Pharmacokinetics of Ovarian Steroids in Sprague-Dawley Rats After Acute Exposure to 2,3,7,8-Tetrachlorodibenzo- P-Dioxin (TCDD)

Total Page:16

File Type:pdf, Size:1020Kb

Pharmacokinetics of Ovarian Steroids in Sprague-Dawley Rats After Acute Exposure to 2,3,7,8-Tetrachlorodibenzo- P-Dioxin (TCDD) Vol. 3, No. 2 131 ORIGINAL PAPER Pharmacokinetics of ovarian steroids in Sprague-Dawley rats after acute exposure to 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) Brian K. Petroff 1,2,3 and Kemmy M. Mizinga4 2Department of Molecular and Integrative Physiology,Physiology, 3Center for Reproductive Sciences, University of Kansas Medical Center, Kansas City, KS 66160. 4Department of Pharmacology,Pharmacology, University of Health Sciences, Kansas City,City, MO 64106 Received: 3 June 2003; accepted: 28 June 2003 SUMMARY 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces abnormalities in ste- roid-dependent processes such as mammary cell proliferation, gonadotropin release and maintenance of pregnancy. In the current study, the effects of TCDD on the pharmacokinetics of 17ß-estradiol and progesterone were examined. Adult Sprague-Dawley rats were ovariectomized and pretreated with TCDD (15 µg/kg p.o.) or vehicle. A single bolus of 17ß-estradiol (E2, 0.3 µmol/kg i.v.) or progesterone (P4, 6 µmol/kg i.v.) was administered 24 hours after TCDD and blood was collected serially from 0-72 hours post- injection. Intravenous E2 and P4 in DMSO vehicle had elimination half-lives of approximately 10 and 11 hours, respectively. TCDD had no signifi cant effect on the pharmacokinetic parameters of P4. The elimination constant 1Corresponding author: Center for Reproductive Sciences, Department of Molecular and Integra- tive Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA; e-mail: [email protected] Copyright © 2003 by the Society for Biology of Reproduction 132 TCDD and ovarian steroid pharmacokinetics and clearance of E2 were decreased by TCDD while the elimination half-life, volume of distribution and area under the time*concentration curve were not altered signifi cantly. Overall, these results indicate that diminished serum progesterone and estradiol concentrations following exposure to TCDD are due primarily to actions on steroid synthesis and release rather than any al- terations in pharmacokinetics. Reproductive Biology 2003 3 (2): 131-141. Key words: TCDD, dioxin, ovary, progesterone, estradiol, pharmacokinetics INTRODUCTION The environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has been shown to block ovulation through alterations in preovulatory go- nadotropin release [4, 5, 15, 17] and has direct actions on the ovary as well [3, 4, 8, 9, 13, 14, 15, 20]. This blockade of ovulation following exposure to TCDD is accompanied by aberrations in serum concentrations of ovarian steroids [17]. This is in agreement with other studies that have found de- creases in serum estradiol and progesterone following acute exposure to TCDD [13, 14, 17]. Decreased steroid concentrations induced by TCDD may be due to alterations in gonadotropic support of ovarian steroidogenesis [17], direct impact on steroid synthesis [3, 8, 9, 13, 14] or altered distribu- tion, metabolism and elimination [16]. In this study, we tested the effects of TCDD on the pharmacokinetics of intravenously administered 17β-estradiol (E2) and progesterone (P4) in ovariectomized adult rats. MATERIALS AND METHODS Animals and Treatments Adult female Sprague-Dawley rats (80-100 days old, 250-350 g, Charles River Laboratories, USA) were supplied by a commercial vendor and housed under scheduled lighting (12:12 hours light:dark) and provided with rodent chow (Purina, USA) and water ad libitum. Chemicals were supplied by Sigma (St. Louis, MO, USA) unless stated otherwise. TCDD was graciously donated by Dr. Karl K. Rozman (University of Kansas