedation with dexmedetomidine, but not propofol, significantly inhibited gastrointestinal peristalsis in endotoxemic mice while the inhibitory effect disappeared 24 h after sedation. These data suggested that both propofol and dexmedetomidine could be applied in septic patients while dexmedetomidine should be used cautiously in patients with cardiac disease or ileus.
Collapse
Affiliation(s)
- Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiwen Miao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hui Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hongli Ma
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yanfeng Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
21
|
Huuskonen V, Restitutti F, Honkavaara JM, Raekallio MR, Männikkö S, Scheinin M, Vainio OM. Investigation of the effects of vatinoxan on somatic and visceral antinociceptive efficacy of medetomidine in dogs. Am J Vet Res 2020; 81:299-308. [PMID: 32228259 DOI: 10.2460/ajvr.81.4.299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine whether concurrent vatinoxan administration affects the antinociceptive efficacy of medetomidine in dogs at doses that provide circulating dexmedetomidine concentrations similar to those produced by medetomidine alone. ANIMALS 8 healthy Beagles. PROCEDURES Dogs received 3 IV treatments in a randomized crossover-design trial with a 2-week washout period between experiments (medetomidine [20 μg/kg], medetomidine [20 μg/kg] and vatinoxan [400 μg/kg], and medetomidine [40 μg/kg] and vatinoxan [800 μg/kg]; M20, M20V400, and M40V800, respectively). Sedation, visceral and somatic nociception, and plasma drug concentrations were assessed. Somatic and visceral nociception measurements and sedation scores were compared among treatments and over time. Sedation, visceral antinociception, and somatic antinociception effects of M20V400 and M40V800 were analyzed for noninferiority to effects of M20, and plasma drug concentration data were assessed for equivalence between treatments. RESULTS Plasma dexmedetomidine concentrations after administration of M20 and M40V800 were equivalent. Sedation scores, visceral nociception measurements, and somatic nociception measurements did not differ significantly among treatments within time points. Overall sedative effects of M20V400 and M40V800 and visceral antinociceptive effects of M40V800 were noninferior to those produced by M20. Somatic antinociception effects of M20V400 at 10 minutes and M40V800 at 10 and 55 minutes after injection were noninferior to those produced by M20. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested coadministration with vatinoxan did not substantially diminish visceral antinociceptive effects of medetomidine when plasma dexmedetomidine concentrations were equivalent to those produced by medetomidine alone. For somatic antinociception, noninferiority of treatments was detected at some time points.
Collapse
|
22
|
Organ-Protective Effects and the Underlying Mechanism of Dexmedetomidine. Mediators Inflamm 2020; 2020:6136105. [PMID: 32454792 PMCID: PMC7232715 DOI: 10.1155/2020/6136105] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective α2 adrenergic receptor (α2AR) agonist currently used in clinical settings. Because DEX has dose-dependent advantages of sedation, analgesia, antianxiety, inhibition of sympathetic nervous system activity, cardiovascular stabilization, and significant reduction of postoperative delirium and agitation, but does not produce respiratory depression and agitation, it is widely used in clinical anesthesia and ICU departments. In recent years, much clinical study and basic research has confirmed that DEX has a protective effect on a variety of organs, including the nervous system, heart, lungs, kidneys, liver, and small intestine. It acts by reducing the inflammatory response in these organs, activating antiapoptotic signaling pathways which protect cells from damage. Therefore, based on wide clinical application and safety, DEX may become a promising clinical multiorgan protection drug in the future. In this article, we review the physiological effects related to organ protection in α2AR agonists along with the organ-protective effects and mechanisms of DEX to understand their combined application value.
