Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.689
Filter
1.
Int J Pharm Compd ; 28(4): 293-294, 2024.
Article in English | MEDLINE | ID: mdl-39094042

ABSTRACT

The Alliance for Pharmacy Compounding recently released four resource documents aimed at shaping compounding best practices and regulatory compliance.


Subject(s)
Drug Compounding , Ketamine , Drug Compounding/standards , Ketamine/administration & dosage , Humans , Peptides
2.
Int J Pharm Compd ; 28(4): 278-281, 2024.
Article in English | MEDLINE | ID: mdl-39094048

ABSTRACT

The author has been using ketamine to treat hospice patients for several years, with varying degrees of success, and reports being most successful with the transdermal-gel form. He has also had success with ketamine administered as a nasal spray. In addition to providing general comments on the use of ketamine in this context, he presents four brief case reports demonstrating the use of ketamine and other medications in treating pain associated with various types of cancer.


Subject(s)
Administration, Cutaneous , Analgesics , Gels , Ketamine , Ketamine/administration & dosage , Humans , Male , Analgesics/administration & dosage , Hospice Care , Middle Aged , Aged , Pain/drug therapy , Female , Cancer Pain/drug therapy
3.
Adv Emerg Nurs J ; 46(3): 195-206, 2024.
Article in English | MEDLINE | ID: mdl-39094079

ABSTRACT

Procedural sedation and analgesia is an essential activity in the emergency department for managing pain and anxiety during a variety of medical procedures. Various pharmacotherapy options, including opioid analgesics, antiemetics, anticholinergics, sedatives, and ketamine have been utilized, all with their unique efficacy and safety profiles. This review highlights the challenges associated with using certain agents and discusses emerging trends such as the use of newer synthetic opioids and the expanding use of dexmedetomidine. Overall, the selection of the optimal agents for procedural sedation and analgesia should be guided based on the unique characteristics of each agent tailored to the needs of the specific procedure, along with consideration for individual patient characteristics.


Subject(s)
Analgesics, Opioid , Hypnotics and Sedatives , Humans , Hypnotics and Sedatives/therapeutic use , Analgesics, Opioid/therapeutic use , Dexmedetomidine/therapeutic use , Conscious Sedation/methods , Ketamine/therapeutic use , Emergency Service, Hospital , Antiemetics/therapeutic use , Pain Management/methods , Cholinergic Antagonists/therapeutic use
4.
Nat Commun ; 15(1): 7025, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147749

ABSTRACT

Scale-free statistics of coordinated neuronal activity, suggesting a universal operating mechanism across spatio-temporal scales, have been proposed as a necessary condition of healthy resting-state brain activity. Recent studies have focused on anesthetic agents to induce distinct neural states in which consciousness is altered to understand the importance of critical dynamics. However, variation in experimental techniques, species, and anesthetics, have made comparisons across studies difficult. Here we conduct a survey of several common anesthetics (isoflurane, pentobarbital, ketamine) at multiple dosages, using calcium wide-field optical imaging of the mouse cortex. We show that while low-dose anesthesia largely preserves scale-free statistics, surgical plane anesthesia induces multiple dynamical modes, most of which do not maintain critical avalanche dynamics. Our findings indicate multiple pathways away from default critical dynamics associated with quiet wakefulness, not only reflecting differences between these common anesthetics but also showing significant variations in individual responses. This is suggestive of a non-trivial relationship between criticality and the underlying state of the subject.


Subject(s)
Anesthetics , Ketamine , Pentobarbital , Wakefulness , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Mice , Anesthetics/pharmacology , Pentobarbital/pharmacology , Male , Wakefulness/drug effects , Wakefulness/physiology , Isoflurane/pharmacology , Isoflurane/administration & dosage , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Consciousness/drug effects , Consciousness/physiology , Calcium/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cerebral Cortex/diagnostic imaging , Anesthesia , Optical Imaging
5.
Clin Toxicol (Phila) ; 62(8): 477-482, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39091275

