Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Anaesthesia ; 67(3): 226-31, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22321076

ABSTRACT

The Airtraq(™) optical laryngoscope became available in paediatric sizes in the UK in May 2008. We conducted a randomised, controlled trial comparing the Airtraq with conventional laryngoscopy during routine anaesthesia in children. We hypothesised that the Airtraq laryngoscope would perform as well as conventional laryngoscopy. Sixty patients (20 infants and 40 children) were recruited. The mean (SD) intubation time using the Airtraq was longer than conventional laryngoscopy overall (47.3 (32.6) vs 26.3 (11.5) s; p=0.002), though the difference was only significant for children (p=0.003) and not for infants (p=0.29). The Airtraq provided a better view of the larynx compared with conventional laryngoscopy (in infants (percentage of glottic opening scores 100 (95-100 [90-100]) vs 77 (50-90 [40-100]), respectively; p=0.001; visual analogue scores for field of view 9.2 (9.2-9.5 [8.2-10.0]) vs 6.8 (5.1-8.0 [4.7-10.0]), respectively; p=0.001). In children, the Airtraq provided a similar view of the larynx (percentage of glottic opening scores 100 (100-100 [40-100]) vs 100 (90-100 [50-100]), respectively; visual analogue scores for field of view 9.2 (8.6-10.0 [7.0-10.0]) vs 9.2 (8.6-10.0 [5.6-10.0]), respectively; both p>0.05), compared with conventional laryngoscopy.


Subject(s)
Laryngoscopes , Child , Child, Preschool , Humans , Infant , Intubation, Intratracheal/instrumentation , Laryngoscopy
2.
Anaesthesia ; 57(5): 520-1, 2002 May.
Article in English | MEDLINE | ID: mdl-12004825
3.
Proc Soc Exp Biol Med ; 217(3): 317-21, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9492341

ABSTRACT

There is considerable interest in the role(s) of plant-derived compounds such as bioflavonoids in regulating steroid hormone action in mammals, and in particular, the possible effects of the bioflavonoids on the growth of steroid-dependent breast and prostate tumors and on possible abnormal development of steroid-sensitive tissues. Studies of the hormone-like actions of bioflavonoids often use fetal or neonatal rats, which contain high levels of serum alpha-fetoprotein (AFP), a protein that binds estradiol with a Kd approximately 5 x 10(-9) M. Interaction of bioflavonoids with AFP could affect the availability of estrogens to estrogen-responsive cells, as well as the actions of bioflavonoids. These considerations motivated us to study the effect of several flavonoids (quercetin, rutin, naringenin, chrysin, apigenin, kaempferol, myricetin, morin, fisetin) and isoflavonoids (daidzein, genistein) on estrogen binding to rat AFP. We found that naringenin, a flavanone, and quercetin and kaempferol, flavonols, inhibit estrogen binding to AFP with apparent Kds of about 5 x 10(-7) M. To our surprise, the two isoflavonoids, daidzein and genistein, have Kds of about 5 x 10(-6) M for AFP. This 10-fold [correction of 1Q-fold] difference in affinity for AFP between flavonoids and isoflavonoids suggests that AFP has a specificity for the flavonoid structure. Moreover, the affinities of bioflavonoids for rat AFP are sufficiently high to suggest that flavonoids and isoflavonoids could modulate estradiol and estrone binding to rat AFP in vivo, when present at dietary levels. Additionally, the potency of the plant estrogens may be altered by binding to AFP. The flavonoids that we tested have different hydroxyl and glucoside substituents on the A, B, and C rings, which allows us to define some of the spatial requirements for binding to AFP. We find that 5,7-hydroxyl groups in ring A and a 4'-hydroxyl group in ring B are important for binding to AFP. This information, combined with molecular modeling studies, may elucidate the molecular basis for recognition of flavonoids and estrogens by AFP. Also, these findings indicate that the flavonoid levels in the diet need to be considered in studies of the effects of various xenobiotics and endocrine manipulations on experimental animals, particularly during development when serum estrogen binding protein concentrations are often elevated. Finally, bioflavonoids should be useful tools for understanding the variety of estrogen actions initiated by different structural classes of estrogens.


