Anabolic steroid

Chemical structure of the natural anabolic hormone testosterone, 17β-hydroxy-4-androsten-3-one
Chemical structure of the synthetic steroid metandienone (Dianabol). 17α-Methylation (upper-right corner) enhances oral bioavailability.
This article is about androgens as medications. For androgens as natural hormones, see Androgen.

Anabolic steroids, also known more properly as anabolic-androgenic steroids (AAS),[1] are steroidal androgens that are structurally related to and have similar effects as testosterone in the body. They are anabolic and increase protein within cells, especially in skeletal muscles. AAS also have androgenic and virilizing effects, including induction of the development and maintenance of masculine secondary sexual characteristics such as the growth of the vocal cords and body hair. The word anabolic, referring to anabolism, comes from the Greek ἀναβολή anabole, "that which is thrown up, mound."

AAS were synthesized in the 1930s, and are now used therapeutically in medicine to stimulate muscle growth and appetite, induce male puberty and treat chronic wasting conditions, such as cancer and AIDS. The American College of Sports Medicine acknowledges that AAS, in the presence of adequate diet, can contribute to increases in body weight, often as lean mass increases and that the gains in muscular strength achieved through high-intensity exercise and proper diet can be additionally increased by the use of AAS in some individuals.[2]

Health risks can be produced by long-term use or excessive doses of AAS.[3][4] These effects include harmful changes in cholesterol levels (increased low-density lipoprotein and decreased high-density lipoprotein), acne, high blood pressure, liver damage (mainly with most oral AAS), and dangerous changes in the structure of the left ventricle of the heart.[5] Conditions pertaining to hormonal imbalances such as gynecomastia and testicular size reduction may also be caused by AAS.

Ergogenic uses for AAS in sports, racing, and bodybuilding as performance-enhancing drugs are controversial because of their adverse effects and the potential to gain unfair advantage in competitive physical competitions. Their use is referred to as doping and banned by most major sporting bodies. For many years, AAS have been by far the most detected doping substances in IOC-accredited laboratories.[6][7] In countries where AAS are controlled substances, there is often a black market in which smuggled, clandestinely manufactured or even counterfeit drugs are sold to users.

Examples of AAS

The most commonly used AAS in medicine are testosterone and its various esters (but most commonly testosterone undecanoate, testosterone enanthate, testosterone cypionate, and testosterone propionate),[8] nandrolone (as, most commonly, nandrolone decanoate or nandrolone phenylpropionate), stanozolol, and metandienone (methandrostenolone).[1] Others also available and used commonly but to a lesser extent include methyltestosterone, oxandrolone, mesterolone, and oxymetholone, as well as drostanolone propionate (dromostanolone propionate), metenolone (methylandrostenolone), and fluoxymesterone.[1] Boldenone undecylenate and trenbolone acetate are used in veterinary medicine.[1]

Uses

Medical

Various anabolic steroids and related compounds

Since the discovery and synthesis of testosterone in the 1930s, anabolic steroids have been used by physicians for many purposes, with varying degrees of success, for the treatment of:

Enhancing performance

Numerous vials of injectable anabolic steroids

Most steroid users are not athletes.[32] Between 1 million and 3 million people (1% of the population) are thought to have misused AAS in the United States.[33] Studies in the United States have shown that anabolic steroid users tend to be mostly middle-class heterosexual men with a median age of about 25 who are noncompetitive bodybuilders and non-athletes and use the drugs for cosmetic purposes.[34] "Among 12- to 17-year-old boys, use of steroids and similar drugs jumped 25 percent from 1999 to 2000, with 20 percent saying they use them for looks rather than sports, a study by insurer Blue Cross Blue Shield found."(Eisenhauer) Another study found that non-medical use of AAS among college students was at or less than 1%.[35] According to a recent survey, 78.4% of steroid users were noncompetitive bodybuilders and non-athletes, while about 13% reported unsafe injection practices such as reusing needles, sharing needles, and sharing multidose vials,[36] though a 2007 study found that sharing of needles was extremely uncommon among individuals using anabolic steroids for non-medical purposes, less than 1%.[37] Another 2007 study found that 74% of non-medical anabolic steroid users had post-secondary degrees and more had completed college and fewer had failed to complete high school than is expected from the general populace.[37] The same study found that individuals using anabolic steroids for non-medical purposes had a higher employment rate and a higher household income than the general population.[37] Anabolic steroid users tend to research the drugs they are taking more than other controlled-substance users; however, the major sources consulted by steroid users include friends, non-medical handbooks, internet-based forums, blogs, and fitness magazines, which can provide questionable or inaccurate information.[38]

Anabolic steroid users tend to be disillusioned by the portrayal of anabolic steroids as deadly in the media and in politics.[39] According to one study, AAS users also distrust their physicians and in the sample 56% had not disclosed their AAS use to their physicians.[40] Another 2007 study had similar findings, showing that, while 66% of individuals using anabolic steroids for non-medical purposes were willing to seek medical supervision for their steroid use, 58% lacked trust in their physicians, 92% felt that the medical community's knowledge of non-medical anabolic steroid use was lacking, and 99% felt that the public has an exaggerated view of the side-effects of anabolic steroid use.[37] A recent study has also shown that long term AAS users were more likely to have symptoms of muscle dysmorphia and also showed stronger endorsement of more conventional male roles.[41] A recent study in the Journal of Health Psychology showed that many users believed that steroids used in moderation were safe.[42]

Anabolic steroids have been used by men and women in many different kinds of professional sports to attain a competitive edge or to assist in recovery from injury. These sports include bodybuilding, weightlifting, shot put and other track and field, cycling, baseball, wrestling, mixed martial arts, boxing, football, and cricket. Such use is prohibited by the rules of the governing bodies of most sports. Anabolic steroid use occurs among adolescents, especially by those participating in competitive sports. It has been suggested that the prevalence of use among high-school students in the U.S. may be as high as 2.7%.[43] Male students used anabolic steroids more frequently than female students and, on average, those that participated in sports used steroids more often than those that did not.

Available forms

A vial of injectable testosterone cypionate

There are four common forms in which anabolic steroids are administered: oral pills; injectable steroids; creams/gels for topical application; and skin patches. Oral administration is the most convenient. Testosterone administered by mouth is rapidly absorbed, but it is largely converted to inactive metabolites, and only about 1/6 is available in active form. In order to be sufficiently active when given by mouth, testosterone derivatives are alkylated at the 17 position, e.g. methyltestosterone and fluoxymesterone. This modification reduces the liver's ability to break down these compounds before they reach the systemic circulation.

Testosterone can be administered parenterally, but it has more irregular prolonged absorption time and greater activity in muscle in enanthate, undecanoate, or cypionate ester form. These derivatives are hydrolyzed to release free testosterone at the site of injection; absorption rate (and thus injection schedule) varies among different esters, but medical injections are normally done anywhere between semi-weekly to once every 12 weeks. A more frequent schedule may be desirable in order to maintain a more constant level of hormone in the system.[44] Injectable steroids are typically administered into the muscle, not into the vein, to avoid sudden changes in the amount of the drug in the bloodstream. In addition, because estered testosterone is dissolved in oil, intravenous injection has the potential to cause a dangerous embolism (clot) in the bloodstream.

Transdermal patches (adhesive patches placed on the skin) may also be used to deliver a steady dose through the skin and into the bloodstream. Testosterone-containing creams and gels that are applied daily to the skin are also available, but absorption is inefficient (roughly 10%, varying between individuals) and these treatments tend to be more expensive. Individuals who are especially physically active and/or bathe often may not be good candidates, since the medication can be washed off and may take up to six hours to be fully absorbed. There is also the risk that an intimate partner or child may come in contact with the application site and inadvertently dose himself or herself; children and women are highly sensitive to testosterone and can suffer unintended masculinization and health effects, even from small doses. Injection is the most common method used by individuals administering anabolic steroids for non-medical purposes.[37]

The traditional routes of administration do not have differential effects on the efficacy of the drug. Studies indicate that the anabolic properties of anabolic steroids are relatively similar despite the differences in pharmacokinetic principles such as first-pass metabolism. However, the orally available forms of AAS may cause liver damage in high doses.[7][45]

Adverse effects

Addiction experts in psychiatry, chemistry, pharmacology, forensic science, epidemiology, and the police and legal services engaged in delphic analysis regarding 20 popular recreational drugs. Anabolic steroids were ranked 19th in dependence, 9th in physical harm, and 15th in social harm.[46]

Neuropsychiatric

A 2005 review in CNS Drugs determined that "significant psychiatric symptoms including aggression and violence, mania, and less frequently psychosis and suicide have been associated with steroid abuse. Long-term steroid abusers may develop symptoms of dependence and withdrawal on discontinuation of AAS".[47] High concentrations of AAS, comparable to those likely sustained by many recreational AAS users, produce apoptotic effects on neurons , raising the specter of possibly irreversible neurotoxicity. Recreational AAS use appears to be associated with a range of potentially prolonged psychiatric effects, including dependence syndromes, mood disorders, and progression to other forms of substance abuse, but the prevalence and severity of these various effects remains poorly understood.[48] There is no evidence that steroid dependence develops from therapeutic use of anabolic steroids to treat medical disorders, but instances of AAS dependence have been reported among weightlifters and bodybuilders who chronically administered supraphysiologic doses.[49] Mood disturbances (e.g. depression, [hypo-]mania, psychotic features) are likely to be dose- and drug-dependent, but AAS dependence or withdrawal effects seem to occur only in a small number of AAS users.[6]

Large-scale long-term studies of psychiatric effects on AAS users are not currently available.[48] In 2003, the first naturalistic long-term study on ten users, seven of which having completed the study, found a high incidence of mood disorders and substance abuse, but few clinically relevant changes in physiological parameters or laboratory measures were noted throughout the study, and these changes were not clearly related to periods of reported AAS use.[50] A 13-month study, which was published in 2006 and which involved 320 body builders and athletes suggests that the wide range of psychiatric side-effects induced by the use of AAS is correlated to the severity of abuse.[51]

Diagnostic Statistical Manual assertion

DSM-IV lists General diagnostic criteria for a personality disorder guideline that "The pattern must not be better accounted for as a manifestation of another mental disorder, or to the direct physiological effects of a substance (e.g. drug or medication) or a general medical condition (e.g. head trauma).". As a result, anabolic steroid users may get misdiagnosed by a psychiatrist not told about their habit.[52]

Personality profiles

Cooper, Noakes, Dunne, Lambert, and Rochford identified that anabolic–androgenic steroid (AAS) using individuals are more likely to score higher on borderline (4.7 times), antisocial (3.8 times), paranoid (3.4 times), schizotypal (3.1 times), histrionic (2.9 times), passive-aggressive (2.4 times), and narcissistic (1.6 times) personality profiles than non-users.[53] Other studies have suggested that antisocial personality disorder is slightly more likely among anabolic steroid users than among non-users (Pope & Katz, 1994).[52] Bipolar dysfunction,[54] substance dependency, and conduct disorder have also been associated with AAS use.[55]

Mood and anxiety

Affective disorders have long been recognised as a complication of anabolic steroid use. Case reports describe both hypomania and mania, along with irritability, elation, recklessness, racing thoughts and feelings of power and invincibility that did not meet the criteria for mania/hypomania.[56] Of 53 bodybuilders who used anabolic steroids, 27 (51%) reported unspecified mood disturbance.[57]

Aggression and hypomania

From the mid-1980s onward, the media reported "roid rage" as a side-effect of AAS.[58]:23

A 2005 review determined that some, but not all, randomized controlled studies have found that anabolic steroid use correlates with hypomania and increased aggressiveness, but pointed out that attempts to determine whether AAS use triggers violent behavior have failed, primarily because of high rates of non-participation.[59] A 2008 study on a nationally representative sample of young adult males in the United States found an association between lifetime and past-year self-reported anabolic-androgenic steroid use and involvement in violent acts. Compared with individuals that did not use steroids, young adult males that used anabolic-androgenic steroids reported greater involvement in violent behaviors even after controlling for the effects of key demographic variables, previous violent behavior, and polydrug use.[60] A 1996 review examining the blind studies available at that time also found that these had demonstrated a link between aggression and steroid use, but pointed out that with estimates of over one million past or current steroid users in the United States at that time, an extremely small percentage of those using steroids appear to have experienced mental disturbance severe enough to result in clinical treatments or medical case reports.[61]

A 1996 randomized controlled trial, which involved 43 men, did not find an increase in the occurrence of angry behavior during 10 weeks of administration of testosterone enanthate at 600 mg/week, but this study screened out subjects that had previously abused steroids or had any psychiatric antecedents.[62][63] A trial conducted in 2000 using testosterone cypionate at 600 mg/week found that treatment significantly increased manic scores on the YMRS, and aggressive responses on several scales. The drug response was highly variable. However: 84% of subjects exhibited minimal psychiatric effects, 12% became mildly hypomanic, and 4% (2 subjects) became markedly hypomanic. The mechanism of these variable reactions could not be explained by demographic, psychological, laboratory, or physiological measures.[64]

A 2006 study of two pairs of identical twins, in which one twin used anabolic steroids and the other did not, found that in both cases the steroid-using twin exhibited high levels of aggressiveness, hostility, anxiety, and paranoid ideation not found in the "control" twin.[65] A small-scale study of 10 AAS users found that cluster B personality disorders were confounding factors for aggression.[66]

Androgenic steroids are known to increase aggression with greater likelihood when compared to more anabolic steroids. Trenbolone is frequently noted for its increases in aggression. Equipoise is known to increase anxiety in some users.

