Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Trends Pharmacol Sci ; 10(4): 149-53, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2665247

ABSTRACT

Certain members of the cytochrome P-450 family are expressed at different levels in the livers of male and female rats. Although little is known of the functional significance of these sex differences, progress has been made towards the understanding of the endocrine control of hepatic sex differences in cytochrome P-450 levels. Jan-Ake Gustafsson and colleagues describe a subpopulation of hepatic sexually differentiated P-450s that is regulated by sex differences in growth hormone (GH) secretory pattern. This secretory pattern is in turn regulated by gonadal steroids. These studies demonstrate a novel action of GH and suggest that the hormonal secretory rhythm is pivotal in determination of biological effects.


Subject(s)
Pharmaceutical Preparations/metabolism , Steroids/metabolism , Animals , Female , Humans , Male , Sex Differentiation , Sex Factors
2.
Biochem Pharmacol ; 69(12): 1805-13, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15885657

ABSTRACT

The use of adult primary hepatocytes in culture is of importance for the understanding of hepatic processes at the cellular and molecular levels, and the possibility to employ transient transfection of reporter constructs is invaluable for mechanistic studies on hepatic gene regulation. Although frequently used, there is a lack of reports addressing optimization and characterization of transfection of primary rodent hepatocytes. Here, we have shown that the efficiency of biochemical transfection reagents varies significantly and that Lipofectamine2000 was a superior transfection reagent for adult primary rat hepatocytes when using luciferase reporter vectors. The efficiency increased when the cells were allowed ample time to adapt to the in vitro milieu. Cotransfection of a second reporter gene indicated a risk for promoter competition, and we found that relating reporter activity to total cellular protein content gave consistent and reliable results. Differentiation of the cells, achieved by including biomatrix from the Engelbreth-Holm-Swarm mouse sarcoma in the culture system, was to a larger extent required for hormonal/drug responses of transfected constructs than for responses of endogenous genes and assured responses of transfected constructs. Dexamethasone (Dex) is most often included in hepatocyte culture media, but we could not demonstrate a general beneficial effect of Dex on expression of luciferease reporter contructs. Using the established protocol, we have demonstrated responses of transfected constructs to growth hormone, glucocorticoid and LXR stimuli.


Subject(s)
Cell Culture Techniques/methods , Hepatocytes/cytology , Hepatocytes/metabolism , Transfection/methods , Animals , Cytochromes/genetics , Female , Gene Expression Regulation , Genes, Reporter , Hepatocytes/enzymology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
3.
Mol Endocrinol ; 4(12): 1934-42, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2082191

ABSTRACT

We have studied the GH-dependent expression of cytochrome P-450IIC12 (P-450(15)beta) mRNA and insulin-like growth factor-I (IGF-I) mRNA in primary adult rat hepatocytes. The GH receptor (GHR), being the common denominator for the GH response, was also studied. The respective mRNA levels were measured with specific solution hybridization assays. By investigating the effects of insulin, IGF-I, T3, and corticosterone, alone or in combinations, in the presence or absence of GH we concluded that GH is indeed the inducer of P-450(15)beta mRNA and IGF-I mRNA. However, insulin and IGF-I exerted a 2-fold potentiation of the GH-induced expression of the P-450(15)beta and IGF-I mRNA species. No significant effect of insulin was observed on GHR mRNA expression, but a translational or posttranslational effect on GHR was seen, in that insulin increased the binding of GH to the cells 4-fold. Furthermore, T3 caused a 9-fold increase in the GH-induced expression of IGF-I mRNA. These observations led us to postulate a possible mechanism of hormonal interplay between GH, thyroid hormone, and IGF-I in vivo, i.e. a thyroid hormone potentiation of the GH-induced IGF-I expression, which, in turn, leads to an increased GHR level and thereby a potentiation of the GH-induced expression of P-450(15)beta and, at least transiently, of IGF-I. A transcriptional mechanism of GH action on P-450(15)beta and IGF-I mRNA induction was indicated by the similar half-lives of respective mRNAs in the presence or absence of GH in cell cultures treated with actinomycin-D.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Gene Expression/drug effects , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/genetics , RNA, Messenger/genetics , Receptors, Somatotropin/genetics , Animals , Cells, Cultured , Corticosterone/pharmacology , Half-Life , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Liver/metabolism , Male , Nucleic Acid Hybridization , RNA Probes , Rats , Rats, Inbred Strains , Transcription, Genetic/drug effects , Triiodothyronine/pharmacology
4.
Mol Endocrinol ; 3(7): 1142-7, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2797005

