Categories

The Science of Trenbolone v2 – Part Two

In the last article, we covered a lot of ground, focusing primarily on the history of trenbolone as well as its metabolic fate in mammals.  If you have not already read part one, I’d urge you to go back and give it a read before moving on, as some of the topics covered there will be expanded upon in this article.

VI. Effects on HPG Axis

In vertebrates, the hypothalamic-pituitary-gonadal (HPG) axis controls reproductive processes through a variety of hormones which act on target tissues either directly or indirectly. At a high level, in males, gonadotropin-releasing hormone (GnRH) released from the hypothalamus stimulates the pituitary to release luteinizing hormone (LH) and follicle stimulating hormone (FSH). These, in turn, stimulate the release of sex-hormones from the testes [1-2]. The strongly interwoven nature of the HPG axis means that no single component of the system operates in isolation. Upon entering circulation, both androgenic and estrogenic hormones are capable of crossing the blood-brain barrier and exerting negative feedback inhibition on the pituitary and hypothalamus, thereby downregulating GnRH release and suppressing the entire axis [3].

The administration of trenbolone is associated with numerous types of HPG axis disruptions, which is in line with what has been witnessed with various other androgen treatments over the years [4]. Some of the trenbolone-induced disruptions seen over the years include reduced levels of serum LH [5-10], reduced serum FSH levels [11], reduced testosterone levels [5-10,12-16], reduced DHT levels [11], reduced estradiol levels [13,15], testicular atrophy [7,17-18], and a delayed onset of puberty [19]. These effects occur quite rapidly as, was demonstrated in one trial, within ten days of trenbolone enanthate administration, castrated rats had 80% suppression rates of serum testosterone and 70% suppression rates of DHT as compared to control animals [20]. It is worth noting, enanthate is a long ester variant of trenbolone (TBE) so the use of acetate (TBA) would have produced these effects even more rapidly.

It is not precisely understood what mechanisms are behind trenbolone’s suppressive effects on the HPG axis, however there have certainly been some trials over the years which provide clues. One popular hypothesis involves direct hypothalamic feedback inhibition, as evidenced by reduced GnRH transcription seen in the brains of fish models. This may be additive to its direct effects on testicular steroid biosynthesis, as supported by downregulated expression of testicular CYP17 [21]. CYP17 is a very important enzyme in steroid biosynthesis, and sequentially catalyzes two key reactions in the production of sex steroids in males.

It is also interesting to note that whatever the mechanism is, it does not appear to be androgen receptor (AR) dependent [22-23]. Further supporting this line of thought, in ovary tissue cultures from fish, non-aromatizable androgens such as trenbolone had direct and non-genomic, anti-androgen-insensitive inhibitory, effects on estrogen production [24]. It is highly likely that the underlying feedback mechanisms at work are similar to other androgens, resulting in inhibited GnRH levels and ultimately inhibited FSH and LH production [25].

One other potential gene candidate involved in decreased sex steroid concentrations, noted in fish trials in which they had been exposed to the strong exogenous androgen 17-trenbolone, is hydroxysteroid (17β) dehydrogenase 12a (hsd17b12a). Hsd17b12a catalyzes the conversion of androstenedione to testosterone which, in turn, is converted to 17β-estradiol by the aromatase enzymes. Thus, down-regulation of hsd17b12a, as seen in these trenbolone-exposed fish, is predictably expected to lead to declines in both testosterone and estradiol [14].

VII. Effects on Anabolic Pathways

As we’ve covered previously, trenbolone expresses SARM-like behaviors and I’d like to use this section to discuss them in more depth.

5α REDUCTASE

Despite a structural similarity to testosterone, trenbolone does not undergo 5α reduction due to the presence of a 3-oxotriene structure which prevents A ring reduction [26]. This is the pathway used for converting testosterone into its more potent form dihydrotestosterone (DHT). As trenbolone is not a substrate for 5α reductase, it has been shown to stimulate less pronounced androgenic effects than testosterone in androgen-sensitive tissues which express the 5α reductase enzyme, including the prostate and accessory sex organs [27-32]. To put this statement into perspective, testosterone has an approximately three-fold higher potency in androgenic tissues that express 5α reductase despite having a significantly lower binding affinity to the AR than trenbolone [33]. We’ll discuss how this impacts hypertrophy potential in these tissues a bit later.

As you can already start to imagine, one particular reason trenbolone is beginning to pick up steam in scientific communities is due to its potential to lower the risks associated with prostate cancer in those patients being treated for hypogonadism. The current de facto treatment strategy for these individuals includes providing them with testosterone, in a manner designed to restore hormone levels to natural reference ranges. However, in adult males, benign and malignant growth of the glandular prostate tissue is largely regulated by sex hormones. And furthermore even moderate increases in circulating testosterone have been shown to directly translate into pronounced hyperplastic effects in prostate tissues, mediated via its 5α reduction into DHT [34-35]. Later in the article, we’ll dig deeper into the available literature to see if trenbolone’s potential to lower prostate cancer risk actually pans out.