Collapse
|
23
|
Li M, Wang T, Xiao W, Zhao L, Yao D. Low-Dose Dexmedetomidine Accelerates Gastrointestinal Function Recovery in Patients Undergoing Lumbar Spinal Fusion. Front Pharmacol 2020; 10:1509. [PMID: 31920678 PMCID: PMC6930906 DOI: 10.3389/fphar.2019.01509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Dexmedetomidine possesses sedative, sympatholytic, and opioid-sparing properties, but its impact on postoperative gastrointestinal function is controversial. Methods: This single-center, prospective, randomized study compared low-dose dexmedetomidine and placebo on gastrointestinal function recovery and inflammation after posterior lumbar spinal fusion. Sixty-six patients were randomized into two groups and received normal saline (control group) or dexmedetomidine (DEX group) during posterior lumbar fusion. Blood was taken at five timepoints to measure lipopolysaccharides, tumor necrosis factor-α, and C-reactive protein. The primary outcome was duration to first flatus. The secondary outcomes were inflammatory mediators and determination of correlations between perioperative factors and duration to first flatus. Results: Patients in DEX group showed significantly lower duration to first flatus (15.37 [13.35-17.38] vs 19.58 [17.31-21.86] h; p = 0.006) and overall sufentanil consumption (67.19 [63.78-70.62] vs 74.67 [69.96-79.30] μg; p = 0.011) than controls. Lipopolysaccharides, tumor necrosis factor-α, and C-reactive protein did not differ between the groups at any timepoint (all p > 0.05). Multiple linear regression modeling assessed the ability of independent variables to predict variance in duration to first flatus (adjusted R2 = 0.379, p = 0.000). In the model, age (β = 0.243, p = 0.003), gender (β = -3.718, p = 0.011), BMI (β = -0.913, p = 0.001), operative segments (β = -4.079, p = 0.028), and overall sufentanil consumption (β = 0.426, p = 0.000) contributed significantly. Conclusions: Thus, low-dose dexmedetomidine accelerates gastrointestinal function recovery after lumbar spinal fusion. The effect may be partially produced by opioid-sparing effects rather than inhibition of inflammation. Clinical Trial Registration: www.chictr.org.cn, identifier ChiCTR1800018127.
Collapse
Affiliation(s)
- Meng Li
- Department of Anesthesiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Tianlong Wang
- Department of Anesthesiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Wei Xiao
- Department of Anesthesiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Lei Zhao
- Department of Anesthesiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Dongxu Yao
- Department of Anesthesiology, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
24
|
Pérez-Lara JL, Santana Y, Hernández-Torres J, Díaz-Fuentes G. Acute Colonic Pseudo-Obstruction Caused by Dexmedetomidine: A Case Report and Literature Review. AMERICAN JOURNAL OF CASE REPORTS 2019; 20:278-284. [PMID: 30826812 PMCID: PMC6410604 DOI: 10.12659/ajcr.913645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Patient: Male, 50 Final Diagnosis: Acute colonic pseudo-obstruction Symptoms: Abdominal pain • cough • fever Medication: — Clinical Procedure: Colonoscopy decompression and colectomy Specialty: Critical Care Medicine
Collapse
Affiliation(s)
- Jose L Pérez-Lara
- Department of Internal Medicine, BronxCare Health System, Bronx, NY, USA.,Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Yaneidy Santana
- Department of Internal Medicine, BronxCare Health System, Bronx, NY, USA.,Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Janette Hernández-Torres
- Department of Internal Medicine, BronxCare Health System, Bronx, NY, USA.,Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gilda Díaz-Fuentes
- Department of Internal Medicine, BronxCare Health System, Bronx, NY, USA.,Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| |
Collapse
|
25
|
Wan Q, Ding W, Cui X, Zeng X. CONSORT-epidural dexmedetomidine improves gastrointestinal motility after laparoscopic colonic resection compared with morphine. Medicine (Baltimore) 2018; 97:e11218. [PMID: 29924051 PMCID: PMC6024965 DOI: 10.1097/md.0000000000011218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND We have previously shown that epidural dexmedetomidine, when used as an adjunct to levobupivacaine for control of postoperative pain after open colonic resection, improves recovery of gastrointestinal motility compared with morphine. METHODS Sixty patients undergoing laparoscopic colonic resection were enrolled and allocated randomly to treatment with dexmedetomidine (group D) or morphine (group M). Group D received an epidural loading dose of dexmedetomidine (5 mL, 0.5 μg/kg), followed by continuous epidural administration of dexmedetomidine (80 μg) in 0.125% levobupivacaine (240 mL) at a rate of 5 mL/h for 2 days. Group M received an epidural loading dose of morphine (5 mL, 0.03 mg/kg) followed by continuous epidural administration of morphine (4.5 mg) in 0.125% levobupivacaine (240 mL) at a rate of 5 mL/h for 2 days. Verbal rating score (VRS) of pain, postoperative analgesic requirements, side effects related to analgesia, and time to postoperative first flatus (FFL) and first feces (FFE) were recorded. RESULTS VRS and postoperative analgesic requirements were not significantly different between the treatment groups. In contrast, FFL and FFE were significant delayed in group M compared with group D (P < .05). Patients in group M also had a significantly higher incidence of nausea, vomiting, and pruritus (P < .05). No neurological deficits were observed in either group. CONCLUSIONS Compared with morphine, epidural dexmedetomidine is a better adjunct to levobupivacaine for control of postoperative pain after laparoscopic colonic resection.