ABSTRACT

The female specific cytochrome P-45015 beta and the male specific cytochrome P-45016 alpha are the major constitutive P-450 forms in rat liver. Their expression is pretranslationally regulated by the sex specific plasma pattern of GH. To measure the mRNA levels of these hemoproteins in studies on their regulation and expression we set up specific solution hybridization assays with in vitro transcribed cRNA probes. An extensive tissue distribution study showed that both P-45015 beta and P-45016 alpha are specifically expressed in the liver. Investigation of the developmental regulation of respective mRNA showed that the onset of sexual differentiation of P-45015 beta and P-45016 alpha expression coincides with the start of puberty. To assess the level(s) of the pretranslational GH regulation of P-45015 beta and P-45016 alpha nuclear run-on transcription analysis was performed. Sex characteristic administration of GH to hypophysectomized rats indicated that GH exerts its dual effects via both transcriptional and postranscriptional mechanisms.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation , Growth Hormone/physiology , Sex Characteristics , Animals , Cytochrome P-450 Enzyme System/metabolism , Female , Male , Nucleic Acid Hybridization , RNA Probes/isolation & purification , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Strains , Steroid Hydroxylases/genetics , Transcription, Genetic
5.
Mol Endocrinol ; 5(9): 1351-8, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1770953

ABSTRACT

GH is a major determinant of cytochrome P4502C12 and insulin-like growth factor-I (IGF-I) mRNA expression in rat liver. In the present study, a possible role for protein kinase C (PKC) in the GH-mediated regulation of these two genes was investigated. Addition of bovine GH (bGH) to cultured primary adult rat hepatocytes lead to the formation of diacylglycerol and subsequent induction of P4502C12 and IGF-I mRNA, indicating a PKC-dependent signal transduction. However, stimulation of PKC by phorbol 12-myristate 13-acetate (PMA) or sn-1,2-dioctanoylglycerol treatment, in dose and time-course experiments in the presence or absence of ionomycin, failed to induce either P4502C12 or IGF-I mRNA. On the other hand, down-regulation of PKC by PMA treatment, i.e. 24 h pretreatment, attenuated the bGH induction of both P4502C12 and IGF-I mRNA. One hundred nanomolar PMA reduced the bGH-stimulated expression of both IGF-I mRNA and P4502C12 mRNA (approximately 50%). Treatment with the potent kinase inhibitor staurosporine in combination with bGH caused a dose-dependent decrease of the bGH response with different sensitivities toward the inhibitor for the different mRNA species, IGF-I being less sensitive. These data indicate a permissive role for PKC in the GH-mediated induction of P4502C12 and IGF-I mRNA. When activators of protein kinase A, such as forskolin and 8-Br-cAMP were added to the culture medium opposite effects were observed on the mRNA levels of P4502C12 and IGF-I.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/genetics , Liver/cytology , Protein Kinase C/metabolism , Alkaloids/pharmacology , Animals , Cells, Cultured , Diglycerides/metabolism , Down-Regulation , Enzyme Activation , Gene Expression Regulation , Glucagon/pharmacology , Male , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Staurosporine , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
6.
Mol Endocrinol ; 9(12): 1771-81, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8614413

ABSTRACT

Expression of the rat CYP2C12 gene is liver specific and is induced by GH at the transcriptional level. In primary cultures of rat hepatocytes, GH inducibility of CYP2C12 and the presence of C/EBP alpha protein were demonstrated to be equally dependent on attachment of the cells to an extracellular matrix gel (Matrigel). Transient transfection of a C/EBP alpha expression vector into hepatocytes, cultured without Matrigel, increased the cellular P4502C12 messenger RNA levels 10-fold. Cotransfection studies using deletion constructs of the CYP2C12 promoter fused to the luciferase reporter gene localized the C/EBP alpha response to the region -250 to -180. Sequence comparisons and deoxyribonuclease I footprinting using rat liver nuclear extracts indicated two potential C/EBP binding sites in this region. Mutagenesis of the most upstream element (-229 to -207) abolished transactivation by C/EBP alpha. Using gel mobility supershift assays, this element was demonstrated to bind C/EBP alpha and C/EBP beta in liver nuclear extracts and in lysates from hepatocytes cultured on Matrigel. GH treatment of the cells did not alter the C/EBP protein levels or the C/EBP-binding activity to this element. Neither did GH increase the expression of CYP2C12 reporter gene constructs regardless of the presence of different amounts of cotransfected C/EBP alpha. We conclude that C/EBP alpha is a potent transactivator of the CYP2C12 gene and most likely contributes to its liver-specific expression. Although the results presented here do not exclude the possibility of a GH-enhanced transactivating ability of C/EBP alpha, the mechanism of GH-induced levels of P4502C12 is not through increased levels of C/EBP alpha or via enhanced DNA-binding activity of this transcription factor.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins/pharmacology , Liver/metabolism , Nuclear Proteins/pharmacology , Steroid Hydroxylases/genetics , Transcriptional Activation , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression , Growth Hormone/pharmacology , Luciferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins
7.
Mol Endocrinol ; 6(2): 259-66, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1569969