AROMATASE ENZYME

Seemingly one of the more asked about questions is whether or not trenbolone has impacts on serum estrogen levels, as well as whether or not it can aromatize like testosterone. Popular opinion in the scientific community is that trenbolone and other 19-nor compounds are not substrates for the aromatase enzyme [36-37]. With that said, please understand this is not the same thing as saying they cannot convert to estrogen as C19 norandrogens can induce estrogenic effects [38-39].

Following this line of thought, trenbolone-itself is largely thought to be non-estrogenic [40-41] and there have been numerous animal trials that have demonstrated it reduces serum estradiol concentrations [13-15,42-44]. Keep in mind that there have been a few trials that did not show this suppressive effect on estrogen levels [10,45-46] but, by and large, the body of literature as a whole does support the hypothesis that trenbolone possesses anti-estrogenic effects.

Based upon what we now know about the HPG axis, this would tend to make a lot of sense as the anti-estrogenic effects caused by trenbolone administration likely have to do with its negative feedback on the axis. This negative feedback would cause the inhibition of endogenous testosterone production, thereby leading to suppressed levels of aromatization via the aromatase enzyme, which is necessary for endogenous estrogen biosynthesis in males. This impact on the HPG axis would cause a more severe rate of estrogen inhibition as compared to any potential direct effects trenbolone would have on estrogen receptors and/or the aromatase enzyme [5-6,8,21,47]. There may even be a secondary mechanism at work here which is related to trenbolone’s ability to downregulate expression in both the estrogen alpha and beta receptors [48].

There have been some other interesting discoveries with regard to the mechanisms behind trenbolone’s relationship with estrogen, as well as the compensatory responses associated with suppressed hormone levels. Trenbolone has been shown to reduce tissue concentrations, and gene expression, of VTG (vitellogenin) which is a protein positively associated with exposure to estrogenic compounds [13-14,21,41,47,49-54]. It has also been shown to downregulate brain CYP19B (aromatase B) and upregulate gonadal CYP19A (aromatase A) in female fish, but interestingly not in males [14,54].

Similar to what we’ve seen already in the HPG axis, the impacts of trenbolone on estrogen do not appear to be AR-dependent as trials have shown co-treatment with an AR antagonist (flutamide) resulted in the same anti-estrogenic activity in fish [13]. Interestingly, there has been another fish trial that reported trenbolone to have low-affinity with the estrogen receptor and can potentially even activate it [44]. Whether or not this is species-specific is a matter of debate, as I have not seen this occur in any other trials I’ve reviewed. However, cell culture experiments and bioassays do show that trenbolone and its metabolites have a very low binding affinity with estrogen receptors, roughly 20% of the efficacy of estradiol [40].

So can it aromatize? Although I have found nothing which definitively suggests it can, there has been a hypothesis thrown out there by Holland et al [55] which I find intriguing enough to include in its entirety:

“We previously reported that trenbolone enanthate potently reduced visceral fat mass in young and older ORX animals, indicating that fat loss occurs in response to androgen administration, even in the absence of an androgenic substrate for aromatase. However, our previous work did not account for the possibility that androstenedione (derived from dehydroepiandrosterone) can be aromatized to estrone and, subsequently, converted to E2 by actions of 17β-hydroxysteroid dehydrogenase in tissues, such as fat, expressing the required enzymes”

PROGESTERONE/SHBG

Trenbolone has been shown to have a high affinity for the bovine progestin receptor, and it is assumed that it has a similar affinity to the progesterone receptor as progesterone itself [56]. In vitro analysis has revealed that the relative binding affinity to the bovine progesterone receptor, as compared to progesterone, was 137.4% for 17β-TbOH and 2.1% for 17α-TbOH [57]. And finally, the relative binding affinity of trenbolone to human SHBG, as compared to DHT, is 29.4% for 17β-TbOH and 94.8% for 17α-TbOH.

VIII. Effects on Metabolic Health Markers

One of the primary reasons the anti-trenbolone crowd admonish against its use is related to how harsh the compound seemingly is on one’s health markers. I had hoped to have some actual reference blood work to add to this article, however unfortunately my crowdsourcing efforts were not successful as not many individuals run trenbolone by itself. So what I will do in this section is go over the available animal literature covering various health markers and trenbolone’s impacts upon them.