The relationship between AAS use and depression is inconclusive. There have been anecdotal reports of depression and suicide in teenage steroid users,[67] but little systematic evidence. A 1992 review found that anabolic-androgenic steroids may both relieve and cause depression, and that cessation or diminished use of anabolic-androgenic steroids may also result in depression, but called for additional studies due to disparate data.[68] In the case of suicide, 3.9% of a sample of 77 those classified as AAS users reported attempting suicide during withdrawal (Malone, Dimeff, Lombardo, & Sample, 1995).[69]

Physiological

Depending on the length of drug abuse, there is a chance that the immune system can be damaged. Most of these side-effects are dose-dependent, the most common being elevated blood pressure, especially in those with pre-existing hypertension.[70] In addition to morphological changes of the heart which may alter cardiovascular inefficiency irreversibly.

Anabolic steroids have been shown to alter fasting blood sugar and glucose tolerance tests.[71] Anabolic steroids such as testosterone also increase the risk of cardiovascular disease[3] or coronary artery disease.[72][73] Acne is fairly common among anabolic steroid users, mostly due to stimulation of the sebaceous glands by increased testosterone levels.[6][74] Conversion of testosterone to dihydrotestosterone (DHT) can accelerate the rate of premature baldness for males genetically predisposed, but testosterone itself can produce baldness in females.[75]

A number of severe side-effects can occur if adolescents use anabolic steroids.

For example, the steroids may prematurely stop the lengthening of bones (premature epiphyseal fusion through increased levels of estrogen metabolites), resulting in stunted growth. Other effects include, but are not limited to, accelerated bone maturation, increased frequency and duration of erections, and premature sexual development. Anabolic steroid use in adolescence is also correlated with poorer attitudes related to health.[76]

Cancer

WHO organization International Agency for Research on Cancer (IARC) list Androgenic (anabolic) steroids under Group 2A: Probably carcinogenic to humans.[77]

Cardiovascular

Other side-effects can include alterations in the structure of the heart, such as enlargement and thickening of the left ventricle, which impairs its contraction and relaxation.[5] Possible effects of these alterations in the heart are hypertension, cardiac arrhythmias, congestive heart failure, heart attacks, and sudden cardiac death.[78] These changes are also seen in non-drug-using athletes, but steroid use may accelerate this process.[79][80] However, both the connection between changes in the structure of the left ventricle and decreased cardiac function, as well as the connection to steroid use have been disputed.[81][82]

AAS use can cause harmful changes in cholesterol levels: Some steroids cause an increase in LDL "bad" cholesterol and a decrease in HDL "good" cholesterol.[83] In addition, steroids provoke a rapid increase in body weight and an accompanying rise in blood pressure, both of which leave users more vulnerable to a cardiovascular event.

Growth defects

AAS use in adolescents quickens bone maturation and may reduce adult height in high doses. Low doses of anabolic steroids such as oxandrolone are used in the treatment of idiopathic short stature, but this may only quicken maturation rather than increasing adult height.[84]

Feminization

Male with gynecomastia

There are also sex-specific side effects of anabolic steroids. Development of breast tissue in males, a condition called gynecomastia (which is usually caused by high levels of circulating estradiol), may arise because of increased conversion of testosterone to estradiol by the enzyme aromatase.[85] Reduced sexual function and temporary infertility can also occur in males.[14][86][87] Another male-specific side-effect that can occur is testicular atrophy, caused by the suppression of natural testosterone levels, which inhibits production of sperm (most of the mass of the testes is developing sperm). This side-effect is temporary: The size of the testicles usually returns to normal within a few weeks of discontinuing anabolic steroid use as normal production of sperm resumes.[88]

Masculinization

See also: Virilization

Female-specific side effects include increases in body hair, permanent deepening of the voice, enlarged clitoris, and temporary decreases in menstrual cycles. Alteration of fertility and ovarian cysts can also occur in females.[89] When taken during pregnancy, anabolic steroids can affect fetal development by causing the development of male features in the female fetus and female features in the male fetus.[90]

Kidney problems

Kidney tests revealed that nine of the ten steroid users developed a condition called focal segmental glomerulosclerosis, a type of scarring within the kidneys. The kidney damage in the bodybuilders has similarities to that seen in morbidly obese patients, but appears to be even more severe.[91]

Liver problems

High doses of oral anabolic steroid compounds can cause liver damage, as the steroids are metabolized (17α-alkylated) in the digestive system to increase their bioavailability and stability.[4] Peliosis hepatis has been increasingly recognised with the use of anabolic steroids.

Pharmacology

Mechanism of action

See also: Steroid hormone
The human androgen receptor bound to testosterone[92] The protein is shown as a ribbon diagram in red, green, and blue, with the steroid shown in white.

The pharmacodynamics of anabolic steroids are unlike peptide hormones. Water-soluble peptide hormones cannot penetrate the fatty cell membrane and only indirectly affect the nucleus of target cells through their interaction with the cell’s surface receptors. However, as fat-soluble hormones, anabolic steroids are membrane-permeable and influence the nucleus of cells by direct action. The pharmacodynamic action of anabolic steroids begin when the exogenous hormone penetrates the membrane of the target cell and binds to an androgen receptor located in the cytoplasm of that cell. From there, the compound hormone-receptor diffuses into the nucleus, where it either alters the expression of genes[93] or activates processes that send signals to other parts of the cell.[94] Different types of anabolic steroids bind to the androgen receptor with different affinities, depending on their chemical structure.[6] Some anabolic steroids such as methandrostenolone bind weakly to this receptor in vitro, but still exhibit androgenic effects in vivo. The reason for this discrepancy is not known.[95]

The effect of anabolic steroids on muscle mass is caused in at least two ways:[96] first, they increase the production of proteins; second, they reduce recovery time by blocking the effects of stress hormone cortisol on muscle tissue, so that catabolism of muscle is greatly reduced. It has been hypothesized that this reduction in muscle breakdown may occur through anabolic steroids inhibiting the action of other steroid hormones called glucocorticoids that promote the breakdown of muscles.[97] Anabolic steroids also affect the number of cells that develop into fat-storage cells, by favouring cellular differentiation into muscle cells instead.[98] Anabolic steroids can also decrease fat by increasing basal metabolic rate (BMR), since an increase in muscle mass increases BMR.

Anabolic and androgenic effects

Relative androgenic:anabolic
activity in animals[44]:735
Preparation Ratio
Testosterone 1:1
Testosterone cypionate 1:1
Testosterone enanthate 1:1
Methyltestosterone 1:1
Fluoxymesterone 1:2
Oxymetholone 1:3
Oxandrolone 1:3–1:13
Nandrolone decanoate 1:2.5–1:4

As the name suggests, anabolic-androgenic steroids have two different, but overlapping, types of effects: anabolic, meaning that they promote anabolism (cell growth), and androgenic (or virilising), meaning that they affect the development and maintenance of masculine characteristics.

Some examples of the anabolic effects of these hormones are increased protein synthesis from amino acids, increased appetite, increased bone remodeling and growth, and stimulation of bone marrow, which increases the production of red blood cells. Through a number of mechanisms anabolic steroids stimulate the formation of muscle cells and hence cause an increase in the size of skeletal muscles, leading to increased strength.[99][100][101]

The androgenic effects of AAS are numerous. Depending on the length of use, the side effects of the steroid can be irreversible. Processes affected include pubertal growth, sebaceous gland oil production, and sexuality (especially in fetal development). Some examples of virilizing effects are growth of the clitoris in females and the penis in male children (the adult penis size does not change due to steroids ), increased vocal cord size, increased libido, suppression of natural sex hormones, and impaired production of sperm.[102] Effects on women include deepening of the voice, facial hair growth, and possibly a decrease in breast size. Men may develop an enlargement of breast tissue, known as gynecomastia, testicular atrophy, and a reduced sperm count.[103]

The androgenic:anabolic ratio of an AAS is an important factor when determining the clinical application of these compounds. Compounds with a high ratio of androgenic to an anabolic effects are the drug of choice in androgen-replacement therapy (e.g., treating hypogonadism in males), whereas compounds with a reduced androgenic:anabolic ratio are preferred for anemia and osteoporosis, and to reverse protein loss following trauma, surgery, or prolonged immobilization. Determination of androgenic:anabolic ratio is typically performed in animal studies, which has led to the marketing of some compounds claimed to have anabolic activity with weak androgenic effects. This disassociation is less marked in humans, where all anabolic steroids have significant androgenic effects.[44]

A commonly used protocol for determining the androgenic:anabolic ratio, dating back to the 1950s, uses the relative weights of ventral prostate (VP) and levator ani muscle (LA) of male rats. The VP weight is an indicator of the androgenic effect, while the LA weight is an indicator of the anabolic effect. Two or more batches of rats are castrated and given no treatment and respectively some AAS of interest. The LA/VP ratio for an AAS is calculated as the ratio of LA/VP weight gains produced by the treatment with that compound using castrated but untreated rats as baseline: (LAc,t–LAc)/(VPc,t–VPc). The LA/VP weight gain ratio from rat experiments is not unitary for testosterone (typically 0.3–0.4), but it is normalized for presentation purposes, and used as basis of comparison for other AAS, which have their androgenic:anabolic ratios scaled accordingly (as shown in the table above).[95][104] In the early 2000s, this procedure was standardized and generalized throughout OECD in what is now known as the Hershberger assay.

