Androgen

Last updated

Androgen
Drug class
Testosteron.svg
Testosterone, the major androgen
Class identifiers
Synonyms Androgenic hormone; Testoid
Use Hypogonadism, transgender men, performance enhancement, bodybuilding, others
ATC code G03B
Biological target Androgen receptor, mARs (e.g., GPRC6A, others)
External links
MeSH D000728
Legal status
In Wikidata

An androgen (from Greek andr-, the stem of the word meaning "man") is any natural or synthetic steroid hormone that regulates the development and maintenance of male characteristics in vertebrates by binding to androgen receptors. [1] [2] This includes the embryological development of the primary male sex organs, and the development of male secondary sex characteristics at puberty. Androgens are synthesized in the testes, the ovaries, and the adrenal glands.

Contents

Androgens increase in both males and females during puberty. [3] The major androgen in males is testosterone. [4] Dihydrotestosterone (DHT) and androstenedione are of equal importance in male development. [4] DHT in utero causes differentiation of the penis, scrotum and prostate. In adulthood, DHT contributes to balding, prostate growth, and sebaceous gland activity.

Although androgens are commonly thought of only as male sex hormones, females also have them, but at lower levels: they function in libido and sexual arousal. Androgens are the precursors to estrogens in both men and women.

In addition to their role as natural hormones, androgens are used as medications; for information on androgens as medications, see the androgen replacement therapy and anabolic steroid articles.

Types and examples

The main subset of androgens, known as adrenal androgens, is composed of 19-carbon steroids synthesized in the zona reticularis, the innermost layer of the adrenal cortex. Adrenal androgens function as weak steroids (though some are precursors), and the subset includes dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEA-S), androstenedione (A4), and androstenediol (A5).

Besides testosterone, other androgens include:

Determined by consideration of all biological assay methods (c.1970): [7]

Female ovarian and adrenal androgens

The ovaries and adrenal glands also produce androgens, but at much lower levels than the testes. Regarding the relative contributions of ovaries and adrenal glands to female androgen levels, in a study with six menstruating women the following observations have been made: [8]

Ovarian and adrenal contribution to peripheral androgens during the menstrual cycle [8]
AndrogenOvarian (%) (F, M, L)Adrenal (%)
DHEA2080
DHEA-S4, 10, 490–96
Androstenedione45, 70, 6030–55
Testosterone33, 60, 3340–66
DHT5050
F = early follicular, M = midcycle, L = late luteal phase.

Biological function

Male prenatal development

Testes formation

During mammalian development, the gonads are at first capable of becoming either ovaries or testes. [9] In humans, starting at about week 4, the gonadal rudiments are present within the intermediate mesoderm adjacent to the developing kidneys. At about week 6, epithelial sex cords develop within the forming testes and incorporate the germ cells as they migrate into the gonads. In males, certain Y chromosome genes, particularly SRY, control development of the male phenotype, including conversion of the early bipotential gonad into testes. In males, the sex cords fully invade the developing gonads.

Androgen production

The mesoderm-derived epithelial cells of the sex cords in developing testes become the Sertoli cells, which will function to support sperm cell formation. A minor population of nonepithelial cells appear between the tubules by week 8 of human fetal development. These are Leydig cells. Soon after they differentiate, Leydig cells begin to produce androgens.

Androgen effects

The androgens function as paracrine hormones required by the Sertoli cells to support sperm production. They are also required for the masculinization of the developing male fetus (including penis and scrotum formation). Under the influence of androgens, remnants of the mesonephron, the Wolffian ducts, develop into the epididymis, vas deferens and seminal vesicles. This action of androgens is supported by a hormone from Sertoli cells, Müllerian inhibitory hormone (MIH), which prevents the embryonic Müllerian ducts from developing into fallopian tubes and other female reproductive tract tissues in male embryos. MIH and androgens cooperate to allow for movement of testes into the scrotum.

Early regulation

Before the production of the pituitary hormone luteinizing hormone (LH) by the embryo starting at about weeks 11–12, human chorionic gonadotrophin (hCG) promotes the differentiation of Leydig cells and their production of androgens at week 8. Androgen action in target tissues often involves conversion of testosterone to 5α-dihydrotestosterone (DHT).