Collapse
|
26
|
de Vries A, Pakkanen SAE, Raekallio MR, Ekiri A, Scheinin M, Taylor PM, Vainio OM. Clinical effects and pharmacokinetic variables of romifidine and the peripheral α 2 ‐adrenoceptor antagonist MK‐467 in horses. Vet Anaesth Analg 2016; 43:599-610. [DOI: 10.1111/vaa.12354] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/24/2015] [Indexed: 11/29/2022]
|
27
|
Chen C, Huang P, Lai L, Luo C, Ge M, Hei Z, Zhu Q, Zhou S. Dexmedetomidine improves gastrointestinal motility after laparoscopic resection of colorectal cancer: A randomized clinical trial. Medicine (Baltimore) 2016; 95:e4295. [PMID: 27442674 PMCID: PMC5265791 DOI: 10.1097/md.0000000000004295] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND To investigate the effects of intraoperative application of dexmedetomidine (Dex) on early gastrointestinal motility after laparoscopic resection of colorectal cancer. METHODS In this prospective, randomized double-blind investigation, 60 patients who underwent laparoscopic resection of colorectal cancer were randomly allocated to receive Dex (DEX group, n = 30) or saline (CON group, n = 30). In the DEX group, Dex was loaded (1 μg/kg) before anesthesia induction and was infused (0.3 μg/kg/h) during surgery. Time to postoperative first flatus (FFL) and first feces (FFE), and time to regular diet were recorded. Serum diamine oxidase (DAO) activity and intestinal fatty acid-binding protein (I-FABP) were detected. RESULTS Both the time to the FFL (44.41 ± 4.51 hours vs 61.03 ± 5.16 hours, P = 0.02) and the time to the FFE (60.67 ± 4.94 hours vs 82.50 ± 6.88 hours, P = 0.014) were significantly shorter in the DEX group than the CON group. Furthermore, the time to regular diet of the DEX group was shorter than that of the CON group (76.15 ± 4.11 hours vs 91.50 ± 5.70 hours, P = 0.037). Both DAO and I-FABP increased significantly from beginning of surgery to postoperative day 1 in the CON group (2.49 ± 0.41 ng/mL vs 4.48 ± 0.94 ng/mL for DAO, P = 0.028, 1.32 ± 0.09 ng/mL vs 2.17 ± 0.12 ng/mL for I-FABP, P = 0.045, respectively), whereas no significant change was observed in the DEX group. Furthermore, patients in the DEX group had stable hemodynamics and shorter hospital stay than those in the CON group. CONCLUSION Dex administration intraoperatively benefits recovery of gastrointestinal motility function after laparoscopic resection of colorectal cancer with stable hemodynamics during surgery though further studies are needed to explore the mechanisms of Dex on gastrointestinal motility.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qianqian Zhu
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China
- Correspondence: Shaoli Zhou and Qianqian Zhu, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China (e-mails: ; )
| | - Shaoli Zhou
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China
- Correspondence: Shaoli Zhou and Qianqian Zhu, Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, People's Republic of China (e-mails: ; )
| |
Collapse
|
28
|
Whitehead K, Cortes Y, Eirmann L. Gastrointestinal dysmotility disorders in critically ill dogs and cats. J Vet Emerg Crit Care (San Antonio) 2016; 26:234-53. [PMID: 26822390 DOI: 10.1111/vec.12449] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 07/21/2015] [Accepted: 08/30/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To review the human and veterinary literature regarding gastrointestinal (GI) dysmotility disorders in respect to pathogenesis, patient risk factors, and treatment options in critically ill dogs and cats. ETIOLOGY GI dysmotility is a common sequela of critical illness in people and small animals. The most common GI motility disorders in critically ill people and small animals include esophageal dysmotility, delayed gastric emptying, functional intestinal obstruction (ie, ileus), and colonic motility abnormalities. Medical conditions associated with the highest risk of GI dysmotility include mechanical ventilation, sepsis, shock, trauma, systemic inflammatory response syndrome, and multiple organ failure. The incidence