Body composition and strength improvements

Body weight in men may increase by 2–5 kg as a result of short-term (<10 weeks) AAS use, which may be attributed mainly to an increase of lean mass. Animal studies also found that fat mass was reduced, but most studies in humans failed to elucidate significant fat mass decrements. The effects on lean body mass have been shown to be dose-dependent. Both muscle hypertrophy and the formation of new muscle fibers have been observed. The hydration of lean mass remains unaffected by AAS use, although small increments of blood volume cannot be ruled out.[6]

The upper region of the body (thorax, neck, shoulders, and upper arm) seems to be more susceptible for AAS than other body regions because of predominance of androgen receptors in the upper body. The largest difference in muscle fiber size between AAS users and non-users was observed in type I muscle fibers of the vastus lateralis and the trapezius muscle as a result of long-term AAS self-administration. After drug withdrawal, the effects fade away slowly, but may persist for more than 6–12 weeks after cessation of AAS use.[6]

Strength improvements in the range of 5–20% of baseline strength, depending largely on the drugs and dose used as well as the administration period. Overall, the exercise where the most significant improvements were observed is the bench press.[6] For almost two decades, it was assumed that AAS exerted significant effects only in experienced strength athletes.[105][106] A randomized controlled trial demonstrated, however, that even in novice athletes a 10-week strength training program accompanied by testosterone enanthate at 600 mg/week may improve strength more than training alone does.[6][62] This dose is sufficient to significantly improve lean muscle mass relative to placebo even in subjects that did not exercise at all.[62] The anabolic effects of testosterone enanthate were highly dose dependent.[6][107]

Dissociation of anabolic and androgenic effects

Endogenous androgens like testosterone and DHT and synthetic anabolic-androgenic steroids mediate their effects by binding to and activating the androgen receptor (AR).[1] On the basis of animal bioassays, the effects of these agents have been divided into two partially dissociable types: anabolic (myotrophic) and androgenic.[1] Dissociation between the ratios of these two types of effects is observed in rat bioassays with various anabolic-androgenic steroids relative to the ratio observed with testosterone.[1] Explanations for the dissociation include differences in intracellular metabolism, functional selectivity (recruitment of coactivators), and non-genomic mechanisms (i.e., signaling through non-AR membrane androgen receptors).[1] Support for the latter two explanations is limited and more hypothetical, but there is a good deal of support for the intracellular metabolism explanation.[1]

The measurement of the dissociation between anabolic and androgenic effects among anabolic-androgenic steroids is based largely on a simple although arguably unsophisticated and outdated model involving rat tissue bioassays.[1] It is referred to as the myotrophic-androgenic index.[1] In this model, anabolic (myotrophic) activity is measured by change in the weight of the rat bulbocavernosus/levator ani muscle and androgenic activity is measured by change in the weight of the rat ventral prostate (or, alternatively, the rat seminal vesicles) in response to exposure of the anabolic-androgenic steroid, and the measurements are then compared and used to form a ratio.[1]

Testosterone is metabolized in various tissues by 5α-reductase into DHT, which is 3- to 10-fold more potent as an AR agonist, and by aromatase into estradiol, which is an estrogen and lacks significant AR affinity.[1] In addition, DHT is metabolized by 3α-hydroxysteroid dehydrogenase (3α-HSD) and 3β-hydroxysteroid dehydrogenase (3β-HSD) into metabolites with little or no AR affinity.[1] 5α-Reductase is widely distributed throughout the body, and is concentrated to various extents in skin (particularly the scalp, beard-area of the face, pubic area, and genital area (penis and scrotum)), prostate, seminal vesicles, liver, and the brain.[1] In contrast, expression of 5α-reductase in skeletal muscle is undetectable.[1] Aromatase is highly expressed in adipose tissue and the brain, and is also expressed significantly in skeletal muscle.[1]

Endogenous androgens like testosterone and DHT and synthetic anabolic-androgenic steroids are analogues and are very similar structurally, and due to this structural similarity, have the capacity to bind to and be metabolized by the same enzymes.[1] According to the intracellular metabolism explanation, the androgenic-to-anabolic ratio of a given AR agonist is based on its capacity to be transformed by the aforementioned enzymes in conjunction with the AR activity of any resulting products.[1] As an example, whereas the AR activity of testosterone is greatly potentiated by local conversion via 5α-reductase into DHT in tissues where 5α-reductase is expressed, an anabolic steroid that is not metabolized by 5α-reductase or has already been 5α-reduced, such as DHT itself or a derivative (like mesterolone or drostanolone), would not experience such potentiation in said tissues.[1] Moreover, nandrolone is metabolized by 5α-reductase, but unlike the case of testosterone and DHT, the 5α-reduced metabolite of nandrolone has much lower affinity for the AR than does nandrolone itself, and this results in reduced AR activation in 5α-reductase-expressing tissues.[1] As so-called "androgenic" tissues such as skin/hair follicles and reproductive tissues are very high in 5α-reductase expression, while skeletal muscle is virtually devoid of 5α-reductase, this may primarily explain the high myotrophic-androgenic ratio and dissociation seen with nandrolone, as well as with various other anabolic-androgenic steroids.[1]

Aside from 5α-reductase, aromatase may inactivate testosterone signaling in skeletal muscle and adipose tissue, so anabolic-androgenic steroids that lack aromatase affinity, in addition to being free of the potential side effect of gynecomastia, might be expected to have a higher myotrophic-androgenic ratio in comparison.[1] In addition, DHT is inactivated by high activity of 3α-HSD in skeletal muscle (and cardiac tissue), and anabolic-androgenic steroids that lack affinity for 3α-HSD could similarly be expected to have a higher myotrophic-androgenic ratio (although perhaps also increased long-term cardiovascular risks).[1]

The intracellular metabolism theory explains how and why remarkable dissociation between anabolic and androgenic effects can occur, but why dissociation is invariably incomplete.[1] In support of the model is the rare condition congenital 5α-reductase deficiency, in which the 5α-reductase enzyme is defective, production of DHT is impaired, and DHT levels are very low while testosterone levels are normal.[108][109] Males with this condition are born with ambiguous genitalia and an underdeveloped prostate gland.[108][109] In addition, saliently, such males develop normal musculature, voice changes/deepening, and libido at puberty, but have little to no acne or facial, pubic, or body hair, minimal enlargement of the prostate, and no incidence of male-pattern baldness or prostate cancer.[109][110][111][112][113] They also notably do not develop gynecomastia as a consequence of their condition.[111]

Changes in endogenous testosterone levels may also contribute to differences in myotrophic-androgenic ratio between testosterone and synthetic anabolic-androgenic steroids. Anabolic-androgenic steroids are antigonadotropic – that is, they dose-dependently suppress gonadal testosterone production and hence reduce systemic testosterone concentrations. By suppressing endogenous testosterone levels and effectively replacing AR signaling in the body with that of the exogenous anabolic-androgenic steroid, the myotrophic-androgenic ratio would be expected to be further increased, and this hence may be yet an additional mechanism contributing to the differences in myotrophic-androgenic ratio. In addition, some anabolic-androgenic steroids, such as nandrolone, are also potent progestogens, and activation of the progesterone receptor is antigonadotropic similarly to activation of the AR. As such, combined progestogenic activity might further increase the myotrophic-androgenic ratio for a given anabolic-androgenic steroid.

Interaction with the GABAA receptor

Some anabolic-androgenic steroids, such as testosterone, dihydrotestosterone, stanozolol, and methyltestosterone, have been found to modulate the GABAA receptor similarly to endogenous neurosteroids like allopregnanolone, 3α-androstanediol, dehydroepiandrosterone sulfate, and pregnenolone sulfate.[1] It has been suggested that this may contribute as an alternative or additional mechanism to the neurological and behavioral effects of anabolic-androgenic steroids.[1][114][115][116][117][118][119]

Comparison of AAS

5α-Reductase and androgenicity

Testosterone can be robustly converted by 5α-reductase into dihydrotestosterone (DHT) in so-called androgenic tissues such as skin, scalp, prostate, and seminal vesicles, but not in muscle or bone, where 5α-reductase either is not expressed or is only minimally expressed.[1] As DHT is 3- to 10-fold more potent as an agonist of the androgen receptor (AR) than is testosterone, the AR agonist activity of testosterone is thus markedly and selectively potentiated in such tissues.[1] In contrast to testosterone, DHT and other 4,5α-dihydrogenated AAS are already 5α-reduced, and for this reason, cannot be potentiated in androgenic tissues.[1] 19-Nortestosterone derivatives like nandrolone can be metabolized by 5α-reductase similarly to testosterone, but 5α-reduced metabolites of 19-nortestosterone derivatives tend to have reduced activity as AR agonists, resulting in reduced androgenic activity in tissues that express 5α-reductase.[1] In addition, some 19-nortestosterone derivatives, including trestolone (7α-methyl-19-nortestosterone (MENT)), 11β-methyl-19-nortestosterone (11β-MNT), and dimethandrolone (7α,11β-dimethyl-19-nortestosterone), cannot be 5α-reduced.[120] Conversely, 17α-alkylated AAS can be 5α-reduced and are potentiated in androgenic tissues similarly to testosterone, with an exception being 17α-alkylated AAS that are already 4,5α-reduced.[1]

The capacity to be metabolized by 5α-reductase and the AR activity of the resultant metabolites appears to be one of the major, if not the most important determinant of the androgenic-myotrophic ratio for a given AAS.[1] AAS that are not potentiated by 5α-reductase or that are weakened by 5α-reductase in androgenic tissues have a reduced risk of androgenic side effects such as acne, androgenic alopecia (male-pattern baldness), hirsutism (excessive male-pattern hair growth), benign prostatic hyperplasia (prostate enlargement), and prostate cancer, while incidence and magnitude of other effects such as muscle hypertrophy, bone changes,[121] voice deepening, and changes in sex drive show no difference.[1][122]

Aromatase and estrogenicity

Testosterone can be metabolized by aromatase into estradiol, and many other AAS can be metabolized into their corresponding estrogenic metabolites as well.[1] As an example, the 17α-alkylated AAS methyltestosterone and metandienone are converted by aromatase into methylestradiol.[123] 4,5α-Dihydrogenated derivatives of testosterone such as DHT cannot be aromatized, whereas 19-nortestosterone derivatives like nandrolone can be but to a greatly reduced extent.[1][124] Some 19-nortestosterone derivatives, such as dimethandrolone and 11β-MNT, cannot be aromatized due to steric hindrance provided by their 11β-methyl group, whereas the closely related AAS trestolone (7α-methyl-19-nortestosterone), in relation to its lack of an 11β-methyl group, can be aromatized.[124] AAS that are 17α-alkylated (and not also 4,5α-reduced or 19-demethylated) are also aromatized but to a lesser extent than is testosterone.[1][125] However, it is notable that estrogens that are 17α-substituted (e.g., ethinyl estradiol and methylestradiol) are of markedly increased estrogenic potency due to improved metabolic stability,[123] and for this reason, 17α-alkylated AAS can actually have high estrogenicity and comparatively greater estrogenic effects than testosterone.[123][125]

The major effect of estrogenicity is gynecomastia (woman-like breasts).[1] AAS that have a high potential for aromatization like testosterone and particularly methyltestosterone show a high risk of gynecomastia, while AAS that have a reduced potential for aromatization like nandrolone show a much lower risk (though still significant at high dosages).[1] In contrast, AAS that are 4,5α-reduced, and some other AAS (e.g., 11β-methylated 19-nortestosterone derivatives), have no risk of gynecomastia.[1] In addition to gynecomastia, AAS with high estrogenicity have increased antigonadotropic activity, which results in increased potency in suppression of the hypothalamic-pituitary-gonadal axis and gonadal testosterone production.[126][127]

Progestogenic activity

Some 19-nortestosterone derivatives, including nandrolone, ethylestrenol (ethylnandrol), metribolone (R-1881), trestolone, 11β-MNT, dimethandrolone, and others, are potent agonists of the progesterone receptor (AR) and hence are progestogens in addition to AAS.[1][128] Similarly to the case of estrogenic activity, the progestogenic activity of these drugs serves to augment their antigonadotropic activity.[128] This results in increased potency and effectiveness of these AAS as antispermatogenic agents and male contraceptives (or, put in another way, increased potency and effectiveness in producing azoospermia and male infertility).[128]

Oral activity and hepatotoxicity

Non-17α-alkylated testosterone derivatives such as testosterone itself, DHT, and nandrolone all have poor oral bioavailability due to extensive first-pass hepatic metabolism and hence are not orally active.[1] A notable exception to this are AAS that are androgen precursors or prohormones, including dehydroepiandrosterone (DHEA), androstenediol, androstenedione, boldione (androstadienedione), bolandiol (norandrostenediol), bolandione (norandrostenedione), dienedione, mentabolan (MENT dione, trestione), and methoxydienone (methoxygonadiene) (although these are relatively weak AAS).[129][130] AAS that are not orally active are used almost exclusively in the form of esters administered by intramuscular injection, which act as depots and function as long-acting prodrugs.[1] Examples include testosterone, as testosterone cypionate, testosterone enanthate, and testosterone propionate, and nandrolone, as nandrolone phenylpropionate and nandrolone decanoate, among many others (see here for a full list of testosterone and nandrolone esters).[1] An exception is the very long-chain ester testosterone undecanoate, which is orally active, albeit with only very low oral bioavailability (approximately 3%).[131] In contrast to most other AAS, 17α-alkylated testosterone derivatives show resistance to metabolism due to steric hindrance and are orally active, though they may be esterified and administered via intramuscular injection as well.[1]

In addition to oral activity, 17α-alkylation also confers a high potential for hepatotoxicity, and all 17α-alkylated AAS have been associated, albeit uncommonly and only after prolonged use (different estimates between 1 and 17%),[132][133] with hepatotoxicity.[1][134][135] In contrast, testosterone esters have only extremely rarely or never been associated with hepatotoxicity,[133] and other non-17α-alkylated AAS only rarely, although long-term use may reportedly still increase the risk of hepatic changes (but at a much lower rate than 17α-alkylated AAS, and reportedly not at replacement dosages).[132][136][8] In accordance, D-ring glucuronides of testosterone and DHT have been found to be cholestatic.[137]

Aside from prohormones and testosterone undecanoate, almost all orally active AAS are 17α-alkylated.[138] A few AAS that are not 17α-alkylated are orally active.[1] Some examples include the testosterone 17-ethers cloxotestosterone, quinbolone, and silandrone, which are prodrugs (to testosterone, boldenone1-testosterone), and testosterone, respectively), the DHT 17-ethers mepitiostane, mesabolone, and prostanozol (which are also prodrugs), the 1-methylated DHT derivatives mesterolone and metenolone (although these are relatively weak AAS),[1][8] and the 19-nortestosterone derivatives dimethandrolone and 11β-MNT, which have improved resistance to first-pass hepatic metabolism due to their 11β-methyl groups (in contrast to them, the related AAS trestolone (7α-methyl-19-nortestosterone) is not orally active).[1][128] As these AAS are not 17α-alkylated, they show minimal potential for hepatotoxicity.[1]

Neurosteroid activity

DHT, via its metabolite 3α-androstanediol (produced by 3α-hydroxysteroid dehydrogenase (3α-HSD)), is a neurosteroid that acts via positive allosteric modulation of the GABAA receptor.[1] Testosterone, via conversion into DHT, also produces 3α-androstanediol as a metabolite and hence has similar activity.[1] Some AAS that are or can be 5α-reduced, including testosterone, DHT, stanozolol, and methyltestosterone, among many others, can or may modulate the GABAA receptor, and this may contribute as an alternative or additional mechanism to their central nervous system effects in terms of mood, anxiety, aggression, and sex drive.[1][114][115][116][117][118][119]

Chemistry

AAS are steroids and are derivatives of testosterone with various structural modifications.