ABSTRACT

The onset of the sexually dimorphic pattern of GH secretion and increased hepatic GH-binding capacity in rats at puberty is temporally correlated with the developmental induction of three hepatic cytochrome P-450s with steroid hydroxylase activity, P-450 IIC11, P-450 IIC12, and P-450 IIC13, and one cytochrome P-450 with vitamin A hydroxylase activity, P-450 IIC7. In this study we demonstrate that expression of the 2C11, 2C12, and 2C13 genes is modulated by GH at the level of transcriptional initiation both in vivo and in primary cultures of adult hepatocytes. In an effort to define the minimum sequence responsible for the inductive effects of GH, we have analyzed the ability of a 0.7-kilobase fragment isolated from the 5'-flank of the 2C12 gene, including the natural promoter, to drive transcription of a 320-basepair G-less cassette in vitro. We were unable to detect any substantial difference in RNA polymerase-II-dependent transcriptional efficiency toward the 2C12 promoter between liver nuclear extracts from normal and hypophysectomized rats of both sexes. This observation supports the assumption that the sequence information contained between bases -700 and 1 is sufficient to support basal transcription of the 2C12 gene. Sequence information residing 5' or 3' of the 0.7-kilobase 5'-flank or a higher ordered chromatin structure may be necessary for the sex-specific transcriptional activation of the 2C12 gene.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Growth Hormone/metabolism , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/genetics , Transcription, Genetic , Animals , Cells, Cultured , Female , Growth Hormone/pharmacology , Liver/cytology , Liver/metabolism , Male , Promoter Regions, Genetic , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Sex Characteristics
8.
Mol Endocrinol ; 12(9): 1294-309, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731699

ABSTRACT

In primary hepatocytes, overexpression of an insulin response element-A binding protein (IRE-ABP), a member of the SRY family of high-mobility group (HMG) proteins, inhibits CCAAT/enhancer-binding protein alpha (C/EBPalpha)-mediated activation of the female-specific cytochrome P450 2C12 (CYP2C12) gene, but not the male-specific cytochrome P450 2C11 (CYP2C11) gene. IRE-ABP and C/EBPalpha have overlapping specificity for the C/EBPalpha target site in the CYP2C12 promoter and compete for binding to CYP2C12 DNA in vitro. In contrast, IRE-ABP and C/EBPalpha bind distinct sequences in the CYP2C11 promoter. A single amino acid substitution in the HMG domain of IRE-ABP impairs its ability to bind DNA and to inhibit the effect of C/EBPalpha on CYP2C12 gene expression. Therefore, the ability of IRE-ABP to inhibit C/EBPalpha-stimulated CYP2C12 gene expression requires a functional DNA-binding domain. Taken together, our findings suggest that SRY-like proteins can bind to a subset of sequences recognized by the C/EBP family of DNA-binding proteins and modulate gene transcription in a context-specific manner.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Enzymologic , Nuclear Proteins/metabolism , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/genetics , Transcription Factors/metabolism , 3T3 Cells , Amino Acid Substitution , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , CHO Cells , Cells, Cultured , Cricetinae , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P450 Family 2 , DNA/metabolism , Female , Liver/metabolism , Male , Mice , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Sex Characteristics , Steroid Hydroxylases/biosynthesis , Transcriptional Activation
9.
Endocrinology ; 111(2): 645-9, 1982 Aug.
Article in English | MEDLINE | ID: mdl-6284488

ABSTRACT

The aim of the present study was to identify the primary sites of action of estrogens and androgens in the regulation of hepatic PRL receptors in the rat. Implantation of estradiol benzoate in the pituitary region of male rats caused a feminization (i.e. an increase) of PRL receptors to a concentration typical of that found in female rats. Estrogen implanted in the paraventricular region of male rats was less effective in causing such a feminization. Subcutaneous implantation of estrogen did not affect the PRL receptor concentration. It is concluded that estrogen induces PRL receptors via an action at the hypothalamo-pituitary level, possibly directly on the pituitary. The PRL receptor-suppressive action of sc injected testosterone was also exerted by the synthetic androgen R1881 (17 beta-hydroxy-17 alpha-methylestra-4,9,11-trien-3-one). However, anterior hypothalamic deafferentation rendered both male and female rats insensitive to this action of R1881. It is concluded that an intact hypothalamo-pituitary unit is required for the PRL receptor-suppressive action of R1991. It is possible that the site of action of androgens is in the rostral hypothalamus or in adjacent areas of the brain.