and pathophysiology of GI dysmotility in critically ill small animals is incompletely understood. DIAGNOSIS A presumptive diagnosis of GI dysmotility is often made in high-risk patient populations following detection of persistent regurgitation, vomiting, lack of tolerance of enteral nutrition, abdominal pain, and constipation. Definitive diagnosis is established via radioscintigraphy; however, this diagnostic tool is not readily available and is difficult to perform on small animals. Other diagnostic modalities that have been evaluated include abdominal ultrasonography, radiographic contrast, and tracer studies. THERAPY Therapy is centered at optimizing GI perfusion, enhancement of GI motility, and early enteral nutrition. Pharmacological interventions are instituted to promote gastric emptying and effective intestinal motility and prevention of complications. Promotility agents, including ranitidine/nizatidine, metoclopramide, erythromycin, and cisapride are the mainstays of therapy in small animals. PROGNOSIS The development of complications related to GI dysmotility (eg, gastroesophageal reflux and aspiration) have been associated with increased mortality risk. Institution of prophylaxic therapy is recommended in high-risk patients, however, no consensus exists regarding optimal timing of initiating prophylaxic measures, preference of treatment, or duration of therapy. The prognosis for affected small animal patients remains unknown.
Collapse
Affiliation(s)
- KimMi Whitehead
- Emergency and Critical Care Department, Oradell Animal Hospital, Paramus, NJ, 07452
| | - Yonaira Cortes
- Emergency and Critical Care Department, Oradell Animal Hospital, Paramus, NJ, 07452
| | - Laura Eirmann
- the Nutrition Department (Eirmann), Oradell Animal Hospital, Paramus, NJ, 07452
| |
Collapse
|
29
|
Zeng XZ, Lu ZF, Lv XQ, Guo YP, Cui XG. Epidural Co-Administration of Dexmedetomidine and Levobupivacaine Improves the Gastrointestinal Motility Function after Colonic Resection in Comparison to Co-Administration of Morphine and Levobupivacaine. PLoS One 2016; 11:e0146215. [PMID: 26751791 PMCID: PMC4709108 DOI: 10.1371/journal.pone.0146215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/14/2015] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Gastrointestinal motility may be impaired after intestinal surgery. Epidural morphine is effective in controlling postoperative pain, but can further reduce gastrointestinal motility. Here, we aimed to investigate the effects of epidural dexmedetomidine on gastrointestinal motility in patients undergoing colonic resection. Seventy-four patients undergoing colonic resection were enrolled in this clinical trial and allocated randomly to treatment with dexmedetomidine (D group) or morphine (M group). The D group received a loading dose epidural administration of 3 ml dexmedetomidine (0.5 μg kg(-1)) and then a continuous epidural administration of 80 μg dexmedetomidine in 150 ml levobupivacaine (0.125%) at 3 ml h(-1) for two days. The M group received a loading dose epidural administration of 3 ml morphine (0.03 mg kg(-1)) and then a continuous epidural administration of 4.5 mg morphine in 150 ml levobupivacaine at 3 ml h(-1) for two days. Verbal rating score (VRS), postoperative analgesic requirements, side effects related to analgesia, the time to postoperative first flatus (FFL) and first feces (FFE) were recorded. VRS and postoperative analgesic requirements were not significantly different between treatment groups. In contrast, the time to FFL and time to FFE were significant longer in M group in comparison to D group (P < 0.05). Moreover, patients in M group had a significantly higher incidence of nausea, vomiting, and pruritus (P < 0.05). No patients showed neurologic deficits in either group. In comparison to morphine, epidural dexmedetomidine is safe and beneficial for the recovery of gastrointestinal motility after colonic resection when used as an adjunct with levobupivacaine for postoperative pain control. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR-TRC-14004644.