Male pubertal development

At the time of puberty, androgen levels increase dramatically in males, and androgens mediate the development of masculine secondary sexual characteristics as well as the activation of spermatogenesis and fertility and masculine behavioral changes such as increased sex drive. Masculine secondary sexual characteristics include androgenic hair, voice deepening, emergence of the Adam's apple, broadening of the shoulders, increased muscle mass, and penile growth.

Spermatogenesis

During puberty, androgen, LH and follicle stimulating hormone (FSH) production increase and the sex cords hollow out, forming the seminiferous tubules, and the germ cells start to differentiate into sperm. Throughout adulthood, androgens and FSH cooperatively act on Sertoli cells in the testes to support sperm production. [10] Exogenous androgen supplements can be used as a male contraceptive. Elevated androgen levels caused by use of androgen supplements can inhibit production of LH and block production of endogenous androgens by Leydig cells. Without the locally high levels of androgens in testes due to androgen production by Leydig cells, the seminiferous tubules can degenerate, resulting in infertility. For this reason, many transdermal androgen patches are applied to the scrotum.

Fat deposition

Males typically have less body fat than females. Recent results indicate androgens inhibit the ability of some fat cells to store lipids by blocking a signal transduction pathway that normally supports adipocyte function. [11] Also, androgens, but not estrogens, increase beta adrenergic receptors while decreasing alpha adrenergic receptors- which results in increased levels of epinephrine/ norepinephrine due to lack of alpha-2 receptor negative feedback and decreased fat accumulation due to epinephrine/ norepinephrine then acting on lipolysis-inducing beta receptors.

Muscle mass

Males typically have more skeletal muscle mass than females. Androgens promote the enlargement of skeletal muscle cells in a coordinated manner by acting on several cell types in skeletal muscle tissue. [12] One cell type, called the myoblast, conveys androgen receptors for generating muscle. Fusion of myoblasts generates myotubes, in a process linked to androgen receptor levels. [13] Higher androgen levels lead to increased expression of androgen receptor.

Brain

Circulating levels of androgens can influence human behavior because some neurons are sensitive to steroid hormones. Androgen levels have been implicated in the regulation of human aggression and libido. Indeed, androgens are capable of altering the structure of the brain in several species, including mice, rats, and primates, producing sex differences. [14] Although more recent studies showing the general mood of transgender men, who have undergone transgender hormone replacement therapy replacing estrogens with androgens, do not show any substantial long-term behavioral changes. [15] [16] [17]

Numerous reports have shown androgens alone are capable of altering the structure of the brain, [18] but identification of which alterations in neuroanatomy stem from androgens or estrogens is difficult, because of their potential for conversion.

Evidence from neurogenesis (formation of new neurons) studies on male rats has shown that the hippocampus is a useful brain region to examine when determining the effects of androgens on behavior. To examine neurogenesis, wild-type male rats were compared with male rats that had androgen insensitivity syndrome, a genetic difference resulting in complete or partial insensitivity to androgens and a lack of external male genitalia.

Neural injections of Bromodeoxyuridine (BrdU) were applied to males of both groups to test for neurogenesis. Analysis showed that testosterone and dihydrotestosterone regulated adult hippocampal neurogenesis (AHN). Adult hippocampal neurogenesis was regulated through the androgen receptor in the wild-type male rats, but not in the TMF male rats. To further test the role of activated androgen receptors on AHN, flutamide, an antiandrogen drug that competes with testosterone and dihydrotestosterone for androgen receptors, and dihydrotestosterone were administered to normal male rats. Dihydrotestosterone increased the number of BrdU cells, while flutamide inhibited these cells.

Moreover, estrogens had no effect. This research demonstrates how androgens can increase AHN. [19]

Researchers also examined how mild exercise affected androgen synthesis which in turn causes AHN activation of N-methyl-D-aspartate (NMDA) receptors.

NMDA induces a calcium flux that allows for synaptic plasticity which is crucial for AHN.

Researchers injected both orchidectomized (ORX) (castrated) and sham castrated male rats with BrdU to determine if the number of new cells was increased. They found that AHN in male rats is increased with mild exercise by boosting synthesis of dihydrotestosterone in the hippocampus.