THYROIDAL AXIS

Arguably the most intriguing relationship to me is trenbolone and the thyroidal axis. Although the effects have been a bit inconsistent, there does seem to be a pattern which suggests trenbolone has an overall suppressive effect on the thyroidal axis. In one trial, trenbolone-alone decreased T4 in heifers while trenbolone plus estradiol decreased T4 in steers, while no impact was seen on T3 uptake [58]. In another trial, trenbolone plus estradiol actually increased T3 whereas trenbolone by itself decreased both T3 and T4 [59]. We must remember that estradiol stimulates the GH/IGF axis, which acutely increases the conversion of T4 to T3. This may help to explain why co-treatment with estradiol can result in higher T3 levels whereas trenbolone-only has the opposite effect, as estradiol levels are highly suppressed. Even in trials where thyroid levels are not significantly different, trenbolone decreased fasting metabolic rates, leading to less intake requirements for creating intake surplus [60].

Therefore, it may be reasonable to speculate the increased feed efficiency seen in numerous studies over the years could be related to trenbolone-mediated suppression of metabolic rate. Of course, it should be noted that leaner cattle just tend to grow faster, and use feed more efficiently, so it also may just be a byproduct of this [61]. Before I wrap this article series up, I’ll talk a bit more about the practical applications here and why these impacts may want to be considered when deciding how to use trenbolone for bodybuilding purposes.

CHOLESTEROL

Generally speaking, there is a strong correlation between fat loss and favorable changes in serum lipid levels, particularly in men [62-63]. Therefore, because trenbolone has been consistently shown to improve body composition, it is reasonable to speculate that it may have favorable impacts on lipid markers. So, let’s see what animal trials have shown us.

Studies on rats have shown that both testosterone and trenbolone elicit similar protections against elevated cholesterol despite trenbolone’s ability to elicit more visceral fat loss. This suggests that serum cholesterol levels may be governed primarily by overall body composition, independently of changes in visceral stores. In one trial, serum total cholesterol, HDL, and LDL were all significantly lower in trenbolone-treated intact rats than in control rats (- 62%, – 57%, and – 78% respectively). The byproduct of this was that treated rats had a greater HDL:LDL ratio. Serum triglycerides were also significantly decreased by 51% as compared to control rats [15]. In another trial, both testosterone and trenbolone reduced circulating cholesterol in rats fed a high fat and high sugar diet, but only trenbolone reduced circulating triglyceride levels [64].

It is strongly suggested to keep an eye on cholesterol levels when using supraphysiological doses of androgens, as they tend to have the ability to increase catecholamine-stimulated hormone-sensitive lipase (HSL) activity in both the liver and cardiac tissues [65-66]. This increased HSL activity tends to result in an increased rate of triglyceride breakdown and suppressed rates of HDL hydrolyzation. Having chronically suppressed levels of HDL seems to be an independent cardiovascular risk factor so prolonged use of androgens resulting in suppressed HDL levels should be done with extreme caution [67]

LIVER MARKERS

There are quite a few hepatic markers commonly used to assess overall liver functionality and health, as well as hepatic damage. Albumin is a marker indicative of overall liver function while AST, ALT, ALP are all general markers of hepatic damage.

Recent rodent trials have shown that trenbolone does not appear to induce significant damage to hepatic tissues. In one trial, hepatic tissue samples of trenbolone-treated rats showed a similar morphology to those of control rats. AST, ALT, ALP, and albumin were all at similar level in trenbolone-treated rats as compared to control [15]. In a follow-up trial, similar liver enzyme values were seen with rats fed high fat and high sugar diet in all treatment groups, including testosterone and trenbolone treatments [64].

INSULIN

This is another hormone that tends to have a direct correlation with body fat, and specifically visceral fat levels. Visceral fat accumulation and elevated circulating triglyceride levels are both associated with insulin resistance [68]. Conversely, calorie restriction and weight loss in viscerally obese non-diabetics induced significant improvements in insulin sensitivity [69]. In addition to obesity, there is also compelling evidence which shows that low androgen levels also promote insulin resistance [70]. It has been hypothesized that trenbolone treatment in animal models may promote insulin sensitizing effects through similar mechanisms to those achieved by calorie restriction in human males so let’s see what the trials have actually demonstrated.

One trial showed serum insulin to be significantly lower in trenbolone-treated rats (38% reduction) as compared to control rats which translated into a significantly lower HOMA-IR value, a metric used to measure insulin resistance [15]. Fascinatingly, rats being fed high fat and high sugar diets had significantly elevated serum insulin levels that were only partly restored with testosterone, yet trenbolone significantly reduced insulin levels [64]. In fact, trenbolone was also the only treatment group to reduce HOMA-IR values indicating increased beta-cell function and lowered insulin resistance. So albeit limited, the evidence does suggest that trenbolone has superior insulin sensitizing effects as compared to testosterone.

Adiponectin is a 30 kDa insulin sensitizing adipokine that is primarily secreted by visceral adipose tissues [71-72]. Generally speaking, serum adiponectin levels are inversely proportional to fat mass [73]. Testosterone and trenbolone tend to reduce total adiponectin levels to a similar degree in rats [74].