Subject(s)
Estradiol/analogs & derivatives , Estrenes/pharmacology , Liver/metabolism , Receptors, Cell Surface/metabolism , Testosterone/pharmacology , Animals , Delayed-Action Preparations , Estradiol/administration & dosage , Estradiol/pharmacology , Estrenes/administration & dosage , Female , Injections, Subcutaneous , Male , Metribolone , Rats , Receptors, Cell Surface/drug effects , Receptors, Prolactin , Testosterone/administration & dosage
10.
Endocrinology ; 142(6): 2695-701, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356721

ABSTRACT

A sex-specific secretion of GH prevails in the rat. This has bearings on the expression of target genes, particularly in the liver. We have used suppressive subtractive hybridization to search for genes expressed in response to the female-characteristic, near-continuous secretion of GH. One sequence was particularly abundant among the obtained clones. After isolation of the corresponding full-length complementary DNA using rapid amplification of complementary DNA ends, it was found to be homologous to the human alpha1B-glycoprotein. Sequence comparisons suggest that the human alpha1B-glycoprotein and the rat homolog are members of a new family of proteins, of which at least four additional forms were found in the databases of human and mouse expressed sequence tags. In situ hybridization confirmed the female-specific expression, and by RNase protection analysis a liver-specific expression was indicated. Up-regulation of the messenger RNA by continuous exposure to GH, but not to the male-characteristic intermittent exposure, was demonstrated in hypophysectomized rats and in cultured primary hepatocytes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Blood Proteins/genetics , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression Regulation/drug effects , Glycoproteins , Growth Hormone/pharmacology , Immunoglobulins , Androgen-Insensitivity Syndrome/metabolism , Animals , Blood Proteins/chemistry , Blotting, Northern , Cells, Cultured , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , DNA, Complementary/chemistry , Female , Growth Hormone/metabolism , Hepatocytes/chemistry , Hepatocytes/metabolism , Humans , Hypophysectomy , In Situ Hybridization , Liver/chemistry , Male , Nucleic Acid Hybridization , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sequence Alignment , Sequence Homology , Sex Characteristics , Steroid Hydroxylases/chemistry , Steroid Hydroxylases/genetics
11.
Endocrinology ; 108(6): 2103-8, 1981 Jun.
Article in English | MEDLINE | ID: mdl-7227299

ABSTRACT

The metabolism of 4-[4-14C]androstene-3,17-dione was studied in the microsomal fraction of rat livers after continuous administration of human GH (hGH) in Alzet osmotic minipumps under varying conditions. hGH caused a complete feminization of hepatic steroid metabolism (i.e. increased the 5 alpha-reductase and decreased the 6 beta- and 16 alpha-hydroxylase activities) in normal male rats when infused at a rate of 5 microgram/h for 7 days. Hypophysectomy and castration or adrenalectomy and thyroidectomy of male rats did not reduce the feminizing capacity of hGH, indicating that the adrenals and the thyroid gland are not involved in the mediation of the feminizing effect of hGH. The same dose (5 microgram/h for 7 days) of hGH was also able to refeminize the liver steroid metabolism in hypophysectomized-ovariectomized female rats. The effect of the homologous hormones, rat PRL and rat GH on hepatic steroid metabolism was also investigated. Either hormone was infused at a rate of 10 microgram/h for 7 days, a dose which was sufficient to increase the serum level of the hormone in hypophysectomized animals. No feminizing effect was seen after the administration of rat PRL, whereas rat GH caused a partial feminization of hepatic steroid metabolism in hypophysectomized male animals. It is concluded that GH or a hormone related to GH is involved in sexual differentiation of liver steroid metabolism.


Subject(s)
Androstenedione/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System , Growth Hormone/pharmacology , Microsomes, Liver/metabolism , Sex Characteristics , Steroid 16-alpha-Hydroxylase , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Adrenalectomy , Animals , Castration , Female , Humans , Hypophysectomy , Male , Prolactin/pharmacology , Rats , Steroid Hydroxylases/metabolism , Thyroidectomy
12.
Endocrinology ; 134(5): 2135-41, 1994 May.
Article in English | MEDLINE | ID: mdl-8156914

