Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.615
Filter
1.
Acta Vet Scand ; 66(1): 23, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822394

ABSTRACT

BACKGROUND: Alpha2-adrenoceptor agonists (α2-agonists) are widely used in animals as sedatives and for pre-anaesthetic medication. Medetomidine has often been given subcutaneously (SC) to rats, although its absorption rate is slow and the individual variation in serum drug concentrations is high via this route. In addition, α2-agonists have various effects on metabolic and endocrine functions such as hypoinsulinaemia, hyperglycaemia and diuresis. Vatinoxan is a peripherally acting α2-adrenoceptor antagonist that, as a hydrophilic molecule, does not cross the blood-brain barrier in significant quantities and thus alleviates peripheral cardiovascular effects and adverse metabolic effects of α2-agonists. Aim of this study was to evaluate the effects of vatinoxan on sedation, blood glucose concentration, voiding and heart and respiratory rates and arterial oxygen saturation in rats sedated with subcutaneous medetomidine, midazolam and fentanyl. RESULTS: Onset of sedation and loss of righting reflex occurred significantly faster with vatinoxan [5.35 ± 1.08 (mean ± SD) versus 12.97 ± 6.18 min and 6.53 ± 2.18 versus 14.47 ± 7.28 min, respectively]. No significant differences were detected in heart and respiratory rates and arterial oxygen saturation between treatments. Blood glucose concentration (18.3 ± 3.6 versus 11.8 ± 1.2 mmol/L) and spontaneous urinary voiding [35.9 (15.1-41.6), range (median) versus 0.9 (0-8.0) mL /kg/min] were significantly higher without vatinoxan. CONCLUSIONS: Acceleration of induction of sedation, alleviation of hyperglycaemia and prevention of profuse diuresis by vatinoxan may be beneficial when sedating rats for clinical and experimental purposes with subcutaneous medetomidine, midazolam and fentanyl.


Subject(s)
Fentanyl , Hypnotics and Sedatives , Medetomidine , Midazolam , Animals , Medetomidine/pharmacology , Medetomidine/administration & dosage , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Fentanyl/pharmacology , Fentanyl/administration & dosage , Rats , Male , Midazolam/pharmacology , Midazolam/administration & dosage , Quinolizines/pharmacology , Quinolizines/administration & dosage , Blood Glucose/drug effects , Heart Rate/drug effects , Rats, Sprague-Dawley , Rats, Wistar
2.
Can Vet J ; 65(6): 574-580, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38827599

ABSTRACT

Background: As a major animal control service provider in the city of Guelph and Wellington County in Ontario, the Guelph Humane Society transports and presents injured or ill raccoons requiring humane euthanasia to the Ontario Veterinary College Health Sciences Centre (OVC-HSC). Issues around handling, transportation, and delays before euthanasia have recently raised some concerns for welfare and the need for means of improving this process. Objective: Investigation of a noncontrolled sedation and analgesia protocol for injured or ill raccoons intended to improve animal welfare by allowing humane handling, transport, and euthanasia following administration by an animal protection officer (APO). Animals and procedure: Twenty-seven injured or ill raccoons requiring transport and euthanasia, as determined by the Guelph Humane Society APOs, were included in the study. Each raccoon was administered acepromazine (0.05 mg/kg), alfaxalone (4 mg/kg), and medetomidine (0.15 mg/kg), intramuscularly, before being transported to the OVC-HSC for humane euthanasia. Results: The combination of acepromazine, alfaxalone, and medetomidine was suitable for administration by APOs and provided the desired sedation depth to allow transport and humane euthanasia. Transit time was the only predictor of sedation depth upon arrival at the OVC-HSC. Two raccoons showed mild physical response to intracardiac injection for euthanasia. Numerical cutoff points of an in-hospital visual analog score of sedation of ≥ 70/100 and duration of sedation of < 62 min showed zero probability of response to euthanasia. Conclusion and clinical relevance: Administration of acepromazine, alfaxalone, and medetomidine at the stated doses provided acceptable sedation and analgesia to improve animal welfare during transport and eventual euthanasia of raccoons.


Évaluation d'un protocole médicamenteux sans groupe témoin de sédation intramusculaire, pré-euthanasie, comprenant de l'alfaxalone 4 %, de la médétomidine et de l'acépromazine pour les ratons laveurs blessés ou malades. Contexte: En tant que fournisseur majeur de services de contrôle des animaux dans la ville de Guelph et dans le comté de Wellington en Ontario, la Guelph Humane Society transporte et présente les ratons laveurs blessés ou malades nécessitant une euthanasie sans cruauté au Ontario Veterinary College Health Sciences Centre (OVC-HSC). Les problèmes liés à la manutention, au transport et aux délais avant l'euthanasie ont récemment soulevé des inquiétudes quant au bien-être et à la nécessité de trouver des moyens d'améliorer ce processus. Objectif: Enquête sur un protocole de sédation et d'analgésie sans groupe témoin pour les ratons laveurs blessés ou malades destiné à améliorer le bien-être des animaux en permettant une manipulation, un transport et une euthanasie sans cruauté après administration par un agent de protection des animaux (APO). Animaux et procédure: Vingt-sept ratons laveurs blessés ou malades nécessitant un transport et une euthanasie, tel que déterminé par les APO de la Guelph Humane Society, ont été inclus dans l'étude. Chaque raton laveur a reçu de l'acépromazine (0,05 mg/kg), de l'alfaxalone (4 mg/kg) et de la médétomidine (0,15 mg/kg), par voie intramusculaire, avant d'être transporté à l'OVC-HSC pour une euthanasie sans cruauté. Résultats: La combinaison d'acépromazine, d'alfaxalone et de médétomidine convenait à l'administration par un APO et fournissait la profondeur de sédation souhaitée pour permettre le transport et l'euthanasie sans cruauté. Le temps de transit était le seul prédicteur de la profondeur de la sédation à l'arrivée à l'OVC-HSC. Deux ratons laveurs ont montré une légère réponse physique à une injection intracardiaque pour l'euthanasie. Les seuils numériques d'un score analogique visuel de sédation à l'hôpital ≥ 70/100 et d'une durée de sédation < 62 min ont montré une probabilité nulle de réponse à l'euthanasie. Conclusion et pertinence clinique: L'administration d'acépromazine, d'alfaxalone et de médétomidine aux doses indiquées a fourni une sédation et une analgésie acceptables pour améliorer le bien-être des animaux pendant le transport et l'euthanasie éventuelle des ratons laveurs.(Traduit par Dr Serge Messier).