Again it was noted that AHN was not increased via activation of the estrogen receptors. [20]

Androgen regulation decreases the likelihood of depression in males. In preadolescent male rats, neonatal rats treated with flutamide developed more depression-like symptoms compared to control rats.

Again BrdU was injected into both groups of rats in order to see if cells were multiplying in the living tissue. These results demonstrate how the organization of androgens has a positive effect on preadolescent hippocampal neurogenesis that may be linked with lower depression-like symptoms. [21]

Social isolation has a hindering effect in AHN whereas normal regulation of androgens increases AHN. A study using male rats showed that testosterone may block social isolation, which results in hippocampal neurogenesis reaching homeostasis—regulation that keeps internal conditions stable. A Brdu analysis showed that excess testosterone did not increase this blocking effect against social isolation; that is, the natural circulating levels of androgens cancel out the negative effects of social isolation on AHN. [22]

Female-specific effects

Androgens have potential roles in relaxation of the myometrium via non-genomic, androgen receptor-independent pathways, preventing premature uterine contractions in pregnancy. [23]

Androgen insensitivity

Reduced ability of an XY-karyotype fetus to respond to androgens can result in one of several conditions, including infertility and several forms of intersex conditions.

Miscellaneous

Yolk androgen levels in certain birds have been positively correlated to social dominance later in life. See American coot.

Biological activity

Androgens bind to and activate androgen receptors (ARs) to mediate most of their biological effects.

Relative potency

Determined by consideration of all biological assay methods (c.1970): [7]

AndrogenPotency (%)
Testosterone40
5α-Dihydrotestosterone (DHT)100
Androstenediol.0008
Androstenedione.04
Dehydroepiandrosterone.02
Androsterone.06

5α-Dihydrotestosterone (DHT) was 2.4 times more potent than testosterone at maintaining normal prostate weight and duct lumen mass (this is a measure of epithelial cell function stimulation). Whereas DHT was equally potent as testosterone at preventing prostate cell death after castration. [24] One of the 11-oxygenated androgens, namely 11-ketotestosterone, has the same potency as testosterone. [25]

Non-genomic actions

Androgens have also been found to signal through membrane androgen receptors, which are distinct from the classical nuclear androgen receptor. [26] [27] [28]

Biochemistry

Steroidogenesis, showing the relation between several androgens, is at bottom left. Estrone and estradiol, in contrast, are estrogens. Steroidogenesis.svg
Steroidogenesis, showing the relation between several androgens, is at bottom left. Estrone and estradiol, in contrast, are estrogens.

Biosynthesis

Androgens are synthesized from cholesterol and are produced primarily in the gonads (testicles and ovaries) and also in the adrenal glands. The testicles produce a much higher quantity than the ovaries. Conversion of testosterone to the more potent DHT occurs in prostate gland, liver, brain and skin.

