Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Am Assoc Lab Anim Sci ; 61(1): 89-95, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34847984

ABSTRACT

Precise oral dosing in rodents is usually achieved by intragastric gavage. If performed incorrectly due to technical difficulties, inexperience, or animal resistance, oral gavage may have animal welfare implications such as esophageal and gastric rupture and aspiration. The stress that is induced by this procedure can also lead to confounding results. In several animal models, drug vehicles must be sugar-free, deliver drugs in a specific formulation, and sometimes supply water. Gelatin has all of these properties. The current study aimed to evaluate the use of gelatin vehicles with different sensory features as an alternative to oral gavage. We investigated the time taken by 2 different inbred mouse strains, FVB/N and C57BL/6J, to ingest sugar-free gelatin pellets of varying flavors. Results showed that FVB/N mice took more time to eat the unflavored, strawberry and diet-flavored gelatin pellets than did C57BL/6J mice. Both strains showed low preference for lemon flavor, with the same ingestion times after the second day. This study showed that the C57BL/6J mice are more likely to eat gelatin than are FVB/N mice, and that the 2 strains of mice show a lower preference for lemon flavoring as compared with other flavors. This method of voluntarily oral administration offers an alternative to gavage for studies that use oral dosing studies.


Subject(s)
Flavoring Agents , Food , Gelatin , Administration, Oral , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred Strains
2.
Article in English | MEDLINE | ID: mdl-32585370

ABSTRACT

MS-222, the most widely used anaesthetic in fish, has been shown to induce embryotoxic effects in zebrafish. However, the underlying molecular effects are still elusive. This study aimed to investigate the effects of MS-222 exposure during early developmental stages by evaluating biochemical and molecular changes. Embryos were exposed to 50, 100 or 150 mg L-1 MS-222 for 20 min at one of three developmental stages (256-cell, 50% epiboly, or 1-4 somite stage) and oxidative-stress, cell proliferation and apoptosis-related parameters were determined at two time-points (8 and 26 hpf). Following exposure during the 256-cell stage, the biochemical redox balance was not affected. The genes associated with glutathione homeostasis (gstpi and gclc) were affected at 8 hpf, while genes associated with apoptosis (casp3a and casp6) and cellular proliferation (pcna) were found affected at 26 hpf. An inverted U-shaped response was observed at 8 hpf for catalase activity. After exposure at the 50% epiboly stage, the gclc gene associated with oxidative stress was found upregulated at 8 hpf, while gstpi was downregulated and casp6 was upregulated later on, coinciding with a decrease in glutathione peroxidase (GPx) activity and a non-monotonic elevation of protein carbonyls and casp3a. Additionally, MS-222 treated embryos showed a decrease in DCF-staining at 26 hpf. When exposure was performed at the 1-4 somite stage, a similar DCF-staining pattern was observed. The activity of GPx was also affected whereas RT-qPCR showed that caspase transcripts were dose-dependently increased (casp3a, casp6 and casp9). The pcna mRNA levels were also found to be upregulated while gclc was changed by MS-222. These results highlight the impact of MS-222 on zebrafish embryo development and its interference with the antioxidant, cell proliferation and cellular death systems by mechanisms still to be explained; however, the outcomes point to the Erk/Nrf2 signalling pathway as a target candidate.


Subject(s)
Aminobenzoates/toxicity , Anesthetics/toxicity , Embryonic Development/drug effects , Oxidative Stress/drug effects , Zebrafish/genetics , Zebrafish/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Embryo, Nonmammalian , Gene Expression Profiling , Reactive Oxygen Species/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Chemosphere ; 201: 730-739, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29547861