Subject(s)
Acepromazine , Hypnotics and Sedatives , Medetomidine , Pregnanediones , Raccoons , Animals , Medetomidine/administration & dosage , Pregnanediones/administration & dosage , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Acepromazine/administration & dosage , Male , Female , Euthanasia, Animal , Injections, Intramuscular/veterinary , Animal Welfare
3.
BMC Anesthesiol ; 24(1): 167, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702608

ABSTRACT

The exact mechanisms and the neural circuits involved in anesthesia induced unconsciousness are still not fully understood. To elucidate them valid animal models are necessary. Since the most commonly used species in neuroscience are mice, we established a murine model for commonly used anesthetics/sedatives and evaluated the epidural electroencephalographic (EEG) patterns during slow anesthesia induction and emergence. Forty-four mice underwent surgery in which we inserted a central venous catheter and implanted nine intracranial electrodes above the prefrontal, motor, sensory, and visual cortex. After at least one week of recovery, mice were anesthetized either by inhalational sevoflurane or intravenous propofol, ketamine, or dexmedetomidine. We evaluated the loss and return of righting reflex (LORR/RORR) and recorded the electrocorticogram. For spectral analysis we focused on the prefrontal and visual cortex. In addition to analyzing the power spectral density at specific time points we evaluated the changes in the spectral power distribution longitudinally. The median time to LORR after start anesthesia ranged from 1080 [1st quartile: 960; 3rd quartile: 1080]s under sevoflurane anesthesia to 1541 [1455; 1890]s with ketamine. Around LORR sevoflurane as well as propofol induced a decrease in the theta/alpha band and an increase in the beta/gamma band. Dexmedetomidine infusion resulted in a shift towards lower frequencies with an increase in the delta range. Ketamine induced stronger activity in the higher frequencies. Our results showed substance-specific changes in EEG patterns during slow anesthesia induction. These patterns were partially identical to previous observations in humans, but also included significant differences, especially in the low frequencies. Our study emphasizes strengths and limitations of murine models in neuroscience and provides an important basis for future studies investigating complex neurophysiological mechanisms.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Electroencephalography , Ketamine , Propofol , Sevoflurane , Animals , Mice , Ketamine/pharmacology , Ketamine/administration & dosage , Sevoflurane/pharmacology , Sevoflurane/administration & dosage , Dexmedetomidine/pharmacology , Electroencephalography/drug effects , Electroencephalography/methods , Propofol/pharmacology , Propofol/administration & dosage , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Reflex, Righting/drug effects , Reflex, Righting/physiology , Mice, Inbred C57BL , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/administration & dosage , Anesthesia/methods
4.
Drug Des Devel Ther ; 18: 1743-1754, 2024.
Article in English | MEDLINE | ID: mdl-38803562

ABSTRACT

Purpose: Remimazolam tosilate is a novel ultrafast-acting benzodiazepine that has a rapid emergence even after continuous infusion when using flumazenil. So far, relatively few articles are still focusing on the quality of recovery after general anesthesia with remimazolam, especially in day surgery. This study aimed to compare the early postoperative quality of recovery of remimazolam tosilate with flumazenil and propofol in patients undergoing day surgery. Patients and Methods: 137 patients scheduled for day surgery were randomly divided into the remimazolam tosilate or propofol group. The primary endpoint was the incidence of overall recovery assessed with the early postoperative quality of recovery scale (PostopQRS) on postoperative day 1 (POD 1). The Richmond Agitation-Sedation Scale (RASS) scores in the post-anesthesia care unit (PACU), extubation time, postoperative recovery profiles, and perioperative data were documented. Any adverse events were recorded. Results: The incidence of overall recovery on POD1 was 47.7% in the remimazolam tosilate group and 65.1% in the propofol group (odds ratio, 0.52; 95% confidence interval (CI) 0.26 to 1.06; P = 0.072). In general, the overall recovery of the PostopQRS increased over time, and its interaction between time and group was significant (P = 0.003). Among the five dimensions of PostopQRS, there exist statistical differences between groups including emotional state and cognitive recovery. Upon arrival at the PACU, the remimazolam group was more sedated and took longer to recover to a RASS score similar to propofol. The frequency of application of vasoactive drugs during anesthesia was similar in both groups (P = 0.119). Despite rapid emergence with remimazolam after flumazenil reversal, re-sedation (10.8%) or somnolence (60%) in the PACU was observed, and the length of PACU stay in patients treated with remimazolam tosilate was longer than that of the propofol (35 min vs 30 min, P<0.001). Conclusion: General anesthesia with remimazolam tosilate in conjunction with flumazenil reversal permits rapid recovery of consciousness in day surgery, but there was a notable occurrence of re-sedation or somnolence observed in PACU.