Medical Center). Petroff and Mizinga 133 Animal protocols were approved by the Institutional Animal Care and Use Committee at the University of Kansas Medical Center. Rats were ovariectomized under a surgical plane of anesthesia (5 mg xy- lazine and 50 mg ketamine/kg i.p., Abbott Laboratories, North Chicago, IL, USA) via a paralumbar approach. A jugular cannula was placed to the level of the right atrium, fl ushed with heparinized saline and fi lled with a heparin- ized glycerol (50 IU/ml) lock solution to maintain cannula patency during the 3 day recovery period. At the end of this period rats (n = 4 per group) were pretreated with TCDD (15 µg/kg p.o.) or corn oil vehicle and 24 hours later received a single dose of E2 (0.3 µmol/kg i.v.) or P4 (6 µmol/kg i.v.) in DMSO vehicle. Steroids were delivered in 200 µl of DMSO (approximately 0.8 ml DMSO/kg i.v.), a dose below the commonly used therapeutic range for this compound [1]. These doses of E2 and P4 were chosen based on the administration of estradiol cypionate and P4 in previous studies [5, 15, 16] and were intended to provide steroid concentrations across the physiological range. The dose of TCDD chosen induced alterations in steroid concentra- tions in previous studies [15, 16, 17]. Blood (100-300 µl) was collected at 0, 0.25, 0.75, 2.5, 7.5, 24 and 72 hours post-injection. Blood removal was limited to less than 5% of estimated total blood volume and hematocrit was measured following each blood collection to insure that iatrogenic anemia was avoided. Serum was stored at –20oC until hormone measurement by previously validated radioimmunoassay. Radioimmunoassay Serum concentrations of E2 and P4 were measured by specifi c radioimmu- noassay following extraction with diethyl ether as described previously [16, 27]. Both intrassay and interassay coeffi cients of variation were less than 10% for progesterone and estradiol. Data Analysis Serum E2 and P4 concentration data were subjected to noncompartmental analysis for initial determination of pharmacokinetic parameters. Briefl y, 134 TCDD and ovarian steroid pharmacokinetics area under the concentration vs. time curve (AUC0-inf) andand areaarea uundernder tthehe fi r srstt moment curve (AUMC0-inf) werewere calculatedcalculated usingusing thethe linearlinear trapezoidaltrapezoidal rulerule [19, 29] applied through the computer program “PK Functions for Microsoft Excel” (J.I. Usansky et al., Dept. of Pharmacokinetics and Drug metabolism, Allergan, Irvine, CA). Half-life (t1⁄2) and elimination rate constants (Kelim) were calculated by regression of the semi-logarithmic concentration vs. time data [7] using the same computer program. Total systemic clearance (CL), calculated from Dose/AUC0-inf, meanmean residenceresidence timetime (MRT),(MRT), calculatedcalculated fromfrom AUMC/AUC, and steady state volume of distribution (Vss), calculated from MRT*CL and other noncompartmental pharmacokinetic parameters [22] were evaluated but not reported because they yielded similar conclusions to those based on subsequent compartmental analysis. Serum E2 and P4 concentration data were then subjected to compartmental analysis to obtain micro-rate constants and volumes of distribution. Both E2 and P4 concentra- tion vs. time curves exhibited bi-exponential decline best described by two- compartment models represented by the equation: C = Ae-αt + Be-βt (where C = concentration of E2 or P4 in serum, t = time, A = ordinate intercept of distribution curve, α = hybrid rate constant (slope) of distribution curve, B = ordinate intercept of elimination curve, β = hybrid rate constant (slope) of elimination curve) [2, 11]. Volume of distribution (Vd) was calculated from Dose/(A+B) [30]. Serum steroid concentrations were analyzed by ANOVA for a repeated measures design. Effects tested included time and TCDD and their interaction. Comparisons