Subject(s)
Estrogens/metabolism , Flavonoids/pharmacology , alpha-Fetoproteins/metabolism , Animals , Rats , Structure-Activity Relationship
4.
Biol Reprod ; 56(5): 1239-44, 1997 May.
Article in English | MEDLINE | ID: mdl-9160724

ABSTRACT

In the developing rodent uterus, the estrogen agonist activity of triphenylethylene antiestrogens such as tamoxifen alters uterine luminal epithelium morphology and inhibits uterine gland genesis. We examined uterine growth and differentiation in female offspring from date-mated Sprague-Dawley rats given the structurally related antiestrogen, toremifene, by s.c. injection in 10 microl of sesame oil on postnatal days (PND) 1-5, 10-14, or 20-24. Toremifene given on PND 10-14, a period of rapid uterine gland differentiation, caused a dose-related increase in uterine weight, tripled luminal epithelium cell height, and completely inhibited uterine gland development on PND 14 at doses of 10 microg or higher. Based on this dose-response analysis, a 10-microg dose of toremifene was chosen to assess uterine development after neonatal exposure (PND 1-5). Uterine weights and luminal epithelium cell heights were significantly increased by toremifene on PND 5 but returned to control levels by PND 26. Uterine gland numbers were reduced to 50% those of controls on PND 26. Dose-related uterine weight and luminal epithelium cell height increases were also observed in rats given toremifene on PND 20-24. This estrogen agonist activity of toremifene, revealed primarily in the uterine luminal epithelium, indicates that toremifene is developmentally toxic.


Subject(s)
Estrogen Antagonists/pharmacology , Toremifene/pharmacology , Uterus/drug effects , Uterus/growth & development , Age Factors , Animals , Animals, Newborn , Epithelium/drug effects , Epithelium/growth & development , Estrogen Antagonists/administration & dosage , Female , Rats , Rats, Sprague-Dawley , Toremifene/administration & dosage
5.
Biol Reprod ; 54(1): 160-7, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8838013

ABSTRACT

To assess the effects of the steroidal antiestrogen ICI 182,780 on postnatal uterine development, female Sprague-Dawley rats were given s.c. injections of ICI 182,780 (0.1-100 micrograms/rat) on each of postnatal days (PND) 10-14. ICI 182,780 inhibited uterine growth, as measured by uterine weight, in a dose-dependent manner but had no effect on either uterine luminal epithelium hypertrophy or gland genesis. Immunohistochemical analysis revealed that ICI 182,780 (10 micrograms) markedly reduced uterine estrogen receptor (ER) immunoreactivity in all uterine cell types while tamoxifen (10 micrograms) increased ER immunoreactivity, most notably in the luminal epithelium. In addition, tamoxifen increased uterine weight and induced luminal epithelium hypertrophy but inhibited uterine gland genesis--outcomes also seen with synthetic estrogens such as diethylstilbestrol. To test the hypothesis that these effects are a consequence of the estrogen agonist activity of tamoxifen, rats were cotreated with ICI 182,780 (10 micrograms, PND 8-14) and tamoxifen (10 micrograms, PND 10-14). ICI 182,780 greatly reduced or completely blocked tamoxifen-induced uterine weight gain, luminal epithelium hypertrophy, tamoxifen-induced ER immunoreactivity, and the inhibition of uterine gland genesis. ICI 182,780 given daily on PND 1-5 did not alter PND 5 uterine weight or uterine differentiation on PND 26. We conclude that postnatal exposure to ICI 182,780 does not affect uterine growth or differentiation at an age when the uterus is not dependent on estrogen for growth, i.e., PND 1-5, but does inhibit later endogenous estrogen-dependent uterine growth. The blockade of tamoxifen-induced uterine developmental alterations by ICI 182,780 demonstrates that these tamoxifen effects result from its estrogen agonist activity.


Subject(s)
Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogens/physiology , Tamoxifen/pharmacology , Uterus/drug effects , Uterus/growth & development , Animals , Epithelium/metabolism , Estradiol/pharmacology , Female , Fulvestrant , Immunohistochemistry , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Uterus/metabolism
6.
Proc Soc Exp Biol Med ; 208(3): 307-13, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7878071