Production rates, secretion rates, clearance rates, and blood levels of major sex hormones
SexSex hormoneReproductive
phase
Blood
production rate
Gonadal
secretion rate
Metabolic
clearance rate
Reference range (serum levels)
SI unitsNon-SI units
Men Androstenedione
2.8 mg/day1.6 mg/day2200 L/day2.8–7.3 nmol/L80–210 ng/dL
Testosterone
6.5 mg/day6.2 mg/day950 L/day6.9–34.7 nmol/L200–1000 ng/dL
Estrone
150 μg/day110 μg/day2050 L/day37–250 pmol/L10–70 pg/mL
Estradiol
60 μg/day50 μg/day1600 L/day<37–210 pmol/L10–57 pg/mL
Estrone sulfate
80 μg/dayInsignificant167 L/day600–2500 pmol/L200–900 pg/mL
Women Androstenedione
3.2 mg/day2.8 mg/day2000 L/day3.1–12.2 nmol/L89–350 ng/dL
Testosterone
190 μg/day60 μg/day500 L/day0.7–2.8 nmol/L20–81 ng/dL
Estrone Follicular phase110 μg/day80 μg/day2200 L/day110–400 pmol/L30–110 pg/mL
Luteal phase260 μg/day150 μg/day2200 L/day310–660 pmol/L80–180 pg/mL
Postmenopause40 μg/dayInsignificant1610 L/day22–230 pmol/L6–60 pg/mL
Estradiol Follicular phase90 μg/day80 μg/day1200 L/day<37–360 pmol/L10–98 pg/mL
Luteal phase250 μg/day240 μg/day1200 L/day699–1250 pmol/L190–341 pg/mL
Postmenopause6 μg/dayInsignificant910 L/day<37–140 pmol/L10–38 pg/mL
Estrone sulfate Follicular phase100 μg/dayInsignificant146 L/day700–3600 pmol/L250–1300 pg/mL
Luteal phase180 μg/dayInsignificant146 L/day1100–7300 pmol/L400–2600 pg/mL
Progesterone Follicular phase2 mg/day1.7 mg/day2100 L/day0.3–3 nmol/L0.1–0.9 ng/mL
Luteal phase25 mg/day24 mg/day2100 L/day19–45 nmol/L6–14 ng/mL
Notes and sources
Notes: "The concentration of a steroid in the circulation is determined by the rate at which it is secreted from glands, the rate of metabolism of precursor or prehormones into the steroid, and the rate at which it is extracted by tissues and metabolized. The secretion rate of a steroid refers to the total secretion of the compound from a gland per unit time. Secretion rates have been assessed by sampling the venous effluent from a gland over time and subtracting out the arterial and peripheral venous hormone concentration. The metabolic clearance rate of a steroid is defined as the volume of blood that has been completely cleared of the hormone per unit time. The production rate of a steroid hormone refers to entry into the blood of the compound from all possible sources, including secretion from glands and conversion of prohormones into the steroid of interest. At steady state, the amount of hormone entering the blood from all sources will be equal to the rate at which it is being cleared (metabolic clearance rate) multiplied by blood concentration (production rate = metabolic clearance rate × concentration). If there is little contribution of prohormone metabolism to the circulating pool of steroid, then the production rate will approximate the secretion rate." Sources: See template.

Metabolism

Androgens are metabolized mainly in the liver.

Medical uses

A low testosterone level (hypogonadism) in men may be treated with testosterone administration. Prostate cancer may be treated by removing the major source of testosterone: testicle removal (orchiectomy); or agents which block androgens from accessing their receptor: antiandrogens.

See also

Related Research Articles

<span class="mw-page-title-main">Dehydroepiandrosterone</span> Chemical compound

Dehydroepiandrosterone (DHEA), also known as androstenolone, is an endogenous steroid hormone precursor. It is one of the most abundant circulating steroids in humans. DHEA is produced in the adrenal glands, the gonads, and the brain. It functions as a metabolic intermediate in the biosynthesis of the androgen and estrogen sex steroids both in the gonads and in various other tissues. However, DHEA also has a variety of potential biological effects in its own right, binding to an array of nuclear and cell surface receptors, and acting as a neurosteroid and modulator of neurotrophic factor receptors.

<span class="mw-page-title-main">Testosterone</span> Primary male sex hormone

Testosterone is the primary male sex hormone and androgen in males. In humans, testosterone plays a key role in the development of male reproductive tissues such as testicles and prostate, as well as promoting secondary sexual characteristics such as increased muscle and bone mass, and the growth of body hair. It is associated with increased aggression, sex drive, dominance, courtship display, and a wide range of behavioral characteristics. In addition, testosterone in both sexes is involved in health and well-being, where it has a significant effect on overall mood, cognition, social and sexual behavior, metabolism and energy output, the cardiovascular system, and in the prevention of osteoporosis. Insufficient levels of testosterone in men may lead to abnormalities including frailty, accumulation of adipose fat tissue within the body, anxiety and depression, sexual performance issues, and bone loss.

<span class="mw-page-title-main">Luteinizing hormone</span> Gonadotropin secreted by the adenohypophysis

Luteinizing hormone is a hormone produced by gonadotropic cells in the anterior pituitary gland. The production of LH is regulated by gonadotropin-releasing hormone (GnRH) from the hypothalamus. In females, an acute rise of LH known as an LH surge, triggers ovulation and development of the corpus luteum. In males, where LH had also been called interstitial cell–stimulating hormone (ICSH), it stimulates Leydig cell production of testosterone. It acts synergistically with follicle-stimulating hormone (FSH).