ERYTHROPOIESIS

Erythropoiesis is just a fancy term for the body’s production of red blood cells (RBCs). One of the most commonly reported side effects of TRT treatments tends to be elevated levels of hematocrit and hemoglobin. Specifically, androgen deprivation reduces both hematocrit and hemoglobin whereas testosterone administration results in a dose-dependent increase in both [75-76].

The mechanisms by which androgens augment RBC production may be directly related to stimulation of kidney erythropoietin secretion or even bone marrow [77]. And based upon existing evidence, it would appear as if androgens directly elevate erythropoiesis via AR-mediated mechanisms [11]. It does not appear as if the aromatization of testosterone is required for erythropoiesis as DHT administration also increases the process in male subjects [78]. Furthermore, it has also been demonstrated to occur in male subjects with aromatase-deficiencies [79]. Similarly, 5α reduction of testosterone does not appear to be required for erythropoiesis as the co-administration of testosterone and finasteride (5α reductase inhibitor) increased both hematocrit and hemoglobin to the same extent as testosterone alone despite 65% lower DHT concentrations in the finasteride group [80].

If trenbolone can reduce the elevations of hematocrit and hemoglobin seen with traditional TRT treatments, then this would be another potential reason it could be an interesting candidate for HRT. Let’s see what the trials indicate.

Preliminary evidence indicates that trenbolone increases hemoglobin in male rodents in a dose-dependent manner, and to a slightly greater extent than supraphysiological testosterone (8-10%), despite DHT being suppressed by over 70% following administration [20]. In another trial, at administered doses which were seven times higher than testosterone, trenbolone treated rats had nearly identical levels of hemoglobin, although both were significantly elevated as compared to controls [81].

Okay, I think this is a natural stopping point for part two. In part three, we’re going to begin to dive into the more exciting stuff as we cover anabolism and hypertrophy. Depending on how deeply we dive on that topic, that may be all we cover in part three. However, we also may cover lipolysis and some other fun topics if time permits. So, until next time…