yielding p≤0.05 were considered signifi cant. The elimination half-life, elimination constant, volume of distribution and clearance of P4 or E2 in vehicle and TCDD-treated rats following adminis- tration of E2 or P4 were compared using a student t –test. RESULTS 17ß-estradiol Concentrations of E2 in serum as measured by radioimmunoassay rose abruptly following intravenous administration of E2, reaching a measured peak of approximately 7000 pg/ml at 15 minutes post-administration Petroff and Mizinga 135 Fig. 1. Serum concentrations of 17β-estradiol (E2, log pg/ml) in ovariectomized rats receiving TCDD (15 µg/kg p.o.) or vehicle followed by E2, (n = 4). (fi g. 1). Data from one rat in the control group were excluded due to cannula failure. E2 was undetectable in these animals by 72 hours post-administration and was eliminated with an elimination half-life of 8-13 hours for E2. While the calculated elimination constant and clearance of E2 were decreased in TCDD-treated animal, the dioxin had no signifi cant impact on the elimina- tion half-life, area under the curve and volume of distribution of serum E2 following the intravenous administration of estradiol-17ß (tab. 1). Progesterone Serum P4 rose abruptly following intravenous administration in DMSO vehicle and reached a measured peak of approximately 400 ng/ml by 15 136 TCDD and ovarian steroid pharmacokinetics Table 1. Pharmacokinetic parameters of serum estradiol following intravenous administration of 17ß-estradiol to ovariectomized female rats pretreated with TCDD or vehicle Control TCDD (15 µg/kg p.o.) Mean SEM Mean SEM t1/2 8.48 3.63 13.81 0.84 AUC (pg*h/ml) 17848 4197 22300 2106 CL(ml/h) 1613 574 986* 83 -1 Kelim (hr ) 0.11 0.04 0.051* 0.003 Vd (l/kg) 11.01 0.65 11.39 0.38 t1/2 = half life; AUC = area under curve; CL = clearance; Kelim = eliminationelimination cconstant;onstant; TCDD = 2,3,7,8-tetrachlorodibenzo-p-dioxin; Vd = volume of distribution *a signifi cant (p<0.05) difference between controls and TCDD-treated animals minutes post-administration (fi g. 2). The elimination half-life of intrave- nously administered P4
Recommended publications
  • Depo-Estradiol (Estradiol Cypionate) Injection
    Market Applicability Market DC GA KY MD NJ NY WA Applicable X X X X X X NA Depo-Estradiol (estradiol cypionate) injection Override(s) Approval Duration Prior Authorization 1 year Medications Depo-Estradiol (estradiol cypionate) injection APPROVAL CRITERIA Requests for Depo-Estradiol (estradiol cypionate) injection may be approved when the following criteria are met: I. Individual is using to treat moderate to severe vasomotor symptoms associated with menopause; OR II. Individual has been diagnosed with hypoestrogenism due to hypogonadism; OR III. Individual has a diagnosis of gender dysphoria or gender identity disorder (WPATH 2012, Endocrine Society 2017) ; AND IV. Individual is 16 years of age or older; AND V. The goal of treatment is male-to-female gender reassignment. Requests for Depo-Estradiol (estradiol cypionate) injection may not be approved for any of the following: I. Individual has undiagnosed abnormal genital bleeding; OR II. Individual has known or suspected cancer of the breast; OR III. Individual has known or suspected estrogen-dependent neoplasia; OR IV. Individual has active deep vein thrombosis, pulmonary embolism or history of these conditions; OR V. Individual has active or recent (within the past year) arterial thromboembolic disease (such as stroke or myocardial infarction); OR VI. Individual has liver dysfunction or disease. PAGE 1 of 2 02/24/2020 New Program Date 02/24/2020 CRX-ALL-0519-20 This policy does not apply to health plans or member categories that do not have pharmacy benefits, nor does it apply to Medicare. Note that market specific restrictions or transition-of-care benefit limitations may apply.