Subject(s)
Benzodiazepines , Hypnotics and Sedatives , Propofol , Humans , Propofol/administration & dosage , Female , Male , Prospective Studies , Middle Aged , Benzodiazepines/administration & dosage , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Adult , Ambulatory Surgical Procedures , Anesthesia Recovery Period , Aged , Flumazenil/administration & dosage , Flumazenil/pharmacology , Flumazenil/therapeutic use
5.
PeerJ ; 12: e17342, 2024.
Article in English | MEDLINE | ID: mdl-38737745

ABSTRACT

Background: N-Ethylmaleimide (NEM), an agonist of the potassium chloride cotransporters 2 (KCC2) receptor, has been correlated with neurosuppressive outcomes, including decreased pain perception and the prevention of epileptic seizures. Nevertheless, its relationship with sleep-inducing effects remains unreported. Objective: The present study aimed to investigate the potential enhancement of NEM on the sleep-inducing properties of alprazolam (Alp). Methods: The test of the righting reflex was used to identify the appropriate concentrations of Alp and NEM for inducing sleep-promoting effects in mice. Total sleep duration and sleep quality were evaluated through EEG/EMG analysis. The neural mechanism underlying the sleep-promoting effect was examined through c-fos immunoreactivity in the brain using immunofluorescence. Furthermore, potential CNS-side effects of the combination Alp and NEM were assessed using LABORAS automated home-cage behavioral phenotyping. Results: Combination administration of Alp (1.84 mg/kg) and NEM (1.0 mg/kg) significantly decreased sleep latency and increased sleep duration in comparison to administering 1.84 mg/kg Alp alone. This effect was characterized by a notable increase in REM duration. The findings from c-fos immunoreactivity indicated that NEM significantly suppressed neuron activation in brain regions associated with wakefulness. Additionally, combination administration of Alp and NEM showed no effects on mouse neural behaviors during automated home cage monitoring. Conclusions: This study is the first to propose and demonstrate a combination therapy involving Alp and NEM that not only enhances the hypnotic effect but also mitigates potential CNS side effects, suggesting its potential application in treating insomnia.


Subject(s)
Alprazolam , Drug Synergism , Sleep , Animals , Alprazolam/pharmacology , Alprazolam/administration & dosage , Mice , Male , Sleep/drug effects , Electroencephalography/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Brain/drug effects , Brain/metabolism , Reflex, Righting/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage
6.
Drug Des Devel Ther ; 18: 1613-1625, 2024.
Article in English | MEDLINE | ID: mdl-38774484

ABSTRACT

Purpose: Remimazolam, an ultra-short-acting and fast-metabolized sedative, has only been sporadically investigated in children. This study was performed to determine the beneficial effects of intranasal remimazolam or dexmedetomidine on preoperative anxiety in children undergoing general surgeries. Patients and Methods: Ninety children were randomly and equally assigned to Group R (intranasal remimazolam 1.5mg kg-1), Group D (intranasal dexmedetomidine 2 mcg kg-1), and Group C (intranasal distilled water). The primary outcomes were the preoperative anxiety scores using the modified Yale preoperative anxiety scale (m-Ypas). The secondary outcomes included the cooperation behaviour of intranasal drug application, preoperative sedation levels, parental separation anxiety scores (PSAS), and mask acceptance scores (MAS). Results: Group R showed a significant low anxiety at 10 min after intranasal premedication (vs group C, P=0.010; vs group D, P = 0.002) and at anaesthesia induction (vs group C, P = 0.004). Group D showed a significantly low anxiety score only prior to anaesthesia induction (vs group C, P = 0.005). Most children in group R achieved mild sedation at 10 min (vs group C, P < 0.001; vs group D, P < 0.001), with a few progressing to deep sedation afterwards, while group D tended toward deep sedation. Compared to Group C, patients in Group R performed significantly better on the MAS (P = 0.014) and PSAS (P = 0.008). However, remimazolam did cause poor cooperation behavior to the intranasal application due to its mucosal irritation (vs group C, P = 0.001; vs group D, P = 0.010). Conclusion: Both intranasal remimazolam and dexmedetomidine can effectively alleviate preoperative anxiety in children. While intranasal remimazolam has a rapid onset, it produces only mild sedation and causes substantial nasal irritation. Trial Registration: NCT04720963, January 22, 2021, ClinicalTrials.Gov.


Subject(s)
Administration, Intranasal , Anti-Anxiety Agents , Anxiety , Dexmedetomidine , Hypnotics and Sedatives , Humans , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacology , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Male , Female , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Child , Child, Preschool , Anxiety/drug therapy , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacology , Double-Blind Method
7.
Bioorg Chem ; 148: 107435, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38762999