ABSTRACT

Hepatic mRNA transcripts for the steroid-metabolizing enzymes cytochrome P4502C11 (male specific) and P4502C12 (female specific) differ in abundance by 10- to 20-fold in male and female rats and are regulated by their different patterns of GH secretion. This sex difference is also found in dwarf rats with low GH secretion, implying that these transcripts may be very sensitive to low level GH exposure. This has now been characterized in normal and dwarf rats. Continuous i.v. infusion of recombinant human (h) GH (0, 3, 12, and 48 micrograms/day) in both dwarf and normal male rats caused a dose-dependent decrease in P4502C11 and an increase in P4502C12, so that the 2C11/2C12 ratio fell from 17.9 +/- 1.3 to 1.5 +/- 1.0 in normal males and from 6.5 +/- 0.9 to 0.4 +/- 0.3 in dwarf males (0 vs. 48 micrograms hGH/day); over this dose range of hGH, body weight gain, total hepatic insulin-like growth factor-I mRNA levels, and plasma GHBP levels were largely unaffected. These effects of hGH were pattern dependent. The 2C11/2C12 ratio in dwarf males was feminized (from 11.9 +/- 1.3 to 0.08 +/- 0.03) by continuous infusion of hGH (36 micrograms/day), whereas a pulsatile infusion (3-min pulses every 3 h) of the same daily hGH dose was much less effective. Neither continuous nor pulsatile hGH affected P4502C11 or P4502C12 transcripts in dwarf females, although pulsatile hGH infusion caused a significant weight gain. To test whether baseline GH levels could be modified by circulating GH-binding protein (GHBP), hGH infusions were given with and without recombinant hGHBP in different patterns. Pulsatile infusions of recombinant hGHBP (42 micrograms/day, i.v.) did not prevent the feminizing effect of continuously infused hGH (36 micrograms/day, sc) in dwarf males (2C11/2C12 ratios were 0.08 +/- 0.01 and 0.09 +/- 0.01 for hGH vs. hGH plus hGHBP, respectively). This suggested that intermittent complex formation with GHBP did not prevent continuous access of hGH to the hepatic GH receptors. Furthermore, pulses of hGH complexed with GHBP significantly reduced the 2C11/2C12 ratio in dwarf males (from 21.5 +/- 3.9 with pulsatile hGH alone to 9.2 +/- 2.5 with pulses of hGH plus hGHBP), indicating that GHBP prolongs the exposure to hGH. Thus, 2C11/2C12 expression is very sensitive to basal GH levels in dwarf rats, and GHBP can alter hepatic gene expression by modifying the pattern of GH exposure.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Carrier Proteins/pharmacology , Cytochrome P-450 Enzyme System/genetics , Gene Expression/drug effects , Growth Hormone/pharmacology , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/genetics , Animals , Carrier Proteins/administration & dosage , Female , Growth Hormone/administration & dosage , Male , Periodicity , RNA, Messenger/metabolism , Rats
13.
Endocrinology ; 113(4): 1250-60, 1983 Oct.
Article in English | MEDLINE | ID: mdl-6311514

ABSTRACT

The identity of the pituitary factor responsible for the maintenance of a female pattern of hepatic steroid metabolism and a female level of PRL receptors has been established. Fractionation of pituitary extracts revealed that only the GH fraction had the capacity to feminize liver metabolism of androstenedione (i.e. increase 5 alpha-reductase activity and decrease 16 alpha-hydroxylase activity) and to induce PRL receptors to a female level in hypophysectomized animals. The purification of pituitary GH was performed by chromatofocusing followed by gel filtration on Sephadex G-75. GH obtained from male or female pituitary glands showed an identical chromatographic behavior and both preparations had a mol wt of 22,000 and an isoelectric point of 6.1 when analyzed by analytical sodium dodecyl sulfate-gel electrophoresis and isoelectric focusing, respectively. The degree of homogeneity of GH varied between 93% and 97% as judged from sodium dodecyl sulfate-gel electrophoresis. Purified male and female GH were equally efficient in feminizing the liver metabolism. Since degradation of the native mol wt 22,000 form reduced the feminizing capacity, we believe that the intact hormone is needed for the feminization.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Growth Hormone/isolation & purification , Liver/metabolism , Pituitary Gland/analysis , Steroid 16-alpha-Hydroxylase , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Androstenedione/metabolism , Animals , Female , Growth Hormone/pharmacology , Hypophysectomy , Liver/drug effects , Male , Rats , Rats, Inbred Strains , Receptors, Cell Surface/metabolism , Receptors, Prolactin , Sex Factors , Steroid Hydroxylases/metabolism
14.
Endocrinology ; 117(5): 1881-9, 1985 Nov.
Article in English | MEDLINE | ID: mdl-4042968