ABSTRACT

Ketamine is a widely used pharmaceutical that has been detected in water sources worldwide. Zebrafish embryos were used in this study to investigate the oxidative stress and apoptotic signals following a 24h exposure to different ketamine concentrations (0, 50, 70 and 90 mg L-1). Early blastula embryos (∼2 h post fertilisation-hpf) were exposed for 24 h and analysed at 8 and 26 hpf. Reactive oxygen species and apoptotic cells were identified in vivo, at 26 hpf. Enzymatic activities (superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), lactate dehydrogenase (LDH) and acetylcholinesterase (AChE)), glutathione levels (oxidised (GSSG) and reduced (GSH)), oxidative damage (lipid peroxidation (LPO) and protein carbonyls (CO)) as well as oxidative stress (gclc, gstp1, sod1 and cat), apoptosis (casp3a, casp6, casp8, casp9, aifm1 and tp53) and cell proliferation (pcna) related-genes were evaluated at 8 and 26 hpf. Caspase (3 and 9) activity was also determined at both time-points by colorimetric methods. Superoxide dismutase (SOD), catalase (CAT), glutathione levels (GSSG), caspase-9 and reactive oxygen species (ROS) were shown to be affected by ketamine exposure while in vivo analysis showed no difference in ROS. A significant up-regulation of superoxide dismutase (sod1) and catalase (cat) genes expression was also perceived. Ketamine-induced apoptosis was observed in vivo and confirmed by the apoptotic-related genes up-regulation. The overall results suggest that ketamine induced oxidative stress and apoptosis through the involvement of p53-dependent pathways in zebrafish embryos which could be important for the evaluation of the overall risk of ketamine in aquatic environments.


Subject(s)
Apoptosis/drug effects , Embryo, Nonmammalian/drug effects , Ketamine/toxicity , Oxidative Stress/drug effects , Tumor Suppressor Protein p53/metabolism , Water Pollutants, Chemical/toxicity , Zebrafish , Animals , Antioxidants/metabolism , Apoptosis/genetics , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Embryonic Development/genetics , Gene Expression/drug effects , Oxidative Stress/genetics , Zebrafish/embryology
4.
J Sci Food Agric ; 98(8): 2837-2844, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29363750

ABSTRACT

Obesity is linked to the onset of many diseases such as diabetes mellitus, cardiovascular diseases and cancer, among others. The prevalence of obesity nearly doubled worldwide between 1980 and 2014. Simultaneously, in the last decade, the effects of sulforaphane as a potential treatment for obesity have been investigated, with promising results. Fruits and vegetables and their processed agri-food co-products are good sources of natural health-promoting compounds. Brassica crops are among the most produced crops in the world and are a good source of glucoraphanin, which, following hydrolysis, releases sulforaphane. The Brassicaceae family generates large amounts of co-products with no intended use, causing negative economic and environmental impact. Valorization of these co-products could be achieved through their exploitation for the extraction of bioactive compounds such as sulforaphane. However, the extraction process still needs further improvement for its economic feasibility. This article reviews the potential effects of sulforaphane in the treatment of obesity, linked to the relevance of giving Brassica co-products added value, which is of key importance for the competitiveness of farmers and the agri-food industry. © 2018 Society of Chemical Industry.


Subject(s)
Anti-Obesity Agents/administration & dosage , Brassica/chemistry , Isothiocyanates/administration & dosage , Obesity/drug therapy , Plant Extracts/administration & dosage , Animals , Anti-Obesity Agents/isolation & purification , Humans , Isothiocyanates/isolation & purification , Obesity/metabolism , Plant Extracts/isolation & purification , Sulfoxides
5.
Toxicol Lett ; 279: 1-8, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28716577

ABSTRACT

Increasing evidence supports that ketamine, a widely used anaesthetic, potentiates apoptosis during development through the mitochondrial pathway of apoptosis. Defects in the apoptotic machinery can cause or contribute to the developmental abnormalities previously described in ketamine-exposed zebrafish. The involvement of the apoptotic machinery in ketamine-induced teratogenicity was addressed by assessing the apoptotic signals at 8 and 24 hpf following 20min exposure to ketamine at three stages of early zebrafish embryo development (256 cell, 50% epiboly and 1-4 somites stages). Exposure at the 256-cell stage to ketamine induced an up-regulation of casp8 and pcna at 8 hpf while changes in pcna at the mRNA level were observed at 24 hpf. After the 50% epiboly stage exposure, the mRNA levels of casp9 were increased at 8 and 24 hpf while aifm1 was affected at 24 hpf. Both tp53 and pcna expressions were increased at 8 hpf. After exposure during the 1-4 somites stage, no meaningful changes on transcript levels were observed. The distribution of apoptotic cells and the caspase-like enzymatic activities of caspase-3 and -9 were not affected by ketamine exposure. It is proposed that ketamine exposure at the 256-cell stage induced a cooperative mechanism between proliferation and cellular death while following exposure at the 50% epiboly, a p53-dependent and -independent caspase activation may occur. Finally, at the 1-4 somites stage, the defence mechanisms are already fully in place to protect against ketamine-insult. Thus, ketamine teratogenicity seems to be dependent on the functional mechanisms present in each developmental stage.


