Intrinsic signals in the sexually dimorphic circulating growth hormone profiles of the rat

https://doi.org/10.1016/S0303-7207(00)00401-9Get rights and content

Abstract

Male rats secrete growth hormone (GH) in episodic bursts every 3.5–4 h. Between the peaks, GH levels are undetectable. In females, GH secretory profiles are characterized as continuous because hormone concentrations are always measurable in the circulation. These gender differences in the circulating GH profiles are responsible, to varying degrees, for observed sexual dimorphisms ranging from body growth to the expression of hepatic cytochrome P450 (P450, CYP) isoforms. Using hypophysectomized rats in which restored gender-dependent plasma GH profiles were manipulated, we have investigated the importance of the interpulse period in the masculine episodic plasma GH profile in regulating expression (mRNA, protein and/or specific catalytic activity) of male-specific CYP2A2, 2C11, 2C13 and 3A2, female-specific CYP2C12 and female-predominant CYP2A1, 2C6 and 2C7. We observed that some isoforms were induced or suppressed by discerning the length of the GH-devoid interpulse period, others responded to the pulse amplitudes, still others recognized the mean circulating concentrations of GH and some were regulated by a combination of these signals. We conclude that concealed in the gender-dependent circulating GH profiles are numerous intrinsic signals, both inductive and repressive, individually “tailored” to be recognized by each isoform of P450. There would appear to be no one signal in each gender-dependent GH profile responsible, in toto, for the characteristic sexually dimorphic expression of some dozen hepatic P450s in male and female rats.

Introduction

Circulating GH profiles in rats as well as other species have been shown to be sexually dimorphic (Shapiro et al., 1995). Male rats secrete GH in episodic bursts (∼200–300 ng/ml of plasma) every 3.5–4 h. Between the peaks, GH levels are undetectable. In females the hormone pulses are more frequent and irregular and are of lower magnitude than those in males, whereas the interpulse concentrations of GH are always measurable (Legraverend et al., 1992a, Shapiro et al., 1995). These gender differences in the circulating GH profiles, and not sexual differences in GH concentrations, per se, are responsible for observed sexual dimorphisms ranging from body growth to the expression of hepatic enzymes (Legraverend et al., 1992a, Shapiro et al., 1995). In this regard, rat, as well as murine liver each contain at least a dozen sex-dependent isoforms of P450 that are regulated by the gender-dependent profiles of circulating GH (Legraverend et al., 1992a, Waxman, 1992, Shapiro et al., 1995).2

Whereas much less is known about GH regulation of murine P450s, in the rat, P450 responses to GH regulation are almost as variable as the number of GH-dependent isoforms. That is, expression of the major female-specific CYP2C12 (as well as the non-P450 5α-reductase) is dependent upon the feminine profile of continuous GH secretion. Exposure to the masculine profile of episodic hormone release, as well as the absence of the hormone from the circulation (e.g., hypophysectomy), completely prevents expression of CYP2C12 (Ram and Waxman, 1990, Legraverend et al., 1992b). In a somewhat similar vein, female-predominant CYP2C7 expression is also dependent on the feminine GH profile and is completely suppressed in the hypophysectomized rat. However, exposure to the masculine profile allows expression of CYP2C7 at 25–40% of normal female levels (Ram and Waxman, 1990, Westin et al., 1990). Expression of the major male-specific CYP2C11 requires the episodic ‘on/off’ masculine profile of GH secretion. Although the feminine pattern of continuous hormone secretion blocks CYP2C11 expression, total GH depletion from the circulation allows CYP2C11 expression at 20–30% of intact male levels (Morgan et al., 1985, Janeczko et al., 1990, Legraverend et al., 1992b). After hypophysectomy, female-predominant CYP2A1 (male/female, ∼1:3) concentrations decline to around male levels and are restored to intact female-like levels with continuously administered GH (Waxman et al., 1990b, Yamazoe et al., 1990). Although the expression levels of CYP2C7, CYP2C11, CYP2C12, and CYP2A1 are greatest when exposed to their gender-dependent GH profiles, other isoforms are optimally expressed in the absence of GH. Male-specific CYP2A2 and CYP3A2 are maximally expressed in the hypophysectomized rat, disappear when GH is secreted constantly, but are only partially suppressed under the influence of episodic GH (Waxman et al., 1988, Waxman et al., 1995). Male-specific CYP2C13 is optimally expressed when exposed to the masculine hormone profile or under conditions of no GH, whereas the feminine GH profile completely suppresses CYP2C13 (Legraverend et al., 1992a, Legraverend et al., 1992b).