ABSTRACT

BACKGROUND: Pyridine and its derivatives play a vital role in medicinal chemistry, serving as key scaffolds for drugs. The ability to bind to biological targets makes pyridine compounds significant, sparking interest in creating new pyridine-based drugs. Thus, the purpose of the research is to synthesize new thioalkyl derivatives of pyridine, predict their biological spectrum, study their psychotropic properties, and based on these findings, perform structure-activity relationships to assess pharmacophore functional groups. METHODS: Classical organic methods were employed for synthesizing new thioalkyl derivatives of pyridine, with a multifaceted pharmacological profiles. Various software packages and methods were employed to evaluate the biological spectrum of the newly synthesized compounds. For the evaluation of neurotropic activity of new synthesized compounds, some biological methods were used according to indicators characterizing anticonvulsant, sedative and antianxiety activity as well as side effects. RESULTS: Effective synthetic methods for 6-amino-4-phenyl-2-thio-2H-thiopyran-5-carboxylic acid ethyl ester, 2-amino substituted thiopyridine derivatives and 6-cycloamino-2-thioalkyl-4-phenylnicotinate derivatives were obtained in high yield. Predicted biological spectra and pharmacokinetic data indicated high gastrointestinal absorption and low blood-brain barrier passage for most compounds and demonstrated potential various biological effects, particularly psychotropic properties. Studied compounds demonstrated high anticonvulsant activity through antagonism with pentylenetetrazole. They exhibited low toxicity without inducing muscle relaxation in the studied doses. In psychotropic studies, the compounds displayed activating, sedative, and anxiolytic effects. Notably, the 6-amino-2-thioalkyl-4-phenylnicotinate derivatives demonstrated significant anxiolytic activity (about four times more compared to diazepam). They also exhibited pronounced sedative effects. Ethyl 2-({2-[(diphenylmethyl)amino]-2-oxoethyl}thio)-4-phenyl-6-pyrrolidin-1-ylnicotinate exhibited anxiolytic activity even two times greater than diazepam. Moreover, all studied compounds showed statistically significant antidepressant effects. Noteworthy ethyl 2-({2-oxo-2-[(tetrahydrofuran-2-ylmethyl)amino]ethyl}thio)-4-phenyl-6-pyrrolidin-1-ylnicotinate showcasing its unique psychotropic effect. CONCLUSIONS: The selected compounds demonstrate anticonvulsant properties, activating behavior, and anxiolytic effects, while simultaneously exhibiting antidepressant effects and these compounds as promising candidates for further exploration in the development of therapeutics with a broad spectrum of neuropsychiatric applications.


Subject(s)
Anti-Anxiety Agents , Anticonvulsants , Pyridines , Structure-Activity Relationship , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/chemical synthesis , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacology , Anticonvulsants/chemistry , Mice , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/chemical synthesis , Anti-Anxiety Agents/chemistry , Molecular Structure , Dose-Response Relationship, Drug , Male , Seizures/drug therapy , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/chemical synthesis , Hypnotics and Sedatives/chemistry , Pentylenetetrazole
8.
ACS Chem Neurosci ; 15(11): 2091-2098, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38747710

ABSTRACT

Xylazine (also known as "tranq") is a potent nonopioid veterinary sedative that has recently experienced a surge in use as a drug adulterant, most often combined with illicitly manufactured fentanyl. This combination may heighten the risk of fatal overdose. Xylazine has no known antidote approved for use in humans, and age-adjusted overdose deaths involving xylazine were 35 times higher in 2021 than 2018. In April 2023, the Biden Administration declared xylazine-laced fentanyl an emerging drug threat in the United States. In 2022, the Drug Enforcement Agency (DEA) reported nearly a quarter of seized fentanyl powder contained xylazine. This dramatic increase in prevalence has solidified the status of xylazine as an emerging drug of abuse and an evolving threat to public health. The following narrative review outlines the synthesis, pharmacokinetics, pharmacodynamics, and adverse effects of xylazine, as well as the role it may play in the ongoing opioid epidemic.


Subject(s)
Xylazine , Xylazine/pharmacology , Humans , Animals , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/chemistry , Fentanyl/pharmacology , Fentanyl/chemistry , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Drug Overdose/epidemiology
9.
Biol Pharm Bull ; 47(4): 785-790, 2024.
Article in English | MEDLINE | ID: mdl-38583949

ABSTRACT

Midazolam (MDZ) is clinically used for its sedative and anticonvulsant properties. However, its prolonged or potentiated effects are sometimes concerning. The main binding protein of MDZ is albumin, and reduced serum albumin levels could lead to MDZ accumulation, thereby potentiating or prolonging its effects. Previous investigations have not thoroughly examined these phenomena from a behavioral pharmacology standpoint. Consequently, this study aimed to evaluate both the prolonged and potentiated effects of MDZ, as well as the effects of serum albumin levels on the action of MDZ in low-albumin rats. Male Wistar rats were classified into control (20% protein diet), low-protein (5% protein), and non-protein groups (0% protein diet) and were fed the protein-controlled diets for 30 d to obtain low-albumin rats. The locomotor activity and muscle relaxant effects of MDZ were evaluated using the rotarod, grip strength, and open-field tests conducted 10, 60, and 120 min after MDZ administration. Serum albumin levels decreased significantly in the low-protein and non-protein diet groups compared with those in the control group. Compared with the control rats, low-albumin rats demonstrated a significantly shorter time to fall, decreased muscle strength, and a significant decrease in the distance traveled after MDZ administration in the rotarod, grip strength, and open-field tests, respectively. Decreased serum albumin levels potentiated and prolonged the effects of MDZ. Hence, serum albumin level is a critical parameter associated with MDZ administration, which should be monitored, and any side effects related to decreased albumin levels should be investigated.