Subject(s)
Anesthetics, Dissociative/toxicity , Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , Blastula/drug effects , Gene Expression Regulation, Developmental/drug effects , Ketamine/toxicity , Teratogens/toxicity , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Blastula/metabolism , Blastula/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Assessment , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
6.
Eur J Pharmacol ; 803: 48-56, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28341344

ABSTRACT

The molecular mechanisms of hepatotoxicity after propofol anaesthesia have not been fully elucidated, although there is a relation with mitochondrial dysfunction. The action of propofol on mitochondrial hepatic functions in a rat model was evaluated by infusion for 4h with 25 and 62.5mg/kg/h propofol or 3.125ml/kg/h (vehicle). Liver mitochondrial respiratory rates were evaluated as well as mitochondrial transmembrane potential (ΔΨ), calcium fluxes, mitochondrial enzymatic activities (Complex I-V) and oxidative stress biomarkers (superoxide dismutase, catalase, glutathione reductase, glutathione S-transferase, lipid peroxidation and the oxidised/reduced glutathione ratio). Biophysical interactions with membrane models were also performed. The mitochondrial transmembrane potential was decreased and the opening time of the mitochondrial permeability transition pore was slightly reduced for the highest dose. The activity of complex II was stimulated by propofol, which also causes fluctuations on some respiratory parameters, whereas the antioxidant system was affected in a nonspecific manner. Fluorescence quenching studies suggested that propofol is preferably located in deeper regions of the bilayer and has a high affinity to mitochondrial membranes. It is suggested that propofol interacts with liver mitochondrial membranes with mild modification in mitochondrial function.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Propofol/metabolism , Propofol/pharmacology , Animals , Biomarkers/metabolism , Energy Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Oxidative Stress/drug effects , Permeability/drug effects , Rats , Rats, Wistar
7.
Toxicol Appl Pharmacol ; 321: 27-36, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28215996

ABSTRACT

Ketamine, one anesthetic used as an illicit drug, has been detected both in freshwater and marine ecosystems. However, knowledge of its impact on aquatic life is still limited. This study aimed to test its effects in zebrafish embryos by analyzing its time- and dose-dependent developmental toxicity and long-term behavioral changes. The 24h-LC50 was calculated from percent survival using probit analysis. Based on the 24h-LC50 (94.4mgL-1), embryos (2hour post-fertilization - hpf) were divided into four groups, including control, and exposed for 24h to ketamine concentrations of 50, 70 or 90mgL-1. Developmental parameters were evaluated on the course of the experimental period, and anatomical abnormalities and locomotor deficits were analyzed at 144hpf. Although the portion of ketamine transferred into the embryo was higher in the lowest exposed group (about 0.056±0.020pmol per embryo), the results showed that endpoints such as increased mortality, edema, heart rate alterations, malformation and abnormal growth rates were significantly affected. At 144hpf, the developmental abnormalities included thoracic and trunk abnormalities in the groups exposed to 70 and 90mgL-1. Defects in cartilage (alcian blue) and bone (calcein) elements also corroborated the craniofacial anomalies observed. A significant up-regulation of the development-related gene nog3 was detected by qRT-PCR at 8 hpf. Early exposure to ketamine also resulted in long-term behavioral changes, such as an increase in thigmotaxis and disruption of avoidance behavior at 144 hpf. Altogether, this study provides new evidence on the ketamine teratogenic potential, indicating a possible pharmacological impact of ketamine in aquatic environments.


Subject(s)
Anesthetics, Dissociative/toxicity , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Embryonic Development/drug effects , Ketamine/toxicity , Locomotion/drug effects , Animals , Embryo, Nonmammalian/physiology , Embryonic Development/physiology , Female , Locomotion/physiology , Male , Social Behavior , Zebrafish
8.
Psychopharmacology (Berl) ; 234(4): 549-558, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27933364