Although there are additional examples, it becomes clear that the expression or suppression of each isoform of P450 is likely to be regulated by a different “signal” or perhaps, a differential sensitivity to the ‘signal’ in the sexually dimorphic GH profiles. These signals may be recognized by the hepatocyte in the frequencies and/or durations of the pulse and interpulse periods. Alternatively, perhaps the hepatocyte can monitor the mean plasma concentration of the hormone. In the latter case, using the selective-GH deficient monosodium glutamate-treated rat, we reported that a 70–85% reduction in the feminine GH profile has little, if any, effect on the levels of gender-dependent P450 isoforms normally expressed in the female liver (Waxman et al., 1990a, Pampori and Shapiro, 1994a). More recently, using the euthyroid, hypophysectomized rat (Pampori and Shapiro, 1996), we observed that a renaturalized feminine GH secretory pattern of only 2–3% of its normal level (the lowest values we examined) remained completely suppressive of male-specific CYP2A2, CYP2C11, CYP2C13 and CYP3A2 expression. In contrast, the female-dependent isoforms exhibited a variable responsiveness to the restored hormonal profile. Female-predominant CYP2A1 and CYP2C6 were restored to pre-hypophysectomy levels when the feminine GH profile was renaturalized to just 3% of normal. While this level of GH restored female-specific CYP2C12 and 5α-reductase to ∼50% and ∼70% of normal, respectively, restoration to intact expression levels required a circulating feminine profile of 12–25% of normal. Female-predominant CYP2C7 appeared to be the least GH-sensitive isoform. Although exhibiting a commensurate increase in mRNA and protein concentrations with increasing levels of GH, the feminine GH profile had to be restored to near 100% (i.e. physiologic) to induce normal female-like expression levels of CYP2C7. Gender is an additional factor that influences the differential responsiveness of P450s to GH. That is, the renaturalized feminine GH profile is far more effective in restoring the female pattern of P450 expression when infused in females than in males (Pampori and Shapiro, 1999) and vice versa (Shapiro et al., 1993). Lastly, less well-defined, multitropic endpoints like body and organ weights also exhibit a differential sensitivity to the feminine growth hormone profile requiring replacement of 25–50% of the profile for normal maintenance (Pampori and Shapiro, 1996).

While these studies have identified ‘concentration’ as the intrinsic signal in the feminine GH profile regulating the expression of hepatic P450 isoforms observed in female rats, the present study has examined the important properties in the episodic masculine growth hormone profile, in particular, the interpulse period devoid of GH, in regulating the expression of sex-dependent hepatic P450s found in the male rat.

Section snippets

Animals

Animals were housed in the University of Pennsylvania Laboratory Animal Resources facility under the supervision of certified laboratory animal medicine veterinarians and were treated according to a research protocol approved by the University's Institutional Animal Care and Use Committee. Male rats [Crl:CD(SD)BR] were hypophysectomized by the vendor (Charles River Laboratories, Wilmington, MA) between 7 and 8 weeks of age and were observed in our facilities for 5 weeks. The effectiveness of

Hormone delivery systems

Whereas almost all reports examining the effects of GH replacement on P450 expression do not verify the resulting plasma profiles of the hormone, we considered it essential to do so in the present study. In addition to actually measuring the circulating GH profiles in the rGH-replaced hypophysectomized rats (Fig. 1 and Table 1), we also evaluated the accuracy and precision of the GH delivery system. Initially, each morning when the pump syringes were replaced with new sterile syringes

Discussion

The hypophysectomized rat has proven to be a useful model in our studies to identify the fundamental signaling elements in the plasma GH profile (which could be considered the initial signal, albeit extrahepatic, in GH-initiated signal transduction pathways) but, these studies require that certain procedures, which are often overlooked, be followed.