Subject(s)
Hypoalbuminemia , Midazolam , Rats , Male , Animals , Midazolam/pharmacology , Rats, Wistar , Hypnotics and Sedatives/pharmacology , Serum Albumin
10.
Drug Des Devel Ther ; 18: 1349-1368, 2024.
Article in English | MEDLINE | ID: mdl-38681208

ABSTRACT

Background: Sepsis is recognized as a multiorgan and systemic damage caused by dysregulated host response to infection. Its acute systemic inflammatory response highly resembles that of lipopolysaccharide (LPS)-induced endotoxemia. Propofol and dexmedetomidine are two commonly used sedatives for mechanical ventilation in critically ill patients and have been reported to alleviate cognitive impairment in many diseases. In this study, we aimed to explore and compare the effects of propofol and dexmedetomidine on the encephalopathy induced by endotoxemia and to investigate whether ferroptosis is involved, finally providing experimental evidence for multi-drug combination in septic sedation. Methods: A total of 218 C57BL/6J male mice (20-25 g, 6-8 weeks) were used. Morris water maze (MWM) tests were performed to evaluate whether propofol and dexmedetomidine attenuated LPS-induced cognitive deficits. Brain injury was evaluated using Nissl and Fluoro-Jade C (FJC) staining. Neuroinflammation was assessed by dihydroethidium (DHE) and DCFH-DA staining and by measuring the levels of three cytokines. The number of Iba1+ and GFAP+ cells was used to detect the activation of microglia and astrocytes. To explore the involvement of ferroptosis, the levels of ptgs2 and chac1; the content of iron, malondialdehyde (MDA), and glutathione (GSH); and the expression of ferroptosis-related proteins were investigated. Conclusion: The single use of propofol and dexmedetomidine mitigated LPS-induced cognitive impairment, while the combination showed poor performance. In alleviating endotoxemic neural loss and degeneration, the united sedative group exhibited the most potent capability. Both propofol and dexmedetomidine inhibited neuroinflammation, while propofol's effect was slightly weaker. All sedative groups reduced the neural apoptosis, inhibited the activation of microglia and astrocytes, and relieved neurologic ferroptosis. The combined group was most prominent in combating genetic and biochemical alterations of ferroptosis. Fpn1 may be at the core of endotoxemia-related ferroptosis activation.


Subject(s)
Dexmedetomidine , Endotoxemia , Ferroptosis , Lipopolysaccharides , Mice, Inbred C57BL , Propofol , Dexmedetomidine/pharmacology , Animals , Propofol/pharmacology , Ferroptosis/drug effects , Mice , Male , Endotoxemia/drug therapy , Endotoxemia/metabolism , Endotoxemia/chemically induced , Lipopolysaccharides/pharmacology , Dose-Response Relationship, Drug , Brain Diseases/drug therapy , Brain Diseases/metabolism , Brain Diseases/pathology , Hypnotics and Sedatives/pharmacology
12.
Vet Anaesth Analg ; 51(3): 253-265, 2024.
Article in English | MEDLINE | ID: mdl-38580536

ABSTRACT

OBJECTIVE: To evaluate cardiovascular effects of oral tasipimidine on propofol-isoflurane anaesthesia with or without methadone and dexmedetomidine at equianaesthetic levels. STUDY DESIGN: Prospective, placebo-controlled, blinded, experimental trial. ANIMALS: A group of seven adult Beagle dogs weighing (mean ± standard deviation) 12.4 ± 2.6 kg and a mean age of 20.6 ± 1 months. METHODS: The dogs underwent four treatments 60 minutes before induction of anaesthesia with propofol. PP: placebo orally and placebo (NaCl 0.9%) intravenously (IV); TP: tasipimidine 30 µg kg-1 orally and placebo IV; TMP: tasipimidine 30 µg kg-1 orally and methadone 0.2 mg kg-1 IV; and TMPD: tasipimidine 30 µg kg-1 orally with methadone 0.2 mg kg-1 and dexmedetomidine 1 µg kg-1 IV followed by 1 µg kg-1 hour-1. Isoflurane in oxygen was maintained for 120 minutes at 1.2 individual minimum alveolar concentration preventing motor movement. Cardiac output (CO), tissue blood flow (tbf), tissue oxygen saturation (stO2) and relative haemoglobin content were determined. Arterial and mixed venous blood gases, arterial and pulmonary artery pressures and heart rate (HR) were measured at baseline; 60 minutes after oral premedication; 5 minutes after IV premedication; 15, 30, 60, 90 and 120 minutes after propofol injection; and 30 minutes after switching the vaporiser off. Data were analysed by two-way anova for repeated measures; p < 0.05. RESULTS: Tasipimidine induced a significant 20-30% reduction in HR and CO with decreases in MAP (10-15%), tbf (40%) and stO2 (43%). Blood pressure and oxygenation variables were mainly influenced by propofol-isoflurane-oxygen anaesthesia, preceded by short-lived alterations related to IV methadone and dexmedetomidine. CONCLUSIONS AND CLINICAL RELEVANCE: Tasipimidine induced mild to moderate cardiovascular depression. It can be incorporated into a common anaesthetic protocol without detrimental effects in healthy dogs, when anaesthetics are administered to effect and cardiorespiratory function is monitored.


