AnesthesiologyPub Date : 2025-03-10DOI: 10.1097/ALN.0000000000005378
Yuriy S Bronshteyn, Diana Hsu, Ashley Vincent, Sophia Dunworth
{"title":"Discordance between Chest Radiography and Lung Ultrasound in the Evaluation of Intraoperative Hypoxemia.","authors":"Yuriy S Bronshteyn, Diana Hsu, Ashley Vincent, Sophia Dunworth","doi":"10.1097/ALN.0000000000005378","DOIUrl":"https://doi.org/10.1097/ALN.0000000000005378","url":null,"abstract":"","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-03-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143584390","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-07DOI: 10.1097/ALN.0000000000005336
Martina Baiardo Redaelli, Fabrizio Monaco, Nikola Bradic, Anna Mara Scandroglio, Lian Kah Ti, Alessandro Belletti, Cristina Viscido, Margherita Licheri, Fabio Guarracino, Alessandro Pruna, Antonio Pisano, Domenico Pontillo, Francesco Federici, Rosario Losiggio, Giovanni Serena, Enrico Tomasi, Simona Silvetti, Marco Ranucci, Luca Brazzi, Andrea Cortegiani, Giovanni Landoni, Pasquale Mastroroberto, Gianluca Paternoster, Mario F L Gaudino, Alberto Zangrillo, Rinaldo Bellomo
{"title":"Amino Acid Infusion for Kidney Protection in Cardiac Surgery Patients with Chronic Kidney Disease: A Secondary Analysis of the PROTECTION Trial.","authors":"Martina Baiardo Redaelli, Fabrizio Monaco, Nikola Bradic, Anna Mara Scandroglio, Lian Kah Ti, Alessandro Belletti, Cristina Viscido, Margherita Licheri, Fabio Guarracino, Alessandro Pruna, Antonio Pisano, Domenico Pontillo, Francesco Federici, Rosario Losiggio, Giovanni Serena, Enrico Tomasi, Simona Silvetti, Marco Ranucci, Luca Brazzi, Andrea Cortegiani, Giovanni Landoni, Pasquale Mastroroberto, Gianluca Paternoster, Mario F L Gaudino, Alberto Zangrillo, Rinaldo Bellomo","doi":"10.1097/ALN.0000000000005336","DOIUrl":"10.1097/ALN.0000000000005336","url":null,"abstract":"<p><strong>Background: </strong>In the PROTECTION trial (Intravenous Amino Acid Therapy for Kidney Protection in Cardiac Surgery), intravenous amino acids decreased the occurrence of acute kidney injury in cardiac surgery patients with cardiopulmonary bypass. Recruitment of renal functional reserve may be responsible for such protection. However, patients with chronic kidney disease have diminished renal functional reserve, and amino acids may be less protective in such patients. Thus, a separate investigation of such patients is warranted.</p><p><strong>Methods: </strong>For this study chronic kidney disease was defined as an estimated glomerular filtration rate of less than 60 ml · min -1 · 1.73 m -2 , and patients with estimated glomerular filtration rates greater than or equal to 60 ml · min -1 · 1.73 m -2 served as controls. The primary outcome was the occurrence of acute kidney injury. Secondary outcomes included severity of acute kidney injury, need for and duration of renal replacement therapy, and all-cause mortality.</p><p><strong>Results: </strong>Among chronic kidney disease patients (n = 812), compared with placebo, amino acids significantly decreased the rate of acute kidney injury (43.1% vs 50.3%; relative risk, 0.86; 95% CI, 0.74 to 0.99; P = 0.041; number needed to treat = 14) with a median percentage increase in estimated glomerular filtration rate from baseline to postoperative day 3 of 12.7% versus 6.5% ( P = 0.002). In estimated glomerular filtration rate-based chronic kidney disease subgroups (30 to 39, 40 to 49, and 50 to 59 ml · min -1 · 1.73 m -2 ), the amino acid effect was similar (interaction P = 0.50). Finally, amino acid infusion decreased the occurrence of severe (stage 3) acute kidney injury (2.7% vs . 5.6%; relative risk 0.48; 95% CI, 0.24 to 0.98; P = 0.038).</p><p><strong>Conclusions: </strong>Amino acid infusion protected chronic kidney disease patients undergoing cardiopulmonary bypass from developing acute kidney injury, with an absolute risk reduction of 7% and a number needed to treat of 14 in a cohort with a greater than 45% rate of acute kidney injury. Moreover, it delivered a greater than 50% relative risk reduction in severe acute kidney injury.</p>","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-03-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142869296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-05DOI: 10.1097/ALN.0000000000005434
Ru Li, Yujie Huang, Kevin Lin, Xuri Li, James P Dilger, Jun Lin