ABSTRACT

RATIONALE: Ketamine has been associated with pediatric risks that include neurocognitive impairment and long-term behavioral disorders. However, the neurobehavioral effects of ketamine exposure in early development remain uncertain. OBJECTIVES: This study aimed to test stage- and dose-dependent effects of ketamine exposure on certain brain functions by evaluating alterations in locomotion, anxiety-like and avoidance behaviors, as well as socialization. METHODS: Embryos were exposed to different concentrations of ketamine (0, 0.2, 0.4, and 0.8 mg mL-1) for 20 min during the 256-cell (2.5 h post fertilization-hpf), 50% epiboly (5.5 hpf), and 1-4 somites (10.5 hpf) stages. General exploratory activities, natural escape-like responses, and social interactions were analyzed under continuous light or under a moving light stimulus. RESULTS: A dose-dependent decrease in the overall mean speed was perceived in the embryos exposed during the 256-cell stage. These results were related to previously observed head and eye malformations, following ketamine exposure at this stage and may indicate possible neurobehavioral disorders when ketamine exposure is performed at this stage. Results also showed that ketamine exposure during the 50% epiboly and 1-4 somites stages induced a significant increment of the anxiety-like behavior and a decrease in avoidance behavior in all exposed groups. CONCLUSIONS: Overall, the results validate the neurodevelopmental risks of early-life exposure to ketamine.


Subject(s)
Anxiety , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Locomotion/drug effects , Animals , Zebrafish
9.
Lab Anim ; 50(6): 476-488, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27909198

ABSTRACT

Anaesthesia is used daily in fish experimental procedures; however, the use of an inadequate anaesthetic protocol can compromise not only the animal's welfare but also the reliability of results. The use of zebrafish (Danio rerio) in biomedical research has increased in the last decades, highlighting the importance of appropriate anaesthetic regimens for this species. This article reviews the main anaesthetic agents and protocols used in laboratory adult zebrafish, and some of the analgesic methods to be used in this species that still need more research. In addition, a systematized observation of signs is proposed to evaluate adult zebrafish welfare to reduce pain and distress.


Subject(s)
Analgesia/statistics & numerical data , Anesthesia/statistics & numerical data , Animal Welfare , Pain Measurement/methods , Research Design , Zebrafish/physiology , Anesthetics/therapeutic use , Animals
10.
Exp Toxicol Pathol ; 68(9): 521-531, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27531257

ABSTRACT

BACKGROUND: Propofol biotransformation occurs in the liver via hydroxylation by CYP450 enzymatic complex and by glucuronidation, however extra-hepatic metabolism has also been described. OBJECTIVES: To better understand the metabolic pathways involved in propofol biotransformation, the expression of CYP1A1, CYP1A2, the enzymatic and non-enzymatic antioxidant activity and the amount of propofol and its non-conjugated metabolites were investigated. METHODS: Twenty-one NewZealand rabbits were allocated into three groups continuously treated for 20h. Each group received: NaCl 0.9%, vehicle (SMOFlipid) and propofol 2% (Lipuro). At the end, liver and kidney samples were collected for histopathology and immunohistochemistry and plasma for quantification of propofol and its metabolites. RESULTS: CYP1A1 and CYP1A2 were observed in zone 1 and zone 3 regions of the liver acinus. The propofol and saline groups showed a higher expression of CYP1A1 when compared to vehicle group. Propofol significantly increased CYP1A2 expression, compared to saline. CYP1A1 and CYP1A2 immunoexpression were observed in the kidney but no differences were registered between groups. CONCLUSIONS: This suggests that propofol may act as selective inhibitor of CYP1A1 and an inducer of CYP1A2 expression in different regions of the liver. Propofol seems to have an antioxidative protective effect on liver parenchyma, comparatively to the emulsion alone. In the rabbit, extra-hepatic propofol biotransformation may also occur in the kidney.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Hypnotics and Sedatives/metabolism , Kidney/drug effects , Liver/drug effects , Propofol/metabolism , Animals , Hypnotics and Sedatives/pharmacology , Immunohistochemistry , Kidney/metabolism , Liver/metabolism , Propofol/pharmacology , Rabbits
11.
Chem Res Toxicol ; 29(8): 1298-309, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27359275