ABSTRACT

INTRODUCTION: Recent decades have witnessed an extraordinary global crisis of drug misuse. Although opioid analgesics receive the most attention, numerous other drugs have increased rates of misuse. KETAMINE AND ESKETAMINE: Ketamine and esketamine offer a unique natural experiment to explore two medications that are similar pharmacologically but differ in their availability to users and in their regulation by government agencies. MISUSE AND ABUSE OF KETAMINE AND ESKETAMINE: Multisystem "mosaic" surveillance of many drugs using real-world data has emerged in recent years. Ketamine and esketamine have been monitored concurrently. Ketamine is much more widely available than esketamine and shows clear signs of increasing misuse and abuse. In contrast, esketamine is difficult to detect in postmarket surveillance even though availability is increasing. DISCUSSION: Ketamine and esketamine offer insights regarding the safety of prescription medications with the potential for misuse. Since the pharmacology of ketamine and esketamine are similar, the regulatory apparatus may be the primary difference that limits misuse. Ketamine has few restrictions and can be prescribed or administered by many healthcare providers, and is available as an illicit drug. In contrast, the product labeling for esketamine has rigorous restrictions on its use. Many important issues remain to be addressed. We need a more rigorous evaluation of the natural experiment of ketamine and esketamine. How does this experience relate to the introduction of new psychedelics? CONCLUSIONS: Ketamine misuse use and misuse are increasing while esketamine use in increasing, but misuse is not increasing. It is reasonable to reevaluate the regulatory controls on ketamine to reduce its misuse and abuse.


Subject(s)
Ketamine , Ketamine/adverse effects , Humans , Substance-Related Disorders/prevention & control , Prescription Drug Misuse/prevention & control , Prescription Drugs/adverse effects , Illicit Drugs
7.
Science ; 385(6709): eado7010, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39116252

ABSTRACT

Ketamine has been found to have rapid and potent antidepressant activity. However, despite the ubiquitous brain expression of its molecular target, the N-methyl-d-aspartate receptor (NMDAR), it was not clear whether there is a selective, primary site for ketamine's antidepressant action. We found that ketamine injection in depressive-like mice specifically blocks NMDARs in lateral habenular (LHb) neurons, but not in hippocampal pyramidal neurons. This regional specificity depended on the use-dependent nature of ketamine as a channel blocker, local neural activity, and the extrasynaptic reservoir pool size of NMDARs. Activating hippocampal or inactivating LHb neurons swapped their ketamine sensitivity. Conditional knockout of NMDARs in the LHb occluded ketamine's antidepressant effects and blocked the systemic ketamine-induced elevation of serotonin and brain-derived neurotrophic factor in the hippocampus. This distinction of the primary versus secondary brain target(s) of ketamine should help with the design of more precise and efficient antidepressant treatments.


Subject(s)
Antidepressive Agents , Depression , Habenula , Ketamine , Receptors, N-Methyl-D-Aspartate , Animals , Male , Mice , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Depression/drug therapy , Depression/metabolism , Habenula/drug effects , Habenula/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Ketamine/pharmacology , Ketamine/administration & dosage , Mice, Inbred C57BL , Mice, Knockout , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Serotonin/metabolism
9.
Transl Psychiatry ; 14(1): 327, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122686

ABSTRACT

Ketamine has gained attention for its effective treatment for patients with major depressive disorder (MDD) and suicidal ideation; Despite numerous studies presenting the rapid efficacy, long-term benefit in real-world populations remains poorly characterized. This is a retrospective cohort study using TriNetX US Collaborative Network, a platform aggregating electronic health records (EHRs) data from 108 million patients from 62 health care organizations in the US, and the study population includes 514,988 patients with a diagnosis of recurrent MDD who were prescribed relevant treatment in their EHRs. The prescription of ketamine was associated with significantly decreased risk of suicidal ideation compared to the prescription of other common antidepressants: HR = 0.63 (95% CI: 0.53-0.76) at 1 day - 7 days, 0.67 (95% CI: 0.59-0.77) at 1 day - 30 days, 0.69 (95% CI: 0.62-0.77) at 1 day - 90 days, 0.74 (95% CI: 0.67-0.81) at 1 day - 180 days, and 0.78 (95% CI: 0.69-0.83) at 1 day - 270 days. This trend was especially robust among adults over 24 years of age, females, males, and White patients with recurrent MDD. This study provides real-world evidence that ketamine has long-term benefits in mitigating suicidal ideation in patients with recurrent MDD. Future work should focus on optimizing dosage regimens for ketamine, understanding the mechanism, and the difference in various demographic subpopulations.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Ketamine , Recurrence , Suicidal Ideation , Humans , Ketamine/therapeutic use , Depressive Disorder, Major/drug therapy , Male , Female , Adult , Middle Aged , Retrospective Studies , Antidepressive Agents/therapeutic use , United States , Young Adult , Aged , Adolescent
10.
BMC Surg ; 24(1): 228, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127614