ABSTRACT

The influence of endogenous sex steroids and exogenous testosterone treatment on pulsatile GH secretion, body weight, longitudinal bone growth, and hepatic steroid metabolism was studied in male and female adult rats. Blood samples were obtained from the tip of the tail, and maximum and minimum GH levels were determined in individual rats to evaluate pulse heights and baseline levels. Longitudinal bone growth was measured using the intravital marker tetracycline, and hepatic steroid metabolism was evaluated by determining the enzyme activities of 16 alpha-hydroxylase and 5 alpha-reductase. Neonatal, but not prepubertal, gonadectomy of male rats suppressed maximum and mean plasma GH levels during adult life. The body weight and the rate of longitudinal bone growth were also decreased. Testosterone treatment neonatally reversed all of these effects. Neonatal gonadectomy of male rats also caused an elevation of minimum plasma GH levels, an effect, however, which was not reversed by testosterone replacement during neonatal life. Neonatally gonadectomized females treated with testosterone neonatally or during adult life increased their maximum and decreased their minimum GH levels. Their longitudinal bone growth was increased. The body weight of these rats was increased by neonatal, but not adult, testosterone treatment. There was no effect of neonatal ovariectomy on plasma GH levels in 3- to 4-month-old female rats. However, neonatal ovariectomy did increase the maximum and mean plasma GH levels immediately postpubertally, suggesting that the effect of the ovaries on GH secretion differs among mature female rats of different ages. Prepubertal gonadectomy of male rats feminized their hepatic steroid metabolism by decreasing 16 alpha-hydroxylase and increasing 5 alpha-reductase activities. Neonatal gonadectomy caused an even more pronounced feminization, which was partly reversed in rats given testosterone replacement therapy neonatally. In neonatally gonadectomized female rats, treatment with testosterone during adult life increased 16 alpha-hydroxylase and decreased 5 alpha-reductase to levels seen in intact male rats. The present results indicate that neonatally secreted testicular androgens imprint the high amplitude pulses characteristic of GH secretion in adult male rats. Neonatal androgens also stimulate somatic growth and partially account for the masculinized hepatic steroid metabolism in the adult animal. It is proposed that imprinting of the GH secretory pattern contributes to the influence of neonatal testicular androgens on body growth and hepatic steroid-metabolizing enzymes.


Subject(s)
Growth Hormone/metabolism , Growth , Liver/metabolism , Sex Differentiation , Steroids/metabolism , Testosterone/physiology , Animals , Animals, Newborn/physiology , Female , Growth/drug effects , Male , Orchiectomy , Ovariectomy , Rats , Steroid 16-alpha-Hydroxylase , Testosterone/blood
15.
Endocrinology ; 112(3): 1076-90, 1983 Mar.
Article in English | MEDLINE | ID: mdl-6129964

ABSTRACT

The regulation of the sexually differentiated metabolism of 4-[4-14C]androstene-3,17-dione and the presence of PRL receptors in rat liver were studied. Electrolytic lesions in male rats placed in a restricted area in the anterior hypothalamic periventricular area caused a feminization of hepatic steroid metabolism (i.e. increased the 5 alpha-reductase and decreased the 6 beta- and 16 alpha-hydroxylase activities) and of the levels of PRL receptors (increased binding of [125I]-labeled human PRL). After periventricular lesions, histochemical analysis revealed a decrease in somatostatin-like immunoreactive cell bodies in the periventricular area. Also the number of immunoreactive somatostatin fibers in the median eminence was dramatically reduced. Somatostatin levels in the median eminence, as measured by RIA, were reduced to approximately 2-10% of control values after periventricular lesions. Large lesions in the amygdaloid complex in male rats caused a partial feminization of hepatic steroid metabolism and PRL receptors. Passive immunization during 4 days by multiple injections of an antiserum generated against somatostatin resulted in a partial feminization of the male rat liver. When somatostatin was injected into female rats, the PRL receptors were reduced to approximately 60% of the control female receptor levels. The present study indicates that the anterior periventricular hypothalamic area is important in the control of the sexually differentiated steroid metabolism and PRL receptors in the liver and that the amygdaloid complex also may have regulatory influences on this system. A possible central neuro-endocrine mediator of these sex differences in the liver could be somatostatin or a related compound.


Subject(s)
Androstenedione/metabolism , Hypothalamus/physiology , Liver/metabolism , Prolactin/metabolism , Receptors, Cell Surface/metabolism , Somatostatin/physiology , Animals , Anterior Hypothalamic Nucleus/physiology , Female , Immune Sera , Male , Rats , Rats, Inbred Strains , Receptors, Prolactin , Sex Factors , Steroid Hydroxylases/metabolism
16.
Endocrinology ; 137(2): 447-54, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8593788

ABSTRACT

Human GH (hGH) acts by dimerizing two hGH receptors that bind to different sites in hGH. G120RhGH, an analog mutated in the second binding site to prevent receptor dimerization, acts as an antagonist in vitro. We have now tested the activity of this analog in vivo in rats with low or absent endogenous GH secretion. Surprisingly, treatment with G120RhGH failed to antagonize the effects of infusions or injections of hGH in hypophysectomized (Hx) rats and had little effect on hepatic GH-sensitive CYP2C transcripts in GH-deficient dwarf (dw) rats. Paradoxically, G120RhGH stimulated skeletal growth when infused into Hx rats; a pulsatile iv infusion was more effective than a continuous pattern. Coinfusion of G120RhGH with hGH produced an additive effect on growth. In addition, continuous, but not pulsatile, G120RhGH infusion elevated hepatic 2C12 messenger RNA (mRNA) expression and reduced 2C11 mRNA expression in Hx rats. The direct effects of G120RhGH on hepatic CYP2C transcripts were confirmed in cultured hepatocytes in vitro, which also revealed a significant action of PRL in elevating 2C12 mRNA expression. Binding studies revealed that G120RhGH bound preferentially to hepatic PRL receptors, as [125I]G120hGH was completely displaced by ovine PRL but was unaffected by bGH, a specific GH receptor ligand. The weak growth-promoting effects of G120RhGH were similar to those induced by recombinant hPRL in Hx rats. Our results show that G120RhGH is a poor in vivo GH antagonist in the rat, but shows a paradoxical agonist effect, probably mediated by PRL receptors in this species.