ABSTRACT

Phytoestrogens found in clover, alfalfa, and soybeans have caused reproductive toxicity in several mammalian species. Other estrogens, such as diethylstilbestrol (DES), are developmental toxicants, reducing uterine estrogen receptor (ER) concentration, altering uterine growth, and eliciting reproductive tract abnormalities in the rat. The present study examines the effects of the phytoestrogens coumestrol and equol on the developing rat uterus. Various doses of these compounds were injected sc on postnatal days (PND) 1-5 or 1-10 to ascertain their effects on uterine weight and ER levels, and on PND 10-14 to determine their effects on uterine weight and gland genesis. Coumestrol (PND 1-5) was about 10(-3) as potent as DES in increasing uterine weight (wet or dry) while equol increased dry weight only, with a potency of 10(-5) that of DES. Although the 10 and 100 micrograms doses of coumestrol (PND 1-5 or 1-10) initially increased uterine wet weight, by PND 20 uterine weights either equaled or fell significantly below controls. The 100-micrograms dose of coumestrol (PND 1-5 or 1-10) reduced ER levels at all ages, while the 10-micrograms dose was not as effective. Equol (PND 1-5 or 1-10) did not affect ER levels. Premature uterine gland genesis occurred by PND 9 for the PND 1-5 100-micrograms coumestrol dose. When given on PND 10-14 (the critical period of gland genesis), 10 micrograms and 100 micrograms of coumestrol and 10 micrograms DES greatly increased uterine weight, while no effect was elicited by equol. Although coumestrol and equol inhibited uterine gland genesis in a dose-dependent manner, neither abolished gland genesis as did 10 micrograms of DES or tamoxifen. These data demonstrate that coumestrol elicits uterine biochemical and morphological toxicity much like DES. Equol decreased uterine gland number without increasing uterine wet weight or luminal epithelial hypertrophy, which is inconsistent with either an estrogenic or antiestrogenic action in the uterus.


Subject(s)
Chromans/toxicity , Coumestrol/toxicity , Isoflavones , Uterus/drug effects , Animals , Animals, Newborn , Diethylstilbestrol/toxicity , Dose-Response Relationship, Drug , Equol , Female , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/analysis , Uterus/growth & development
7.
Proc Soc Exp Biol Med ; 208(1): 67-71, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7892298

ABSTRACT

The phytoestrogens, coumestrol and equol, are weakly estrogenic. Here, we have examined their ability to induce responses in the neonatal rat uterus. Potent estrogens such as diethylstilbestrol (DES) and 17 beta-estradiol which initially double uterine weight on postnatal Day (PND) 5 when given on PND 1-5 subsequently reduce both uterine growth and gland development at later ages. In this study, Sprague-Dawley pups were treated neonatally (PND 1-5) with various doses of coumestrol and equol, and sacrificed at different ages to determine alterations in biochemical and morphological endpoints. Other rats were injected with the same compounds during the critical period of gland genesis (PND 10-14) to examine their effects on gland development. At the 100 micrograms coumestrol dose, on PND 1-5, premature gland development and increased uterine weight were observed. However, at later ages, uterine weight was significantly lowered and there was a severe suppression in the estrogen receptor (ER) levels. Equol lowered uterine weight at the later ages but did not affect ER levels. When given on PND 10-14, both coumestrol and equol caused a dose-dependent inhibition of gland genesis though not as severe as either DES or tamoxifen. Coumestrol was about 10(3) more potent than equol as an estrogen and behaved much like DES with respect to its effects on uterine weight, glands, and ER levels. At the doses used in this study, equol failed to demonstrate either estrogenic or antiestrogenic activity.


Subject(s)
Chromans/pharmacology , Coumestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Isoflavones , Uterus/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Equol , Female , Hypertrophy , Organ Size , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/biosynthesis , Uterus/growth & development , Uterus/pathology
8.
Proc Soc Exp Biol Med ; 208(1): 116-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7534422

ABSTRACT

Specific gene hypermethylation has been shown in DNA from neonatal rats exposed to the phytoestrogens, coumestrol, and equol. The pancreas is an organ in which estrogen receptors have been shown to be present. Studies have correlated the development of acute pancreatitis with rising levels of human estrogen binding proteins. Neonatal rats were dosed with 10 or 100 micrograms of coumestrol or equol on postnatal day (PND) 1-10. The animals were sacrificed at Day 15. The pancreas was excised and pancreatic acinar cells isolated for molecular analysis. DNA was isolated from the cells by lysis in TEN-9 buffer supplemented with proteinase K and 0.1% SDS. High molecular weight (HMW) DNA was digested with the methylated DNA specific restriction enzymes, Hpa II and Msp I, for determination of methylation profiles. Both coumestrol and equol at high doses caused hypermethylation of the c-H-ras proto-oncogene. No hypermethylation or hypomethylation was observed in the proto-oncogenes, c-myc or c-fos. Methylation is thought to be an epigenetic mechanism involved in the activation (hypomethylation) or inactivation (hypermethylation) of cellular genes which are known to play a role in carcinogenesis. Epidemiology studies have shown that equol may have anti-carcinogenic effects on some hormone-dependent cancers. Additional studies are needed to further understand the role of phytoestrogens and methylation in relation to pancreatic disorders.