ABSTRACT

BACKGROUND: OFA (Opioid-free anesthesia) has the potential to reduce the occurrence of opioid-related adverse events and enhance postoperative recovery. Our research aimed to investigate whether OFA, combining esketamine and dexmedetomidine, could serve as an alternative protocol to traditional OBA (opioid-based anesthesia) in shoulder arthroscopy, particularly in terms of reducing PONV (postoperative nausea and vomiting). METHODS: A total of 60 patients treated with shoulder arthroscopy from September 2021 to September 2022 were recruited. Patients were randomly assigned to the OBA group (n = 30) and OFA group (n = 30), receiving propofol-remifentanil TIVA (total intravenous anesthesia) and esketamine-dexmedetomidine intravenous anesthesia, respectively. Both groups received ultrasound-guided ISBPB(interscalene brachial plexus block)for postoperative analgesia. RESULTS: The incidence of PONV on the first postoperative day in the ward (13.3% vs. 40%, P < 0.05) was significantly lower in the OFA group than in the OBA group. Moreover, the severity of PONV was less severe in the OFA group than in the OBA group in PACU (post-anesthesia care unit) (0 [0, 0] vs. 0 [0, 3], P<0.05 ) and in the ward 24 h postoperatively ( 0 [0, 0] vs. 0 [0, 2.25], P<0.05). Additionally, the OFA group experienced a significantly shorter length of stay in the PACU compared to the OBA group (39.4 ± 6.76 min vs. 48.7 ± 7.90 min, P < 0.001). CONCLUSIONS: Compared to the OBA with propofol-remifentanil, the OFA with esketamine- dexmedetomidine proved to be feasible for shoulder arthroscopy, resulting in a reduced incidence of PONV and a shorter duration of stay in the PACU. TRIAL REGISTRATION: The Chinese Clinical Trial Registry (No: ChiCTR2100047355), 12/06/2021.


Subject(s)
Analgesics, Opioid , Anesthetics, Intravenous , Arthroscopy , Dexmedetomidine , Ketamine , Postoperative Nausea and Vomiting , Propofol , Remifentanil , Humans , Ketamine/administration & dosage , Ketamine/therapeutic use , Dexmedetomidine/administration & dosage , Male , Remifentanil/administration & dosage , Propofol/administration & dosage , Female , Arthroscopy/methods , Middle Aged , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Adult , Postoperative Nausea and Vomiting/prevention & control , Postoperative Nausea and Vomiting/epidemiology , Postoperative Nausea and Vomiting/etiology , Anesthetics, Intravenous/administration & dosage , Pain, Postoperative/prevention & control , Pain, Postoperative/etiology , Pain, Postoperative/diagnosis , Anesthesia, Intravenous/methods , Brachial Plexus Block/methods
11.
Iran J Med Sci ; 49(7): 421-429, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39114639

ABSTRACT

Background: The management of preoperative anxiety in pediatric patients, as well as its implications, has remained challenging for anesthesiologists. In this study, we compared the safety and efficacy of intranasal dexmedetomidine, midazolam, and ketamine as surgical premedication in children. Methods: This double-blinded randomized clinical trial was conducted at two tertiary hospitals in January 2014, on 90 children aged between 2-7 years old. The participants' American Society of Anesthesiologists (ASA) physical status was I or II, and they were scheduled for elective unilateral inguinal herniorrhaphy. Using the block randomization method, the patients were randomly assigned to three groups, each receiving intranasal dexmedetomidine (2 µg/Kg), midazolam (0.2 mg/Kg), and ketamine (8 mg/Kg) 60 min before induction of anesthesia. Anxiety and sedation state were evaluated before drug administration, and then every 10 min for the next 50 min. Parental separation anxiety, mask acceptance, postoperative agitation, pain, nausea, and vomiting were also recorded and compared between these groups. All the statistical analyses were performed using SPSS software (version 21.0). P<0.05 was considered statistically significant. Results: Ketamine indicated the strongest sedative effect 10, 20, and 30 min after administration of premedication (P<0.001, P=0.03, P=0.01, respectively). However, dexmedetomidine was more effective than other drugs after 40 and 50 min (P<0.001). Other variables indicated no statistically significant difference. Conclusion: In case of emergencies, intranasal ketamine, with the shortest time of action, could be administered. Intranasal dexmedetomidine, which was revealed to be the most potent drug in this study, could be administrated 40-50 min before elective pediatric surgeries.Trial registration number: IRCT2013081614372N1.