ABSTRACT

Ketamine, a widely used anesthetic, has been shown to have NMDA receptor dependent and independent actions during zebrafish (Danio rerio) embryogenesis. Notwithstanding, the effects of developmental toxicity and the mechanisms of ketamine action on fish embryos are still not well understood, and its implications for early vertebrate development remains to be clarified. In this work, zebrafish embryos were exposed to ketamine (0.2, 0.4, and 0.8 mg mL(-1)) in order to study the stage-developmental toxicity of this pharmaceutical. During 256-cell (2.5 h post-fertilization, hpf), 50% epiboly (5.5 hpf) and 1-4 somites (10.5 hpf), embryos were exposed to the referred ketamine concentrations for a period of 20 min and were allowed to grow until 144 hpf. Both lethal and nonlethal parameters were evaluated. Skeletal development was assessed by alcian blue and calcein staining. Additionally, the expression of the developmental genes sonic hedgehog a (shh a) and noggin 3 (nog3) was evaluated. Similar to our previous work, bone and cartilage malformations were observed after 256-cell exposure. During 50% epiboly, ketamine exposure induced concentration-dependent mortality and malformations, such as lordosis and/or kyphosis and microcephaly, namely, at higher concentrations. Conversely, exposure during 1-4 somites showed the induction of nonspecific effects with no rise in mortality. The quantitative real-time polymerase chain reaction (qRT-PCR) analysis showed differences in shh a and nog3 expressions comparatively to the control group. Overall, this study shows that the ketamine toxic profile is developmental phase-dependent with 256-cell being the most susceptible phase. The effects observed may result from ketamine interaction with cellular signaling pathways that merits further investigation.


Subject(s)
Ketamine/toxicity , Teratogens/toxicity , Zebrafish/embryology , Animals , Chromatography, High Pressure Liquid , Spectrophotometry, Ultraviolet
12.
PLoS One ; 11(1): e0147747, 2016.
Article in English | MEDLINE | ID: mdl-26808508

ABSTRACT

BACKGROUND: The increasing use of zebrafish model has not been accompanied by the evolution of proper anaesthesia for this species in research. The most used anaesthetic in fishes, MS222, may induce aversion, reduction of heart rate, and consequently high mortality, especially during long exposures. Therefore, we aim to explore new anaesthetic protocols to be used in zebrafish by studying the quality of anaesthesia and recovery induced by different concentrations of propofol alone and in combination with different concentrations of lidocaine. MATERIAL AND METHODS: In experiment A, eighty-three AB zebrafish were randomly assigned to 7 different groups: control, 2.5 (2.5P), 5 (5P) or 7.5 µg/ml (7.5P) of propofol; and 2.5 µg/ml of propofol combined with 50, (P/50L), 100 (P/100L) or 150 µg/ml (P/150L) of lidocaine. Zebrafish were placed in an anaesthetic water bath and time to lose the equilibrium, reflex to touch, reflex to a tail pinch, and respiratory rate were measured. Time to gain equilibrium was also assessed in a clean tank. Five and 24 hours after anaesthesia recovery, zebrafish were evaluated concerning activity and reactivity. Afterwards, in a second phase of experiments (experiment B), the best protocol of the experiment A was compared with a new group of 8 fishes treated with 100 mg/L of MS222 (100M). RESULTS: In experiment A, only different concentrations of propofol/lidocaine combination induced full anaesthesia in all animals. Thus only these groups were compared with a standard dose of MS222 in experiment B. Propofol/lidocaine induced a quicker loss of equilibrium, and loss of response to light and painful stimuli compared with MS222. However zebrafish treated with MS222 recovered quickly than the ones treated with propofol/lidocaine. CONCLUSION: In conclusion, propofol/lidocaine combination and MS222 have advantages in different situations. MS222 is ideal for minor procedures when a quick recovery is important, while propofol/lidocaine is best to induce a quick and complete anaesthesia.


Subject(s)
Anesthetics/administration & dosage , Lidocaine/administration & dosage , Models, Animal , Propofol/administration & dosage , Animals , Behavior, Animal , Zebrafish
13.
Anesth Analg ; 120(5): 1130-1137, 2015 May.
Article in English | MEDLINE | ID: mdl-25658314