<span class="mw-page-title-main">Adrenal cortex</span> Cortex of the adrenal gland

The adrenal cortex is the outer region and also the largest part of the adrenal gland. It is divided into three separate zones: zona glomerulosa, zona fasciculata and zona reticularis. Each zone is responsible for producing specific hormones. It is also a secondary site of androgen synthesis.

<span class="mw-page-title-main">Androstenedione</span> Endogenous weak androgen

Androstenedione, or 4-androstenedione, also known as androst-4-ene-3,17-dione, is an endogenous weak androgen steroid hormone and intermediate in the biosynthesis of estrone and of testosterone from dehydroepiandrosterone (DHEA). It is closely related to androstenediol (androst-5-ene-3β,17β-diol).

<span class="mw-page-title-main">Dihydrotestosterone</span> Human hormone

Dihydrotestosterone is an endogenous androgen sex steroid and hormone primarily involved in the growth and repair of the prostate and the penis, as well as the production of sebum and body hair composition.

<span class="mw-page-title-main">Sex hormone</span> Type of steroid hormone

Sex hormones, also known as sex steroids, gonadocorticoids and gonadal steroids, are steroid hormones that interact with vertebrate steroid hormone receptors. The sex hormones include the androgens, estrogens, and progestogens. Their effects are mediated by slow genomic mechanisms through nuclear receptors as well as by fast nongenomic mechanisms through membrane-associated receptors and signaling cascades. The polypeptide hormones luteinizing hormone, follicle-stimulating hormone and gonadotropin-releasing hormone – each associated with the gonadotropin axis – are usually not regarded as sex hormones, although they play major sex-related roles.

<span class="mw-page-title-main">Lipoid congenital adrenal hyperplasia</span> Medical condition

Lipoid congenital adrenal hyperplasia is an endocrine disorder that is an uncommon and potentially lethal form of congenital adrenal hyperplasia (CAH). It arises from defects in the earliest stages of steroid hormone synthesis: the transport of cholesterol into the mitochondria and the conversion of cholesterol to pregnenolone—the first step in the synthesis of all steroid hormones. Lipoid CAH causes mineralocorticoid deficiency in affected infants and children. Male infants are severely undervirilized causing their external genitalia to look feminine. The adrenals are large and filled with lipid globules derived from cholesterol.

<span class="mw-page-title-main">Testosterone enanthate</span> Chemical compound

Testosterone enanthate is an androgen and anabolic steroid (AAS) medication which is used mainly in the treatment of low testosterone levels in men. It is also used in hormone therapy for transgender men. It is given by injection into muscle or subcutaneously usually once every one to four weeks.

<span class="mw-page-title-main">Dehydroepiandrosterone sulfate</span> Chemical compound

Dehydroepiandrosterone sulfate, abbreviated as DHEA sulfate or DHEA-S, also known as androstenolone sulfate, is an endogenous androstane steroid that is produced by the adrenal cortex. It is the 3β-sulfate ester and a metabolite of dehydroepiandrosterone (DHEA) and circulates in far greater relative concentrations than DHEA. The steroid is hormonally inert and is instead an important neurosteroid and neurotrophin.

<span class="mw-page-title-main">Sexual differentiation in humans</span> Process of development of sex differences in humans

Sexual differentiation in humans is the process of development of sex differences in humans. It is defined as the development of phenotypic structures consequent to the action of hormones produced following gonadal determination. Sexual differentiation includes development of different genitalia and the internal genital tracts and body hair plays a role in sex identification.

<span class="mw-page-title-main">Estrogen insensitivity syndrome</span> Medical condition

Estrogen insensitivity syndrome (EIS), or estrogen resistance, is a form of congenital estrogen deficiency or hypoestrogenism which is caused by a defective estrogen receptor (ER) – specifically, the estrogen receptor alpha (ERα) – that results in an inability of estrogen to mediate its biological effects in the body. Congenital estrogen deficiency can alternatively be caused by a defect in aromatase, the enzyme responsible for the biosynthesis of estrogens, a condition which is referred to as aromatase deficiency and is similar in symptomatology to EIS.