Subject(s)
Administration, Intranasal , Dexmedetomidine , Hypnotics and Sedatives , Ketamine , Midazolam , Humans , Ketamine/therapeutic use , Ketamine/pharmacology , Ketamine/administration & dosage , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Dexmedetomidine/administration & dosage , Midazolam/therapeutic use , Midazolam/pharmacology , Midazolam/administration & dosage , Child, Preschool , Male , Female , Child , Administration, Intranasal/methods , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/therapeutic use , Hypnotics and Sedatives/administration & dosage , Double-Blind Method , Ambulatory Surgical Procedures/methods , Anxiety/drug therapy
12.
Sci Rep ; 14(1): 18867, 2024 08 14.
Article in English | MEDLINE | ID: mdl-39143187

ABSTRACT

Ketamine has recently become an anesthetic drug used in human and veterinary clinical medicine for illicit abuse worldwide, but the detection of illicit abuse and inference of time intervals following ketamine abuse are challenging issues in forensic toxicological investigations. Here, we developed methods to estimate time intervals since ketamine use is based on significant metabolite changes in rat serum over time after a single intraperitoneal injection of ketamine, and global metabolomics was quantified by ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF-MS). Thirty-five rats were treated with saline (control) or ketamine at 3 doses (30, 60, and 90 mg/kg), and the serum was collected at 21 time points (0 h to 29 d). Time-dependent rather than dose-dependent features were observed. Thirty-nine potential biomarkers were identified, including ketamine and its metabolites, lipids, serotonin and other molecules, which were used for building a random forest model to estimate time intervals up to 29 days after ketamine treatment. The accuracy of the model was 85.37% in the cross-validation set and 58.33% in the validation set. This study provides further understanding of the time-dependent changes in metabolites induced by ketamine abuse.


Subject(s)
Ketamine , Machine Learning , Substance-Related Disorders , Animals , Rats , Male , Substance-Related Disorders/metabolism , Metabolomics/methods , Rats, Sprague-Dawley , Biomarkers/blood
13.
Commun Biol ; 7(1): 946, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103539

ABSTRACT

Consciousness has been proposed to be supported by electrophysiological patterns poised at criticality, a dynamical regime which exhibits adaptive computational properties, maximally complex patterns and divergent sensitivity to perturbation. Here, we investigate dynamical properties of the resting-state electroencephalogram (EEG) of healthy subjects undergoing general anesthesia with propofol, xenon or ketamine. Importantly, all participants were unresponsive under anesthesia, while consciousness was retained only during ketamine anesthesia (in the form of vivid dreams), enabling an experimental dissociation between unresponsiveness and unconsciousness. For each condition, we measure (i) avalanche criticality, (ii) chaoticity, and (iii) criticality-related metrics, revealing that states of unconsciousness are characterized by a distancing from both avalanche criticality and the edge of chaos. We then ask whether these same dynamical properties are predictive of the perturbational complexity index (PCI), a TMS-based measure that has shown remarkably high sensitivity in detecting consciousness independently of behavior. We successfully predict individual subjects' PCI values with considerably high accuracy from resting-state EEG dynamical properties alone. Our results establish a firm link between perturbational complexity and criticality, and provide further evidence that criticality is a necessary condition for the emergence of consciousness.