    [Show full text]
  • Pms-ESTRADIOL VALERATE INJECTION
    PRODUCT MONOGRAPH Prpms-ESTRADIOL VALERATE INJECTION Estradiol Valerate 10 mg/mL Estrogen PHARMASCIENCE INC. Date of Revision: 6111 Royalmount Avenue, Suite 100 June 30, 2009 Montreal, Quebec H4P 2T4 Control Number: 120632 Table of Contents PART I: HEALTH PROFESSIONAL INFORMATION……………………………...3 SUMMARY PRODUCT INFORMATION……………………………………….3 INDICATIONS AND CLINICAL USE…………………………………………...3 CONTRAINDICATIONS…………………………………………………………4 WARNINGS AND PRECAUTIONS…………………………………….……….5 ADVERSE REACTIONS………………………………………………………...13 DRUG INTERACTIONS………………………………………………………...15 DOSAGE AND ADMINISTRATION…………………………………………...17 OVERDOSAGE………………………………………………………………….19 ACTION AND CLINICAL PHARMACOLOGY……………………………….19 STORAGE AND STABILITY…………………………………………………...22 DOSAGE FORMS, COMPOSITION AND PACKAGING……………………..22 PART II: SCIENTIFIC INFORMATION…………………………………………….23 PHARMACEUTICAL INFORMATION………………………………………...23 CLINICAL TRIALS……………………………………………………………...24 DETAILED PHARMACOLOGY………………………………………………..24 REFERENCES…………………………………………………………………...25 PART III: CONSUMER INFORMATION……………………………………………………27 2 PRODUCT MONOGRAPH Prpms- ESTRADIOL VALERATE INJECTION (Estradiol Valerate) 10 mg/mL PART I: HEALTH PROFESSIONAL INFORMATION SUMMARY PRODUCT INFORMATION Route of Dosage Form / Strength Clinically Relevant Nonmedicinal Administration Ingredients Intramuscular Injection / 10 mg/mL Sesame Oil For a complete listing see Dosage Forms, Composition and Packaging section. INDICATIONS AND CLINICAL USE pms-ESTRADIOL VALERATE INJECTION is indicated in the treatment of: I. amenorrhea (primary
    [Show full text]
  • Estradiol-17Β Pharmacokinetics and Histological Assessment Of
    animals Article Estradiol-17β Pharmacokinetics and Histological Assessment of the Ovaries and Uterine Horns following Intramuscular Administration of Estradiol Cypionate in Feral Cats Timothy H. Hyndman 1,* , Kelly L. Algar 1, Andrew P. Woodward 2, Flaminia Coiacetto 1 , Jordan O. Hampton 1,2 , Donald Nickels 3, Neil Hamilton 4, Anne Barnes 1 and David Algar 4 1 School of Veterinary Medicine, Murdoch University, Murdoch 6150, Australia; [email protected] (K.L.A.); [email protected] (F.C.); [email protected] (J.O.H.); [email protected] (A.B.) 2 Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne 3030, Australia; [email protected] 3 Lancelin Veterinary Hospital, Lancelin 6044, Australia; [email protected] 4 Department of Biodiversity, Conservation and Attractions, Locked Bag 104, Bentley Delivery Centre 6983, Australia; [email protected] (N.H.); [email protected] (D.A.) * Correspondence: [email protected] Received: 7 September 2020; Accepted: 17 September 2020; Published: 21 September 2020 Simple Summary: Feral cats (Felis catus) have a devastating impact on Australian native fauna. Several programs exist to control their numbers through lethal removal, using tools such as baiting with toxins. Adult male cats are especially difficult to control. We hypothesized that one way to capture these male cats is to lure them using female cats. As female cats are seasonal breeders, a method is needed to artificially induce reproductive (estrous) behavior so that they could be used for this purpose year-round (i.e., regardless of season).