Subject(s)
Dexmedetomidine , Isoflurane , Methadone , Propofol , Pyrazoles , Animals , Dogs , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacology , Propofol/administration & dosage , Propofol/pharmacology , Methadone/administration & dosage , Methadone/pharmacology , Female , Isoflurane/administration & dosage , Isoflurane/pharmacology , Heart Rate/drug effects , Male , Blood Pressure/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Quinolizines/pharmacology , Quinolizines/administration & dosage , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Premedication/veterinary
13.
J Ethnopharmacol ; 330: 118252, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38663782

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Atractylis aristata batt., as an endemic plant from the Asteraceae family, holds a significant position in the Ahaggar region of southern Algeria's traditional medicine. The aerial parts of Atractylis aristata was used to cure inflammation, fever, and stomach disorders. AIM OF THE STUDY: The objective of the present investigation was to ascertain the overall bioactive components and phytochemical components and examine the antioxidant, antidiabetic, anti-inflammatory, acute toxicity, and sedative properties of the crude extract obtained from the aerial portions of Atractylis aristata (AaME). MATERIALS AND METHODS: The AaME's antioxidant activity was assessed by the use of pyrogallol autoxidation, (1,1 diphenyl-2-picrylhydrazyl) (DPPH), 2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonic acid (ABTS), and reducing power (RP) techniques. 1 mg/mL of AaME was used to evaluate the antidiabetic activity by applying the enzyme α-amylase inhibitory power test. At the same time, the bovine serum albumin (BSA) denaturation method was employed to quantify the in vitro anti-inflammatory activity at different concentrations (1.5625, 0.78125, 0.390625, 0.1953125 and 0.09765625 mg/mL). In contrast, following the Organization for Economic Co-operation and Development (OECD) guideline No. 423, which covers acute oral toxicity testing protocols, the limit dosage test was employed to assess in vivo acute toxicity. At the dose of 0.08 mg/mL, the carrageenan-induced paw edema approach was used to assess the anti-inflammatory efficacy in vivo, and the sedative activity was carried out at the dose of 0.08 mg/mL using the measurement of the locomotor method. Different bioactive compounds were identified within AaME using LC-MS/MS and HPLC-UV analysis. RESULTS: The acute toxicity study showed no fatalities or noticeable neurobehavioral consequences at the limit test; this led to their classification in Globally Harmonized System (GHS) category Five, as the OECD guideline No 423 recommended. At a concentration of 0.08 mg/mL (2000 mg/kg), AaME showed apparent inhibition of paw edema and a significant (p = 0.01227) reduction in locomotor activity compared to the control animals. Our findings showed that AaME exhibited considerable antioxidant (IC50 = 0.040 ± 0.003 mg/mL (DPPH), IC50 = 0.005 ± 5.77 × 10-5 mg/mL (ABTS), AEAC = 91.15 ± 3.921 mg (RP) and IR% = 23.81 ± 4.276 (Inhibition rate of pyrogallol) and rebuts antidiabetic activities (I% = 57.6241% ± 2.81772). Our findings revealed that the maximum percentage of BSA inhibition (70.84 ± 0.10%) was obtained at 1.562.5 mg/mL. Thus, the AaME phytochemical profile performed using phytochemical screening, HPLC-UV, and LC-MS/MS analysis demonstrated that A. aristata can be a valuable source of chemicals with biological activity for pharmaceutical manufacturers. CONCLUSION: The phytochemical profiling, determined through HPLC-UV and LC-MS/MS applications, reveals this plant's therapeutic value. The aerial parts of Atractylis aristata contain bioactive molecules such as gallic acid, ascorbic acid, and quercetin, contributing to its significant antioxidant capabilities. Furthermore, identifying alizarin, the active compound responsible for its anti-inflammatory properties, could provide evidence supporting the anti-inflammatory capabilities of this subspecies.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Hypnotics and Sedatives , Hypoglycemic Agents , Phenols , Plant Extracts , Animals , Antioxidants/pharmacology , Antioxidants/isolation & purification , Antioxidants/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Extracts/toxicity , Male , Phenols/pharmacology , Phenols/analysis , Phenols/isolation & purification , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/isolation & purification , Hypnotics and Sedatives/toxicity , Mice , Asteraceae/chemistry , Rats, Wistar , Rats , Edema/drug therapy , Edema/chemically induced , Female , Plant Components, Aerial/chemistry
14.
J Med Chem ; 67(9): 7112-7129, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38647397

ABSTRACT

Research into kappa opioid receptor (KOR) agonists with attenuated central-nervous-system side effects is a critical focus for developing productive and safe analgesics. Herein, a series of ortho-substituted N-cyclopropylmethyl-7α-phenyl-6,14-endoethano-tetrahydronorthebaines were designed, synthesized, and subjected to bioassays. Compound 7a exhibited high subtype selectivity and potent agonistic activity toward KOR (KOR, Ki = 3.9 nM, MOR/KOR = 270, DOR/KOR = 1075; [35S]GTPγS binding, EC50 = 3.4 nM). Additionally, this compound exhibited robust and persistent antinociceptive effects in rodent models with different animal strains (hot plate test, ED50 = 0.20-0.30 mg/kg, i.p.; abdominal constriction test, ED50 = 0.20-0.60 mg/kg, i.p.), with its KOR-mediated mechanism for antinociception firmly established. Notably, compound 7a, unlike conventional KOR agonists, displayed minimal sedation and aversion at the antinociceptive ED50 dose. This feature addresses a crucial limitation in existing KOR agonists, positioning compound 7a as a promising novel therapeutic agent.