ABSTRACT

BACKGROUND: α2-Adrenoceptor agonists are used frequently in human and veterinary anesthesia as sedative/analgesic drugs. However, they can impair cognition. Little is known about the concentration-dependent effects of α2-adrenoceptor agonists on synaptic plasticity, the neurophysiological basis of learning and memory. Therefore, we investigated the effects of different concentrations of medetomidine, an α2-adrenoceptor agonist, on basal excitatory synaptic transmission and on 2 forms of synaptic plasticity: paired-pulse facilitation (PPF) and long-term potentiation (LTP). METHODS: Evoked field excitatory postsynaptic potentials were recorded in Schaffer fibers-CA1 pyramidal cell synapses of mouse hippocampal slices, and the initial field excitatory postsynaptic potentials slope was measured. For basal synaptic transmission and PPF, increasing concentrations of medetomidine (1-200 µM) were applied to each slice. For LTP experiments, individual slices were used for each tested concentration of medetomidine (0.1-0.4 µM), where LTP induction and LTP maintenance were measured. RESULTS: The lower tested concentrations of medetomidine decreased LTP in a concentration-dependent manner, whereas greater concentrations were required to decrease fiber volley amplitude and basal excitatory synaptic transmission. PPF was only affected by the greatest concentration (200 µM). CONCLUSIONS: Medetomidine decreased LTP in the mouse hippocampus, in accordance with the ability of medetomidine to induce memory deficits.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , CA1 Region, Hippocampal/drug effects , Excitatory Postsynaptic Potentials/drug effects , Medetomidine/pharmacology , Neuronal Plasticity/drug effects , Pyramidal Cells/drug effects , Synaptic Transmission/drug effects , Adrenergic alpha-2 Receptor Agonists/toxicity , Age Factors , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Dose-Response Relationship, Drug , Evoked Potentials/drug effects , Female , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Medetomidine/toxicity , Memory/drug effects , Mice , Mice, Inbred BALB C , Pyramidal Cells/physiology , Time Factors
14.
Lab Anim (NY) ; 43(10): 353-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25238524

ABSTRACT

Ketamine is frequently used to induce analgesia or anesthesia in laboratory animals, but its effects on learning and memory are poorly characterized. Long-term potentiation (LTP) is considered a cellular mechanism for learning and memory. Ketamine administration immediately abolishes hippocampal LTP in vivo, but whether this effect persists is not known. The authors administered one of two doses of ketamine to adult male C57BL/6 mice and measured LTP in hippocampal slices from the mice 24 h later. Neither LTP induction nor LTP maintenance differed significantly in mice that were administered ketamine compared with mice that were administered saline. The findings suggest that a single intraperitoneal dose of ketamine does not persistently alter LTP in adult male mice.


Subject(s)
Anesthesia/veterinary , Hippocampus/drug effects , Ketamine/pharmacology , Long-Term Potentiation/drug effects , Animals , Electric Stimulation , Male , Mice , Mice, Inbred C57BL
15.
Brain Res ; 1560: 10-7, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24637258

ABSTRACT

Ketamine, an analgesic/anesthetic drug, is increasingly popular in clinical practice due to its analgesic properties and importance for emergency procedures. The impact of ketamine on basal excitatory synaptic transmission and synaptic plasticity are not yet fully understood. Therefore we investigated the effects of different concentrations of ketamine on basal excitatory synaptic transmission and on two forms of synaptic plasticity: paired-pulse facilitation (PPF) and long-term potentiation (LTP). Evoked field excitatory postsynaptic potentials (fEPSP) were recorded in Schaffer fiber - CA1 pyramid synapses of mouse hippocampal slices and the initial slope of the fEPSP was measured to estimate the percentage of inhibition of the basal synaptic transmission. Presynaptic volley amplitude, PPF and LTP induction and maintenance were also calculated. For basal synaptic transmission and PPF increasing concentrations of ketamine (1, 3, 10, 30, 100, 200, 300 and 600µM) were applied to each slice and for LTP individual slices were used for each concentration (3, 10, 30 or 100µM). Clinically relevant concentrations of ketamine decreased LTP in a concentration-dependent manner without changing PPF, whereas basal excitatory synaptic transmission and presynaptic volley amplitude was affected only with high concentrations of ketamine (300 and 600µM). These results allow dissociating the blockade of LTP from a reduced synaptic input in the action of clinically relevant concentrations of ketamine in the CA1 region of the mouse hippocampus. Moreover, this work shows that the effects of ketamine on LTP and on basal synaptic transmission are dependent of the concentration used.