Subject(s)
Consciousness , Electroencephalography , Unconsciousness , Humans , Unconsciousness/chemically induced , Unconsciousness/physiopathology , Male , Adult , Female , Consciousness/drug effects , Consciousness/physiology , Ketamine/pharmacology , Propofol/pharmacology , Young Adult , Anesthesia, General
14.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125900

ABSTRACT

The effects of exposure to environmental pollutants on neurological processes are of increasing concern due to their potential to induce oxidative stress and neurotoxicity. Considering that many industries are currently using different types of plastics as raw materials, packaging, or distribution pipes, microplastics (MPs) have become one of the biggest threats to the environment and human health. These consequences have led to the need to raise the awareness regarding MPs negative neurological effects and implication in neuropsychiatric pathologies, such as schizophrenia. The study aims to use three zebrafish models of schizophrenia obtained by exposure to ketamine (Ket), methionine (Met), and their combination to investigate the effects of MP exposure on various nervous system structures and the possible interactions with oxidative stress. The results showed that MPs can interact with ketamine and methionine, increasing the severity and frequency of optic tectum lesions, while co-exposure (MP+Met+Ket) resulted in attenuated effects. Regarding oxidative status, we found that all exposure formulations led to oxidative stress, changes in antioxidant defense mechanisms, or compensatory responses to oxidative damage. Met exposure induced structural changes such as necrosis and edema, while paradoxically activating periventricular cell proliferation. Taken together, these findings highlight the complex interplay between environmental pollutants and neurotoxicants in modulating neurotoxicity.


Subject(s)
Brain , Disease Models, Animal , Microplastics , Oxidative Stress , Schizophrenia , Zebrafish , Zebrafish/metabolism , Animals , Oxidative Stress/drug effects , Microplastics/toxicity , Schizophrenia/metabolism , Schizophrenia/chemically induced , Schizophrenia/pathology , Schizophrenia/etiology , Brain/metabolism , Brain/drug effects , Brain/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Ketamine/adverse effects , Ketamine/toxicity , Methionine/metabolism , Immunohistochemistry
15.
PLoS One ; 19(8): e0308413, 2024.
Article in English | MEDLINE | ID: mdl-39116153

ABSTRACT

BACKGROUND: Ketamine has recently attracted considerable attention for its rapid effects on patients with major depressive disorder, including treatment-resistant depression (TRD). Despite ketamine's promising results in treating depression, a significant number of patients do not respond to the treatment, and predicting who will benefit remains a challenge. Although its antidepressant effects are known to be linked to its action as an antagonist of the N-methyl-D-aspartate (NMDA) receptor, the precise mechanisms that determine why some patients respond and others do not are still unclear. OBJECTIVE: This study aims to understand the computational mechanisms underlying changes in the auditory mismatch negativity (MMN) response following treatment with intravenous ketamine. Moreover, we aim to link the computational mechanisms to their underlying neural causes and use the parameters of the neurocomputational model to make individual treatment predictions. METHODS: This is a prospective study of 30 patients with TRD who are undergoing intravenous ketamine therapy. Prior to 3 out of 4 ketamine infusions, EEG will be recorded while patients complete the auditory MMN task. Depression, suicidality, and anxiety will be assessed throughout the study and a week after the last ketamine infusion. To translate the effects of ketamine on the MMN to computational mechanisms, we will model changes in the auditory MMN using the hierarchical Gaussian filter, a hierarchical Bayesian model. Furthermore, we will employ a conductance-based neural mass model of the electrophysiological data to link these computational mechanisms to their neural causes. CONCLUSION: The findings of this study may improve understanding of the mechanisms underlying response and resistance to ketamine treatment in patients with TRD. The parameters obtained from fitting computational models to EEG recordings may facilitate single-patient treatment predictions, which could provide clinically useful prognostic information. TRIAL REGISTRATION: Clinicaltrials.gov NCT05464264. Registered June 24, 2022.


Subject(s)
Depressive Disorder, Treatment-Resistant , Electroencephalography , Ketamine , Ketamine/therapeutic use , Ketamine/pharmacology , Ketamine/administration & dosage , Humans , Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/physiopathology , Adult , Prospective Studies , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Male , Female , Evoked Potentials, Auditory/drug effects , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/physiopathology , Treatment Outcome , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Middle Aged , Young Adult
16.
Transl Psychiatry ; 14(1): 324, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107286