Subject(s)
DNA/metabolism , Estrogens, Non-Steroidal/pharmacology , Gene Amplification/drug effects , Genes, ras/genetics , Isoflavones , Pancreas/drug effects , Animals , Animals, Newborn , Cells, Cultured , Chromans/pharmacology , Coumestrol/pharmacology , Deoxyribonuclease HpaII , Deoxyribonucleases, Type II Site-Specific , Equol , Female , Methylation/drug effects , Pancreas/chemistry , Pancreas/cytology , Phytoestrogens , Plant Preparations , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley
9.
Proc Soc Exp Biol Med ; 205(2): 146-53, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8108464

ABSTRACT

Autologous down-regulation of hormone receptors has been shown for several steroid hormones. We have previously shown estradiol (E2) regulates estrogen receptor (ER) in ovariectomized (OVX) rats. These studies have been extended to investigate the interaction between progesterone (P) and E2 in the regulation of ER and uterine weight. We implanted Silastic capsules containing varying concentrations of E2 (0.0005 mg E2/ml to 5.0 mg E2/ml of sesame oil) into adult female Sprague-Dawley rats one week after OVX. Simultaneously with implantation, P injections were started (sc in sesame oil) at doses of 1-40 mg/day for three days. In the absence of P, the 0.05 mg E2/ml implant significantly increased total ER levels (measured by cytosol and nuclear exchange assays) by 25%, while the two highest concentrations of E2 (0.5 and 5.0 mg E2/ml) significantly decreased cytosol and total ER levels by at least 40%. No P dose altered ER levels in OVX rats or in rats given E2 implants of 0.01 mg E2/ml or lower. At E2 implant concentrations of 0.05 mg E2/ml and higher, P decreased total ER levels 30%-50% compared to the appropriate E2-only controls. P increased uterine weight by 25% in OVX controls and in rats treated with E2 implants of 0.01 mg E2/ml and below. In contrast, P inhibited uterine weight gain induced by 0.05-5.0 mg/E2 ml implants by 20%-30%; maximal inhibition occurred at 5 mg/day of P and above. These data demonstrate that P increases uterine weight but does not alter ER concentration in rats with low E2 levels (OVX or low E2 concentration implants) but decreases uterine weight and down-regulates ER at higher E2 levels, regardless of whether ER is up-regulated or down-regulated by E2.


Subject(s)
Down-Regulation , Estradiol/pharmacology , Progesterone/pharmacology , Receptors, Estrogen/drug effects , Uterus/drug effects , Animals , Dose-Response Relationship, Drug , Drug Implants , Drug Interactions , Estradiol/administration & dosage , Estradiol/blood , Female , Injections, Subcutaneous , Organ Size/drug effects , Ovariectomy , Progesterone/administration & dosage , Progesterone/blood , Rats , Receptors, Estrogen/metabolism , Uterus/anatomy & histology , Uterus/metabolism
10.
J Steroid Biochem Mol Biol ; 42(1): 23-8, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1558818

ABSTRACT

Diethylstilbestrol (DES) treatment of female rats on postnatal days (PND) 1-5 reduces uterine growth, estrogen receptor (ER) level and gland number by PND 25, while daily DES treatment on PND 1-25 increases uterine growth 4-fold, further reduces ER level and completely suppresses gland formation. We now report the persistence of these effects in adults. By PND 60, uterine weight was 70% of controls in rats injected with DES on PND 1-5 but only 10% of controls in rats injected PND 1-10 or longer. In fact, uterine weights were the same on PND 10 and 60. Uterine gland numbers were reduced to 30% of controls in all DES-treated rats regardless of exposure length; however, luminal and glandular epithelial cell heights were reduced to less than 50 and 70%, respectively, of controls when DES was given on PND 1-25 but not when given on PND 1-5. Ovariectomy 7 days prior to sacrifice on PND 60 reduced uterine weight in controls by 67% and in rats injected with DES on PND 1-5 by 53%, but had no effect in rats injected with DES on PND 1-10. DES exposure at either PND 1-5 or 1-10 lowered ER levels by 35-50% at both 60 and 90 days. Treatment with a high dose of estradiol (E2) 1 week before sacrifice significantly down-regulated ER to the same concentration in all treatment groups at PND 60 and 90. Following E2 treatment, all groups also showed increased uterine weight at PND 60 and 90. These data show there is a short period of development (PND 5-10) in which further DES exposure indirectly inhibits uterine growth.