Collapse
Affiliation(s)
- Xian-Zhang Zeng
- Department of Anesthesiology, Second Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, China
| | - Zhi-Fang Lu
- Department of Anesthesiology, Second Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, China
| | - Xiang-Qi Lv
- Department of Anesthesiology, Second Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, China
| | - Yue-Ping Guo
- Department of Anesthesiology, Second Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, China
| | - Xiao-Guang Cui
- Department of Anesthesiology, Second Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, China
| |
Collapse
|
30
|
Abstract
OBJECTIVE We aimed to review gastric dysmotility in critically ill children: 1) its pathophysiology, with a focus on critical care diseases and therapies that affect gastric motility, 2) diagnostic methodologies, and 3) current and future potential therapies. DATA SOURCES Eligible studies were identified from PubMed and MEDLINE. STUDY SELECTION Literature search included the following key terms: "gastric emptying," "gastric motility/dysmotility," "gastrointestinal motility/dysmotility," "nutrition intolerance," and "gastric residual volume." DATA EXTRACTION Studies since 1995 were extracted and reviewed for inclusion by the authors related to the physiology, pathophysiology, diagnostic methodologies, and available therapies for gastric emptying. DATA SYNTHESIS Delayed gastric emptying, a common presentation of gastric dysmotility, is present in up to 50% of critically ill children. It is associated with the potential for aspiration, ventilator-associated pneumonia, and inadequate delivery of enteral nutrition and may affect the efficacy of enteral medications, all of which may be result in poor patient outcomes. Gastric motility is affected by critical illness and its associated therapies. Currently available diagnostic tools to identify gastric emptying at the bedside have not been systematically studied and applied in this cohort. Gastric residual volume measurement, used as an indirect marker of delayed gastric emptying in PICUs around the world, may be inaccurate. CONCLUSIONS Gastric dysmotility is common in critically ill children and impacts patient safety and outcomes. However, it is poorly understood, inadequately defined, and current therapies are limited and based on scant evidence. Understanding gastric motility and developing accurate bedside measures and novel therapies for gastric emptying are highly desirable and need to be further investigated.
Collapse
|
31
|
Kanaeda T, Ueda M, Arai M, Ishimura M, Kajiyama T, Hashiguchi N, Nakano M, Kondo Y, Hiranuma Y, Oyamada A, Yokosuka O, Kobayashi Y. Evaluation of periesophageal nerve injury after pulmonary vein isolation using the (13)C-acetate breath test. J Arrhythm 2015; 31:364-70. [PMID: 26702316 PMCID: PMC4672080 DOI: 10.1016/j.joa.2015.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/25/2015] [Accepted: 06/08/2015] [Indexed: 01/30/2023] Open
Abstract
Background Pulmonary vein isolation (PVI) has become an important option for treating patients with atrial fibrillation (AF). Periesophageal nerve (PEN) injury after PVI causes pyloric spasms and gastric hypomotility. This study aimed to clarify the impact of PVI on gastric motility and assess the prevalence of gastric hypomotility after PVI. Methods Thirty consecutive patients with AF underwent PVI under luminal esophageal temperature (LET) monitoring. The 13C-acetate breath test was conducted before and after the procedure for all patients (PVI group). Gastric emptying was evaluated using the time to peak concentration of 13CO2 (Tmax). This test was also conducted in another 20 patients who underwent catheter ablation procedures other than PVI (control group). Results The number of patients with abnormal Tmax (≥75 min) increased from seven (23%) to 13 (43%) and from three (15%) to five (25%) after the procedure in the PVI group and control group, respectively. The mean Tmax was longer after PVI than before PVI (64±14 min vs. 57±15 min, p=0.006), whereas there was no significant difference before and after the procedure in the control group. However, no significant difference in ΔTmax was observed between the two groups (p=0.27). No patients suffered from symptomatic gastric hypomotility. Conclusions Asymptomatic gastric hypomotility occurred more often after PVI. However, the average impact of PVI on gastric motility was minimal.