REFERENCES

  1. Simoni M, Weinbauer GF, Gromoll J, Nieschlag E. Role of FSH in male gonadal function. Ann Endocrinol (Paris). 1999 Jul;60(2):102-6. Review.
  2. Liu PY, Iranmanesh A, Nehra AX, Keenan DM, Veldhuis JD. Mechanisms of hypoandrogenemia in healthy aging men. Endocrinol Metab Clin North Am. 2005 Dec;34(4):935-55, ix. Review.
  3. Kumar TR, Low MJ. Hormonal regulation of human follicle-stimulating hormone-beta subunit gene expression: GnRH stimulation and GnRH-independent androgen inhibition. Neuroendocrinology. 1995 Jun;61(6):628-37.
  4. Tan RS, Scally MC. Anabolic steroid-induced hypogonadism–towards a unified hypothesis of anabolic steroid action. Med Hypotheses. 2009 Jun;72(6):723-8.
  5. Fabry J, Renaville R, Halleux V, Burny A. Plasma testosterone and LH responses to LHRH in double-muscled bulls treated with trenbolone acetate and zeranol. J Anim Sci. 1983 Nov;57(5):1138-45.
  6. Gettys TW, D’Occhio MJ, Henricks DM, Schanbacher BD. Suppression of LH secretion by oestradiol, dihydrotestosterone and trenbolone acetate in the acutely castrated bull. J Endocrinol. 1984 Jan;100(1):107-12.
  7. Silcox RW, Keeton JT, Johnson BH. Effects of zeranol and trenbolone acetate on testis function, live weight gain and carcass traits of bulls. J Anim Sci. 1986 Aug;63(2):358-68.
  8. Renaville R, Burny A, Sneyers M, Rochart S, Portetelle D, Théwis A. Effects of an anabolic treatment before puberty with trenbolone acetate-oestradiol or oestradiol alone on growth rate, testicular development and luteinizing hormone and testosterone plasma concentrations. Theriogenology. 1988 Feb;29(2):461-76.
  9. Lee CY, Henricks DM, Skelley GC, Grimes LW. Growth and hormonal response of intact and castrate male cattle to trenbolone acetate and estradiol. J Anim Sci. 1990 Sep;68(9):2682-9.
  10. Hunt DW, Henricks DM, Skelley GC, Grimes LW. Use of trenbolone acetate and estradiol in intact and castrate male cattle: effects on growth, serum hormones, and carcass characteristics. J Anim Sci. 1991 Jun;69(6):2452-62.
  11. Yarrow JF, McCoy SC, Borst SE. Tissue selectivity and potential clinical applications of trenbolone (17beta-hydroxyestra-4,9,11-trien-3-one): A potent anabolic steroid with reduced androgenic and estrogenic activity. Steroids. 2010 Jun;75(6):377-89.
  12. Galbraith, H. (1982). Growth, hormonal and metabolic response of post-pubertal entire male cattle to trenbolone acetate and hexoestrol. Animal Science, 35(2), 269-276.
  13. Ankley GT, Defoe DL, Kahl MD, Jensen KM, Makynen EA, Miracle A, Hartig P, Gray LE, Cardon M, Wilson V. Evaluation of the model anti-androgen flutamide for assessing the mechanistic basis of responses to an androgen in the fathead minnow (Pimephales promelas). Environ Sci Technol. 2004 Dec 1;38(23):6322-7.
  14. Dorts J, Richter CA, Wright-Osment MK, Ellersieck MR, Carter BJ, Tillitt DE. The genomic transcriptional response of female fathead minnows (Pimephales promelas) to an acute exposure to the androgen, 17beta-trenbolone. Aquat Toxicol. 2009 Jan 18;91(1):44-53.
  15. Donner DG, Beck BR, Bulmer AC, Lam AK, Du Toit EF. Improvements in body composition, cardiometabolic risk factors and insulin sensitivity with trenbolone in normogonadic rats. Steroids. 2015 Feb;94:60-9.
  16. Ma F, Liu D. 17β-trenbolone, an anabolic-androgenic steroid as well as an environmental hormone, contributes to neurodegeneration. Toxicol Appl Pharmacol. 2015 Jan 1;282(1):68-76.
  17. O’Lamhna M, Roche JF. Effect of repeated implantation with anabolic agents on growth rate, carcase weight, testicular size and behaviour of bulls. Vet Rec. 1983 Dec 3;113(23):531-4.
  18. López-Bote C, Sancho G, Martínez M, Ventanas J, Gázquez A, Roncero V. Trenbolone acetate induced changes in the genital tract of male pigs. Zentralbl Veterinarmed B. 1994 Mar;41(1):42-8.
  19. Moran C, Prendiville DJ, Quirke JF, Roche JF. Effects of oestradiol, zeranol or trenbolone acetate implants on puberty, reproduction and fertility in heifers. J Reprod Fertil. 1990 Jul;89(2):527-36.
  20. Yarrow JF, Conover CF, McCoy SC, Lipinska JA, Santillana CA, Hance JM, Cannady DF, VanPelt TD, Sanchez J, Conrad BP, Pingel JE, Wronski TJ, Borst SE. 17β-Hydroxyestra-4,9,11-trien-3-one (trenbolone) exhibits tissue selective anabolic activity: effects on muscle, bone, adiposity, hemoglobin, and prostate. Am J Physiol Endocrinol Metab. 2011 Apr;300(4):E650-60.