Subject(s)
Diethylstilbestrol/pharmacology , Receptors, Estrogen/metabolism , Uterus/drug effects , Aging , Animals , Diethylstilbestrol/administration & dosage , Down-Regulation/drug effects , Female , Organ Size/drug effects , Ovariectomy , Rats , Rats, Inbred Strains , Uterus/growth & development , Uterus/metabolism
11.
Proc Soc Exp Biol Med ; 196(3): 293-300, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1998006

ABSTRACT

We have previously shown that neonatal exposure of rats to pharmacologic doses of diethylstilbestrol via daily injections resulted in a significant decrease in the estrogen-binding capacity of the uterine estrogen receptor (ER). In this study, we examined the effects of physiologic and pharmacologic doses of estradiol (E2) administered to adult ovariectomized rats via Silastic implants. Two days after implantation, uteri were removed, weighted, and homogenized, and ER levels were determined in the supernatant (hydroxylapatite assay) and low-speed pellet (nuclear exchange assay). Implants containing E2 concentrations of 0.005 or 0.05 mg/ml increased cytosolic but not total ER-binding capacity, whereas 0.5 or 5.0 mg of E2/ml implants decreased the binding capacity of cytosol ER to 40% and total ER to 50% of control values. The 0.005-mg/ml dose increased cytosol ER without increasing uterine weight; all higher doses significantly increased uterine weight. Determination of ER protein by an ER radioimmunoassay showed the same extent of reduction of ER concentration as the binding assays, demonstrating that the loss in E2 binding capacity is homologous down-regulation. The down-regulation of ER was maximal at 24 hr and was completely reversible after implant removal, although the time required to recover from down-regulation was dose dependent. Uterine weight also returned to control levels slowly after implant removal. Neither the sedimentation rate of the down-regulated ER nor the Kd of the cytosolic ER changed following long-term implantation; however, the Kd of the nuclear ER decreased significantly. This is the first demonstration of in vivo homologous down-regulation of uterine ER. ER down-regulation may play a role in several biologic processes.


Subject(s)
Down-Regulation , Estrogens/pharmacology , Receptors, Estrogen/metabolism , Animals , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Drug Implants , Estrogens/administration & dosage , Female , Organ Size , Radioimmunoassay , Rats , Rats, Inbred Strains , Time Factors , Uterus/anatomy & histology , Uterus/metabolism
12.
J Recept Res ; 11(5): 743-56, 1991.
Article in English | MEDLINE | ID: mdl-1920275

ABSTRACT

Estradiol (E2) regulation of estrogen receptor (ER) concentrations has been shown to be both time- and dose-dependent. E2 concentrations of 0.5 mg/ml or greater contained in Silastic capsules suppressed uterine ER concentrations after one day's exposure. In this study, we looked at the effects of physiological (1.0 and 10.0 micrograms subcutaneous injections) and pharmacological (5.0 mg/ml implants) doses of E2 on ER concentrations at times less than 24 hours. The implanted rats had maximum E2 plasma levels of approximately 2000 pg/ml for at least six hours which fell to around 800 pg/ml by 12 hours where they remained up to 24 hours. The physiological doses resulted in plasma levels at one hour of 2000 pg/ml (10 micrograms dose) and 250 pg/ml (1 microgram dose) both of which fell to less than 60 pg/ml by six hours. All treatments caused maximal ER suppression by six hours; however, the implants caused a greater reduction in ER levels than either of the physiological doses. The reduction of ER levels was due primarily to a decrease in the "cytosolic" receptor. Despite the decrease in ER, all doses caused a significant and equivalent increase in uterine weight at six hours, however, only the implanted animals maintained the maximal uterine weight gain through 24 hours. This maintenance of uterine weight appears to be correlated with a small but significant increase in the nuclear ER level over this same time period. Thus, while E2 can cause a short-term suppression of its receptor concentration with no effect on short-term uterine weight gain, uterine growth is positively correlated with the level of nuclear ER.