17β-Hydroxysteroid dehydrogenases, also 17-ketosteroid reductases (17-KSR), are a group of alcohol oxidoreductases which catalyze the reduction of 17-ketosteroids and the dehydrogenation of 17β-hydroxysteroids in steroidogenesis and steroid metabolism. This includes interconversion of DHEA and androstenediol, androstenedione and testosterone, and estrone and estradiol.

<span class="mw-page-title-main">Inborn errors of steroid metabolism</span> Medical condition

An inborn error of steroid metabolism is an inborn error of metabolism due to defects in steroid metabolism.

<span class="mw-page-title-main">3β-Androstanediol</span> Chemical compound

3β-Androstanediol, also known as 5α-androstane-3β,17β-diol, and sometimes shortened in the literature to 3β-diol, is an endogenous steroid hormone and a metabolite of androgens like dehydroepiandrosterone (DHEA) and dihydrotestosterone (DHT).

<span class="mw-page-title-main">Leydig cell hypoplasia</span> Medical condition

Leydig cell hypoplasia (LCH), also known as Leydig cell agenesis, is a rare autosomal recessive genetic and endocrine syndrome affecting an estimated 1 in 1,000,000 genetic males. It is characterized by an inability of the body to respond to luteinizing hormone (LH), a gonadotropin which is normally responsible for signaling Leydig cells of the testicles to produce testosterone and other androgen sex hormones. The condition manifests itself as pseudohermaphroditism, hypergonadotropic hypogonadism, reduced or absent puberty, and infertility.

Follicle-stimulating hormone (FSH) insensitivity, or ovarian insensitivity to FSH in females, also referable to as ovarian follicle hypoplasia or granulosa cell hypoplasia in females, is a rare autosomal recessive genetic and endocrine syndrome affecting both females and males, with the former presenting with much greater severity of symptomatology. It is characterized by a resistance or complete insensitivity to the effects of follicle-stimulating hormone (FSH), a gonadotropin which is normally responsible for the stimulation of estrogen production by the ovaries in females and maintenance of fertility in both sexes. The condition manifests itself as hypergonadotropic hypogonadism, reduced or absent puberty, amenorrhea, and infertility in females, whereas males present merely with varying degrees of infertility and associated symptoms.

Adrenal steroids are steroids that are derived from the adrenal glands. They include corticosteroids, which consist of glucocorticoids like cortisol and mineralocorticoids like aldosterone, adrenal androgens like dehydroepiandrosterone (DHEA), DHEA sulfate (DHEA-S), and androstenedione (A4), and neurosteroids like DHEA and DHEA-S, as well as pregnenolone and pregnenolone sulfate (P5-S). Adrenal steroids are specifically produced in the adrenal cortex.

<span class="mw-page-title-main">Pharmacology of bicalutamide</span>

The pharmacology of bicalutamide is the study of the pharmacodynamic and pharmacokinetic properties of the nonsteroidal antiandrogen (NSAA) bicalutamide. In terms of pharmacodynamics, bicalutamide acts as a selective antagonist of the androgen receptor (AR), the biological target of androgens like testosterone and dihydrotestosterone (DHT). It has no capacity to activate the AR. It does not decrease androgen levels and has no other important hormonal activity. The medication has progonadotropic effects due to its AR antagonist activity and can increase androgen, estrogen, and neurosteroid production and levels. This results in a variety of differences of bicalutamide monotherapy compared to surgical and medical castration, such as indirect estrogenic effects and associated benefits like preservation of sexual function and drawbacks like gynecomastia. Bicalutamide can paradoxically stimulate late-stage prostate cancer due to accumulated mutations in the cancer. When used as a monotherapy, bicalutamide can induce breast development in males due to its estrogenic effects. Unlike other kinds of antiandrogens, it may have less adverse effect on the testes and fertility.

<span class="mw-page-title-main">Androgen backdoor pathway</span> Series of interconnected biochemical reactions

The androgen backdoor pathway synthesizes physiologically relevant androgens from 21-carbon steroids (pregnanes) via 5α-reduction, bypassing testosterone. This differs from the conventional, canonical androgenic pathway, which involves testosterone.