{"title":"Distinct effects of sevoflurane and propofol on vascular permeability in vitro and in a mouse model.","authors":"Ru Li, Yujie Huang, Kevin Lin, Xuri Li, James P Dilger, Jun Lin","doi":"10.1097/ALN.0000000000005434","DOIUrl":"https://doi.org/10.1097/ALN.0000000000005434","url":null,"abstract":"<p><strong>Background: </strong>General anesthetics may substantially influence endothelium function, potentially affecting outcomes of surgical patients, but their effects are unclear. Here, we studied a commonly used inhaled anesthetic, sevoflurane, and an intravenous anesthetic, propofol, on vascular endothelial permeability using multiple in vitro assays and a mouse model.</p><p><strong>Methods: </strong>Human umbilical vein endothelial cells (HUVECs) and mouse pulmonary endothelial cells (MPECs) were used for in vitro models to test the effect of anesthetics on endothelial permeability. The effect of anesthetics on pulmonary vascular leakage was analyzed using AngioSense® 750 fluorescent tracer and rhodamine-labeled 3-kD dextran in a mouse model. Downstream targets were identified using RNA sequencing and confirmed by qRT-PCR and western blot.</p><p><strong>Results: </strong>Sevoflurane at clinically relevant concentrations disrupted the endothelial monolayer formed by HUVECs and MPECs in transwell permeability models. Sevoflurane, but not propofol, induced 1.8-fold increase of AngioSense® dye accumulation in mouse lung than control, indicating pulmonary vascular leakage in sevoflurane group. RNA sequencing analysis, qRT-PCR, and western blot analysis revealed that sevoflurane induced the expression and activation of hypoxic-induced factor 1α (HIF-1α) in vitro and in vivo. The activation of HIF-1α led to the increased expression of its downstream vascular endothelial growth factor (VEGF). The knockdown of HIF-1α restored the change of endothelial permeability and abolished the increase of VEGF induced by sevoflurane in MPECs.</p><p><strong>Conclusions: </strong>Our results demonstrate that sevoflurane increased endothelial and pulmonary vascular permeability via HIF-1α and VEGF. Propofol had no significant effect on the permeability of endothelium.</p>","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-05DOI: 10.1097/ALN.0000000000005435
Emmanuel Gonzalez, Tali Sahar, May Haddad, Sylvie Toupin, Ramzi Zioud, Muhammad Zoabi, Lilach Eyal Waldman, Zohar Tal Leshinsky, Maayan Ben Sasson, Vibhu Kumar, Yosefa Marom, Ayelet Midbari, Nicholas Jb Brereton, Yoram Shir, Amir Minerbi
{"title":"Altered gut microbiome composition and function in individuals with complex regional pain syndrome.","authors":"Emmanuel Gonzalez, Tali Sahar, May Haddad, Sylvie Toupin, Ramzi Zioud, Muhammad Zoabi, Lilach Eyal Waldman, Zohar Tal Leshinsky, Maayan Ben Sasson, Vibhu Kumar, Yosefa Marom, Ayelet Midbari, Nicholas Jb Brereton, Yoram Shir, Amir Minerbi","doi":"10.1097/ALN.0000000000005435","DOIUrl":"https://doi.org/10.1097/ALN.0000000000005435","url":null,"abstract":"<p><strong>Background: </strong>Complex regional pain syndrome (CRPS) is a chronic pain syndrome typically affecting a limb. It is characterized by severe spontaneous and evoked pain, along with vasomotor, autonomic, and motor signs and symptoms. Although dysregulation in several physiologic systems has been suggested in CRPS, including aberrant inflammatory and immune responses, vasomotor dysfunction, and nervous system changes, the pathophysiologic mechanisms underlying the syndrome remain elusive. Effective treatment options are also limited. Previous research has highlighted the role of the gut microbiome in chronic pain, prompting us to investigate the composition and function of the gut microbiome in CRPS.</p><p><strong>Methods: </strong>The gut microbiomes of individuals with CRPS to age-, gender- and ethnicity-matched pain-free control participants were compared using 16S rRNA gene amplification. To minimize environmental confounders, participants were recruited from two geographically independent regions. To explore potential changes in gut-bacteria-derived metabolites targeted metabolomic analysis of feces and plasma was performed. Finally, machine learning algorithms were trained to identify the gut microbiome composition specific to CRPS patients and were tested on a validation cohort.</p><p><strong>Results: </strong>In this study, differential abundance analysis revealed significant differences in several bacterial taxa when comparing 53 CRPS patients to 52 unrelated controls, including alterations in short-chain fatty acid (SCFA) metabolizing species. Targeted stool and plasma metabolite analysis confirmed differences in fecal and plasma SCFA levels between CRPS patients and controls. Notably, the microbiome composition alone allowed accurate classification of patients and controls in a geographically independent test cohort.</p><p><strong>Conclusions: </strong>These findings highlight unique compositional and functional changes in the gut microbiome of individuals with CRPS, thus contributing to the growing body of evidence supporting the role of the gut microbiome in chronic pain syndromes. Furthermore, they pave the way for further studies elucidating the pathophysiology of CRPS and exploring new diagnostic aids and treatment modalities.</p>","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555649","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-04DOI: 10.1097/ALN.0000000000005433