  21. Zhang X, Hecker M, Park JW, Tompsett AR, Jones PD, Newsted J, Au DW, Kong R, Wu RS, Giesy JP. Time-dependent transcriptional profiles of genes of the hypothalamic-pituitary-gonadal axis in medaka (Oryzias latipes) exposed to fadrozole and 17beta-trenbolone. Environ Toxicol Chem. 2008 Dec;27(12):2504-11.
  22. Heinlein CA, Chang C. The roles of androgen receptors and androgen-binding proteins in nongenomic androgen actions. Mol Endocrinol. 2002 Oct;16(10):2181-7. Review.
  23. Martinović D, Blake LS, Durhan EJ, Greene KJ, Kahl MD, Jensen KM, Makynen EA, Villeneuve DL, Ankley GT. Reproductive toxicity of vinclozolin in the fathead minnow: confirming an anti-androgenic mode of action. Environ Toxicol Chem. 2008 Feb;27(2):478-88.
  24. Braun AM, Thomas P. Androgens inhibit estradiol-17beta synthesis in Atlantic croaker (Micropogonias undulatus) ovaries by a nongenomic mechanism initiated at the cell surface. Biol Reprod. 2003 Nov;69(5):1642-50.
  25. MacIndoe JH, Perry PJ, Yates WR, Holman TL, Ellingrod VL, Scott SD. Testosterone suppression of the HPT axis. J Investig Med. 1997 Oct;45(8):441-7.
  26. Pottier J, Cousty C, Heitzman RJ, Reynolds IP. Differences in the biotransformation of a 17 beta-hydroxylated steroid, trenbolone acetate, in rat and cow. Xenobiotica. 1981 Jul;11(7):489-500.
  27. Wilson VS, Lambright C, Ostby J, Gray LE Jr. In vitro and in vivo effects of 17beta-trenbolone: a feedlot effluent contaminant. Toxicol Sci. 2002 Dec;70(2):202-11.
  28. Ashby J, Lefevre PA, Tinwell H, Odum J, Owens W. Testosterone-stimulated weanlings as an alternative to castrated male rats in the Hershberger anti-androgen assay. Regul Toxicol Pharmacol. 2004 Apr;39(2):229-38.
  29. Freyberger A, Hartmann E, Krötlinger F. Evaluation of the rodent Hershberger bioassay using three reference (anti)androgens. Arh Hig Rada Toksikol. 2005 Jun;56(2):131-9.
  30. Freyberger A, Ellinger-Ziegelbauer H, Krötlinger F. Evaluation of the rodent Hershberger bioassay: testing of coded chemicals and supplementary molecular-biological and biochemical investigations. Toxicology. 2007 Sep 24;239(1-2):77-88.
  31. Owens W, Gray LE, Zeiger E, Walker M, Yamasaki K, Ashby J, Jacob E. The OECD program to validate the rat Hershberger bioassay to screen compounds for in vivo androgen and antiandrogen responses: phase 2 dose-response studies. Environ Health Perspect. 2007 May;115(5):671-8.
  32. Moon HJ, Kang TS, Kim TS, Kang IH, Ki HY, Kim SH, Han SY. OECD validation of phase 3 Hershberger assay in Korea using surgically castrated male rats with coded chemicals. J Appl Toxicol. 2009 May;29(4):350-5.
  33. Wilson JD. The role of 5alpha-reduction in steroid hormone physiology. Reprod Fertil Dev. 2001;13(7-8):673-8. Review.
  34. Borst SE, Lee JH, Conover CF. Inhibition of 5alpha-reductase blocks prostate effects of testosterone without blocking anabolic effects. Am J Physiol Endocrinol Metab. 2005 Jan;288(1):E222-7.
  35. Vargas RA, Oliveira LP, Frankenfeld S, Souza DB, Costa WS, Favorito LA, Sampaio FJ. The prostate after administration of anabolic androgenic steroids: a morphometrical study in rats. Int Braz J Urol. 2013 Sep-Oct;39(5):675-82.
  36. Donaldson IA, Hart IC, Heitzman RJ. Growth hormone, insulin, prolactin and total thyroxine in the plasma of sheep implanted with the anabolic steroid trenbolone acetate alone or with oestradiol. Res Vet Sci. 1981 Jan;30(1):7-13.
  37. Quinn MJ Jr, Lavoie ET, Ottinger MA. Reproductive toxicity of trenbolone acetate in embryonically exposed Japanese quail. Chemosphere. 2007 Jan;66(7):1191-6.
  38. Kuhl H, Wiegratz I. Can 19-nortestosterone derivatives be aromatized in the liver of adult humans? Are there clinical implications? Climacteric. 2007 Aug;10(4):344-53. Review.
  39. Attardi BJ, Pham TC, Radler LC, Burgenson J, Hild SA, Reel JR. Dimethandrolone (7alpha,11beta-dimethyl-19-nortestosterone) and 11beta-methyl-19-nortestosterone are not converted to aromatic A-ring products in the presence of recombinant human aromatase. J Steroid Biochem Mol Biol. 2008 Jun;110(3-5):214-22.
  40. Le Guevel R, Pakdel F. Assessment of oestrogenic potency of chemicals used as growth promoter by in-vitro methods. Hum Reprod. 2001 May;16(5):1030-6.
  41. Hemmer MJ, Cripe GM, Hemmer BL, Goodman LR, Salinas KA, Fournie JW, Walker CC. Comparison of estrogen-responsive plasma protein biomarkers and reproductive endpoints in sheepshead minnows exposed to 17beta-trenbolone. Aquat Toxicol. 2008 Jun 23;88(2):128-36.
  42. Neumann F. Pharmacological and endocrinological studies on anabolic agents. Environ Qual Saf Suppl. 1976;(5):253-64. Review.