    [Show full text]
  • Drug and Chemical Reference Guideline
    WISCONSIN DEPARTMENT OF AGRICULTURE, TRADE AND CONSUMER PROTECTION DIVISION OF FOOD AND RECREATIONAL SAFETY TITLE: Drug and Chemical Reference Guideline Document #: Replaces: New Revision Date: 1/21/2020 Page 1 of 6 General Guidelines: These drugs and substances are not to be used or • Drugs and administration equipment stored to not stored on dairy farms. These drugs are not eligible for contaminate milk or milk contact surfaces ELU privileges by veterinarians. • Non-lactating drugs separated from lactating drugs • Chloramphenicol using separate shelves or cabinets • Clenbuterol • Drugs properly labeled (see label requirements • Colloidal Silver below) • Diethylstilbestrol (DES) • Locked drug cabinets must be made accessible for • Dimethysulfoxide (DMSO) inspection • Dimetridazole • Drugs for animal species other than cattle must not • Dipyrone (Novine, No Pain) be stored in dairy facility • Estradiol Cypionate (ECP) • All bottles or packages in case lots properly labeled • Fluoroquinolones (sarafloxacin-Saraflox, orbafloxacin-Orbox, danofloxacin mesylate-A 180) Drug Labeling Requirements: • Furazolidone, Nitrofurazone, other Nitrofurans OTC Drugs: (over the counter) • Glycopeptides (vancomycin) • Name of Drug • Iononphores (Lasalocid) permitted for non-lactating • Active ingredients • Ipronidazole • Directions for use • Metronidazole and other Nitroimidazoles • Withholding/withdrawal or discard time for meat or • Phenylbutazone, “bute” (prohibited in dairy animals milk (even if zero) 20 months of age or older - no ELU allowed.) • Name
    [Show full text]
  • Euthanasia of Experimental Animals
    EUTHANASIA OF EXPERIMENTAL ANIMALS • *• • • • • • • *•* EUROPEAN 1COMMISSIO N This document has been prepared for use within the Commission. It does not necessarily represent the Commission's official position. A great deal of additional information on the European Union is available on the Internet. It can be accessed through the Europa server (http://europa.eu.int) Cataloguing data can be found at the end of this publication Luxembourg: Office for Official Publications of the European Communities, 1997 ISBN 92-827-9694-9 © European Communities, 1997 Reproduction is authorized, except for commercial purposes, provided the source is acknowledged Printed in Belgium European Commission EUTHANASIA OF EXPERIMENTAL ANIMALS Document EUTHANASIA OF EXPERIMENTAL ANIMALS Report prepared for the European Commission by Mrs Bryony Close Dr Keith Banister Dr Vera Baumans Dr Eva-Maria Bernoth Dr Niall Bromage Dr John Bunyan Professor Dr Wolff Erhardt Professor Paul Flecknell Dr Neville Gregory Professor Dr Hansjoachim Hackbarth Professor David Morton Mr Clifford Warwick EUTHANASIA OF EXPERIMENTAL ANIMALS CONTENTS Page Preface 1 Acknowledgements 2 1. Introduction 3 1.1 Objectives of euthanasia 3 1.2 Definition of terms 3 1.3 Signs of pain and distress 4 1.4 Recognition and confirmation of death 5 1.5 Personnel and training 5 1.6 Handling and restraint 6 1.7 Equipment 6 1.8 Carcass and waste disposal 6 2. General comments on methods of euthanasia 7 2.1 Acceptable methods of euthanasia 7 2.2 Methods acceptable for unconscious animals 15 2.3 Methods that are not acceptable for euthanasia 16 3. Methods of euthanasia for each species group 21 3.1 Fish 21 3.2 Amphibians 27 3.3 Reptiles 31 3.4 Birds 35 3.5 Rodents 41 3.6 Rabbits 47 3.7 Carnivores - dogs, cats, ferrets 53 3.8 Large mammals - pigs, sheep, goats, cattle, horses 57 3.9 Non-human primates 61 3.10 Other animals not commonly used for experiments 62 4.