Subject(s)
Estradiol/pharmacology , Estradiol/physiology , Receptors, Estrogen/drug effects , Uterus/growth & development , Animals , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Administration Schedule , Estradiol/blood , Female , Organ Size/drug effects , Ovariectomy , Rats , Rats, Inbred Strains , Uterus/drug effects
13.
Histochemistry ; 96(2): 157-62, 1991.
Article in English | MEDLINE | ID: mdl-1917572

ABSTRACT

Application of Sternberger's unlabeled antibody enzyme method for detection of the estrogen receptor (ER) using a rat primary antibody with rat tissues has been discouraged, presumably because nonspecific staining of endogenous IgG was expected with the required anti-rat IgG bridging antibody. Because the blood-brain barrier greatly reduces immunoglobulin infiltration into the brain, we hypothesized that rat brain tissue could be specifically immunostained using rat IgG primary antibodies. A rat monoclonal anti-ER antibody (H222) specifically stained ERs in the brains of ovariectomized but not in ovariectomized estrogen-treated rats. In contrast, the uterus, a well-perfused target organ stained intensely in a nonspecific fashion. Dense populations of estrogen receptors were observed in the medial preoptic area, the bed nucleus of the stria terminalis, and the arcuate and ventromedial nuclei of the hypothalamus. A monoclonal rat IgG directed against alpha-tubulin labeled primarily cortical dendrites quite distinct from the neuronal nuclei that are the primary antigenic sites for the estrogen receptor antibody. These results confirm that the sensitive unlabeled antibody method can be applied to rat brain tissues, even when the primary antibody is rat IgG and that labeling of endogenous IgG may be used as a simple method to evaluate the integrity of the blood brain barrier.


Subject(s)
Brain/metabolism , Immunohistochemistry/methods , Receptors, Estrogen/analysis , Animals , Antibodies, Monoclonal , Brain Chemistry , Female , Hypothalamus/metabolism , Rats , Rats, Inbred Strains , Receptors, Estrogen/immunology , Tubulin/analysis , Tubulin/immunology , Uterus/metabolism
14.
Endocrinology ; 125(5): 2773-9, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2792007

ABSTRACT

Previous studies from several laboratories have demonstrated that estradiol treatment resulted in an increase in nuclear type II binding sites. Our previous data suggest that this increase was due to the estradiol-stimulated influx of circulating eosinophils. Therefore, we suggested that the uterine nuclear type II estrogen-binding sites were not of uterine origin. In this report we present further evidence to support this hypothesis. Treatment of immature rats with estradiol resulted in the stimulation of several uterine parameters, namely wet weight, protein synthesis, eosinophil number, peroxidase activity, nuclear type II binding sites, and the synthesis and secretion of a 180-kDa protein. The coadministration of pertussigen had no effect on the estradiol-stimulated increase in wet weight, protein synthesis, or the synthesis and secretion of the 180-kDa protein. However, pertussigen did prevent the estradiol-stimulated increase in eosinophils, peroxidase activity, and nuclear type II binding sites, demonstrating a coordinated response. Since peroxidase activity is known to be contained int he eosinophil, these data are consistent with our earlier demonstration that the type II sites are of eosinophil origin. These data also support and extend our previous findings in neonatal animals that estradiol can stimulate a growth response without a corresponding increase in the nuclear type II binding sites. These results further indicate that the estradiol-stimulated increase in eosinophils does not appear to play a key role in the control of uterine growth.


Subject(s)
Cell Nucleus/metabolism , Eosinophils/physiology , Estradiol/pharmacology , Pertussis Toxin , Receptors, Estrogen/physiology , Uterus/physiology , Virulence Factors, Bordetella/pharmacology , Animals , Cell Nucleus/drug effects , Eosinophils/drug effects , Estrogen Antagonists/pharmacology , Female , Kinetics , Peroxidases/metabolism , Rats , Rats, Inbred Strains , Receptors, Estrogen/drug effects , Reference Values , Uterus/drug effects , Uterus/metabolism
15.
Biochem Biophys Res Commun ; 157(1): 232-7, 1988 Nov 30.
Article in English | MEDLINE | ID: mdl-3143361

ABSTRACT

It has been hypothesized that complex sphingolipids may serve as another "lipid second messenger" system via their hydrolysis to free sphingosine, which inhibits protein kinase C and affects multiple cellular functions. To investigate sphingolipid turnover, Chinese hamster ovary cells were pulse labelled with [14C]serine and the [14C]sphingosine in cellular sphingolipids was determined over time. Much of the radiolabelled sphingosine was initially seen in ceramides and was incorporated into sphingomyelin during the 5-hour chase. A major portion of the radiolabel that was initially seen in other sphingolipids disappeared over time. Overall, about half of the total long-chain bases made during this pulse were degraded within 2 to 5 h, depending on the method of analysis. Hence, a substantial portion of the sphingosine synthesized de novo by these cells is turned over fairly quickly. Since the doubling time of these cells is 12 h, this rapid turnover may reflect the remodelling of the cell surface, or the utilization of the free sphingosine derived from sphingolipid turnover, as part of the control of cell growth and division.