ABSTRACT

There is insufficient evidence to guide dose and frequency optimization with repeated-dose ketamine for depression. This study assessed the value of symptomatic non-improvement after the first few ketamine infusions as a predictor of overall non-response in depression for early decision-making to discontinue treatment. A total of 135 individuals with major depressive disorder or bipolar disorder experiencing a current major depressive episode were administered six repeated doses of intravenous ketamine. Depressive symptoms were assessed using the Montgomery-Åsberg Depression Rating Scale (MADRS) at baseline, 4 h after the first infusion, and 24 h after each infusion. Improvement, partial response, and response were defined as a reduction rate of ≥ 20%, 30%, and 50% in MADRS scores, respectively. This study examined the relationship between improvement (as opposed to non-improvement after each infusion or consecutive non-improvements after the first few infusions) and partial response and response after the sixth infusion. This analysis was summarized using sensitivity, specificity, and other diagnostic test parameters. The sensitivities of improvement at 24 h post-infusion 4 and improvement at 24 h post-infusion 3, vs. three consecutive non-improvements, as predictors for overall partial response and response exceeded 90%. No significant reduction in depressive symptoms was seen in non-improvers following the remaining infusions after the above-identified point. Our study suggests that non-improvement after four infusions, or more conservatively three consecutive non-improvements after three infusions, could serve as a signal of overall non-response to repeated-dose intravenous ketamine for depression and that subsequent treatments would not be warranted.


Subject(s)
Bipolar Disorder , Depressive Disorder, Major , Ketamine , Humans , Ketamine/administration & dosage , Bipolar Disorder/drug therapy , Female , Male , Adult , Depressive Disorder, Major/drug therapy , Middle Aged , Infusions, Intravenous , Psychiatric Status Rating Scales , Treatment Outcome , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use
17.
Open Vet J ; 14(7): 1634-1643, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39175964

ABSTRACT

Background: Chronic toxoplasmosis has been strongly implicated in the development of psychosis and schizophrenia. Additionally, the understanding of schizophrenia has been significantly reshaped by insights into N-methyl-D-aspartate receptor (NMDAR) hypofunction. Aim: This study aimed to compare the behavioral, antioxidant, and NMDAR changes in mice subjected to Toxoplasma gondii infection and those treated with ketamine to induce schizophrenia-like symptoms. Methods: Sixty male BALB/c mice were divided into six groups: toxoplasmosis (TOXO) (infected), ketamine-induced schizophrenia (KET), TOXO+KET, TOXO+sulfadiazine-trimethoprim treatment (SDT), TOXO+KET+SDT, and control (CON) (uninfected). After 10 weeks post-infection, behavioral tests were conducted, brain antioxidant status and lipid peroxidation were analyzed, and NMDA-NR1/NR2A expressions were assessed. TOXO and KET induced distinct behaviors: hyperlocomotion, anxiety, and memory impairment. Results: Antioxidant enzyme levels decreased, and lipid peroxidation increased in TOXO and schizophrenic mice brains. NMDAR downregulation, especially NR-1 and NR2A, was evident due to T. gondii and ketamine. Sulfadiazine-trimethoprim ameliorated NMDAR downregulation, but not all of the behavioral alterations. Conclusion: Further studies are needed to elucidate specific NMDAR subunit roles in toxoplasmosis-induced pathophysiology, offering potential therapeutic insights. This investigation highlights the intricate relationship between chronic toxoplasmosis, NMDAR dysfunction, and schizophrenia-like behaviors. Insights gained could pave the way for innovative interventions targeting both cognitive and neurological impairments associated with these conditions.


Subject(s)
Ketamine , Mice, Inbred BALB C , Receptors, N-Methyl-D-Aspartate , Schizophrenia , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Male , Schizophrenia/drug therapy , Schizophrenia/metabolism , Mice , Ketamine/administration & dosage , Ketamine/pharmacology , Ketamine/therapeutic use , Toxoplasmosis/drug therapy , Behavior, Animal/drug effects , Chronic Disease , Toxoplasmosis, Animal/drug therapy
18.
Vet Anaesth Analg ; 51(5): 449-457, 2024.
Article in English | MEDLINE | ID: mdl-39142984