    [Show full text]
  • Pp375-430-Annex 1.Qxd
    ANNEX 1 CHEMICAL AND PHYSICAL DATA ON COMPOUNDS USED IN COMBINED ESTROGEN–PROGESTOGEN CONTRACEPTIVES AND HORMONAL MENOPAUSAL THERAPY Annex 1 describes the chemical and physical data, technical products, trends in produc- tion by region and uses of estrogens and progestogens in combined estrogen–progestogen contraceptives and hormonal menopausal therapy. Estrogens and progestogens are listed separately in alphabetical order. Trade names for these compounds alone and in combination are given in Annexes 2–4. Sales are listed according to the regions designated by WHO. These are: Africa: Algeria, Angola, Benin, Botswana, Burkina Faso, Burundi, Cameroon, Cape Verde, Central African Republic, Chad, Comoros, Congo, Côte d'Ivoire, Democratic Republic of the Congo, Equatorial Guinea, Eritrea, Ethiopia, Gabon, Gambia, Ghana, Guinea, Guinea-Bissau, Kenya, Lesotho, Liberia, Madagascar, Malawi, Mali, Mauritania, Mauritius, Mozambique, Namibia, Niger, Nigeria, Rwanda, Sao Tome and Principe, Senegal, Seychelles, Sierra Leone, South Africa, Swaziland, Togo, Uganda, United Republic of Tanzania, Zambia and Zimbabwe America (North): Canada, Central America (Antigua and Barbuda, Bahamas, Barbados, Belize, Costa Rica, Cuba, Dominica, El Salvador, Grenada, Guatemala, Haiti, Honduras, Jamaica, Mexico, Nicaragua, Panama, Puerto Rico, Saint Kitts and Nevis, Saint Lucia, Saint Vincent and the Grenadines, Suriname, Trinidad and Tobago), United States of America America (South): Argentina, Bolivia, Brazil, Chile, Colombia, Dominican Republic, Ecuador, Guyana, Paraguay,
    [Show full text]
  • Estradiol Cypionate: Summary Report
    Estradiol Cypionate: Summary Report Item Type Report Authors Yuen, Melissa V.; Gianturco, Stephanie L.; Pavlech, Laura L.; Storm, Kathena D.; Yoon, SeJeong; Mattingly, Ashlee N. Publication Date 2019-12 Keywords Estradiol cypionate; Compounding; Food, Drug, and Cosmetic Act, Section 503B; Food and Drug Administration; Outsourcing facility; Drug compounding; Legislation, Drug; United States Food and Drug Administration Rights Attribution-NoDerivatives 4.0 International Download date 27/09/2021 15:15:19 Item License http://creativecommons.org/licenses/by-nd/4.0/ Link to Item http://hdl.handle.net/10713/12106 Summary Report Estradiol cypionate Prepared for: Food and Drug Administration Clinical use of bulk drug substances nominated for inclusion on the 503B Bulks List Grant number: 2U01FD005946 Prepared by: University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI) University of Maryland School of Pharmacy December 2019 This report was supported by the Food and Drug Administration (FDA) of the U.S. Department of Health and Human Services (HHS) as part of a financial assistance award (U01FD005946) totaling $2,342,364, with 100 percent funded by the FDA/HHS. The contents are those of the authors and do not necessarily represent the official views of, nor an endorsement by, the FDA/HHS or the U.S. Government. 1 Table of Contents REVIEW OF NOMINATIONS ................................................................................................... 4 METHODOLOGY ...................................................................................................................
    [Show full text]
  • The 3,4-Quinones of Estrone and Estradiol Are the Initiators of Cancer Whereas Resveratrol and N-Acetylcysteine Are the Preventers
    International Journal of Molecular Sciences Review The 3,4-Quinones of Estrone and Estradiol Are the Initiators of Cancer whereas Resveratrol and N-acetylcysteine Are the Preventers Ercole Cavalieri 1 and Eleanor Rogan 2,* 1 Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA; [email protected] 2 Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, 984388 Nebraska Medical Center, Omaha, NE 68198-4388, USA * Correspondence: [email protected] Abstract: This article reviews evidence suggesting that a common mechanism of initiation leads to the development of many prevalent types of cancer. Endogenous estrogens, in the form of catechol estrogen-3,4-quinones, play a central role in this pathway of cancer initiation. The catechol estrogen- 3,4-quinones react with specific purine bases in DNA to form depurinating estrogen-DNA adducts that generate apurinic sites. The apurinic sites can then lead to cancer-causing mutations. The process of cancer initiation has been demonstrated using results from test tube reactions, cultured mammalian cells, and human subjects. Increased amounts of estrogen-DNA adducts are found not only in people with several different types of cancer but also in women at high risk for breast cancer, indicating that the formation of adducts is on the pathway to cancer initiation. Two compounds, resveratrol, and N-acetylcysteine, are particularly good at preventing the formation of estrogen-DNA Citation: Cavalieri, E.; Rogan, E. The adducts in humans and are, thus, potential cancer-prevention compounds. 3,4-Quinones of Estrone and Estradiol Are the Initiators of Cancer whereas Keywords: cancer initiation; cancer prevention; estrogens; estrogen-DNA adducts; Resveratrol and N-acetylcysteine Are N-acetylcysteine; resveratrol the Preventers.