Nasir Hussain, Jarod Speer, Ryan S D'Souza, Marilly Palettas, Mahmoud Abdel-Rasoul, Alberto Uribe, Tristan Weaver, Michael Kushelev, John Coffman, Faraj W Abdallah
{"title":"EXPLORING THE ADDITIVE OR SYNERGISTIC EFFECTS OF THE SYSTEMIC AND PERINEURAL ROUTES OF DEXAMETHASONE AS ADJUNCTS TO SUPRACLAVICULAR BLOCK: A RANDOMIZED CONTROLLED TRIAL.","authors":"Nasir Hussain, Jarod Speer, Ryan S D'Souza, Marilly Palettas, Mahmoud Abdel-Rasoul, Alberto Uribe, Tristan Weaver, Michael Kushelev, John Coffman, Faraj W Abdallah","doi":"10.1097/ALN.0000000000005433","DOIUrl":"https://doi.org/10.1097/ALN.0000000000005433","url":null,"abstract":"<p><strong>Background: </strong>Both perineural and intravenous dexamethasone have been proposed as effective adjuncts that prolong the duration of peripheral nerve blocks. We sought to explore whether combining systemic with perineural dexamethasone yields any additive or synergistic effect on the characteristics and analgesic effects of peripheral nerve blocks.</p><p><strong>Methods: </strong>Adult patients having distal radius open reduction and internal fixation and/or carpometacarpal arthroplasty under supraclavicular block were randomized to either intravenous dexamethasone; combination of perineural+intravenous dexamethasone; or no dexamethasone (control). Sensory block duration was set as the primary outcome. Secondary outcomes included motor block duration; post-operative rebound pain scores as well as worst pain at 8, 16, 24, 32, 40, and 48-hours; opioid consumption at 0-24 and 25-48 hours; incidence of nausea/vomiting; and presence of burning sensation in the blocked limb at 24 and 48-hours. Our null hypothesis was lack of difference in sensory block duration between the three groups.</p><p><strong>Results: </strong>A total of 104 patients were included in the analysis (37: intravenous dexamethasone; 34: intravenous+perineural dexamethasone; 33: control). Compared to intravenous dexamethasone alone, adding perineural dexamethasone did not yield any incremental benefits in any of the outcomes examined. The mean [SD] of sensory block duration was 21.3 [7.3] hours in the intravenous dexamethasone group, 20.6 [6.1] hours in the perineural+intravenous group, and 16.8 [6.8] hours in the Control group. The mean difference [95% CI] of sensory block duration was significantly prolonged by 4.5 hours [1.3, 7.7] (P=0.006) in intravenous dexamethasone group and 3.4 hours [0.8, 6.8] (P=0.015) in the perineural+intravenous dexamethasone group, when compared to control; however, no difference was observed when the two dexamethasone groups were compared to each other (0.7 hours [-2.5, 3.9](P=0.670)). Compared to control, both intravenous and intravenous+perineural dexamethasone similarly reduced 24-hour pain scores and opioid consumption and decreased incidence of rebound pain.</p><p><strong>Conclusion: </strong>Our findings suggest that intravenous dexamethasone alone is sufficient to improve analgesic outcomes for patients receiving supraclavicular block for upper extremity surgery. Combining the intravenous and perineural dexamethasone routes does not yield additive or synergistic effect on the characteristics and analgesic effects of supraclavicular block.</p>","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":""},"PeriodicalIF":9.1,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143539994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-01Epub Date: 2024-11-27DOI: 10.1097/ALN.0000000000005314
Laura K Schnider, Marta Ratajczak, Rafael Wespi, Jacqueline G Kientsch, Francesco Bavato, Laurenz Marten, Jonas Kost, Maxim Puchkov, Corinne Eicher, Martina Boxler, Clarissa D Voegel, Oliver G Bosch, Eus van Someren, Dario A Dornbierer, Hans-Peter Landolt