Detection in body fluids

The most commonly employed human physiological specimen for detecting anabolic steroid usage is urine, although both blood and hair have been investigated for this purpose. The anabolic steroids, whether of endogenous or exogenous origin, are subject to extensive hepatic biotransformation by a variety of enzymatic pathways. The primary urinary metabolites may be detectable for up to 30 days after the last use, depending on the specific agent, dose and route of administration. A number of the drugs have common metabolic pathways, and their excretion profiles may overlap those of the endogenous steroids, making interpretation of testing results a very significant challenge to the analytical chemist. Methods for detection of the substances or their excretion products in urine specimens usually involve gas chromatography–mass spectrometry or liquid chromatography-mass spectrometry.[139][140][141][142]

History

Isolation of gonadal anabolic-androgenic steroids

The use of gonadal steroids pre-dates their identification and isolation. Medical use of testicle extract began in the late 19th century while its effects on strength were still being studied.[102] The isolation of gonadal steroids can be traced back to 1931, when Adolf Butenandt, a chemist in Marburg, purified 15 milligrams of the male hormone androstenone from tens of thousands of litres of urine. This steroid was subsequently synthesized in 1934 by Leopold Ruzicka, a chemist in Zurich.[143]

In the 1930s, it was already known that the testes contain a more powerful androgen than androstenone, and three groups of scientists, funded by competing pharmaceutical companies in the Netherlands, Germany, and Switzerland, raced to isolate it.[143][144] This hormone was first identified by Karoly Gyula David, E. Dingemanse, J. Freud and Ernst Laqueur in a May 1935 paper "On Crystalline Male Hormone from Testicles (Testosterone)."[145] They named the hormone testosterone, from the stems of testicle and sterol, and the suffix of ketone. The chemical synthesis of testosterone was achieved in August that year, when Butenandt and G. Hanisch published a paper describing "A Method for Preparing Testosterone from Cholesterol."[146] Only a week later, the third group, Ruzicka and A. Wettstein, announced a patent application in a paper "On the Artificial Preparation of the Testicular Hormone Testosterone (Androsten-3-one-17-ol)."[147] Ruzicka and Butenandt were offered the 1939 Nobel Prize in Chemistry for their work, but the Nazi government forced Butenandt to decline the honor, although he accepted the prize after the end of World War II.[143][144]

Clinical trials on humans, involving either oral doses of methyltestosterone or injections of testosterone propionate, began as early as 1937.[143] Testosterone propionate is mentioned in a letter to the editor of Strength and Health magazine in 1938; this is the earliest known reference to an anabolic steroid in a U.S. weightlifting or bodybuilding magazine.[143] There are often reported rumors that German soldiers were administered anabolic steroids during the Second World War, the aim being to increase their aggression and stamina, but these are, as yet, unproven.[58]:6 Adolf Hitler himself, according to his physician, was injected with testosterone derivatives to treat various ailments.[148] AAS were used in experiments conducted by the Nazis on concentration camp inmates,[148] and later by the allies attempting to treat the malnourished victims that survived Nazi camps.[58]:6 President John F. Kennedy was administered steroids both before and during his presidency.[149]

Development of synthetic AAS

The development of muscle-building properties of testosterone was pursued in the 1940s, in the Soviet Union and in Eastern Bloc countries such as East Germany, where steroid programs were used to enhance the performance of Olympic and other amateur weight lifters. In response to the success of Russian weightlifters, the U.S. Olympic Team physician John Ziegler worked with synthetic chemists to develop an anabolic steroid with reduced androgenic effects.[150] Ziegler's work resulted in the production of methandrostenolone, which Ciba Pharmaceuticals marketed as Dianabol. The new steroid was approved for use in the U.S. by the Food and Drug Administration (FDA) in 1958. It was most commonly administered to burn victims and the elderly. The drug's off-label users were mostly bodybuilders and weight lifters. Although Ziegler prescribed only small doses to athletes, he soon discovered that those having abused Dianabol suffered from enlarged prostates and atrophied testes.[151] AAS were placed on the list of banned substances of the IOC in 1976, and a decade later the committee introduced 'out-of-competition' doping tests because many athletes used AAS in their training period rather than during competition.[6]

Three major ideas governed modifications of testosterone into a multitude of AAS: Alkylation at 17-alpha position with methyl or ethyl group created orally active compounds because it slows the degradation of the drug by the liver; esterification of testosterone and nortestosterone at the 17-beta position allows the substance to be administered parenterally and increases the duration of effectiveness because agents soluble in oily liquids may be present in the body for several months; and alterations of the ring structure were applied for both oral and parenteral agents to seeking to obtain different anabolic to androgenic effect ratios.[6]

Society and culture

Legal status

Various compounds with anabolic and androgenic effects, their relation with anabolic steroids

The legal status of anabolic steroids varies from country to country: some have stricter controls on their use or prescription than others though in many countries they are not illegal. In the U.S., anabolic steroids are currently listed as Schedule III controlled substances under the Controlled Substances Act, which makes simple possession of such substances without a prescription a federal crime punishable by up to one year in prison for the first offense. Unlawful distribution or possession with intent to distribute anabolic steroids as a first offense is punished by up to ten years in prison.[152] In Canada, anabolic steroids and their derivatives are part of the Controlled drugs and substances act and are Schedule IV substances, meaning that it is illegal to obtain or sell them without a prescription; however, possession is not punishable, a consequence reserved for schedule I, II, or III substances. Those guilty of buying or selling anabolic steroids in Canada can be imprisoned for up to 18 months.[153] Import and export also carry similar penalties. In Canada, researchers have concluded that steroid use among student athletes is extremely widespread. A study conducted in 1993 by the Canadian Centre for Drug-Free Sport found that nearly 83,000 Canadians between the ages of 11 and 18 use steroids.[154] Anabolic steroids are also illegal without prescription in Australia,[155] Argentina, Brazil and Portugal,[156] and are listed as Class C Controlled Drugs in the United Kingdom. Anabolic steroids are readily available without a prescription in some countries such as Mexico and Thailand.

United States

Steroid pills intercepted by the US Drug Enforcement Administration during the Operation Raw Deal bust in September 2007.

The history of the U.S. legislation on anabolic steroids goes back to the late 1980s, when the U.S. Congress considered placing anabolic steroids under the Controlled Substances Act following the controversy over Ben Johnson's victory at the 1988 Summer Olympics in Seoul. Anabolic steroids were added to Schedule III of the Controlled Substances Act in the Anabolic Steroids Control Act of 1990.[157]

The same act also introduced more stringent controls with higher criminal penalties for offenses involving the illegal distribution of anabolic steroids and human growth hormone. By the early 1990s, after anabolic steroids were scheduled in the U.S., several pharmaceutical companies stopped manufacturing or marketing the products in the U.S., including Ciba, Searle, Syntex, and others. In the Controlled Substances Act, anabolic steroids are defined to be any drug or hormonal substance chemically and pharmacologically related to testosterone (other than estrogens, progestins, and corticosteroids) that promote muscle growth. The act was amended by the Anabolic Steroid Control Act of 2004, which added prohormones to the list of controlled substances, with effect from January 20, 2005.[158]

United Kingdom

In the United Kingdom, anabolic steroids are classified as class C drugs for their illegal abuse potential, which puts them in the same class as benzodiazepines. Anabolic steroids are in Schedule 4, which is divided in 2 parts; Part 1 contains most of the benzodiazepines and Part 2 contains the anabolic and androgenic steroids.

Part 1 drugs are subject to full import and export controls with possession being an offence without an appropriate prescription. There is no restriction on the possession when it is part of a medicinal product. Part 2 drugs require a Home Office licence for importation and export unless the substance is in the form of a medicinal product and is for self-administration by a person.[159]

Status in sports

Legal status of anabolic steroids and other compounds with anabolic effects in Western countries

Anabolic steroids are banned by all major sports bodies including Association of Tennis Professionals, Major League Baseball, Fédération Internationale de Football Association[160] the Olympics,[161] the National Basketball Association,[162] the National Hockey League,[163] World Wrestling Entertainment and the National Football League.[164] The World Anti-Doping Agency (WADA) maintains the list of performance-enhancing substances used by many major sports bodies and includes all anabolic agents, which includes all anabolic steroids and precursors as well as all hormones and related substances.[165][166] Spain has passed an anti-doping law creating a national anti-doping agency.[167] Italy passed a law in 2000 where penalties range up to three years in prison if an athlete has tested positive for banned substances.[168] In 2006, Russian President Vladimir Putin signed into law ratification of the International Convention Against Doping in Sport which would encourage cooperation with WADA. Many other countries have similar legislation prohibiting anabolic steroids in sports including Denmark,[169] France,[170] the Netherlands[171] and Sweden.[172]

Usage

Law enforcement

United States federal law enforcement officials have expressed concern about AAS use by police officers. "It's a big problem, and from the number of cases, it's something we shouldn't ignore. It's not that we set out to target cops, but when we're in the middle of an active investigation into steroids, there have been quite a few cases that have led back to police officers," says Lawrence Payne, a spokesman for the United States Drug Enforcement Administration.[173] The FBI Law Enforcement Bulletin stated that “Anabolic steroid abuse by police officers is a serious problem that merits greater awareness by departments across the country".[174] It is also believed that police officers across the United Kingdom "are using criminals to buy steroids" which he claims to be a top risk factor for police corruption.[175]

Professional wrestling

Following the murder-suicide of Chris Benoit in 2007, the Oversight and Government Reform Committee investigated steroid usage in the wrestling industry.[176] The Committee investigated WWE and Total Nonstop Action Wrestling (TNA), asking for documentation of their companies' drug policies. WWE CEO and Chairman, Linda and Vince McMahon respectively, both testified. The documents stated that 75 wrestlers—roughly 40 percent—had tested positive for drug use since 2006, most commonly for steroids.[177][178]

Economics

Several large buckets containing tens of thousands of anabolic steroid vials confiscated by the DEA during Operation Raw Deal in 2007.