Subject(s)
Anesthetics/pharmacology , Hippocampus/drug effects , Ketamine/pharmacology , Long-Term Potentiation/drug effects , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Female , Hippocampus/physiology , Mice, Inbred BALB C , Microelectrodes , Neuronal Plasticity/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Tissue Culture Techniques
16.
Neurotoxicol Teratol ; 41: 27-34, 2014.
Article in English | MEDLINE | ID: mdl-24287188

ABSTRACT

Concerns have been raised that the effect of anaesthetic drugs on the central nervous system may result in long-term impairment, namely when ketamine is used during embryogenesis. In addition, the cell and molecular basis of anaesthetics teratology and toxicity are still uncertain and its implications in the development remain to be clarified. More recently, the potential risks for human, and animal, exposure through environmental contamination also became an important question. In this study, the effects of sub- and over anaesthetic doses of ketamine were investigated during zebrafish (Danio rerio) embryonic development by exposing zebrafish embryos to ketamine concentrations (0.2, 0.4 and 0.8 mg mL(-1)) for a period of 20 min during the blastula stage. Ethanol 2% was used as a positive control. Morphological parameters, the overall pattern of cell death using acridine orange and overall degree of oxidative stress levels by 2,7-dichlorodihydrofluorescein-diacetate were determined. Lethality and/or developmental anomalies were measured based on specific time endpoints until 144 h post fertilisation. Results showed a concentration-dependent increase in anomalies and mortality. Cephalic disorders, enlarged organs and tail/spine anomalies were the most prominent deformities observed at 144 hpf. Acridine orange images revealed no differences in cellular death pattern in exposed embryos at 24 hpf. At the same time point, the cellular redox processes were found to be similar among groups. In summary, this study shows that ketamine is teratogen and toxic, interfering with the normal developmental pathways of embryogenesis, suggesting that ketamine exerts an independent NMDA receptor action during the zebrafish blastula stage.


Subject(s)
Embryonic Development/physiology , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Age Factors , Analysis of Variance , Animals , Apoptosis/drug effects , Cell Death/drug effects , Dose-Response Relationship, Drug , Embryo, Nonmammalian , Embryonic Development/drug effects , Larva , Mortality , Reactive Oxygen Species/metabolism , Zebrafish/embryology
17.
Eur J Anaesthesiol ; 30(10): 618-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23736087

ABSTRACT

BACKGROUND: Ketamine is an anaesthetic and analgesic drug used in research and clinical practice. Little is known about the effects of different doses of this drug on memory and brain cellular death. OBJECTIVE: To study the effects of different doses of ketamine on working and reference memory, and neurodegeneration in adult mice. DESIGN: A randomised study. SETTINGS: The study was carried out in a basic science laboratory, between March 2011 and August 2012. ANIMALS: Forty-eight 7-month-old, male C57BL/6 mice were used. INTERVENTION: Animals received a single intraperitoneal injection of physiological saline solution or one of three doses of ketamine (25, 75 or 150 mg kg(-1)). Each group consisted of 12 animals (seven animals for behavioural tests and five animals for histopathological and immunohistochemical studies). The animals used for histopathology studies were sacrificed 3 h after anaesthesia. MAIN OUTCOME MEASURES: Working and reference memories were assessed using the radial-maze test over 12 consecutive days. The equilibrium was tested using the vertical pole (4 and 24 h after injection), whereas locomotion was assessed using the open field (24, 48 and 72 h after injection). Histopathological (haematoxylin-eosin staining) and immunohistochemical analyses (procaspase-3 and activated caspase-3 detections) were performed 3 h after injection to assess neurodegeneration in the retrosplenial and visual cortices, pyramidal cell layer of the cornu Ammonis 1 and cornu Ammonis 3 areas of the hippocampus, in the granular layer of the dentate gyrus, in the laterodorsal thalamic nucleus, striatum and accumbens nucleus. RESULTS: No significant differences were observed between the groups regarding the number of dead cells and cells showing positive immune-reactivity in the different regions of the brain studied. The performance in the vertical pole test and the number of reference and working memory errors in the radial-maze were similar in all groups. Nevertheless, the animals treated with ketamine 75 mg kg(-1) were transiently more active, walking a greater total distance at a greater speed in the open field than other groups (power of 0.96). CONCLUSION: These data indicate that a single intraperitoneal injection of ketamine at subanaesthetic and anaesthetic doses does not impair working memory, reference memory or neurodegeneration in adult mice, but an intermediate dose of ketamine produces transitory hyperlocomotion.