Subject(s)
Serine/metabolism , Sphingolipids/metabolism , Sphingosine/metabolism , Cell Line , Ceramides/metabolism , Chromatography, Thin Layer , Sphingomyelins/metabolism
16.
Biochemistry ; 27(18): 7079-84, 1988 Sep 06.
Article in English | MEDLINE | ID: mdl-3143407

ABSTRACT

The effects of beta-chloroalanine (beta-Cl-alanine) on serine palmitoyltransferase activity and the de novo biosynthesis of sphinganine and sphingenine were investigated in vitro with rat liver microsomes and in vivo with intact Chinese hamster ovary (CHO) cells. The inhibition in vitro was rapid (5 mM beta-Cl-alanine caused complete inactivation in 10 min), irreversible, and concentration and time dependent and apparently involved the active site because inactivation only occurred with beta-Cl-L-alanine (not beta-Cl-D-alanine) and was blocked by L-serine. These are characteristics of mechanism-based ("suicide") inhibition. Serine palmitoyltransferase (SPT) was also inhibited when intact CHO cells were incubated with beta-Cl-alanine (complete inhibition occurred in 15 min with 5 mM), and this treatment inhibited [14C]serine incorporation into long-chain bases by intact cells. The concentration dependence of the loss of SPT activity and of long-chain base synthesis was identical. The effects of beta-Cl-L-alanine appeared to occur with little perturbation of other cell functions: the cells exhibited no loss in cell viability, [14C]serine uptake was not blocked, total lipid biosynthesis from [14C]acetic acid was not decreased (nor was the appearance of radiolabel in cholesterol and phosphatidylcholine), and [3H]thymidine incorporation into DNA was not affected. There appeared to be little effect on protein synthesis based on the incorporation of [3H]leucine, which was only decreased by 14%. Although beta-Cl-L-alanine is known to inhibit other pyridoxal 5'-phosphate dependent enzymes, alanine and aspartate transaminases were not inhibited under these conditions. These results establish the close association between the activity of serine palmitoyltransferase and the cellular rate of long-chain base formation and indicate that beta-Cl-alanine and other mechanism-based inhibitors might be useful to study alterations in cellular long-chain base synthesis.


Subject(s)
Acyltransferases/antagonists & inhibitors , Alanine/analogs & derivatives , Sphingolipids/biosynthesis , beta-Alanine/analogs & derivatives , Animals , Biological Transport, Active , Cells, Cultured , Cricetinae , Female , In Vitro Techniques , Lipids/biosynthesis , Microsomes, Liver/metabolism , Rats , Serine/pharmacokinetics , Serine C-Palmitoyltransferase , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , beta-Alanine/pharmacokinetics , beta-Alanine/pharmacology
17.
J Steroid Biochem ; 29(5): 527-32, 1988 May.
Article in English | MEDLINE | ID: mdl-3379960

ABSTRACT

The neonatal rodent appears to be an appropriate animal model for estrogen toxicity in the developing reproductive tract. Newborn rats were treated with diethylstilbestrol (DES) at human therapeutic doses (approx 1 mg/kg) during two ontogenetic periods (postnatal days 1-5 and 1-25). Treatment on days 1-5 doubled uterine wt by day 5; however, these uteri failed to grow after discontinuation of DES treatment. In contrast, uterine wt was 4-fold higher and DNA content was 2-fold higher than controls on days 10-25 with continued DES treatment. Total uterine estrogen receptor levels, depressed 60% by day 5 of DES treatment, partially recovered after discontinuation of DES treatment but remained 25% below controls on day 25. Receptor levels following DES on days 1-25 decreased to about 15% of the controls by day 15. Short-term DES treatment approximately halved uterine gland content while continued treatment almost completely inhibited gland appearance. DES effects on glands appear related to continued hypertrophy of the luminal epithelium, from which uterine glands are derived. Subsequent failure of uterine growth caused by DES treatment on days 1-5 is similar to clinical findings of hypoplastic uteri in DES-treated patients. Disruption of the normal ontogenetic patterns of estrogen receptor by DES may be involved. These data demonstrate abnormal patterns of growth, estrogen receptor levels and morphogenesis in uteri of rats treated postnatally with DES.