Anabolic steroids are frequently produced in pharmaceutical laboratories, but, in nations where stricter laws are present, they are also produced in small home-made underground laboratories, usually from raw substances imported from abroad.[179] In these countries, the majority of steroids are obtained illegally through black market trade.[180][181] These steroids are usually manufactured in other countries, and therefore must be smuggled across international borders. As with most significant smuggling operations, organized crime is involved.[182]

In the late 2000s, the worldwide trade in illicit AAS increased significantly, and authorities announced record captures on three continents. In 2006, Finnish authorities announced a record seizure of 11.8 million AAS tablets. A year later, the DEA seized 11.4 million units of AAS in the largest U.S seizure ever. In the first three months of 2008, Australian customs reported a record 300 seizures of AAS shipments.[183]

In the U.S., Canada, and Europe, illegal steroids are sometimes purchased just as any other illegal drug, through dealers who are able to obtain the drugs from a number of sources. Illegal anabolic steroids are sometimes sold at gyms and competitions, and through the mail, but may also be obtained through pharmacists, veterinarians, and physicians.[184] In addition, a significant number of counterfeit products are sold as anabolic steroids, in particular via mail order from websites posing as overseas pharmacies. In the U.S., black-market importation continues from Mexico, Thailand, and other countries where steroids are more easily available, as they are legal.[185]