{"title":"Effects of Subanesthetic Oromucosal Dexmedetomidine on Sleep in Humans: A Randomized, Controlled Pharmacokinetics-Pharmacodynamics Study.","authors":"Laura K Schnider, Marta Ratajczak, Rafael Wespi, Jacqueline G Kientsch, Francesco Bavato, Laurenz Marten, Jonas Kost, Maxim Puchkov, Corinne Eicher, Martina Boxler, Clarissa D Voegel, Oliver G Bosch, Eus van Someren, Dario A Dornbierer, Hans-Peter Landolt","doi":"10.1097/ALN.0000000000005314","DOIUrl":"10.1097/ALN.0000000000005314","url":null,"abstract":"<p><strong>Background: </strong>The locus coeruleus noradrenergic system may provide a potential new target for pharmacologic insomnia treatment, particularly in patients suffering from elevated distress. The selective α 2 -noradrenergic agonist dexmedetomidine attenuates locus coeruleus activity in subanesthetic doses, yet no adequate nonparental delivery systems of dexmedetomidine are currently available. To examine the feasibility of oromucosal dexmedetomidine administration, the authors developed two distinct-one sublingual and one buccal-oromucosal, fast-disintegrating dexmedetomidine formulas tailored for self-administration. Here, the authors established the formulas' pharmacokinetic and pharmacodynamic profiles.</p><p><strong>Methods: </strong>In a pilot study (sublingual formulation; n = 8 good sleepers) and a main study (buccal formulation; n = 17 poor sleepers), each following a randomized, double-blind, placebo-controlled crossover design, the authors investigated subanesthetic doses (20 and 40 µg) of the two formulas. They complemented the pharmacokinetic assessments with all-night polysomnography, nocturnal cortisol and melatonin measurements, assessments of cardiovascular functions during and after sleep, cortisol awakening response, and postawakening examination of subjective state and vigilance.</p><p><strong>Results: </strong>Particularly buccal dexmedetomidine was rapidly absorbed and exhibited excellent dose proportionality with minimal between-subject variation in exposure. In poor sleepers, 40 µg buccal dexmedetomidine shortened the sleep latency by 11.5 min, increased the time spent in non-rapid eye movement sleep by 37.2 min, and elevated non-rapid eye movement sleep electroencephalographic slow-wave energy (0.75 to 4.0 Hz) in the first half of the night by roughly 23%. Rapid eye movement sleep latency was dose-dependently prolonged (20 µg, 55.0 min; 40 µg, 115.3 min). Nocturnal cortisol, melatonin and heart rate, and morning cortisol were not significantly affected by dexmedetomidine, nor did postawakening orthostatic regulation, subjective sleepiness and mood, and psychomotor vigilance differ among the conditions.</p><p><strong>Conclusions: </strong>The favorable pharmacokinetic and pharmacodynamic profile of oromucosal dexmedetomidine delivery warrants further dose-finding and clinical studies to establish the exact roles of α 2 receptor agonism in pharmacologic sleep enhancement and as a possible novel mechanism to alleviate stress-related insomnia.</p>","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":" ","pages":"476-487"},"PeriodicalIF":9.1,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11801451/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142724987","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-01Epub Date: 2025-02-11DOI: 10.1097/ALN.0000000000005331
Sirkku Ahlström, Hanna Granroth-Wilding, Paula Reiterä, Klaus Olkkola, Ritva Jokela, Mari Kaunisto, Eija Kalso
{"title":"Genome-wide Study on Propofol Requirements: Reply.","authors":"Sirkku Ahlström, Hanna Granroth-Wilding, Paula Reiterä, Klaus Olkkola, Ritva Jokela, Mari Kaunisto, Eija Kalso","doi":"10.1097/ALN.0000000000005331","DOIUrl":"10.1097/ALN.0000000000005331","url":null,"abstract":"","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":"142 3","pages":"583-584"},"PeriodicalIF":9.1,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11801418/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143389797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-01Epub Date: 2025-02-11DOI: 10.1097/ALN.0000000000005310
Alexander Sartorius, Sebastian Karl, Christoph Janke, Grietje Beck
{"title":"Intersections of Anesthesiology and Psychiatry: Comment.","authors":"Alexander Sartorius, Sebastian Karl, Christoph Janke, Grietje Beck","doi":"10.1097/ALN.0000000000005310","DOIUrl":"10.1097/ALN.0000000000005310","url":null,"abstract":"","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":"142 3","pages":"577-578"},"PeriodicalIF":9.1,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143389803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AnesthesiologyPub Date : 2025-03-01Epub Date: 2025-02-11DOI: 10.1097/ALN.0000000000005372
{"title":"Science, Medicine, and the Anesthesiologist.","authors":"","doi":"10.1097/ALN.0000000000005372","DOIUrl":"10.1097/ALN.0000000000005372","url":null,"abstract":"","PeriodicalId":7970,"journal":{"name":"Anesthesiology","volume":"142 3","pages":"A13-A15"},"PeriodicalIF":9.1,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143389810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}