ABSTRACT

OBJECTIVE: To compare the effects of propofol, ketamine-propofol and isoflurane, at similar anesthetic depth, on cardiopulmonary variables in unpremedictated chickens. STUDY DESIGN: Prospective, randomized, crossover experimental trial. ANIMALS: A total of 10 male Leghorn domestic chickens, aged 3 months and body mass 1.4-2.0 kg. METHODS: Birds were randomly assigned to each of three anesthetic protocols, 7 days apart: intravenous propofol, intravenous ketamine-propofol or isoflurane. Anesthesia was induced (indicated by loss of righting reflex and tracheal intubation) and maintained with propofol (10 mg kg-1 minute-1, then 1.1 mg kg-1 minute-1), ketamine-propofol (5 mg mL-1 ketamine and 5 mg mL-1 propofol combined; 10 mg kg-1 minute-1, then 1.1 mg kg-1 minute-1) or isoflurane [5% vaporizer setting initially, then end-tidal concentration (Fe'Iso) of 2%] for 65 minutes. Anesthesia was maintained at a similar anesthetic depth based upon positive or negative responses to toe pinch. Heart rate (HR), respiratory rate (fR), noninvasive arterial blood pressure and arterial blood gases were measured during anesthesia. Propofol or ketamine-propofol infusion rates and Fe'Iso required to prevent movement in response to a noxious stimulus and recovery times were recorded. RESULTS: Anesthesia induction dose was 9.0 ± 0.8 (mean ± SD) and 12.2 ± 0.3 mg kg-1 for propofol and ketamine-propofol, respectively. Propofol and ketamine-propofol infusion rates and Fe'Iso required to prevent movement in response to the noxious stimulus were 0.88 ± 0.14 mg kg-1 minute-1, 0.92 ± 0.14 mg kg-1 minute-1 and 1.45 ± 0.28%, respectively. Cardiopulmonary variables remained clinically acceptable, but ketamine-propofol was associated with a significantly higher HR (p = 0.0001) and lower fR (p = 0.0001). Time to extubation did not differ among treatments. CONCLUSIONS AND CLINICAL RELEVANCE: Cardiovascular and respiratory variables were maintained within normal ranges in all treatments. Coadministration of ketamine with propofol significantly reduced the induction and maintenance dose of propofol.


Subject(s)
Anesthetics, Intravenous , Chickens , Heart Rate , Isoflurane , Ketamine , Propofol , Animals , Propofol/pharmacology , Propofol/administration & dosage , Ketamine/administration & dosage , Ketamine/pharmacology , Isoflurane/administration & dosage , Isoflurane/pharmacology , Male , Heart Rate/drug effects , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/administration & dosage , Cross-Over Studies , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Blood Pressure/drug effects , Respiratory Rate/drug effects
19.
Drug Des Devel Ther ; 18: 3629-3641, 2024.
Article in English | MEDLINE | ID: mdl-39161682

ABSTRACT

Purpose: This study aimed to establish the 95% effective dose (ED95) of esketamine in combination with propofol for hysteroscopy and then to evaluate its efficacy and safety profile. Patients and Methods: This prospective, double-blind, randomized controlled trial consisted of two cohorts. In cohort 1, 45 women aged 18-65 years undergoing hysteroscopy were randomly assigned to either group E (esketamine + propofol) or group A (alfentanil + propofol). Dixon's up-and-down method was used to determine the ED95 of esketamine and alfentanil. In cohort 2, 86 patients were randomized to group E and group A, with the calculated ED95 dose of the study drugs used for induction. The success rate of anesthesia using the ED95% dose, along with parameters related to anesthesia induction, recovery, and adverse events were also recorded. Results: The ED95 of esketamine was 0.254 mg/kg (95% CI: 0.214-1.004), while that of alfentanil was 9.121 µg/kg (95% CI: 8.479-13.364). The anesthesia success rate was 93.0% in group E and 95.2% in group A (p = 0.664). After resuscitation, both groups achieved a 100% success rate. The induction time was significantly shorter in group E (60.0 [55.0-70.0] s) compared to group A (67.0 [61.0-79.3] s) (p = 0.006). Group E had lower rates of respiratory depression (p < 0.001), hypoxia (p = 0.006), minimum perioperative SpO2 (p = 0.010), and hypotension (p = 0.001). Esketamine had less effect on respiratory rate, heart rate, mean blood pressure, and end-tidal carbon dioxide compared to alfentanil (all p < 0.001). There were no significant differences in postoperative pain between the two groups. Conclusion: This study determined the ED 95 dose of esketamine for intravenous general anesthesia during hysteroscopy. Esketamine showed less respiratory and hemodynamic depression, as well as fewer adverse effects compared to alfentanil. Esketamine is an ideal anesthetic agent compared to alfentanil for hysteroscopic anesthesia. Trial Registration: www.chictr.org.cn, (ChiCTR2300077283); registered November 3, 2023.