See also

References

  1. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 Kicman, A T (2008). "Pharmacology of anabolic steroids". British Journal of Pharmacology. 154 (3): 502–521. doi:10.1038/bjp.2008.165. ISSN 0007-1188.
  2. Powers M (2011). Houglum J, Harrelson GL, eds. Performance-Enhancing Drugs. Principles of Pharmacology for Athletic Trainers (2nd ed.). SLACK Incorporated. p. 345. ISBN 978-1-55642-901-9.
  3. 1 2 B arrett-Connor EL (1995). "Testosterone and risk factors for cardiovascular disease in men". Diabete Metab. 21 (3): 156–61. PMID 7556805.
  4. 1 2 Yamamoto Y, Moore R, Hess HA, Guo GL, Gonzalez FJ, Korach KS, Maronpot RR, Negishi M (2006). "Estrogen receptor alpha mediates 17alpha-ethynylestradiol causing hepatotoxicity". J Biol Chem. 281 (24): 16625–31. doi:10.1074/jbc.M602723200. PMID 16606610.
  5. 1 2 De Piccoli B, Giada F, Benettin A, Sartori F, Piccolo E (1991). "Anabolic steroid use in body builders: an echocardiographic study of left ventricle morphology and function". Int J Sports Med. 12 (4): 408–12. doi:10.1055/s-2007-1024703. PMID 1917226.
  6. 1 2 3 4 5 6 7 8 9 10 11 Hartgens F, Kuipers H (2004). "Effects of androgenic-anabolic steroids in athletes". Sports Med. 34 (8): 513–54. doi:10.2165/00007256-200434080-00003. PMID 15248788.
  7. 1 2 Kicman AT, Gower DB (July 2003). "Anabolic steroids in sport: biochemical, clinical and analytical perspectives". Ann. Clin. Biochem. 40 (Pt 4): 321–56. doi:10.1258/000456303766476977. PMID 12880534.
  8. 1 2 3 Becker KL (2001). Principles and Practice of Endocrinology and Metabolism. Lippincott Williams & Wilkins. pp. 1185–1186. ISBN 978-0-7817-1750-2.
  9. Basaria S, Wahlstrom JT, Dobs AS (November 2001). "Clinical review 138: Anabolic-androgenic steroid therapy in the treatment of chronic diseases". J. Clin. Endocrinol. Metab. 86 (11): 5108–17. doi:10.1210/jcem.86.11.7983. PMID 11701661.
  10. Ranke MB, Bierich JR (1986). "Treatment of growth hormone deficiency". Clinics in endocrinology and metabolism. 15 (3): 495–510. doi:10.1016/S0300-595X(86)80008-1. PMID 2429792.
  11. Grunfeld C, Kotler DP, Dobs A, Glesby M, Bhasin S (2006). "Oxandrolone in the treatment of HIV-associated weight loss in men: a randomized, double-blind, placebo-controlled study". J. Acquir. Immune Defic. Syndr. 41 (3): 304–14. doi:10.1097/01.qai.0000197546.56131.40. PMID 16540931.
  12. Berger JR, Pall L, Hall CD, Simpson DM, Berry PS, Dudley R (1996). "Oxandrolone in AIDS-wasting myopathy". AIDS. 10 (14): 1657–62. doi:10.1097/00002030-199612000-00010. PMID 8970686.
  13. Arslanian S, Suprasongsin C (1997). "Testosterone treatment in adolescents with delayed puberty: changes in body composition, protein, fat, and glucose metabolism". J. Clin. Endocrinol. Metab. 82 (10): 3213–20. doi:10.1210/jc.82.10.3213. PMID 9329341.
  14. 1 2 Matsumoto AM (1990). "Effects of chronic testosterone administration in normal men: safety and efficacy of high dosage testosterone and parallel dose-dependent suppression of luteinizing hormone, follicle-stimulating hormone, and sperm production". J. Clin. Endocrinol. Metab. 70 (1): 282–7. doi:10.1210/jcem-70-1-282. PMID 2104626.
  15. Aribarg A, Sukcharoen N, Chanprasit Y, Ngeamvijawat J, Kriangsinyos R (1996). "Suppression of spermatogenesis by testosterone enanthate in Thai men". Journal of the Medical Association of Thailand = Chotmaihet thangphaet. 79 (10): 624–9. PMID 8996996.
  16. Kenny AM, Prestwood KM, Gruman CA, Marcello KM, Raisz LG (2001). "Effects of transdermal testosterone on bone and muscle in older men with low bioavailable testosterone levels". J. Gerontol. A Biol. Sci. Med. Sci. 56 (5): M266–72. doi:10.1093/gerona/56.5.M266. PMID 11320105.
  17. Baum NH, Crespi CA (2007). "Testosterone replacement in elderly men". Geriatrics. 62 (9): 14–8. PMID 17824721.
  18. Francis RM (2001). "Androgen replacement in aging men". Calcif. Tissue Int. 69 (4): 235–8. doi:10.1007/s00223-001-1051-9. PMID 11730258.
  19. Nair KS, Rizza RA, O'Brien P, Dhatariya K, Short KR, Nehra A, Vittone JL, Klee GG, Basu A, Basu R, Cobelli C, Toffolo G, Dalla Man C, Tindall DJ, Melton LJ, Smith GE, Khosla S, Jensen MD (October 2006). "DHEA in elderly women and DHEA or testosterone in elderly men". N. Engl. J. Med. 355 (16): 1647–59. doi:10.1056/NEJMoa054629. PMID 17050889.
  20. Shah K, Montoya C, Persons RK (April 2007). "Clinical inquiries. Do testosterone injections increase libido for elderly hypogonadal patients?". J Fam Pract. 56 (4): 301–3. PMID 17403329.
  21. Yassin AA, Saad F (March 2007). "Improvement of sexual function in men with late-onset hypogonadism treated with testosterone only". J Sex Med. 4 (2): 497–501. doi:10.1111/j.1743-6109.2007.00442.x. PMID 17367445.
  22. Arver S, Dobs AS, Meikle AW, Caramelli KE, Rajaram L, Sanders SW, Mazer NA (December 1997). "Long-term efficacy and safety of a permeation-enhanced testosterone transdermal system in hypogonadal men". Clin. Endocrinol. (Oxf). 47 (6): 727–37. doi:10.1046/j.1365-2265.1997.3071113.x. PMID 9497881.
  23. Nieschlag E, Büchter D, Von Eckardstein S, Abshagen K, Simoni M, Behre HM (December 1999). "Repeated intramuscular injections of testosterone undecanoate for substitution therapy in hypogonadal men". Clin. Endocrinol. (Oxf). 51 (6): 757–63. doi:10.1046/j.1365-2265.1999.00881.x. PMID 10619981.
  24. Moore E, Wisniewski A, Dobs A (August 2003). "Endocrine treatment of transsexual people: a review of treatment regimens, outcomes, and adverse effects". J. Clin. Endocrinol. Metab. 88 (8): 3467–73. doi:10.1210/jc.2002-021967. PMID 12915619.
  25. Ask a Gender Therapist: Can I Transition if I’m Non-Binary or Genderfluid? – Dara Hoffman-Fox. May 7, 2014. Retrieved November 17, 2014.
  26. Genderqueer, Pansexual, LGBTQ: Will Gender Exist 100 Years From Now? - Rebooted. October 24, 2013. Retrieved November 17, 2014.
  27. Young people exploring nonbinary gender roles – SF Gate. February 12, 2014. Retrieved November 17, 2014.
  28. Ask Matt: My Genderqueer Child Wants to Use Testosterone – Tranifesto. January 9, 2014. Retrieved November 17, 2014.
  29. Warne GL, Grover S, Zajac JD (2005). "Hormonal therapies for individuals with intersex conditions: protocol for use". Treatments in Endocrinology. 4 (1): 19–29. doi:10.2165/00024677-200504010-00003. PMID 15649098.
  30. What is Intersex – An Intersex FAQ by Inter/Act – Inter/Act Youth. September 12, 2014. Retrieved December 5, 2014.
  31. Gething AD, Grace FM, Davies B, Baker JS (Oct–Dec 2011). "Effects of Long-Term Anabolic Androgenic Steroid Administration on Respiratory Function". Research in Sports Medicine. 19 (4): 231–244. doi:10.1080/15438627.2011.608034 (inactive 2015-02-02). PMID 21988266.
  32. "Most steroid users are not athletes: study". Reuters. 2007-11-21. Retrieved 2014-01-03.
  33. Sjöqvist F, Garle M, Rane A (May 2008). "Use of doping agents, in particular anabolic steroids, in sports and society". Lancet. 371 (9627): 1872–82. doi:10.1016/S0140-6736(08)60801-6. PMID 18514731.
  34. Yesalis CE, Kennedy NJ, Kopstein AN, Bahrke MS (1993). "Anabolic-androgenic steroid use in the United States". JAMA. 270 (10): 1217–21. doi:10.1001/jama.270.10.1217. PMID 8355384.
  35. McCabe SE, Brower KJ, West BT, Nelson TF, Wechsler H (2007). "Trends in non-medical use of anabolic steroids by U.S. college students: Results from four national surveys". Drug and Alcohol Dependence. 90 (2–3): 243–51. doi:10.1016/j.drugalcdep.2007.04.004. PMC 2383927Freely accessible. PMID 17512138.
  36. Parkinson AB, Evans NA (April 2006). "Anabolic androgenic steroids: a survey of 500 users". Med Sci Sports Exerc. 38 (4): 644–51. doi:10.1249/01.mss.0000210194.56834.5d. PMID 16679978.
  37. 1 2 3 4 5 Cohen J, Collins R, Darkes J, Gwartney D (2007). "A league of their own: demographics, motivations and patterns of use of 1,955 male adult non-medical anabolic steroid users in the United States". J Int Soc Sports Nutr. 4: 12. doi:10.1186/1550-2783-4-12. PMC 2131752Freely accessible. PMID 17931410.
  38. Copeland J, Peters R, Dillon P (March 1998). "A study of 100 anabolic-androgenic steroid users". Med. J. Aust. 168 (6): 311–2. PMID 9549549.
  39. Eastley, Tony (January 18, 2006). "Steroid study debunks user stereotypes". ABC. Retrieved 2014-01-03.
  40. Pope HG, Kanayama G, Ionescu-Pioggia M, Hudson JI (September 2004). "Anabolic steroid users' attitudes towards physicians". Addiction. 99 (9): 1189–94. doi:10.1111/j.1360-0443.2004.00781.x. PMID 15317640.
  41. Kanayama G, Barry S, Hudson JI, Pope HG (April 2006). "Body image and attitudes toward male roles in anabolic-androgenic steroid users". Am J Psychiatry. 163 (4): 697–703. doi:10.1176/appi.ajp.163.4.697 (inactive 2015-04-14). PMID 16585446.
  42. Grogan S, Shepherd S, Evans R, Wright S, Hunter G (Nov 2006). "Experiences of Anabolic Steroid Use". Journal of Health Psychology. 11 (6): 845–856. doi:10.1177/1359105306069080. PMID 17035257.
  43. Hickson RC, Czerwinski SM, Falduto MT, Young AP (1990). "Glucocorticoid antagonism by exercise and androgenic-anabolic steroids". Medicine and science in sports and exercise. 22 (3): 331–40. doi:10.1249/00005768-199006000-00010. PMID 2199753.
  44. 1 2 3 Chrousos, George P. (2012). "The Gonadal Hormones & Inhibitors". In Katzung, Bertram G. Basic & Clinical Pharmacology. New York London: McGraw-Hill Medical McGraw-Hill distributor. ISBN 0071764011.
  45. Mutzebaugh C (1998). "Does the choice of alpha-AAS really make a difference?". HIV Hotline. 8 (5–6): 10–1. PMID 11366379.
  46. Nutt, D; King, LA; Saulsbury, W; Blakemore, C (24 March 2007). "Development of a rational scale to assess the harm of drugs of potential misuse.". Lancet (London, England). 369 (9566): 1047–53. doi:10.1016/S0140-6736(07)60464-4. PMID 17382831.
  47. Trenton AJ, Currier GW (2005). "Behavioural manifestations of anabolic steroid use". CNS Drugs. 19 (7): 571–95. doi:10.2165/00023210-200519070-00002. PMID 15984895.
  48. 1 2 Kanayama G, Hudson JI, Pope HG (November 2008). "Long-Term Psychiatric and Medical Consequences of Anabolic-Androgenic Steroid Abuse: A Looming Public Health Concern?". Drug Alcohol Depend. 98 (1–2): 1–12. doi:10.1016/j.drugalcdep.2008.05.004. PMC 2646607Freely accessible. PMID 18599224.
  49. Brower KJ (October 2002). "Anabolic steroid abuse and dependence". Curr Psychiatry Rep. 4 (5): 377–87. doi:10.1007/s11920-002-0086-6. PMID 12230967.
  50. Fudala PJ, Weinrieb RM, Calarco JS, Kampman KM, Boardman C (2003). "An evaluation of anabolic-androgenic steroid abusers over a period of 1 year: seven case studies". Annals of Clinical Psychiatry. 15 (2): 121–30. doi:10.3109/10401230309085677. PMID 12938869.
  51. Pagonis TA, Angelopoulos NV, Koukoulis GN, Hadjichristodoulou CS (2006). "Psychiatric side effects induced by supraphysiological doses of combinations of anabolic steroids correlate to the severity of abuse". Eur. Psychiatry. 21 (8): 551–62. doi:10.1016/j.eurpsy.2005.09.001. PMID 16356691.
  52. 1 2 Rashid H, Ormerod S, Day E (2007). "Anabolic androgenic steroids: What the psychiatrist needs to know". Advances in Psychiatric Treatment. 13 (3): 203–211. doi:10.1192/apt.bp.105.000935.
  53. Cooper CJ, Noakes TD, Dunne T, Lambert MI, Rochford K (September 1996). "A high prevalence of abnormal personality traits in chronic users of anabolic-androgenic steroids". Br J Sports Med. 30 (3): 246–50. doi:10.1136/bjsm.30.3.246. PMC 1332342Freely accessible. PMID 8889121.
  54. "Dr. Ritchi Morris". Vitalquests.org. Retrieved 2013-12-01.
  55. Kanayama G, Brower KJ, Wood RI, Hudson JI, Pope HG (December 2009). "Anabolic-androgenic steroid dependence: an emerging disorder". Addiction. 104 (12): 1966–78. doi:10.1111/j.1360-0443.2009.02734.x. PMC 2780436Freely accessible. PMID 19922565.
  56. Eisenberg ER, Galloway GP. "Anabolic androgenic steroids". In Lowinson JH, Ruiz P, Millman RB. Substance Abuse: A Comprehensive Textbook. Lippincott Williams & Wilkins. ASIN B0049VACMW.
  57. Lindström M, Nilsson AL, Katzman PL, Janzon L, Dymling JF (1990). "Use of anabolic-androgenic steroids among body builders—frequency and attitudes". J. Intern. Med. 227 (6): 407–11. doi:10.1111/j.1365-2796.1990.tb00179.x. PMID 2351927.
  58. 1 2 3 Lenahan P (2003). Anabolic Steroids: And Other Performance-enhancing Drugs. London: Taylor & Francis. ISBN 0-415-28030-3.
  59. Thiblin I, Petersson A (February 2005). "Pharmacoepidemiology of anabolic androgenic steroids: a review". Fundam Clin Pharmacol. 19 (1): 27–44. doi:10.1111/j.1472-8206.2004.00298.x. PMID 15660958.
  60. Beaver KM, Vaughn MG, Delisi M, Wright JP (December 2008). "Anabolic-Androgenic Steroid Use and Involvement in Violent Behavior in a Nationally Representative Sample of Young Adult Males in the United States". Am J Public Health. 98 (12): 2185–7. doi:10.2105/AJPH.2008.137018. PMC 2636528Freely accessible. PMID 18923108.
  61. Bahrke MS, Yesalis CE, Wright JE (1996). "Psychological and behavioural effects of endogenous testosterone and anabolic-androgenic steroids. An update". Sports medicine (Auckland, N.Z.). 22 (6): 367–90. doi:10.2165/00007256-199622060-00005. PMID 8969015.
  62. 1 2 3 Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J, Bunnell TJ, Tricker R, Shirazi A, Casaburi R (July 1996). "The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men". N. Engl. J. Med. 335 (1): 1–7. doi:10.1056/NEJM199607043350101. PMID 8637535.
  63. Tricker R, Casaburi R, Storer TW, Clevenger B, Berman N, Shirazi A, Bhasin S (October 1996). "The effects of supraphysiological doses of testosterone on angry behavior in healthy eugonadal men—a clinical research center study". J. Clin. Endocrinol. Metab. 81 (10): 3754–8. doi:10.1210/jcem.81.10.8855834. PMID 8855834.
  64. Pope HG, Kouri EM, Hudson JI (February 2000). "Effects of supraphysiologic doses of testosterone on mood and aggression in normal men: a randomized controlled trial". Arch. Gen. Psychiatry. 57 (2): 133–40; discussion 155–6. doi:10.1001/archpsyc.57.2.133. PMID 10665615.
  65. Pagonis TA, Angelopoulos NV, Koukoulis GN, Hadjichristodoulou CS, Toli PN (2006). "Psychiatric and hostility factors related to use of anabolic steroids in monozygotic twins". Eur. Psychiatry. 21 (8): 563–9. doi:10.1016/j.eurpsy.2005.11.002. PMID 16529916.
  66. Perry PJ, Kutscher EC, Lund BC, Yates WR, Holman TL, Demers L (May 2003). "Measures of aggression and mood changes in male weightlifters with and without androgenic anabolic steroid use". J. Forensic Sci. 48 (3): 646–51. PMID 12762541.
  67. "Teens & Steroids: A Dangerous Mix". CBS Broadcasting Inc. 2004-06-03. Archived from the original on 10 July 2007. Retrieved 2007-06-27.
  68. Uzych L (February 1992). "Anabolic-androgenic steroids and psychiatric-related effects: a review". Can J Psychiatry. 37 (1): 23–8. PMID 1551042.
  69. "Anabolic Steroids and Suicide – A Brief Review of the Evidence". Thinksteroids.com. 2005-07-12. Retrieved 2013-12-01.
  70. Grace F, Sculthorpe N, Baker J, Davies B (2003). "Blood pressure and rate pressure product response in males using high-dose anabolic-androgenic steroids (AAS)". J Sci Med Sport. 6 (3): 307–12. doi:10.1016/S1440-2440(03)80024-5. PMID 14609147.
  71. "DailyMed: About DailyMed". Dailymed.nlm.nih.gov. Retrieved 2008-11-03.
  72. Bagatell CJ, Knopp RH, Vale WW, Rivier JE, Bremner WJ (1992). "Physiologic testosterone levels in normal men suppress high-density lipoprotein cholesterol levels". Annals of Internal Medicine. 116 (12 Pt 1): 967–73. doi:10.7326/0003-4819-116-12-967. PMID 1586105.
  73. Mewis C, Spyridopoulos I, Kühlkamp V, Seipel L (1996). "Manifestation of severe coronary heart disease after anabolic drug abuse". Clinical Cardiology. 19 (2): 153–5. doi:10.1002/clc.4960190216. PMID 8821428.
  74. Melnik B, Jansen T, Grabbe S (2007). "Abuse of anabolic-androgenic steroids and bodybuilding acne: an underestimated health problem". Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG. 5 (2): 110–7. doi:10.1111/j.1610-0387.2007.06176.x. PMID 17274777.
  75. Vierhapper H, Maier H, Nowotny P, Waldhäusl W (July 2003). "Production rates of testosterone and of dihydrotestosterone in female pattern hair loss". Metab. Clin. Exp. 52 (7): 927–9. doi:10.1016/S0026-0495(03)00060-X. PMID 12870172.
  76. Irving LM, Wall M, Neumark-Sztainer D, Story M (2002). "Steroid use among adolescents: findings from Project EAT". The Journal of Adolescent Health. 30 (4): 243–52. doi:10.1016/S1054-139X(01)00414-1. PMID 11927236.
  77. "Known and Probable Human Carcinogens". American Cancer Society. 2011-06-29.
  78. Sullivan ML, Martinez CM, Gallagher EJ (1999). "Atrial fibrillation and anabolic steroids". The Journal of emergency medicine. 17 (5): 851–7. doi:10.1016/S0736-4679(99)00095-5. PMID 10499702.
  79. Dickerman RD, Schaller F, McConathy WJ (1998). "Left ventricular wall thickening does occur in elite power athletes with or without anabolic steroid Use". Cardiology. 90 (2): 145–8. doi:10.1159/000006834. PMID 9778553.
  80. George KP, Wolfe LA, Burggraf GW (1991). "The 'athletic heart syndrome'. A critical review". Sports medicine (Auckland, N.Z.). 11 (5): 300–30. doi:10.2165/00007256-199111050-00003. PMID 1829849.
  81. Dickerman RD, Schaller F, Zachariah NY, McConathy WJ (1997). "Left ventricular size and function in elite bodybuilders using anabolic steroids". Clin J Sport Med. 7 (2): 90–3. doi:10.1097/00042752-199704000-00003. PMID 9113423.
  82. Salke RC, Rowland TW, Burke EJ (1985). "Left ventricular size and function in body builders using anabolic steroids". Medicine and science in sports and exercise. 17 (6): 701–4. doi:10.1249/00005768-198512000-00014. PMID 4079743.
  83. Tokar, Steve (February 2006). "Liver Damage And Increased Heart Attack Risk Caused By Anabolic Steroid Use". University of California – San Francisco. Retrieved 2007-04-24.
  84. Wit JM, Oostdijk W (2015). "Novel approaches to short stature therapy". Best Practice & Research. Clinical Endocrinology & Metabolism. 29 (3): 353–66. doi:10.1016/j.beem.2015.01.003. PMID 26051296.
  85. Marcus R, Korenman SG (1976). "Estrogens and the human male". Annu Rev Med. 27: 357–70. doi:10.1146/annurev.me.27.020176.002041. PMID 779604.
  86. Hoffman JR, Ratamess NA (June 1, 2006). "Medical Issues Associated with Anabolic Steroid Use: Are they Exaggerated?" (PDF). Journal of Sports Science and Medicine. Archived (PDF) from the original on 20 June 2007. Retrieved 2007-05-08.
  87. Meriggiola MC, Costantino A, Bremner WJ, Morselli-Labate AM (2002). "Higher testosterone dose impairs sperm suppression induced by a combined androgen-progestin regimen". J. Androl. 23 (5): 684–90. PMID 12185103.
  88. Alén M, Reinilä M, Vihko R (1985). "Response of serum hormones to androgen administration in power athletes". Medicine and science in sports and exercise. 17 (3): 354–9. doi:10.1249/00005768-198506000-00009. PMID 2991700.
  89. Franke, Werner W., and Brigitte Berendonk. "Hormonal doping and androgenization of athletes: a secret program of the German Democratic Republic government." Clinical chemistry 43.7 (1997): 1262-1279.
  90. Manikkam M, Crespi EJ, Doop DD, Herkimer C, Lee JS, Yu S, Brown MB, Foster DL, Padmanabhan V (February 2004). "Fetal programming: prenatal testosterone excess leads to fetal growth retardation and postnatal catch-up growth in sheep". Endocrinology. 145 (2): 790–8. doi:10.1210/en.2003-0478. PMID 14576190.