Subject(s)
Receptors, Opioid, kappa , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Animals , Mice , Structure-Activity Relationship , Male , Humans , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/chemical synthesis , Hypnotics and Sedatives/chemistry , Rats , Analgesics/pharmacology , Analgesics/chemical synthesis , Analgesics/chemistry , Drug Discovery , Rats, Sprague-Dawley , Cricetulus
15.
Drug Des Devel Ther ; 18: 1221-1229, 2024.
Article in English | MEDLINE | ID: mdl-38645987

ABSTRACT

Remimazolam is a novel ultrashort-acting benzodiazepine that allosterically modulates γ-aminobutyric acid type A (GABAA) receptors to exert sedative effects. Remimazolam has the properties of controllable sedation, rapid onset, and a short duration of action, along with minor depression of circulation and respiration. Remimazolam has been approved for clinical use since 2020 in Japan, and it has been applied for procedural sedation, general anesthesia induction and maintenance, and sedation in ICU patients, and has been proven to be safe and effective. Currently, no consensus has been reached on the clinical application of remimazolam in pediatric patients. This review introduces the clinical research progress and limitations of remimazolam in recent years, aiming to supply scientific guidance and a theoretical reference for the application of remimazolam in pediatric anaesthesia.


Subject(s)
Benzodiazepines , Hypnotics and Sedatives , Humans , Child , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Benzodiazepines/administration & dosage
16.
Res Vet Sci ; 172: 105254, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582048

ABSTRACT

Two randomized crossover trials evaluated the effects of nicardipine constant rate infusion (CRI) on 1) the anesthetic potency of sevoflurane and 2) the ability to attenuate dexmedetomidine-induced cardiovascular depression in anesthetized dogs. First, six healthy Beagle dogs weighing 11.7 ± 0.9 kg were allocated to one of three treatments that administered a CRI of carrier (saline) or dexmedetomidine 0.5 or 3.0 µg/kg/h following a loading dose. The minimum alveolar concentration (MAC) of sevoflurane was determined utilizing electric stimuli before and after the loading dose of nicardipine (20 µg/kg intravenously for 10 min), followed by CRI at 40 µg/kg/h with 60 min of equilibration. Subsequently, cardiovascular and blood gas variables were evaluated in another trial under sevoflurane anesthesia at the individual 1.5 MAC. After baseline measurements, the dogs were assigned to two treatments (dexmedetomidine CRI at 0.5 or 3.0 µg/kg/h following a loading dose) with sevoflurane doses adjusted to 1.5 times of MAC equivalent, and the measurements were repeated every 15 min for 120 min. After 60 min, nicardipine CRI at 40 µg/kg/h with a loading dose was added to the dexmedetomidine CRI. Dexmedetomidine infusions significantly decreased the sevoflurane MAC but nicardipine did not significantly alter the MAC either with or without dexmedetomidine CRI in dogs. Dexmedetomidine dose-dependently decreased the cardiac index and increased the systemic vascular resistance index; these effects were fully counteracted by concomitant nicardipine CRI. Nicardipine CRI can be useful for controlling the cardiovascular depression elicited by dexmedetomidine in anesthetized dogs without affecting the anesthetic potency of sevoflurane.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Nicardipine , Sevoflurane , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/administration & dosage , Dogs , Sevoflurane/pharmacology , Sevoflurane/administration & dosage , Nicardipine/pharmacology , Nicardipine/administration & dosage , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Male , Cross-Over Studies , Female , Pulmonary Alveoli/drug effects , Infusions, Intravenous/veterinary , Heart Rate/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Blood Pressure/drug effects
17.
Am J Vet Res ; 85(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38569538

ABSTRACT

OBJECTIVE: To assess the efficacy of transmucosal euthanasia solution to induce euthanasia. ANIMALS: 6 bearded dragons (Pogona vitticeps). METHODS: An initial dose of euthanasia solution containing pentobarbital and phenytoin sodium was administered transmucosally in conscious lizards (100 mg/kg pentobarbital dose), followed by a second dose 20 minutes later (400 mg/kg pentobarbital dose). The presence of movement, leakage of euthanasia solution, behaviors consistent with oral irritation, respiratory rate, heart rate, palpebral and corneal reflex, and response to noxious stimuli were recorded until death, confirmed by the absence of Doppler cardiac flow and cardiac electrical activity. The time to loss of all parameters was calculated. Postmortem evaluation allowed for histopathologic evaluation of the oral cavity and gastrointestinal tract to detect potential mucosal damage from the alkaline euthanasia solution. RESULTS: The median time to death was 300 minutes (range, 300 to 360 minutes), median time to respiratory arrest was 30 minutes (range, 30 to 50 minutes), and median time to loss of deep pain response was 30 minutes (range, 20 to 50 minutes). Signs consistent with oral irritation occurred in 4 of 6 (66.7%) lizards, including 2 lizards that exhibited whole-body spasms after euthanasia solution administration. Histopathologic changes indicating peracute mucosal ulceration, suspected to be from caustic causes, were identified in 1 (1/6 [16.7%]) lizard. CLINICAL RELEVANCE: Transmucosal euthanasia solution administration resulted in clinical euthanasia within 6 hours. This method should be utilized only after premedication with analgesic and/or anesthetic medications due to the potential for acute mucosal ulceration and behaviors that may be distressing in client-owned animals.