Subject(s)
Diethylstilbestrol/pharmacology , Receptors, Estrogen/metabolism , Uterus/growth & development , Age Factors , Animals , Animals, Newborn , Cell Nucleus/metabolism , Female , Organ Size/drug effects , Rats , Uterus/metabolism
18.
J Steroid Biochem ; 25(1): 37-43, 1986 Jul.
Article in English | MEDLINE | ID: mdl-3747514

ABSTRACT

Uterine nuclear fractions from estrogen-treated rats contain both the estrogen receptor and a lower affinity estrogen binding site (type II site). In Scatchard plots of estrogen binding, two types of curves are seen. The hook-shaped form is composed of a linear component (the estrogen receptor) and a convex component (the type II site) while the curvilinear form is resolvable into two linear binding species (the estrogen receptor and a secondary site). To clarify the relationship between the two forms, we examined the curvilinear form from immature rats injected for 4 days with estradiol (E2) for type II site properties. Like the hook-shaped type II, this form could be detected in a nuclear exchange assay at both 37 and 4 degrees C, but at neither temperature in the presence of reducing agent. Additionally, the steroid specificity of the curvilinear form was identical to the hook-shaped form. The hook-shaped form was found in both immature and ovariectomized adult rats implanted for 6 days with an E2-releasing Silastic capsule to provide pharmacological E2 levels. When uteri from implanted animals displaying the hook-shaped form were mixed in various ratios with uteri lacking type II sites, the curvilinear form was produced. Animals given an E2 implant for 3 days, followed by a 3 day hormone-free period showed a curvilinear form. In vivo E2 dose-response experiments showed the curvilinear form at low E2 doses and the hook-shaped form at the high dose and in implanted animals. We conclude that curvilinear Scatchard plots result from the presence of authentic type II at lower concentrations than those giving rise to the hook-shaped form.


Subject(s)
Cell Nucleus/analysis , Estrogens/pharmacology , Receptors, Estrogen/analysis , Uterus/analysis , Animals , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Rats , Rats, Inbred Strains
20.
Endocrinology ; 117(5): 2238-48, 1985 Nov.
Article in English | MEDLINE | ID: mdl-4042985

ABSTRACT

We have previously shown that rat uterine gland genesis occurs rapidly and synchronously between postnatal days 9-15. Exogenous estrogens either stimulate or inhibit gland genesis depending on dose and age at administration. We therefore examined the developmental effects of the triphenylethylene antiestrogen tamoxifen, which exhibits both estrogen agonist and antagonist properties, in the postnatal rat uterus. Tamoxifen administered sc in oil on postnatal days 1-5 or days 10-14 caused dose-related inhibition of uterine gland genesis which persisted to day 26 or day 60, respectively. Tamoxifen administered on postnatal days 20-24, which is after the age of normal gland genesis, did not alter the number of preexisting glands. A 24-h exposure to tamoxifen inhibited 17 beta-estradiol (E2)-induced ornithine decarboxylase (ODC) activity measured 6 h after E2 administration in 14-day-old rats. Treatment with tamoxifen before or during the period of gland genesis also reduced uterine responsiveness to a single dose of E2 as measured by both uterine weight gain (after a 24-h exposure on days 14, 19, 22, and 26) and the pattern of E2-induced ODC activity in 26-day-old rats. Control rats respond to E2 with peaks of ODC activity at 6 and 18 h after administration. Treatment with tamoxifen on either postnatal days 1-5 or 10-14 reduced the 18-h peak to approximately half of controls but did not affect the 6-h E2-induced ODC peak. Analysis of both nuclear and translocatable cytosol estrogen receptor in uteri from 26-day-old rats indicate that neither the dissociation constant (KD) nor the number of binding sites was affected by tamoxifen treatment on postnatal days 1-5 or 10-14.


Subject(s)
Tamoxifen/pharmacology , Uterus/growth & development , Age Factors , Animals , Animals, Newborn , Diethylstilbestrol/pharmacology , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Estradiol/pharmacology , Female , Organ Size/drug effects , Ornithine Decarboxylase/metabolism , Rats , Receptors, Estrogen/metabolism , Uterus/anatomy & histology , Uterus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...