Acknowledgements

We appreciate the generosity of Drs Marika Rönnholm, Agneta Mode, and Jan-Åke Gustafsson in supplying the antibody to rat CYP2C12 and Dr Stelvio M. Bandiera in supplying the antibody to rat CYP2C7. Materials used to assay rat growth hormone were obtained through the National Hormone and Pituitary Program and Dr A.F. Parlow. We also thank Alka Agrawal for excellent technical assistance. This work was supported by National Institutes of Health Grant GM45758 and HD16358.

References (40)

  • D.J. Waxman

    Rat hepatic P450IIC subfamily expression using catalytic, immunochemical and molecular probes

    Methods Enzymol.

    (1991)
  • D.J. Waxman

    Regulation of liver specific steroid metabolizing cytochromes P450: cholesterol 7α-hydroxylase, bile acid 6β-hydroxylase, and growth hormone-responsive steroid hormone hydroxylase

    J. Steroid Biochem. Mol. Biol.

    (1992)
  • D.J. Waxman et al.

    Adult male-specific and neonatally programmed rat hepatic P-450 forms RLM2 and 2a are not dependent on pulsatile plasma growth hormone for expression

    J. Biol. Chem.

    (1988)
  • A.K. Agrawal et al.

    Phenobarbital induction of hepatic CYP2B1 and CYP2B2: pretranscriptional and post-transcriptional effects of gender, adult age and phenobarbital dose

    Mol. Pharmacol.

    (1996)
  • A.K. Agrawal et al.

    Differential expression of gender-dependent hepatic isoforms of cytochrome P450 by pulse signals in the circulating masculine episodic growth hormone profile of the rat

    J. Pharmacol. Exp. Ther.

    (2000)
  • C. Carter-Su et al.

    Molecular mechanism of growth hormone action

    Annu. Rev. Physiol.

    (1996)
  • C.H. Emerson et al.

    Serum thyrotropin concentrations are more highly correlated with serum triiodothyronine concentrations than with serum thyroxine concentrations in thyroid hormone-infused thyroidectomized rats

    Endocrinology

    (1989)
  • R. Janeczko et al.

    Hormonal regulation of levels of the messenger RNA encoding hepatic P450 2c (IIC11), a constitutive male-specific form of cytochrome P450

    Mol. Endocrinol.

    (1990)
  • C. Legraverend et al.

    Hepatic steroid hydroxylating enzymes are controlled by the sexually dimorphic pattern of growth hormone secretion in normal and dwarf rats

    FASEB J.

    (1992)
  • C. Legraverend et al.

    Transcriptional regulation of rat P-450 2C gene subfamily members by the sexually dimorphic pattern of growth hormone secretion

    Mol. Endocrinol.

    (1992)
  • Cited by (50)

    • The Involvement of PPARs in the Selective Regulation of Brain CYP2D by Growth Hormone

      2018, Neuroscience
      Citation Excerpt :

      GH pulsatile secretion in male rats bursts every 3.5–4 h (200–300 ng/ml) with low or undetectable levels between the pulses. In contract, GH secretion occurs continuously at a low amplitude in female rats (Agrawal and Shapiro, 2001). Male mice were reported a regular periodicity of GH peaks every 2.5 h with low baseline level between the pulses (MacLeod et al., 1991).

    • Sex differences in constitutive mRNA levels of CYP2B22, CYP2C33, CYP2C49, CYP3A22, CYP3A29 and CYP3A46 in the pig liver: Comparison between Meishan and Landrace pigs

      2016, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      Incidentally, the DNA polymorphisms in human CYP genes, including the CYP2B, CYP2C, and CYP3A subfamily genes, have been reported [48–50], although such DNA polymorphisms in pigs remain unclear. To date, the secretion profile of growth hormone (GH) is considered as one of the endogenous factors responsible for sexual dimorphism in the constitutive levels of several drug-metabolizing enzymes, including CYPs, SULTs and UGTs [51–54]. The secretion pattern of GH and/or the serum androgen level are thought to be one of the physiological factors that contribute to sex differences in the expressions of human CYP2C19 and CYP3A4 in transgenic mice [55,56], although no clear sex differences in the enzyme activities of those CYPs have been reported [53].

    View all citing articles on Scopus
    1

    Present address:Department of Drug Metabolism, Merck Research Laboratories, PO Box 2000, RY80E-200, Rahway, NJ 07065-0900, USA.

    View full text