  91. Herlitz LC, Markowitz GS, Farris AB, Schwimmer JA, Stokes MB, Kunis C, Colvin RB, D'Agati VD (October 29, 2009). Development of FSGS Following Anabolic Steroid Use in Bodybuilders (PDF). 42nd Annual Meeting and Scientific Exposition of the American Society of Nephrology. "Bodybuilding With Steroids Damages Kidneys". Lay summary ScienceDaily (October 30, 2009).
  92. Pereira de Jésus-Tran K, Côté PL, Cantin L, Blanchet J, Labrie F, Breton R (2006). "Comparison of crystal structures of human androgen receptor ligand-binding domain complexed with various agonists reveals molecular determinants responsible for binding affinity". Protein Sci. 15 (5): 987–99. doi:10.1110/ps.051905906. PMC 2242507Freely accessible. PMID 16641486.
  93. Lavery DN, McEwan IJ (2005). "Structure and function of steroid receptor AF1 transactivation domains: induction of active conformations". Biochem. J. 391 (Pt 3): 449–64. doi:10.1042/BJ20050872. PMC 1276946Freely accessible. PMID 16238547.
  94. Cheskis BJ (2004). "Regulation of cell signalling cascades by steroid hormones". J. Cell. Biochem. 93 (1): 20–7. doi:10.1002/jcb.20180. PMID 15352158.
  95. 1 2 Roselli CE (1998). "The effect of anabolic-androgenic steroids on aromatase activity and androgen receptor binding in the rat preoptic area". Brain Res. 792 (2): 271–6. doi:10.1016/S0006-8993(98)00148-6. PMID 9593936.
  96. Brodsky IG, Balagopal P, Nair KS (1996). "Effects of testosterone replacement on muscle mass and muscle protein synthesis in hypogonadal men—a clinical research center study". J. Clin. Endocrinol. Metab. 81 (10): 3469–75. doi:10.1210/jc.81.10.3469. PMID 8855787.
  97. Hickson RC, Czerwinski SM, Falduto MT, Young AP (1990). "Glucocorticoid antagonism by exercise and androgenic-anabolic steroids". Med Sci Sports Exerc. 22 (3): 331–40. doi:10.1249/00005768-199006000-00010. PMID 2199753.
  98. Singh R, Artaza JN, Taylor WE, Gonzalez-Cadavid NF, Bhasin S (2003). "Androgens stimulate myogenic differentiation and inhibit adipogenesis in C3H 10T1/2 pluripotent cells through an androgen receptor-mediated pathway". Endocrinology. 144 (11): 5081–8. doi:10.1210/en.2003-0741. PMID 12960001.
  99. Schroeder ET, Vallejo AF, Zheng L, Stewart Y, Flores C, Nakao S, Martinez C, Sattler FR (2005). "Six-week improvements in muscle mass and strength during androgen therapy in older men". J Gerontol a Biol Sci Med Sci. 60 (12): 1586–92. doi:10.1093/gerona/60.12.1586. PMID 16424293.
  100. Grunfeld C, Kotler DP, Dobs A, Glesby M, Bhasin S (2006). "Oxandrolone in the treatment of HIV-associated weight loss in men: a randomized, double-blind, placebo-controlled study". J Acquir Immune Defic Syndr. 41 (3): 304–14. doi:10.1097/01.qai.0000197546.56131.40. PMID 16540931.
  101. Giorgi A, Weatherby RP, Murphy PW (1999). "Muscular strength, body composition and health responses to the use of testosterone enanthate: a double blind study". Journal of science and medicine in sport / Sports Medicine Australia. 2 (4): 341–55. doi:10.1016/S1440-2440(99)80007-3. PMID 10710012.
  102. 1 2 Kuhn CM (2002). "Anabolic steroids". Recent Prog. Horm. Res. 57: 411–34. doi:10.1210/rp.57.1.411. PMID 12017555.
  103. "How Anabolic Steroids Alter Both Men And Women". Retrieved 2 January 2014.
  104. Hershberger LG, Shipley EG, Meyer RK (1953). "Myotrophic activity of 19-nortestosterone and other steroids determined by modified levator ani muscle method". Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y.). 83 (1): 175–80. doi:10.3181/00379727-83-20301. PMID 13064212.
  105. Hervey GR, Hutchinson I, Knibbs AV, Burkinshaw L, Jones PR, Norgan NG, Levell MJ (October 1976). ""Anabolic" effects of methandienone in men undergoing athletic training". Lancet. 2 (7988): 699–702. doi:10.1016/S0140-6736(76)90001-5. PMID 61389.
  106. Hervey GR, Knibbs AV, Burkinshaw L, Morgan DB, Jones PR, Chettle DR, Vartsky D (April 1981). "Effects of methandienone on the performance and body composition of men undergoing athletic training". Clin. Sci. 60 (4): 457–61. PMID 7018798.
  107. Bhasin S, Woodhouse L, Casaburi R, Singh AB, Bhasin D, Berman N, Chen X, Yarasheski KE, Magliano L, Dzekov C, Dzekov J, Bross R, Phillips J, Sinha-Hikim I, Shen R, Storer TW (December 2001). "Testosterone dose-response relationships in healthy young men". Am. J. Physiol. Endocrinol. Metab. 281 (6): E1172–81. PMID 11701431.
  108. 1 2 Imperato-McGinley J, Peterson RE, Gautier T, Sturla E (May 1979). "Androgens and the evolution of male-gender identity among male pseudohermaphrodites with 5alpha-reductase deficiency". The New England Journal of Medicine. 300 (22): 1233–7. doi:10.1056/NEJM197905313002201. PMID 431680.
  109. 1 2 3 Marks LS (2004). "5α-reductase: history and clinical importance". Rev Urol. 6 Suppl 9: S11–21. PMC 1472916Freely accessible. PMID 16985920.
  110. Sloane E (2002). Biology of Women. Cengage Learning. pp. 160–. ISBN 0-7668-1142-5.
  111. 1 2 Hanno PM, Guzzi TJ, Malkowicz SB, J Wein A (26 January 2014). Penn Clinical Manual of Urology. Elsevier Health Sciences. pp. 782–. ISBN 978-0-323-24466-4.
  112. Jain NK, Siddiqi M, Weisburger JH (2006). Protective Effects of Tea on Human Health. CABI. pp. 95–. ISBN 978-1-84593-113-1.
  113. Harper C (1 August 2007). Intersex. Berg. pp. 123–. ISBN 978-1-84788-339-1.
  114. 1 2 Bitran D, Kellogg CK, Hilvers RJ (1993). "Treatment with an anabolic-androgenic steroid affects anxiety-related behavior and alters the sensitivity of cortical GABAA receptors in the rat". Horm Behav. 27 (4): 568–83. doi:10.1006/hbeh.1993.1041. PMID 8294123.
  115. 1 2 Masonis AE, McCarthy MP (1995). "Direct effects of the anabolic/androgenic steroids, stanozolol and 17 alpha-methyltestosterone, on benzodiazepine binding to the. gamma-aminobutyric acid(a) receptor". Neurosci. Lett. 189 (1): 35–8. doi:10.1016/0304-3940(95)11445-3. PMID 7603620.
  116. 1 2 Masonis AE, McCarthy MP (1996). "Effects of the androgenic/anabolic steroid stanozolol on GABAA receptor function: GABA-stimulated 36Cl- influx and [35S] TBPS binding". J. Pharmacol. Exp. Ther. 279 (1): 186–93. PMID 8858992.
  117. 1 2 Rivera-Arce JC, Morales-Crespo L, Vargas-Pinto N, Velázquez KT, Jorge JC (2006). "Central effects of the anabolic steroid 17alpha methyltestosterone in female anxiety". Pharmacol. Biochem. Behav. 84 (2): 275–81. doi:10.1016/j.pbb.2006.05.009. PMID 16814373.
  118. 1 2 Henderson LP (2007). "Steroid modulation of GABAA receptor-mediated transmission in the hypothalamus: effects on reproductive function". Neuropharmacology. 52 (7): 1439–53. doi:10.1016/j.neuropharm.2007.01.022. PMC 1985867Freely accessible. PMID 17433821.
  119. 1 2 Schwartzer JJ, Ricci LA, Melloni RH (2009). "Interactions between the dopaminergic and GABAergic neural systems in the lateral anterior hypothalamus of aggressive AAS-treated hamsters". Behav. Brain Res. 203 (1): 15–22. doi:10.1016/j.bbr.2009.04.007. PMID 19376158.
  120. Attardi, Barbara J.; Hild, Sheri A.; Koduri, Sailaja; Pham, Trung; Pessaint, Laurent; Engbring, Jean; Till, Bruce; Gropp, David; Semon, Anne; Reel, Jerry R. (October 2010). "The potent synthetic androgens, dimethandrolone (7α,11β-dimethyl-19-nortestosterone) and 11β-methyl-19-nortestosterone, do not require 5α-reduction to exert their maximal androgenic effects". The Journal of Steroid Biochemistry and Molecular Biology. 122 (4): 212–218. doi:10.1016/j.jsbmb.2010.06.009. PMC 2949447Freely accessible. PMID 20599615.
  121. Eric S. Orwoll; John P. Bilezikian; Dirk Vanderschueren (30 November 2009). Osteoporosis in Men: The Effects of Gender on Skeletal Health. Academic Press. pp. 296–. ISBN 978-0-08-092346-8.
  122. Howard M. Fillit; Kenneth Rockwood; Kenneth Woodhouse (10 May 2010). Brocklehurst's Textbook of Geriatric Medicine and Gerontology. Elsevier Health Sciences. pp. 166–167. ISBN 1-4377-2075-7.
  123. 1 2 3 Detlef Thieme; Peter Hemmersbach (18 December 2009). Doping in Sports. Springer Science & Business Media. pp. 470–. ISBN 978-3-540-79088-4.
  124. 1 2 Attardi, Barbara J.; Pham, Trung C.; Radler, Lisa M.; Burgenson, Janet; Hild, Sheri A.; Reel, Jerry R. (June 2008). "Dimethandrolone (7α,11β-dimethyl-19-nortestosterone) and 11β-methyl-19-nortestosterone are not converted to aromatic A-ring products in the presence of recombinant human aromatase". The Journal of Steroid Biochemistry and Molecular Biology. 110 (3–5): 214–222. doi:10.1016/j.jsbmb.2007.11.009. PMC 2575079Freely accessible. PMID 18555683.
  125. 1 2 William Llewellyn (2011). Anabolics. Molecular Nutrition Llc. pp. 533–. ISBN 978-0-9828280-1-4.
  126. Tan RS, Scally MC (2009). "Anabolic steroid-induced hypogonadism--towards a unified hypothesis of anabolic steroid action". Med. Hypotheses. 72 (6): 723–8. doi:10.1016/j.mehy.2008.12.042. PMID 19231088. Regulation of the secretion of GnRH, FSH, and LH occurs partially by the negative feedback of testosterone and estradiol at the level of the hypothalamo-pituitary. Estradiol has a much larger, inhibitory effect than testosterone, being 200-fold more effective in suppressing LH secretion [57–61].
  127. Suvisaari, Janne (2000). 7α-Methyl-19-nortestosterone (MENT) Pharmacokinetics and Antigonadotropic Effects in Men (PDF). Helsinki: University of Helsinki. p. 14. ISBN 952-91-2950-5. Androgens, estrogens and progestins exert a negative feedback effect on the secretion of GnRH and LH by their actions on the pituitary and the hypothalamus. Most of the negative feedback effect of androgens is caused by their estrogenic metabolites produced by aromatization. 5α-Reduction does not seem to be necessary for the negative feedback effect of testosterone. (Rittmaster et al, 1992; Kumar et al, 1995a; Hayes et al, 2000).
  128. 1 2 3 4 Attardi, Barbara J.; Hild, Sheri A.; Reel, Jerry R. (June 2006). "Dimethandrolone Undecanoate: A New Potent Orally Active Androgen with Progestational Activity". Endocrinology. 147 (6): 3016–3026. doi:10.1210/en.2005-1524. ISSN 0013-7227. PMID 16497801.
  129. Michelle P. Warren; Naama W. Constantini (1 May 2000). Sports Endocrinology. Springer Science & Business Media. pp. 458–. ISBN 978-1-59259-016-2.
  130. Haff , G. Gregory; Triplett , N. Travis (23 September 2015). Essentials of Strength Training and Conditioning 4th Edition. Human Kinetics. pp. 233–. ISBN 978-1-4925-0162-6.
  131. Thomas L. Lemke; David A. Williams (24 January 2012). Foye's Principles of Medicinal Chemistry. Lippincott Williams & Wilkins. pp. 1360–. ISBN 978-1-60913-345-0.
  132. 1 2 Steven B. Karch, MD; Olaf Drummer; Steven B. Karch, MD, FFFLM (26 December 2001). Karch's Pathology of Drug Abuse, Third Edition. CRC Press. pp. 489–. ISBN 978-1-4200-4211-5.
  133. 1 2 van Amsterdam J, Opperhuizen A, Hartgens F (2010). "Adverse health effects of anabolic-androgenic steroids". Regul. Toxicol. Pharmacol. 57 (1): 117–23. doi:10.1016/j.yrtph.2010.02.001. PMID 20153798.
  134. Wilson JD (1988). "Androgen abuse by athletes". Endocr. Rev. 9 (2): 181–99. doi:10.1210/edrv-9-2-181. PMID 3042375.
  135. J. Larry Jameson; Leslie J. De Groot (25 February 2015). Endocrinology: Adult and Pediatric. Elsevier Health Sciences. pp. 2391–. ISBN 978-0-323-32195-2.
  136. Eberhard Nieschlag; Hermann M. Behre; Susan Nieschlag (26 July 2012). Testosterone: Action, Deficiency, Substitution. Cambridge University Press. pp. 374–. ISBN 978-1-107-01290-5.
  137. Ross Cameron; George Feuer; Felix de la Iglesia (6 December 2012). Drug-Induced Hepatotoxicity. Springer Science & Business Media. pp. 166–. ISBN 978-3-642-61013-4.
  138. Shahidi NT (2001). "A review of the chemistry, biological action, and clinical applications of anabolic-androgenic steroids". Clin Ther. 23 (9): 1355–90. PMID 11589254.
  139. Mareck U, Geyer H, Opfermann G, Thevis M, Schänzer W (July 2008). "Factors influencing the steroid profile in doping control analysis". J Mass Spectrom. 43 (7): 877–91. doi:10.1002/jms.1457. PMID 18570179.
  140. Fragkaki AG, Angelis YS, Tsantili-Kakoulidou A, Koupparis M, Georgakopoulos C (May 2009). "Schemes of metabolic patterns of anabolic androgenic steroids for the estimation of metabolites of designer steroids in human urine". J. Steroid Biochem. Mol. Biol. 115 (1–2): 44–61. doi:10.1016/j.jsbmb.2009.02.016. PMID 19429460.
  141. Blackledge RD (August 2009). "Bad science: the instrumental data in the Floyd Landis case". Clin. Chim. Acta. 406 (1–2): 8–13. doi:10.1016/j.cca.2009.05.016. PMID 19465014.
  142. Baselt, Randall Clint (2008). Disposition of Toxic Drugs and Chemicals in Man (8th ed.). Foster City, CA: Biomedical Publications. pp. 95, 393, 403, 649, 695, 952, 962, 1078, 1156, 1170, 1442, 1501, 1581. ISBN 978-0-9626523-7-0.
  143. 1 2 3 4 5 Hoberman JM, Yesalis CE (1995). "The history of synthetic testosterone". Scientific American. 272 (2): 76–81. doi:10.1038/scientificamerican0295-76. PMID 7817189.
  144. 1 2 Freeman ER, Bloom DA, McGuire EJ (2001). "A brief history of testosterone". Journal of Urology. 165 (2): 371–373. doi:10.1097/00005392-200102000-00004. PMID 11176375.
  145. David K, Dingemanse E, Freud J, Laqueur L (1935). "Uber krystallinisches mannliches Hormon aus Hoden (Testosteron) wirksamer als aus harn oder aus Cholesterin bereitetes Androsteron". Hoppe Seylers Z Physiol Chem. 233 (5–6): 281–283. doi:10.1515/bchm2.1935.233.5-6.281.
  146. Butenandt A, Hanisch G (1935). "Über die Umwandlung des Dehydro-androsterons in Δ4-Androsten-ol-(17)-0n-(3) (Testosteron); ein Weg zur Darstellung des Testosterons aus Cholesterin (Vorläuf. Mitteil.)" [On the conversion of dehydro-Δ4-androstene androsterons in-ol (17) 0n (3) (testosterone), a way to represent the testosterone from cholesterol (Vorläuf. msgs.)]. Berichte der deutschen chemischen Gesellschaft (A and B Series) (in German). 68 (9): 1859–62. doi:10.1002/cber.19350680937.
  147. Ruzicka L, Wettstein A (1935). "Sexualhormone VII. Uber die kunstliche Herstellung des Testikelhormons. Testosteron (Androsten-3-one-17-ol.)" [Sex hormones VII About the artificial production of testosterone Testikelhormons (androstene-3-one-17-ol)]. Helvetica Chimica Acta (in German). 18: 1264–75. doi:10.1002/hlca.193501801176.
  148. 1 2 Taylor WN (January 1, 2009). Anabolic Steroids and the Athlete. McFarland & Company. p. 181. ISBN 0-7864-1128-7.
  149. Suarez R, Senior Correspondent, Kelman J, physician (2002-11-18). "President Kennedy's Health Secrets". PBS NewsHour. Public Broadcasting System.
  150. Calfee R, Fadale P (2006). "Popular ergogenic drugs and supplements in young athletes". Pediatrics. 117 (3): e577–89. doi:10.1542/peds.2005-1429. PMID 16510635.
  151. Justin Peters The Man Behind the Juice, Slate Friday, Feb. 18, 2005. Retrieved 29 April 2008
  152. "Title 21 United States Code (USC) Controlled Substances Act". US Department of Justice. Archived from the original on 1 August 2009. Retrieved 2009-09-07.
  153. Controlled Drugs and Substances Act, S.C. 1996, c. 19, s. 4(7) (Controlled Drugs and Substances Act at Department of Justice)
  154. Deacon, James (2 May 1994). "Biceps in a bottle". Maclean's: 52.
  155. "Steroids". Australian Institute of Criminology. 2006. Archived from the original on 2007-04-05. Retrieved 2007-05-06.
  156. "Library of congress search". Library of Congress. Archived from the original on May 22, 2011. Retrieved 2007-05-06.
  157. H.R. 4658
  158. "News from DEA, Congressional Testimony, 03/16/04". Archived from the original on February 6, 2007. Retrieved 2007-04-24.
  159. "Patient.info Controlled Drugs". Egton Medical Information Systems Limited. Retrieved 8 August 2013.
  160. "FIFA Anit-Doping Regulations" (PDF). FIFA. Retrieved 2013-12-01.
  161. "Olympic movement anti-doping code" (PDF). International Olympic Committee. 1999. Retrieved 2007-05-06.
  162. "The nba and nbpa anti-drug program". NBA Policy. findlaw.com. 1999. Retrieved 2007-05-06.
  163. "NHL/NHLPA performance-enhancing substances program summary". nhlpa.com. Archived from the original on 2 June 2007. Retrieved 2007-05-06.
  164. "List of Prohibited Substances" (PDF). nflpa.com. 2006. Archived from the original (PDF) on 2007-06-20. Retrieved 2007-05-06.
  165. "World anti-doping code" (PDF). WADA. 2003. Archived (PDF) from the original on 7 August 2007. Retrieved 2007-07-10.
  166. "Prohibited list of 2005" (PDF). WADA. 2005. Retrieved 2007-05-06.
  167. "Spain's senate passes anti-doping law". Herald Tribune. Associated Press. October 5, 2006. Archived from the original on October 12, 2006. Retrieved 2007-05-06.
  168. Johnson, Kevin (2006-02-20). "Italian anti-doping laws could mean 3 years in jail". USA Today. Retrieved 2007-05-06.
  169. "Act on promotion of doping-free sport" (PDF). kum.dk. 2004. Retrieved 2016-09-04.
  170. "Protection of health of athletes and the fight against doping" (PDF). WADA. 2006. Retrieved 2007-05-06.
  171. "Anti-doping legislation in the netherlands" (PDF). WADA. 2006. Retrieved 2007-05-06.
  172. "The Swedish Act prohibiting certain doping substances (1991:1969)" (PDF). WADA. 1991. Retrieved 2007-05-06.
  173. Keeping, Juliana (27 December 2010). "Steroid abuse among law enforcement a problem nationwide". The Ann Arbor News. Retrieved 1 December 2013.
  174. "Anabolic Steroid Use and Abuse by Police Officers: Policy & Prevention". The Police Chief. June 2008. Retrieved 1 December 2013.
  175. "Chief constable admits police officers across UK 'are using criminals to buy steroids and abuse their power for sexual gratification'". Daily Mail. 22 January 2013. Retrieved 1 December 2013.
  176. Lockhart B (2010-03-01). "WWE steroid investigation: A controversy McMahon 'doesn't need'". Greenwich Time. Retrieved 2010-03-01.
  177. documents Archived December 24, 2010, at the Wayback Machine.
  178. "Deposition details McMahon steroid testimony | News from southeastern Connecticut". The Day. 2007-12-13. Retrieved 2010-08-14.
  179. Assael, Shaun (2007-09-24). "'Raw Deal' busts labs across U.S., many supplied by China". ESPN The Magazine. Archived from the original on 14 October 2007. Retrieved 2007-09-24.
  180. Yesalis, C (2000). "Source of Anabolic Steroids". Anabolic Steroids in Sport and Exercise. Champaign, Ill.: Human Kinetics. ISBN 978-0-88011-786-9.
  181. Black, Terry (1996). "Does the Ban on Drugs in Sport Improve Societal Welfare?". Faculty of Business, Queensland University of Technology. Retrieved 2007-04-24.
  182. Pound RW (2006). "Organized Crime". Inside dope : how drugs are the biggest threat to sports, why you should care, and what can be done about them. Mississaug, Ontario: Wiley. p. 175. ISBN 978-0-470-83733-7.
  183. Kanayama G, Hudson JI, Pope HG (November 2008). "Long-term psychiatric and medical consequences of anabolic-androgenic steroid abuse: a looming public health concern?". Drug Alcohol Depend. 98 (1–2): 1–12. doi:10.1016/j.drugalcdep.2008.05.004. PMC 2646607Freely accessible. PMID 18599224.
  184. "Steroids". National Institute on Drug Abuse. GDCADA. Archived from the original on 2007-09-11. Retrieved 2007-09-13.
  185. "The Drug Enforcement Administration's International Operations (Redacted)". Office of the Inspector General. USDOJ. February 2007. Retrieved 2014-01-02.