References

  1. Moini J (2015). Fundamental Pharmacology for Pharmacy Technicians. Cengage Learning. p. 338. ISBN   978-1-30-568615-1. Androgen is the generic term for any natural or synthetic compound, usually a steroid hormone, which stimulates or controls the development of masculine characteristics by binding to androgen receptors.
  2. Gylys BA, Wedding ME (2017). Medical Terminology Systems: A Body Systems Approach. F.A. Davis. p. 82. ISBN   978-0-80-365868-4. Generic term for an agent (usually a hormone, such as testosterone or androsterone) that stimulates development of male characteristics.
  3. "15 Ways To Get Rid of Pimples Overnight Natural". Fast Health Fitness. 17 May 2016.
  4. 1 2 Carlson N (22 January 2012). Physiology of Behavior. Reproductive Behavior. Vol. 11th edition. Pearson. p. 326. ISBN   978-0205239399.
  5. "Androgens". DIAsource. Archived from the original on 8 August 2014. Retrieved 26 June 2013.
  6. 1 2 Adriaansen BP, Oude Alink SE, Swinkels DW, Schröder MA, Span PN, Sweep FC, et al. (January 2024). "Reference intervals for serum 11-oxygenated androgens in children". European Journal of Endocrinology. 190 (1): 96–103. doi: 10.1093/ejendo/lvae008 . PMID   38243909.
  7. 1 2 Briggs MH, Brotherton J (3 February 1970). Steroid Biochemistry and Pharmacology. London: Academic Press. ISBN   978-0-12-134650-8.
  8. 1 2 Abraham GE (August 1974). "Ovarian and adrenal contribution to peripheral androgens during the menstrual cycle". The Journal of Clinical Endocrinology and Metabolism. 39 (2): 340–346. doi:10.1210/jcem-39-2-340. PMID   4278727.
  9. Gilbert SF (2000). Developmental Biology (6th ed.). Sunderland, Massachusetts: Sinauer Associates. ISBN   978-0-87893-243-6.[ page needed ]
  10. Stephen Nussey, Saffron Whitehead (2001). Saffron A. Whitehead, Stephen Nussey (eds.). Endocrinology: an integrated approach. Oxford: British Institute of Organ Studies. ISBN   978-1-85996-252-7.[ page needed ]
  11. Singh R, Artaza JN, Taylor WE, Braga M, Yuan X, Gonzalez-Cadavid NF, Bhasin S (January 2006). "Testosterone inhibits adipogenic differentiation in 3T3-L1 cells: nuclear translocation of androgen receptor complex with beta-catenin and T-cell factor 4 may bypass canonical Wnt signaling to down-regulate adipogenic transcription factors". Endocrinology. 147 (1): 141–154. doi:10.1210/en.2004-1649. PMC   4417624 . PMID   16210377.
  12. Sinha-Hikim I, Taylor WE, Gonzalez-Cadavid NF, Zheng W, Bhasin S (October 2004). "Androgen receptor in human skeletal muscle and cultured muscle satellite cells: up-regulation by androgen treatment". The Journal of Clinical Endocrinology and Metabolism. 89 (10): 5245–5255. doi: 10.1210/jc.2004-0084 . PMID   15472231.
  13. Vlahopoulos S, Zimmer WE, Jenster G, Belaguli NS, Balk SP, Brinkmann AO, et al. (March 2005). "Recruitment of the androgen receptor via serum response factor facilitates expression of a myogenic gene". The Journal of Biological Chemistry. 280 (9): 7786–7792. doi: 10.1074/jbc.M413992200 . PMID   15623502.
  14. Cooke B, Hegstrom CD, Villeneuve LS, Breedlove SM (October 1998). "Sexual differentiation of the vertebrate brain: principles and mechanisms". Frontiers in Neuroendocrinology. 19 (4): 323–362. doi:10.1006/frne.1998.0171. PMID   9799588. S2CID   14372914.
  15. Irwig MS (April 2017). "Testosterone therapy for transgender men". The Lancet. Diabetes & Endocrinology. 5 (4): 301–311. doi:10.1016/S2213-8587(16)00036-X. PMID   27084565.