Subject(s)
Anesthetics, Dissociative/administration & dosage , Behavior, Animal/drug effects , Brain/drug effects , Ketamine/administration & dosage , Memory, Short-Term/drug effects , Nerve Degeneration , Spatial Memory/drug effects , Animals , Brain/pathology , Brain/physiopathology , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Motor Activity/drug effects , Time Factors
18.
Eur J Pharmacol ; 712(1-3): 39-47, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23665495

ABSTRACT

Anesthetics, such as the ketamine/midazolam combination, are used in research with animals and in human clinical practice; thus, it is essential to clarify the potential effects of these anesthetics on memory. This study aimed to evaluate how a low dose of the ketamine/midazolam combination affects the acquisition, consolidation, or recall of a spatial memory task. Thirty-three adult male C57BL/6 mice were divided into four treatment groups: unanesthetized control animals and three groups of animals treated with 40 mg/kg of ketamine and 10mg/kg of midazolam administered in a single intraperitoneal injection. The different treatment groups received the same anesthetic dose at different time points, to study the acquisition, consolidation, and recall of spatial memory in the Y-maze task. The percentage of correct choices was measured. Six mice were killed 4 days and 12 days after anesthesia for histopathological analyses. There were no differences between treatment and control groups regarding the acquisition of spatial memory, measured as the slope of the learning curve, or in the percentage of correct choices in the consolidation or recall periods of the task. Similarly, no differences were detected between groups regarding the number of cells per square millimeter in the visual and retrosplenial cortex, in the dentate gyrus, and in the CA1 and CA3 regions of the hippocampus. Hence, a low dose of the ketamine/midazolam combination did not impair memory processes or brain integrity in adult mice, suggesting that this combination is unlikely to cause cognitive complications.


Subject(s)
Anesthetics/pharmacology , Ketamine/pharmacology , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Midazolam/pharmacology , Spatial Behavior/physiology , Animals , Brain/cytology , Brain/drug effects , Brain/physiology , Choice Behavior/drug effects , Choice Behavior/physiology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Spatial Behavior/drug effects
20.
Vet Anaesth Analg ; 39(1): 21-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22151874

ABSTRACT

OBJECTIVE: To evaluate if the cerebral state index (CSI), measured by a Cerebral State Monitor (CSM), can predict depth of anaesthesia as assessed clinically or by estimated propofol plasma concentrations. STUDY DESIGN: Prospective clinical study. ANIMALS: Fourteen mixed breed dogs, weighing 24.5 ± 4.7 kg, scheduled to undergo neutering procedures. METHODS: Dogs were premedicated with 0.05 mg kg(-1) acepromazine intramuscularly. The CSM and cardiovascular monitoring equipment were attached. Anaesthesia was induced with propofol using a target controlled infusion (TCI) to varying plasma propofol targets (PropCp). Following endotracheal intubation the dogs were ventilated with oxygen. Anaesthetic maintenance was with propofol by TCI. A PropCp of 3 µg dL(-1) was set initially, then PropCps were increased in 1 µg dL(-1) steps to 7, 9 and then 11 µg dL(-1). Each PropCp was held constant for a 5 minute period, at the end of which depth of anaesthesia was classified using a previously evaluated scale of 'planes' based on palpebral and corneal reflexes and eye position. Cerebral state index (CSI), burst suppression (BSR) and electromyogram were measured at these time points. The prediction probability (PK) of these variables, or of the PropCp in predicting depth of anaesthesia was calculated. RESULTS: The PKs for predicting anaesthetic planes were 0.74, 0.91, 0.76 and 0.78 for CSI, BSR, EMG and PropCp, respectively. The PKs for PropCp to predict CSI, BSR and EMG were 0.65, 0.71 and 0.65 respectively. CONCLUSION AND CLINICAL RELEVANCE: The Cerebral State Monitor was able to detect very deep planes of anaesthesia when BSR occurs, but was not able to distinguish between the intermediate anaesthetic planes likely to be used in clinical anaesthesia.


Subject(s)
Anesthesia, Intravenous/veterinary , Anesthetics, Intravenous/administration & dosage , Propofol/administration & dosage , Anesthetics, Intravenous/blood , Anesthetics, Intravenous/pharmacology , Animals , Blinking/drug effects , Consciousness Monitors/veterinary , Dogs , Electroencephalography/drug effects , Electroencephalography/veterinary , Electromyography/drug effects , Electromyography/veterinary , Female , Hemodynamics/drug effects , Infusions, Intravenous/veterinary , Male , Propofol/blood , Propofol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...