Subject(s)
Growth Hormone/analogs & derivatives , Growth Hormone/agonists , Growth Hormone/antagonists & inhibitors , Rats/physiology , Receptors, Prolactin/physiology , Animals , Cells, Cultured , Dwarfism/genetics , Growth Hormone/metabolism , Growth Hormone/pharmacology , Homozygote , Humans , Hypophysectomy , Injections, Intravenous , Injections, Subcutaneous , Liver/cytology , Liver/metabolism , Male , Prolactin/pharmacology , Rats/genetics , Rats, Sprague-Dawley , Recombinant Proteins
17.
Endocrinology ; 139(4): 1815-24, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9528967

ABSTRACT

Signal transducers and activators of transcription (Stat) proteins are latent cytoplasmic transcription factors that are tyrosine phosphorylated by Janus kinases (Jak) in response to GH and other cytokines. GH activates Stat5 by a mechanism that involves tyrosine phosphorylation and nuclear translocation. However, the mechanisms that turn off the GH-activated Jak2/Stat5 pathway are unknown. Continuous exposure to GH of BRL-4 cells, a rat hepatoma cell line stably transfected with rat GH receptor, induces a rapid but transient activation of Jak2 and Stat5. GH-induced Stat5 DNA-binding activity was detected after 2 min and reached a maximum at 10 min. Continued exposure to GH resulted in a desensitization characterized by 1) a rapid decrease in Stat5 DNA-binding activity. The rate of decrease of activity was rapid up to 1 h of GH treatment, and the remaining activity declined slowly thereafter. The activity of Stat5 present after 5 h is still higher than the control levels and almost 10-20% with respect to maximal activity at 10 min; and 2) the inability of further GH treatment to reinduce activation of Stat5. In contrast, with transient exposures of BRL-4 cells to GH, Stat5 DNA-binding activity could repeatedly be induced. GH-induced Jak2 and Stat5 activities were independent of ongoing protein synthesis. However, Jak2 tyrosine phosphorylation and Stat5 DNA-binding activity were prolonged for at least 4 h in the presence of cycloheximide, which suggests that the maintenance of desensitization requires ongoing protein synthesis. Furthermore, inhibition of protein synthesis potentiated GH-induced transcriptional activity in BRL-4 cells transiently transfected with SPIGLE1CAT, a reporter plasmid activated by Stat5. GH-induced Jak2 and Stat5 activation were not affected by D609 or mepacrine, both inhibitors of phospholipase C. However, in the presence of D609 and mepacrine, GH maintained prolonged Jak2 and Stat5 activation. Transactivation of SPIGLE1 by GH was potentiated by mepacrine and D609 but not by the phospholipase A2 inhibitor AACOCF3. Thus, a regulatory circuit of GH-induced transcription through the Jak2/Stat5-signaling pathway includes a prompt GH-induced activation of Jak2/Stat5 followed by a negative regulatory response; ongoing protein synthesis and intracellular signaling pathways, where phospholipase C activity is involved, play a critical role to desensitize the GH-activated Jak2/Stat5-signaling pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Human Growth Hormone/pharmacology , Milk Proteins , Protein Biosynthesis , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Signal Transduction , Trans-Activators/metabolism , Type C Phospholipases/metabolism , Animals , Cycloheximide/pharmacology , DNA/metabolism , Enzyme Inhibitors/pharmacology , Humans , Janus Kinase 2 , Liver Neoplasms, Experimental/metabolism , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Inbred BUF , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Recombinant Proteins/pharmacology , STAT5 Transcription Factor , Transcriptional Activation/drug effects , Tumor Cells, Cultured , Type C Phospholipases/antagonists & inhibitors
18.
J Endocrinol ; 95(2): 181-7, 1982 Nov.
Article in English | MEDLINE | ID: mdl-6983555