  16. Costantino A, Cerpolini S, Alvisi S, Morselli PG, Venturoli S, Meriggiola MC (14 February 2013). "A prospective study on sexual function and mood in female-to-male transsexuals during testosterone administration and after sex reassignment surgery". Journal of Sex & Marital Therapy. 39 (4): 321–335. doi:10.1080/0092623X.2012.736920. PMID   23470169. S2CID   34943756.
  17. Johnson JM, Nachtigall LB, Stern TA (1 November 2013). "The effect of testosterone levels on mood in men: a review". Psychosomatics. 54 (6): 509–514. doi:10.1016/j.psym.2013.06.018. PMID   24016385.
  18. Zuloaga DG, Puts DA, Jordan CL, Breedlove SM (May 2008). "The role of androgen receptors in the masculinization of brain and behavior: what we've learned from the testicular feminization mutation". Hormones and Behavior. 53 (5): 613–626. doi:10.1016/j.yhbeh.2008.01.013. PMC   2706155 . PMID   18374335.
  19. Hamson DK, Wainwright SR, Taylor JR, Jones BA, Watson NV, Galea LA (September 2013). "Androgens increase survival of adult-born neurons in the dentate gyrus by an androgen receptor-dependent mechanism in male rats". Endocrinology. 154 (9): 3294–3304. doi: 10.1210/en.2013-1129 . hdl: 2429/63213 . PMID   23782943.
  20. Okamoto M, Hojo Y, Inoue K, Matsui T, Kawato S, McEwen BS, Soya H (August 2012). "Mild exercise increases dihydrotestosterone in hippocampus providing evidence for androgenic mediation of neurogenesis". Proceedings of the National Academy of Sciences of the United States of America. 109 (32): 13100–13105. Bibcode:2012PNAS..10913100O. doi: 10.1073/pnas.1210023109 . PMC   3420174 . PMID   22807478.
  21. Zhang JM, Tonelli L, Regenold WT, McCarthy MM (August 2010). "Effects of neonatal flutamide treatment on hippocampal neurogenesis and synaptogenesis correlate with depression-like behaviors in preadolescent male rats". Neuroscience. 169 (1): 544–554. doi:10.1016/j.neuroscience.2010.03.029. PMC   3574794 . PMID   20399256.
  22. Spritzer MD, Ibler E, Inglis W, Curtis MG (November 2011). "Testosterone and social isolation influence adult neurogenesis in the dentate gyrus of male rats". Neuroscience. 195: 180–190. doi:10.1016/j.neuroscience.2011.08.034. PMC   3198792 . PMID   21875652.
  23. Makieva S, Saunders PT, Norman JE (2014). "Androgens in pregnancy: roles in parturition". Human Reproduction Update. 20 (4): 542–559. doi:10.1093/humupd/dmu008. PMC   4063701 . PMID   24643344.
  24. Wright AS, Thomas LN, Douglas RC, Lazier CB, Rittmaster RS (December 1996). "Relative potency of testosterone and dihydrotestosterone in preventing atrophy and apoptosis in the prostate of the castrated rat". The Journal of Clinical Investigation. 98 (11): 2558–2563. doi:10.1172/JCI119074. PMC   507713 . PMID   8958218.
  25. Adriaansen BP, Oude Alink SE, Swinkels DW, Schröder MA, Span PN, Sweep FC, et al. (January 2024). "Reference intervals for serum 11-oxygenated androgens in children". European Journal of Endocrinology. 190 (1): 96–103. doi: 10.1093/ejendo/lvae008 . PMID   38243909.
  26. Bennett NC, Gardiner RA, Hooper JD, Johnson DW, Gobe GC (June 2010). "Molecular cell biology of androgen receptor signalling". The International Journal of Biochemistry & Cell Biology. 42 (6): 813–827. doi:10.1016/j.biocel.2009.11.013. PMID   19931639.
  27. Wang C, Liu Y, Cao JM (September 2014). "G protein-coupled receptors: extranuclear mediators for the non-genomic actions of steroids". International Journal of Molecular Sciences. 15 (9): 15412–15425. doi: 10.3390/ijms150915412 . PMC   4200746 . PMID   25257522.
  28. Lang F, Alevizopoulos K, Stournaras C (August 2013). "Targeting membrane androgen receptors in tumors". Expert Opinion on Therapeutic Targets. 17 (8): 951–963. doi:10.1517/14728222.2013.806491. PMID   23746222. S2CID   23918273.