    [Show full text]
  • Diluted Isoflurane As a Suitable Alternative for Diethyl Ether for Rat Anaesthesia in Regular Toxicology Studies
    FULL PAPER Laboratory Aminal Science Diluted Isoflurane as a Suitable Alternative for Diethyl ether for Rat Anaesthesia in Regular Toxicology Studies Toshiaki NAGATE1)*, Tomonobu CHINO1), Chizuru NISHIYAMA1), Daisuke OKUHARA1), Toru TAHARA1), Yoshimasa MARUYAMA1), Hiroko KASAHARA1), Kayoko TAKASHIMA1), Sayaka KOBAYASHI1), Yoshiyuki MOTOKAWA1), Shin-ichi MUTO1) and Junji KURODA1) 1)Toxicology Research Laboratory, R&D, Kissei Pharmaceutical Co., Ltd., 2320–1 Maki, Hotaka, Azumino-City, Nagano-Pref. 399–8305, Japan (Received 6 October 2006/Accepted 20 July 2007) ABSTRACT. Despite its explosive properties and toxicity to both animals and humans, diethyl ether is an agent long used in Japan in the anaesthesia jar method of rat anaesthetises. However, in response to a recent report from the Science Council of Japan condemning diethyl ether as acceptable practice, we searched for an alternative rat anaesthesia method that provided data continuous with pre-existing regular toxicology studies already conducted under diethyl ether anaesthesia. For this, we examined two candidates; 30% isoflurane diluted with propylene glycol and pentobarbitone. Whereas isoflurane is considered to be one of the representatives of modern volatile anaesthetics, the method of propylene glycol-diluted 30% isoflurane used in this study was our modification of a recently reported method revealed to have several advantages as an inhalation anaesthesia. Intraperitoneal pentobarbitone has long been accepted as a humane method in laboratory animal anaesthesiology. These 2 modalities were scrutinized in terms of consistency of haematology and blood chemistry with previous results using ether. We found that pentobarbitone required a much longer induction time than diethyl ether, which is suspected to be the cause of fluctuations in several haematological and blood chemical results.
    [Show full text]
  • Drug Interaction in Anaesthesia a Review
    DRUG INTERACTION IN ANAESTHESIA A REVIEW M. M. GHONEIM, M.B., B.CH., F.F.A.R.C.S. = RECENTLY, THE P~OBLE.',~s and hazards associated with the interaction between drugs have received widespread attention. The potential for the interaction has certainly increased in recent years. It has been demonstrated that the average patient will receive eight different drugs during one hospitalization. 1 In many in- stances, one drug may profoundly modify the action of another. In such drug inter- actions the effect of one may be prevented, or its action may be intensified. Though sometimes beneficial, drug interactions are most often recognized when they in- crease mortality or morbidity. They form around 19-22 per cent of causes of adverse drug reactionsd There are a number of good general reviews on drug interac- tions, ~-6 but there are not many which are concerned primarily with the practice of anaesthesia. 7,8 The anaesthetist uses a wide variety of pharmacologically active drugs which may interact with one another or with other drugs the patient is receiving. The multitudes of possible interactions limit the possibility of reviewing each individual drug interaction. This also entails a lot of repetition and would not keep pace with the number of new drugs introduced into the market every month. Our aim is elucidation of the principles and mechanisms involved with examples which are of interest to the anaesthetist. Several mechanisms of interaction are recognized. 1. A direct physical or chemical interaction A familiar example is the neutralization of heparin with protamine. This is an example, also, of a useful drug interaction.