  43. Hunt DW, Henricks DM, Skelley GC, Grimes LW. Use of trenbolone acetate and estradiol in intact and castrate male cattle: effects on growth, serum hormones, and carcass characteristics. J Anim Sci. 1991 Jun;69(6):2452-62.
  44. Ankley GT, Jensen KM, Makynen EA, Kahl MD, Korte JJ, Hornung MW, Henry TR, Denny JS, Leino RL, Wilson VS, Cardon MC, Hartig PC, Gray LE. Effects of the androgenic growth promoter 17-beta-trenbolone on fecundity and reproductive endocrinology of the fathead minnow. Environ Toxicol Chem. 2003 Jun;22(6):1350-60.
  45. Henricks DM, Edwards RL, Champe KA, Gettys TW, Skelley GC Jr, Gimenez T. Trenbolone, estradiol-17 beta and estrone levels in plasma and tissues and live weight gains of heifers implanted with trenbolone acetate. J Anim Sci. 1982 Nov;55(5):1048-56.
  46. Henricks DM, Brandt RT Jr, Titgemeyer EC, Milton CT. Serum concentrations of trenbolone-17 beta and estradiol-17 beta and performance of heifers treated with trenbolone acetate, melengestrol acetate, or estradiol-17 beta. J Anim Sci. 1997 Oct;75(10):2627-33.
  47. Zhang X, Hecker M, Park JW, Tompsett AR, Jones PD, Newsted J, Au DW, Kong R,
    Wu RS, Giesy JP. Time-dependent transcriptional profiles of genes of the
    hypothalamic-pituitary-gonadal axis in medaka (Oryzias latipes) exposed to
    fadrozole and 17beta-trenbolone. Environ Toxicol Chem. 2008 Dec;27(12):2504-11.
  48. Reiter M, Walf VM, Christians A, Pfaffl MW, Meyer HH. Modification of mRNA
    expression after treatment with anabolic agents and the usefulness for gene
    expression-biomarkers. Anal Chim Acta. 2007 Mar 14;586(1-2):73-81.
  49. Seki M, Fujishima S, Nozaka T, Maeda M, Kobayashi K. Comparison of response to 17 beta-estradiol and 17 beta-trenbolone among three small fish species. Environ Toxicol Chem. 2006 Oct;25(10):2742-52.
  50. Jensen KM, Ankley GT. Evaluation of a commercial kit for measuring vitellogenin in the fathead minnow (Pimephales promelas). Ecotoxicol Environ Saf. 2006 Jun;64(2):101-5. Epub 2006 Apr 17.
  51. Holbech H, Kinnberg K, Petersen GI, Jackson P, Hylland K, Norrgren L, Bjerregaard P. Detection of endocrine disrupters: evaluation of a Fish Sexual Development Test (FSDT). Comp Biochem Physiol C Toxicol Pharmacol. 2006 Sep;144(1):57-66.
  52. Orn S, Yamani S, Norrgren L. Comparison of vitellogenin induction, sex ratio, and gonad morphology between zebrafish and Japanese medaka after exposure to 17alpha-ethinylestradiol and 17beta-trenbolone. Arch Environ Contam Toxicol. 2006 Aug;51(2):237-43.
  53. Park JW, Tompsett A, Zhang X, Newsted JL, Jones PD, Au D, Kong R, Wu RS, Giesy JP, Hecker M. Fluorescence in situ hybridization techniques (FISH) to detect changes in CYP19a gene expression of Japanese medaka (Oryzias latipes). Toxicol Appl Pharmacol. 2008 Oct 15;232(2):226-35.
  54. Zhang X, Hecker M, Park JW, Tompsett AR, Newsted J, Nakayama K, Jones PD, Au D, Kong R, Wu RS, Giesy JP. Real-time PCR array to study effects of chemicals on the Hypothalamic-Pituitary-Gonadal axis of the Japanese medaka. Aquat Toxicol. 2008 Jul 7;88(3):173-82.
  55. Holland AM, Roberts MD, Mumford PW, Mobley CB, Kephart WC, Conover CF, Beggs LA, Balaez A, Otzel DM, Yarrow JF, Borst SE, Beck DT. Testosterone inhibits expression of lipogenic genes in visceral fat by an estrogen-dependent mechanism. J Appl Physiol (1985). 2016 Sep 1;121(3):792-805.
  56. H. H. D. MEYER and M. RAPP Reversible binding of the anabolic steroid trenbolone to steroid receptors Acta Endocrinol 110 S129-S130
  57. Bauer ER, Daxenberger A, Petri T, Sauerwein H, Meyer HH. Characterisation of the affinity of different anabolics and synthetic hormones to the human androgen receptor, human sex hormone binding globulin and to the bovine progestin receptor. APMIS. 2000 Dec;108(12):838-46.
  58. Heitzman RJ, Donaldson IA, Hart IC. Effect of anabolic steroids on plasma thyroid hormones in steers and heifers. Br Vet J. 1980 Mar-Apr;136(2):168-74.
  59. Mader TL, Kreikemeier WM. Effects of growth-promoting agents and season on blood metabolites and body temperature in heifers. J Anim Sci. 2006 Apr;84(4):1030-7.
  60. Hunter RA, Vercoe JE. Reduction of energy requirements of steers fed on low-quality-roughage diets using trenbolone acetate. Br J Nutr. 1987 Nov;58(3):477-83.
  61. Field RA. Effect of castration on meat quality and quantity. J Anim Sci. 1971 May;32(5):849-58.
  62. Leenen R, van der Kooy K, Droop A, Seidell JC, Deurenberg P, Weststrate JA, Hautvast JG. Visceral fat loss measured by magnetic resonance imaging in relation to changes in serum lipid levels of obese men and women. Arterioscler Thromb. 1993 Apr;13(4):487-94.