Collapse
Affiliation(s)
- Tomonori Kanaeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Marehiko Ueda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makoto Arai
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masayuki Ishimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takatsugu Kajiyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naotaka Hashiguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masahiro Nakano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yusuke Kondo
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yasunori Hiranuma
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Arata Oyamada
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
32
|
Kim N, Yoo YC, Lee SK, Kim H, Ju HM, Min KT. Comparison of the efficacy and safety of sedation between dexmedetomidine-remifentanil and propofol-remifentanil during endoscopic submucosal dissection. World J Gastroenterol 2015; 21:3671-3678. [PMID: 25834336 PMCID: PMC4375593 DOI: 10.3748/wjg.v21.i12.3671] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/16/2014] [Accepted: 01/08/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare the efficacy and safety of sedation protocols for endoscopic submucosal dissection (ESD) between dexmedetomidine-remifentanil and propofol-remifentanil.
METHODS: Fifty-nine patients scheduled for ESD were randomly allocated into a dexmedetomidine-remifentanil (DR) group or a propofol-remifentanil (PR) group. To control patient anxiety, dexmedetomidine or propofol was infused to maintain a score of 4-5 on the Modified Observer’s Assessment of Alertness/Sedation scale. Remifentanil was infused continuously at a rate of 6 μg/kg per hour in both groups. The ease of advancing the scope into the throat, gastric motility grading, and satisfaction of the endoscopist and patient were assessed. Hemodynamic variables and hypoxemic events were compared to evaluate patient safety.
RESULTS: Demographic data were comparable between the groups. The hemodynamic variables and pulse oximetry values were stable during the procedure in both groups despite a lower heart rate in the DR group. No oxygen desaturation events occurred in either group. Although advancing the scope into the throat was easier in the PR group (“very easy” 24.1% vs 56.7%, P = 0.010), gastric motility was more suppressed in the DR group (“no + mild” 96.6% vs 73.3%, P = 0.013). The endoscopists felt that the procedure was more favorable in the DR group (“very good + good” 100% vs 86.7%, P = 0.042), whereas patient satisfaction scores were comparable between the groups. En bloc resection was performed 100% of the time in both groups, and the complete resection rate was 94.4% in the DR group and 100% in the PR group (P = 0.477).
CONCLUSION: The efficacy and safety of dexmedetomidine and remifentanil were comparable to propofol and remifentanil during ESD. However, the endoscopists favored dexmedetomidine perhaps due to lower gastric motility.