    [Show full text]
  • Plasma Progesterone Concentrations and Ovarian Histology in Prairie Deermice (Peromyscus Maniculatus Bairdii) from Experimental Laboratory Populations
    W&M ScholarWorks Dissertations, Theses, and Masters Projects Theses, Dissertations, & Master Projects 1973 Plasma Progesterone Concentrations and Ovarian Histology in Prairie Deermice (Peromyscus maniculatus Bairdii) from Experimental Laboratory Populations Barry Douglas Albertson College of William & Mary - Arts & Sciences Follow this and additional works at: https://scholarworks.wm.edu/etd Part of the Biology Commons, and the Endocrinology Commons Recommended Citation Albertson, Barry Douglas, "Plasma Progesterone Concentrations and Ovarian Histology in Prairie Deermice (Peromyscus maniculatus Bairdii) from Experimental Laboratory Populations" (1973). Dissertations, Theses, and Masters Projects. Paper 1539624808. https://dx.doi.org/doi:10.21220/s2-7nsc-4k85 This Thesis is brought to you for free and open access by the Theses, Dissertations, & Master Projects at W&M ScholarWorks. It has been accepted for inclusion in Dissertations, Theses, and Masters Projects by an authorized administrator of W&M ScholarWorks. For more information, please contact [email protected]. PLASMA PPDGESTEPONE CDNCCNTPATIONS AND OVARIAN HISTOLOGY I ’ IN PRAIRIE DEERMICE (PE.RCITYSCUS MANIOJLATUS RAIRDII) FROM EXPERIMENTAL LABORATORY POPULATIONS A Thesis Presented to The Faculty of the Department of Biology The College of William and Mary in Virginia In Partial Fulfillment Of the Requirements for the Degree of Master of Arts by Barry Douglas Albertson APPROVAL SHEET This thesis is submitted in partial fulfillment of the requirements for the degree of Master of Arts t (XATl-L, P. 0 iLiis X]Author Approved, July, 1973 (S- v u . EricOik L. Bradley, Ph. D. C. RicnardTTferriiah, Ph. ■W)D. fl&itjh (- f- Robert E. u. Black, Phi D. ACKNOWLEDGMENTS The author would like to express his appreciation to Dr.
    [Show full text]
  • Diethyl Ether) (CASRN 60-29-7)
    EPA/690/R-09/022F l Final 1-20-2009 Provisional Peer Reviewed Toxicity Values for Ethyl ether (Diethyl ether) (CASRN 60-29-7) Superfund Health Risk Technical Support Center National Center for Environmental Assessment Office of Research and Development U.S. Environmental Protection Agency Cincinnati, OH 45268 ACRONYMS AND ABBREVIATIONS bw body weight cc cubic centimeters CD Caesarean Delivered CERCLA Comprehensive Environmental Response, Compensation and Liability Act of 1980 CNS central nervous system cu.m cubic meter DWEL Drinking Water Equivalent Level FEL frank-effect level FIFRA Federal Insecticide, Fungicide, and Rodenticide Act g grams GI gastrointestinal HEC human equivalent concentration Hgb hemoglobin i.m. intramuscular i.p. intraperitoneal IRIS Integrated Risk Information System IUR inhalation unit risk i.v. intravenous kg kilogram L liter LEL lowest-effect level LOAEL lowest-observed-adverse-effect level LOAEL(ADJ) LOAEL adjusted to continuous exposure duration LOAEL(HEC) LOAEL adjusted for dosimetric differences across species to a human m meter MCL maximum contaminant level MCLG maximum contaminant level goal MF modifying factor mg milligram mg/kg milligrams per kilogram mg/L milligrams per liter MRL minimal risk level MTD maximum tolerated dose MTL median threshold limit NAAQS National Ambient Air Quality Standards NOAEL no-observed-adverse-effect level NOAEL(ADJ) NOAEL adjusted to continuous exposure duration NOAEL(HEC) NOAEL adjusted for dosimetric differences across species to a human NOEL no-observed-effect level OSF
    [Show full text]