Subject(s)
Euthanasia, Animal , Lizards , Pentobarbital , Phenytoin , Animals , Phenytoin/administration & dosage , Pentobarbital/administration & dosage , Euthanasia, Animal/methods , Male , Female , Administration, Mucosal , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology
18.
Drug Alcohol Depend ; 259: 111301, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38640863

ABSTRACT

BACKGROUND: The incidence of combination methamphetamine (METH)-opioid overdose has substantially increased in recent years. While agitation is uncommon after the naloxone (NLX) reversal of opioids, it is a major clinical concern in acute METH intoxication and can be physiologically antagonized by opioid-induced sedation. This study aimed to perform initial preclinical analysis of the safety and efficacy of dexmedetomidine (DEXMED) co-administered with NLX to attenuate METH-induced locomotor activity, as a rat model of agitation, after the reversal of fentanyl (FENT)-induced sedation. METHODS: Male Sprague Dawley rats were administered subcutaneous (SC) 0.1mg/kg FENT ± 1mg/kg METH. Fifteen min later, SC 0.1mg/kg NLX ± an increasing (0, 0.032, 0.056, and 0.1mg/kg) DEXMED dose was administered prior to the measurement of locomotor activity. After a washout period, the FENT ± METH and NLX ± DEXMED administration with the highest dose of DEXMED was administered for measurement of blood oxygen saturation and heart rate. RESULTS: After the NLX reversal of FENT-induced sedation, adjunct DEXMED substantially and significantly reduced METH-induced locomotor activity (p<0.05) at all doses tested. While the addition of DEXMED did not significantly reduce blood oxygenation in METH treated rats, it did so in the absence of METH. Also, DEXMED significantly reduced heart rate compared to non-DEXMED treated groups and resulted in further significant reductions in the animals not exposed to METH (p<0.05). CONCLUSIONS: These data provide preclinical evidence that DEXMED may be a safe and effective chemical restraint for METH-induced agitation after NLX opioid reversal.


Subject(s)
Dexmedetomidine , Fentanyl , Methamphetamine , Naloxone , Rats, Sprague-Dawley , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/administration & dosage , Male , Methamphetamine/administration & dosage , Fentanyl/pharmacology , Fentanyl/administration & dosage , Rats , Naloxone/pharmacology , Naloxone/administration & dosage , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Motor Activity/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Heart Rate/drug effects , Dose-Response Relationship, Drug
19.
J Cardiothorac Vasc Anesth ; 38(5): 1221-1227, 2024 May.
Article in English | MEDLINE | ID: mdl-38443205

ABSTRACT

The highly selective α2-adrenoceptor agonist dexmedetomidine is a commonly used sedative drug for patients undergoing anesthesia and intensive care treatment. Several studies have indicated that dexmedetomidine may have a potential role in preventing and treating perioperative tachyarrhythmias. However, the specific effect and mechanism of action of dexmedetomidine in this context remain unclear. Dexmedetomidine is known to regulate the electrophysiologic function of the myocardium by inhibiting the function of the sinus node and atrioventricular node, as well as affecting myocardial repolarization. This paper aims to provide a theoretical basis for the prevention and treatment of perioperative arrhythmias by summarizing the effects of dexmedetomidine on myocardial electrophysiologic function and its impact on different types of arrhythmias.


Subject(s)
Anesthesia , Dexmedetomidine , Humans , Dexmedetomidine/therapeutic use , Arrhythmias, Cardiac/drug therapy , Hypnotics and Sedatives/pharmacology , Critical Care
20.
Sci Rep ; 14(1): 7071, 2024 03 25.
Article in English | MEDLINE | ID: mdl-38528026

ABSTRACT

Etomidate is a sedative and hypnotic drug through intravenous administration that act on the central nervous system through GABA (Gamma-Amino Butyric Acid) receptors, which is widely used in anesthesia induction and maintenance and long-term sedation in severe patients. The study aimed to evaluate the pharmacokinetic and pharmacodynamic properties of two etomidate fat emulsions after administration through the intravenous infusion pump in healthy Chinese subjects. A randomized, open-label, 2-period crossover study was performed in 52 healthy subjects. The wash-out period was 7 days. Blood samples and pharmacodynamic index values were collected at the specified time points. Etomidate concentrations were measured using validated liquid chromatography-tandem mass spectrometry. Pharmacokinetic parameters were analyzed using a non-compartment model method. Pharmacodynamic parameters were calculated using pharmacodynamic index values. The study also evaluated the safety of the etomidate. Both the pharmacokinetic parameters and pharmacodynamic parameters result of the test and reference formulation were very similar. The 90% confidence intervals (CI) of the geometric least-squares mean (GLSM) ratios of the test to reference formulation were 91.33-104.96% for the maximum plasma concentration (Cmax), 97.21-102.03% for the area under the plasma concentration time curve from time 0 to the time of the last measurable concentration (AUC0-t), and 97.22-102.33% for the area under the plasma concentration time curve from time 0 to infinity (AUC0-∞). Meanwhile, the 90% CI of the GLSM ratios of the test to reference formulation were 102.28-110.69% for the minimal BIS value (BISmin), 99.23-101.17% for the area under the BIS time curve from time 0-60 min after administration (BISAUC0-60 min), respectively. The 90% CI of these pharmacokinetic and pharmacodynamic parameters all fall in the accepted bioequivalence range of 80.00-125.00%. No serious adverse events occurred during the study. This study has shown that the etomidate fat emulsion test and reference formulation had similar pharmacokinetic and pharmacodynamic characteristics in vivo. The two formulations exhibited good safety and well-tolerance.Clinical trials registration number: http://www.chinadrugtrials.org.cn/index.html . # CTR20191836.


Subject(s)
Etomidate , Humans , Area Under Curve , China , Cross-Over Studies , Etomidate/pharmacokinetics , Etomidate/pharmacology , Healthy Volunteers , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Tablets , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL
...