Collapse
|
33
|
Dexmedetomidine sedation after cardiac surgery decreases atrial arrhythmias. J Clin Anesth 2014; 26:634-42. [DOI: 10.1016/j.jclinane.2014.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 11/23/2022]
|
34
|
Hillyard S, Cowman S, Ramasundaram R, Seed P, O'Sullivan G. Does adding milk to tea delay gastric emptying? Br J Anaesth 2014; 112:66-71. [DOI: 10.1093/bja/aet261] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
35
|
Shimizu S, Akiyama T, Kawada T, Kamiya A, Turner MJ, Yamamoto H, Shishido T, Shirai M, Sugimachi M. Medetomidine Suppresses Cardiac and Gastric Sympathetic Nerve Activities but Selectively Activates Cardiac Vagus Nerve. Circ J 2014; 78:1405-13. [DOI: 10.1253/circj.cj-13-1456] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Shuji Shimizu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| | - Tsuyoshi Akiyama
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center
| | - Toru Kawada
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| | - Atsunori Kamiya
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| | - Michael James Turner
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| | - Hiromi Yamamoto
- Division of Cardiology, Department of Medicine, Faculty of Medicine, Kinki University
| | - Toshiaki Shishido
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| | - Mikiyasu Shirai
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center
| | - Masaru Sugimachi
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center
| |
Collapse
|
36
|
Abstract
Dexmedetomidine is a highly selective, potent α₂-adrenoceptor agonist which was approved in 2011 by the European Medicines Agency for sedation of patients in intensive care units (ICU). Dexmedetomidine exhibits sedative as well as analgesic and anxiolytic effects. Recent studies suggest that dexmedetomidine may be an alternative to midazolam in long-term ICU sedation. This review summarizes the pharmacokinetics and pharmacodynamics of dexmedetomidine particularly in ICU patients and with special regard to covariate effects. Although dexmedetomidine is currently approved only for use in adults the pharmacokinetics and pharmacodynamics in children will also be addressed as there are numerous studies on this off-label use.
Collapse
Affiliation(s)
- H Ihmsen
- Anästhesiologische Klinik, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 12, 91054 Erlangen, Deutschland.
| | | |
Collapse
|
37
|
Romera Ortega MA, Chamorro Jambrina C, Lipperheide Vallhonrat I, Fernández Simón I. [Indications of dexmedetomidine in the current sedoanalgesia tendencies in critical patients]. Med Intensiva 2013; 38:41-8. [PMID: 23683866 DOI: 10.1016/j.medin.2013.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/08/2013] [Accepted: 03/12/2013] [Indexed: 11/20/2022]
Abstract
Recently, dexmedetomidine has been marketed in Spain and other European countries. The published experience regarding its use has placed dexmedetomidine on current trends in sedo-analgesic strategies in the adult critically ill patient. Dexmedetomidine has sedative and analgesic properties, without respiratory depressant effects, inducing a degree of depth of sedation in which the patient can open its eyes to verbal stimulation, obey simple commands and cooperate in nursing care. It is therefore a very useful drug in patients who can be maintained on mechanical ventilation with these levels of sedation avoiding the deleterious effects of over or infrasedation. Because of its effects on α2-receptors, it's very useful for the control and prevention of tolerance and withdrawal to other sedatives and psychotropic drugs. The use of dexmedetomidine has been associated with lower incidence of delirium when compared with other sedatives. Moreover, it's a potentially useful drug for sedation of patients in non-invasive ventilation.
Collapse
Affiliation(s)
- M A Romera Ortega
- Servicio de Medicina Intensiva, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, España
| | - C Chamorro Jambrina
- Servicio de Medicina Intensiva, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, España.
| | - I Lipperheide Vallhonrat
- Servicio de Medicina Intensiva, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, España
| | - I Fernández Simón
- Servicio de Medicina Intensiva, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, España
| |
Collapse
|
38
|
Vainionpää MH, Raekallio MR, Pakkanen SAE, Ranta-Panula V, Rinne VM, Scheinin M, Vainio OM. Plasma drug concentrations and clinical effects of a peripheral alpha-2-adrenoceptor antagonist, MK-467, in horses sedated with detomidine. Vet Anaesth Analg 2013; 40:257-64. [DOI: 10.1111/vaa.12012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 10/13/2012] [Indexed: 12/12/2022]
|
39
|
Pichot C, Longrois D, Ghignone M, Quintin L. [Dexmedetomidine and clonidine: a review of their pharmacodynamy to define their role for sedation in intensive care patients]. ACTA ACUST UNITED AC 2012; 31:876-96. [PMID: 23089375 DOI: 10.1016/j.annfar.2012.07.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 07/12/2012] [Indexed: 11/29/2022]
Abstract
Alpha-2 adrenergic agonists ("alpha-2 agonists") present multiple pharmacodynamic effects: rousable sedation, decreased incidence of delirium in the setting of critical care, preservation of respiratory drive, decreased whole body oxygen consumption, decreased systemic and pulmonary arterial impedance, improved left ventricular systolic and diastolic function, preserved vascular reactivity to exogenous catecholamines, preserved vasomotor baroreflex with lowered set point, preserved kidney function, decreased protein catabolism. These pharmacodynamic effects explain the interest for these drugs in the critical care setting. However, their exact role for sedation in critically ill-patients remains open for further studies. Given the few double-blind randomized multicentric trials available, the present non exhaustive analysis of the literature aims at presenting the utilization of alpha-2 agonists as potential first-line sedative agents, in the critical care setting. Suggestions regarding the use of alpha-2 agonists as sedatives are detailed.