Subject(s)
Alfentanil , Hysteroscopy , Ketamine , Propofol , Humans , Alfentanil/administration & dosage , Ketamine/administration & dosage , Ketamine/adverse effects , Double-Blind Method , Female , Middle Aged , Adult , Prospective Studies , Propofol/administration & dosage , Propofol/adverse effects , Propofol/pharmacology , Adolescent , Young Adult , Aged , Dose-Response Relationship, Drug , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/adverse effects
20.
BMC Anesthesiol ; 24(1): 293, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160473

ABSTRACT

BACKGROUND: The development of postpartum depression has been linked to fluctuations in the levels of neurotransmitters in the human body, such as 5-hydroxytryptamine (5-HT), dopamine (DA), noradrenaline (Norepinephrine, NE), and brain derived neurotrophic factor (BDNF). Research has indicated that the antidepressant effect of esketamine are mediated by monoamine transmitters and neurotrophic factors. Therefore, we postulate that intravenous administration of esketamine in patients with postpartum depression may alter the serum concentrations of these neurotransmitters. METHODS: Three hundred fifteen patients with postpartum depression were selected and divided into two groups based on randomized numerical expression: esketamine (E) group (0. 25 mg/kg esketamine) and control (C) group (a same volume of 0.9% saline), all the drugs were pumped for 40 min. After the end of drug pumping, all patients were continuously observed for 2 h. Changes in serum levels of 5-HT, DA, NE, BDNF were recorded before drug administration and on the 3rd day after drug administration. The scores of Edinburgh Postnatal Depression Scale (EPDS) were calculated before drug administration, and on the 3rd day and on the 30th day after drug administration. Dizziness, headache, nausea, vomiting, drowsiness, and feeling of detachment occurred were recorded within 2 h after drug administration. RESULTS: Before drug administration, the serum concentrations of 5-HT,DA,BDNF,NE in Group E and Group C were namely (0. 91 ± 0. 19 vs. 0. 98 ± 0. 21, P = 0. 181), (2. 38 ± 0. 35 vs. 2. 32 ± 0. 32, P = 0. 491), (3. 07 ± 0. 89 vs 3. 02 ± 0. 88, P = 0. 828), (39. 79 ± 7. 78 vs 41. 34 ± 10. 03, P = 0. 506). On the third day post-medication, the serum concentrations of 5-HT,DA,BDNF,NE in Group E and Group C were namely (1. 42 ± 0. 35 vs. 0. 96 ± 0. 24, P < 0. 001), (3. 99 ± 0. 17 vs. 2. 41 ± 0. 28, P < 0. 001),(5. 45 ± 0. 81 vs 3. 22 ± 0. 76, P < 0. 001),(44. 36 ± 9. 98 vs 40. 69 ± 11. 75, P = 0. 198). Before medication, the EPDS scores were (16. 15 ± 3. 02 vs 17. 85 ± 3. 89, P = 0. 064). on the third day after medication, the Group E had significantly reduced scores (12. 98 ± 2. 39 vs 16. 73 ± 3. 52, P < 0. 001). On the 30rd day after medication, EPDS scores between the two groups were (16. 34 ± 3. 43 vs 16. 91 ± 4. 02, p = 0. 203). Within 2 h of medication, the rate of adverse events was similar between the two groups. CONCLUSIONS: Small doses of esketamine can increase the serum concentration of 5-HT,DA,BDNF, and in the short term, decrease EPDS scores, and improve postpartum depressive symptoms. TRIAL REGISTRATION: Retrospectively registered in the Chinese Clinical Trial Registry (ChiCTR2300078343, 2023/12/05).


Subject(s)
Brain-Derived Neurotrophic Factor , Depression, Postpartum , Ketamine , Neurotransmitter Agents , Serotonin , Humans , Female , Ketamine/administration & dosage , Ketamine/pharmacology , Depression, Postpartum/drug therapy , Depression, Postpartum/blood , Adult , Neurotransmitter Agents/blood , Brain-Derived Neurotrophic Factor/blood , Serotonin/blood , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Norepinephrine/blood , Dopamine/blood
SELECTION OF CITATIONS
SEARCH DETAIL