  63. Leenen R, van der Kooy K, Meyboom S, Seidell JC, Deurenberg P, Weststrate JA. Relative effects of weight loss and dietary fat modification on serum lipid levels in the dietary treatment of obesity. J Lipid Res. 1993 Dec;34(12):2183-91.
  64. Donner DG, Elliott GE, Beck BR, Bulmer AC, Lam AK, Headrick JP, Du Toit EF. Trenbolone Improves Cardiometabolic Risk Factors and Myocardial Tolerance to Ischemia-Reperfusion in Male Rats With Testosterone-Deficient Metabolic Syndrome. Endocrinology. 2016 Jan;157(1):368-81.
  65. Rebuffé-Scrive M, Mårin P, Björntorp P. Effect of testosterone on abdominal adipose tissue in men. Int J Obes. 1991 Nov;15(11):791-5.
  66. Langfort J, Jagsz S, Dobrzyn P, Brzezinska Z, Klapcinska B, Galbo H, Gorski J. Testosterone affects hormone-sensitive lipase (HSL) activity and lipid metabolism in the left ventricle. Biochem Biophys Res Commun. 2010 Sep 3;399(4):670-6.
  67. Goldbourt U, Yaari S, Medalie JH. Isolated low HDL cholesterol as a risk factor for coronary heart disease mortality. A 21-year follow-up of 8000 men. Arterioscler Thromb Vasc Biol. 1997 Jan;17(1):107-13.
  68. Katsuki A, Sumida Y, Urakawa H, Gabazza EC, Murashima S, Maruyama N, Morioka K, Nakatani K, Yano Y, Adachi Y. Increased visceral fat and serum levels of triglyceride are associated with insulin resistance in Japanese metabolically obese, normal weight subjects with normal glucose tolerance. Diabetes Care. 2003 Aug;26(8):2341-4.
  69. Bruun JM, Verdich C, Toubro S, Astrup A, Richelsen B. Association between measures of insulin sensitivity and circulating levels of interleukin-8, interleukin-6 and tumor necrosis factor-alpha. Effect of weight loss in obese men. Eur J Endocrinol. 2003 May;148(5):535-42.
  70. Kapoor D, Malkin CJ, Channer KS, Jones TH. Androgens, insulin resistance and vascular disease in men. Clin Endocrinol (Oxf). 2005 Sep;63(3):239-50. Review.
  71. Motoshima H, Wu X, Sinha MK, Hardy VE, Rosato EL, Barbot DJ, Rosato FE, Goldstein BJ. Differential regulation of adiponectin secretion from cultured human omental and subcutaneous adipocytes: effects of insulin and rosiglitazone. J Clin Endocrinol Metab. 2002 Dec;87(12):5662-7.
  72. Swarbrick MM, Havel PJ. Physiological, pharmacological, and nutritional regulation of circulating adiponectin concentrations in humans. Metab Syndr Relat Disord. 2008 Jun;6(2):87-102.
  73. Gavrila A, Chan JL, Yiannakouris N, Kontogianni M, Miller LC, Orlova C, Mantzoros CS. Serum adiponectin levels are inversely associated with overall and central fat distribution but are not directly regulated by acute fasting or leptin administration in humans: cross-sectional and interventional studies. J Clin Endocrinol Metab. 2003 Oct;88(10):4823-31.
  74. Yarrow JF, Beggs LA, Conover CF, McCoy SC, Beck DT, Borst SE. Influence of Androgens on Circulating Adiponectin in Male and Female Rodents. Lobaccaro J-MA, ed. PLoS ONE. 2012;7(10):e47315.
  75. Calof OM, Singh AB, Lee ML, Kenny AM, Urban RJ, Tenover JL, Bhasin S. Adverse events associated with testosterone replacement in middle-aged and older men: a meta-analysis of randomized, placebo-controlled trials. J Gerontol A Biol Sci Med Sci. 2005 Nov;60(11):1451-7.
  76. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab. 2008 Mar;93(3):914-9.
  77. Shahani S, Braga-Basaria M, Maggio M, Basaria S. Androgens and erythropoiesis: past and present. J Endocrinol Invest. 2009 Sep;32(8):704-16.
  78. Sakhri S, Gooren LJ. Safety aspects of androgen treatment with 5alpha-dihydrotestosterone. Andrologia. 2007 Dec;39(6):216-22. Review.
  79. Rochira V, Zirilli L, Madeo B, Maffei L, Carani C. Testosterone action on erythropoiesis does not require its aromatization to estrogen: Insights from the testosterone and estrogen treatment of two aromatase-deficient men. J Steroid Biochem Mol Biol. 2009 Feb;113(3-5):189-94.
  80. Amory JK, Watts NB, Easley KA, Sutton PR, Anawalt BD, Matsumoto AM, Bremner WJ, Tenover JL. Exogenous testosterone or testosterone with finasteride increases bone mineral density in older men with low serum testosterone. J Clin Endocrinol Metab. 2004 Feb;89(2):503-10.
  81. McCoy SC, Yarrow JF, Conover CF, Borsa PA, Tillman MD, Conrad BP, Pingel JE, Wronski TJ, Johnson SE, Kristinsson HG, Ye F, Borst SE. 17β-Hydroxyestra-4,9,11-trien-3-one (Trenbolone) preserves bone mineral density in skeletally mature orchiectomized rats without prostate enlargement. Bone. 2012 Oct;51(4):667-73.

 

 

Comments are closed.