Collapse
Affiliation(s)
- C Pichot
- Physiologie (EA 4612 : neurocardiologie), campus de la Doua, université de Lyon, 8, rue R.-Dubois, 69622 Lyon-Villeurbanne, France
| | | | | | | |
Collapse
|
40
|
Hoy SM, Keating GM. Dexmedetomidine: a review of its use for sedation in mechanically ventilated patients in an intensive care setting and for procedural sedation. Drugs 2011; 71:1481-501. [PMID: 21812509 DOI: 10.2165/11207190-000000000-00000] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dexmedetomidine (Precedex®), a pharmacologically active dextroisomer of medetomidine, is a selective α(2)-adrenergic receptor agonist. It is indicated in the US for the sedation of mechanically ventilated adult patients in an intensive care setting and in non-intubated adult patients prior to and/or during surgical and other procedures. This article reviews the pharmacological properties, therapeutic efficacy and tolerability of dexmedetomidine in randomized, double-blind, placebo-controlled, multicentre studies in these indications. Post-surgical patients in an intensive care setting receiving dexmedetomidine required less rescue sedation with intravenous propofol or intravenous midazolam to achieve and/or maintain optimal sedation during the assisted ventilation period than placebo recipients, according to two randomized, double-blind, multinational studies. Moreover, significantly more dexmedetomidine than placebo recipients acquired and/or maintained optimal sedation without rescue sedation. Sedation with dexmedetomidine was also effective in terms of the total dose of morphine administered, with dexmedetomidine recipients requiring less morphine than placebo recipients; with regard to patient management, dexmedetomidine recipients were calmer and easier to arouse and manage than placebo recipients. Intravenous dexmedetomidine was effective as a primary sedative in two randomized, double-blind, placebo-controlled, multicentre studies in adult patients undergoing awake fibre-optic intubation or a variety of diagnostic or surgical procedures requiring monitored anaesthesia care. In one study, significantly fewer dexmedetomidine than placebo recipients required rescue sedation with intravenous midazolam to achieve and/or maintain optimal sedation; conversely, in another study, rescue sedation with intravenous midazolam was not required by significantly more dexmedetomidine than placebo recipients. Primary sedation with intravenous dexmedetomidine was also effective in terms of the secondary efficacy endpoints, including the mean total dose of midazolam and fentanyl administered and the percentage of patients requiring further sedation (in addition to dexmedetomidine or placebo and midazolam), with, for the most part, significant between-group differences observed in favour of dexmedetomidine over placebo. In general, no significant differences were observed between the dexmedetomidine and placebo treatment groups in the anaesthesiologists' assessment of ease of intubation, haemodynamic stability, patient cooperation and/or respiratory stability. Intravenous dexmedetomidine is generally well tolerated when utilized in mechanically ventilated patients in an intensive care setting and for procedural sedation in non-intubated patients. Dexmedetomidine is associated with a lower rate of postoperative delirium than midazolam or propofol; it is not associated with respiratory depression. While dexmedetomidine is associated with hypotension and bradycardia, both usually resolve without intervention. Thus, intravenous dexmedetomidine provides a further option as a short-term (<24 hours) primary sedative in mechanically ventilated adult patients in an intensive care setting and in non-intubated adult patients prior to and/or during surgical and other procedures.
Collapse
|
41
|
Affiliation(s)
- Jong Wha Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ki-Young Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|