Further reading

  • Yesalis CE (2000). Anabolic Steroids in Sport and Exercise. Human Kinetics. ISBN 0-88011-786-9. 
  • Daniels RC (February 1, 2003). The Anabolic Steroid Handbook. RCD Books. p. 80. ISBN 0-9548227-0-6. 
  • Gallaway S (January 15, 1997). The Steroid Bible (3rd Sprl ed.). Belle Intl. p. 125. ISBN 1-890342-00-9. 
  • Llewellyn W (January 28, 2007). Anabolics 2007 : Anabolic Steroid Reference Manual (6th ed.). Body of Science. p. 988. ISBN 978-0-9679304-6-6. 
  • Roberts A, Clapp B (January 2006). Anabolic Steroids: Ultimate Research Guide. Anabolic Books, LLC. p. 394. ISBN 1-59975-100-3. 
  • Tygart TT (December 2009). "Steroids, the Media, and Youth". Prevention Researcher Integrated Research Services, Inc. SIRS Researcher. 16 (7–9). 
  • Eisenhauer L (Nov 7, 2005). "Do I Look OK?". St. Louis Post-Dispatch (St. Louis, MO). Retrieved 25 Oct 2010. 
  • Fragkaki AG, Angelis YS, Koupparis M, Tsantili-Kakoulidou A, Kokotos G, Georgakopoulos C (2009). "Structural characteristics of anabolic androgenic steroids contributing to binding to the androgen receptor and to their anabolic and androgenic activities. Applied modifications in the steroidal structure". Steroids. 74 (2): 172–97. doi:10.1016/j.steroids.2008.10.016. PMID 19028512. 
  • McRobb L, Handelsman DJ, Kazlauskas R, Wilkinson S, McLeod MD, Heather AK (2008). "Structure-activity relationships of synthetic progestins in a yeast-based in vitro androgen bioassay". J. Steroid Biochem. Mol. Biol. 110 (1-2): 39–47. doi:10.1016/j.jsbmb.2007.10.008. PMID 18395441. 

External links

Wikimedia Commons has media related to Anabolic-androgenic steroids.
This article is issued from Wikipedia - version of the 12/3/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.