ABSTRACT

The site of action of gonadal hormones in the regulation of hepatic steroid metabolism was investigated by measuring the effects of (i) implantation of estradiol into the pituitary gland or anterior hypothalamus of males and (ii) subcutaneous injection of a synthetic androgen in differentiated male and female rats. The hepatic responses measured in vitro were 5 alpha-reduction, and 6 beta- and 16 alpha-hydroxylation of androstenedione. After intrapituitary or intrahypothalamic implantation of oestradiol, 5 alpha-reductase activity increased and 6 beta- and 16 alpha-hydroxylase activity decreased in males relative to the enzyme activities of cholesterol-implanted animals, indicating a feminizing effect of the oestrogen. This effect could not be accomplished by subcutaneous injection of the same oestrogen preparation. Deafferentation had no effect on hepatic steroid metabolism in females, but caused a feminization in males. In addition, subcutaneous treatment of intact females with the synthetic androgen caused masculinization of hepatic steroid metabolism, but was without effect in differentiated animals. Treatment with synthetic androgens had no effect on the hepatic steroid metabolism in differentiated male animals. Subcutaneous injection of a potent synthetic progestagen had little effect on hepatic steroid metabolism in intact females. It is concluded that oestrogen feminizes hepatic steroid metabolism by an action at the hypothalamic-pituitary level and that an intact hypothalamic-pituitary axis is required for the masculinizing action of the synthetic androgen on hepatic steroid metabolism. It is possible that the site of action of androgens is in the anterior hypothalamus or in adjacent areas of the brain.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Estradiol/pharmacology , Hypothalamus/drug effects , Liver/metabolism , Pituitary Gland/drug effects , Testosterone Congeners/pharmacology , Androstenedione/metabolism , Animals , Cytochrome P450 Family 2 , Denervation , Estrenes/pharmacology , Female , Male , Metribolone , Promegestone/pharmacology , Rats , Rats, Inbred Strains , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism
19.
J Endocrinol ; 100(2): 245-8, 1984 Feb.
Article in English | MEDLINE | ID: mdl-6607311

ABSTRACT

Masculine sexual behaviour was induced in castrated sexually inactive but experienced male rats by testosterone-filled constant-release implants or daily injections of the synthetic androgen 17 beta-hydroxy-17 alpha-methyl-estra-4,9,11-triene-3-one (methyltrienolone, R 1881), which resists metabolism by target organs. Feminization of the hepatic androgen metabolism by subcutaneous implantation of osmotic minipumps, which delivered a constant amount of human GH, did not affect the behavioural response of castrated rats to testosterone. Testosterone implants were only minimally effective in inducing male behaviour in ovariectomized female rats, but R 1881 was as effective in stimulating male behaviour in females as in males. Testosterone-treated but not R 1881-treated females showed pronounced female sexual behaviour in response to progesterone treatment despite the absence of measureable amounts of oestradiol-17 beta in peripheral blood. The results provide evidence that masculine sexual behaviour can be activated by an androgen in the absence of oestrogenic stimulation and suggest that the sex difference in the behavioural response to testosterone may be due to a sex difference in the metabolism of androgens by the brain.


Subject(s)
Androgens/metabolism , Estrenes/pharmacology , Sexual Behavior, Animal/drug effects , Testosterone Congeners/pharmacology , Testosterone/pharmacology , Animals , Castration , Female , Growth Hormone/pharmacology , Liver/metabolism , Male , Metribolone , Rats , Rats, Inbred Strains , Testosterone/metabolism
20.
J Endocrinol ; 122(3): 725-32, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2809480

ABSTRACT

The serum concentration of corticosteroid-binding globulin (CBG) is higher in female rats than in males. Combined hypophysectomy and gonadectomy of female rats reduced the serum concentration of CBG as measured by steady-state polyacrylamide gel electrophoresis, whereas hypophysectomy of male rats increased serum CBG. These effects were seen despite replacement therapy with thyroxine and glucocorticoids. Moreover, neither androgen nor oestrogen treatment affected the serum concentrations of CBG in hypophysectomized rats. Continuous infusions of human or bovine GH (1.4 U/kg per day), by means of osmotic minipumps for 1 week, increased serum concentrations of CBG in both hypophysectomized male and female rats. In contrast, intermittent GH replacement therapy by s.c. injections at 12-h intervals either had no effect or suppressed serum CBG levels. In male rats, neonatal (days 1-2) gonadectomy increased CBG levels more than did prepubertal (day 25) gonadectomy, and testosterone replacement therapy reversed these effects. It is concluded that GH increases the serum CBG levels of hypophysectomized rats when it is given in a continuous manner, but not when given intermittently. The sex difference in serum CBG levels of normal rats may, therefore, be attributed to the more continuous secretory pattern of GH previously observed in female rats.


Subject(s)
Growth Hormone/blood , Transcortin/metabolism , Aging/metabolism , Animals , Female , Growth Hormone/administration & dosage , Hypophysectomy , Male , Ovariectomy , Rats , Rats, Inbred Strains , Sex Factors , Testosterone/metabolism , Thyroxine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL