Abstract
Testosterone acting via the androgen receptor, and via aromatisation to oestradiol, an activator of the oestrogen receptor, plays key roles in adipose tissue, bone and skeletal muscle biology. This is reflected in epidemiological studies associating obesity and disordered glucose metabolism with lower serum testosterone concentrations and an increased risk of type 2 diabetes (T2D) in men. Testosterone also modulates erythrocytosis and vascular endothelial and smooth muscle cell function, with potential impacts on haematocrit and the cardiovascular system. The Testosterone for the Prevention of Type 2 Diabetes (T4DM) study enrolled men aged 50 years and over with a waist circumference of 95 cm or over, impaired glucose tolerance or newly diagnosed T2D, and a serum testosterone concentration (as measured by chemiluminescence immunoassay) <14.0 nmol/L. The study reported that a 2-year treatment with testosterone undecanoate 1000 mg, administered 3-monthly intramuscularly, on the background of a lifestyle program, reduced the likelihood of T2D diagnosis by 40% compared to placebo. This effect was accompanied by a decrease in fasting serum glucose and associated with favourable changes in body composition, hand grip strength, bone mineral density and skeletal microarchitecture but not in HbA1c, a red blood cell-dependent measure of glycaemic control. There was no signal for cardiovascular adverse events. With the objective of informing translational science and future directions, this article discusses mechanistic studies underpinning the rationale for T4DM and translational implications of the key outcomes relating to glycaemia, and body composition, together with effects on erythrocytosis, cardiovascular risk and slow recovery of the hypothalamo–pituitary–testicular axis.
Introduction
Testosterone, obesity and male ageing
A decrease in serum testosterone concentration is not inevitable in ageing men (Sartorius et al. 2012, Shi et al. 2013, Handelsman et al. 2015, Marriott et al. 2022). The apparent decrease in testosterone seen in unselected middle aged and older men reflects obesity and the accumulation of chronic conditions (Shi et al. 2013), for example, cardiovascular disease (Regadera et al. 1985) and depression (Finkelstein et al. 2013, Shi et al. 2013), medication use (most commonly opioids), health-related behaviours (e.g. excessive alcohol consumption) and psychosocial factors (Wittert & Grossmann 2022) rather than chronological ageing in itself. However, beyond the age of 70 years, declining testosterone concentrations in unselected men with co-morbidities are accompanied by increases in LH, suggesting possibly impaired Leydig cell function (Yeap et al. 2018). The obesity phenotype associated with a decrease in serum testosterone concentration in middle to older aged men is associated with a reduction serum SHBG with mid-normal serum LH and FSH and is characterised by an accumulation of visceral adipose tissue associated with insulin resistance, aberrant glucose metabolism and dyslipidaemia as features of the metabolic syndrome (Umapathysivam et al. 2022). The proportionate fall in serum testosterone and sex hormone binding globulin (SHBG) concentrations under these circumstances is inherently reversible with weight loss (Grossmann 2018) and does not demonstrate androgen deficiency. Although weight loss will both increase serum testosterone and prevent type 2 diabetes (T2D), it is unclear whether this effect is mediated directly by the increase in testosterone or by beneficial effects of testosterone on body composition.
Mechanistic aspects of testosterone action
Testosterone is both a hormone, acting directly at the androgen receptor (AR), and a prohormone, undergoing 5-alpha reduction to DHT, a more potent AR analogue and being aromatised to oestradiol (E2), which acts via oestrogen receptors (ERs). These sex steroid receptors are ligand-dependent transcription factors, and AR and ERs, upon binding of their cognate hormone, are shuttled to the nucleus where they bind on specific DNA sequences (response elements), in gene promotor regions to regulate gene transcription in concert with transcriptional cofactors. In addition, there may be expression of cell membrane-bound sex steroid receptors, which may mediate rapid (non-classical) actions of testosterone and E2 (Handelsman 2000).
Testosterone effects on body composition: basic and translational considerations
There is a positive relationship between circulating testosterone concentration with muscle mass and strength and an inverse relationship with fat mass (Bhasin et al. 2001, Finkelstein et al. 2013). Testosterone treatment decreases fat mass and increases lean body mass (skeletal muscle and bone) and muscle strength in both healthy and hypogonadal men irrespective of age (Bhasin et al. 2001).
In skeletal muscle, testosterone, via the AR, induces hypertrophy of both type I and type II muscle fibres, increases muscle satellite cells, myogenic differentiation of muscle progenitor cells and muscle protein synthesis and inhibits adipogenic differentiation of pluripotent stem cells (Herbst & Bhasin 2004). Testosterone, following aromatisation to E2, increases growth hormone (GH) secretion from the pituitary (Eakman et al. 1996) and expression of insulin-like growth factor I (IGF-I) in skeletal muscle (Ferrando et al. 2002). However, the trophic effects of testosterone on skeletal muscle do not require GH and IGF-I (Serra et al. 2011).
In males, E2 is predominantly synthesised by aromatisation of testosterone in a tissue-specific manner (Russell et al. 2019). In females, but not males, E2 has a protective effect on functional skeletal muscle mass (McMillin et al. 2022, Pellegrino et al. 2022). The effect of testosterone to decrease fat mass is mediated at least in part by aromatisation to E2. Men or male mice with genetic inactivation of ER-alpha or aromatase have increased fat mass (Simpson et al. 2005) and, in healthy men, increasing serum testosterone by administration of aromatase inhibitors does not lead to a reduction in fat mass (Burnett-Bowie et al. 2009). Any beneficial effect of E2 on fat mass appears to require the presence of testosterone because in men who are androgen deficient, supplemental E2 leads to an increase in fat mass (Russell et al. 2022).
In males, E2 promotes insulin sensitivity and glucose metabolism in skeletal muscle via an E2 receptor alpha-mediated mechanism (Inada et al. 2016). E2 may also improve skeletal muscle glucose metabolism indirectly by increasing overnight pulsatile GH secretion from the pituitary while inhibiting the hepatic IGF-1 response to GH (Russell et al. 2019). In middle aged and older males, higher fat mass (particularly visceral far accumulation) increases the risk of T2D, an effect reversed with reduction in fat mass, and there is an inverse relationship between skeletal muscle mass and strength and risk of T2D (Atlantis et al. 2009, Li et al. 2016, Qadir et al. 2021).
Applying both epidemiological and mechanistic perspectives implicating testosterone in adipose and skeletal muscle biology, and diabetes risk, we designed a randomised controlled trial (RCT), ‘The Testosterone for the Prevention of Type 2 Diabetes (T4DM) study’. T4DM sought to determine whether treatment with testosterone, on the background of a lifestyle program, would prevent the progression of prediabetes to T2D or reverse newly diagnosed T2D in men aged 50 and over with visceral obesity as reflected by a waist circumference (WC) of 95 cm or greater and mild lowering of circulating testosterone but excluded those with pathological hypogonadism.
All men in T4DM were enrolled in a lifestyle program as this is the standard of care for T2D prevention and management of T2D. The lifestyle intervention delivered by WW (formerly Weight Watchers) was selected because it is standardised, widely accessible nationally and evidence based for the cost-effective prevention of T2D in men (Fuller et al. 2013).
Design of T4DM
The serum testosterone cut-off for inclusion
To reflect the typical at-risk population, we excluded men with pathological hypogonadism, where the benefit and safety of testosterone is widely accepted. Ultimately, an upper limit of serum testosterone concentration of 14 nmol/L, as measured by a CLIA was adopted as a screening compromise between treatment thresholds of serum testosterone advocated elsewhere with our observations that the association between serum testosterone concentration and T2D risk in men with obesity and the metabolic syndrome, persists up to about 16 nmol/L (Atlantis et al. 2016), implying that higher testosterone concentrations are associated with protection from T2D. After enrolment in the study, serum testosterone was measured at baseline by LCMS resulting in serum concentrations from 4 to 30 nmol/L, a finding confirming the unreliability of the testosterone immunoassays (Sikaris et al. 2005, Rosner et al. 2007). The absence of a relationship between baseline concentration of testosterone regardless of the method of measurement and the primary outcome of the study highlights the pharmacological effect of testosterone treatment in this study.
Waist circumference to assess obesity
The details of the design of the study are published (Wittert et al. 2019). WC rather than body mass index (BMI) was used as the anthropometric criterion for enrolment because an increase, reflecting abdominal obesity, is associated with T2D risk even if BMI is normal (Janiszewski et al. 2007). Abdominal obesity, where fat accumulates around the viscera and in the liver and infiltrates skeletal muscles mechanistically underpins the metabolic syndrome, which is characterised by insulin resistance, dyslipidaemia (typically raised triglycerides and reduced high-density lipoprotein concentrations), abnormal glucose metabolism and hypertension. The metabolic syndrome is associated with a reduction in serum testosterone, but normal LH and FSH concentrations most likely reflecting a reduction in SHBG (Brand et al. 2014). SHBG is synthesised in the liver and tightly coupled to de novo lipogenesis. When de novo lipogenesis increases, SHBG synthesis decreases (Selva et al. 2007). All testosterone assays measure total testosterone which predominantly reflects SHBG-bound testosterone, and therefore, if SHBG decreases, this will be reflected in a decrease in total testosterone concentration (Gleicher et al. 2020, Kim et al. 2020, Zhou et al. 2020). The extent to which, and under what circumstances, there are direct defects of excess adipose tissue on the regulation of the hypothalamic–pituitary–testicular (HPT) axis is not completely resolved, but there is some evidence for mediation by hypothalamic leptin resistance, central effects of inflammatory cytokines and inhibitory effects of increased oxidised low-density lipoprotein, adenosine and dysregulated iron metabolism on Leydig cell function in the testis (Umapathysivam et al. 2022).
Beyond the age of 60 years, skeletal muscle mass decreases and visceral lipid-filled adipose tissue increases. Unless there is a very substantial increase in fat mass, BMI tends to remain stable and may even decrease therefore not reflecting the increase in viscerally located adipose tissue. Furthermore, with advancing age, the risks (including T2D) associated with higher BMI flatten. WC which is simple to perform and reproducible when done correctly (Ross et al. 2020) provides an approximation of visceral fat and on average increases with increasing age; values 95 cm or above predict increased risk for T2D in middle-aged and older Caucasian men (Siren et al. 2012).
Outcome measures
The primary outcome of T4DM was based on an oral glucose tolerance test (OGTT). Although inherently variable, the OGTT has better sensitivity and specificity for diagnosing T2D compared with fasting glucose or with HbA1c, which at its standard cut-point has high specificity but low sensitivity (Jesudason et al. 2003). The OGTT was used to define the primary outcome in the diabetes prevention program (DPP) (Knowler et al. 2002) and remains generally accepted as the gold standard diagnostic test for T2D (Collaboration 2015). Secondary outcome measures relevant to this paper included fasting serum glucose, HbA1c, body weight, WC, body composition (fat mass, skeletal muscle mass and bone density) by dual-xray absorptiometry (DXA) and muscle strength by peak non-dominant handgrip. Haematocrit was measured and data relating to cardiovascular events collected at each clinic visit. Other outcome and safety measures have previously been described in detail (Wittert et al. 2019).
Glycaemic status: main findings and questions arising
Testosterone and prevention/remission of type 2 diabetes
The T4DM study achieved its recruitment target, enrolling 1007 men, across six centres in Australia. All were enrolled in the WW lifestyle programme and randomly allocated to be treated with either 3 monthly intramuscular TU or placebo for 2 years, making it the largest and longest RCT of testosterone treatment at the time it was done. Outcome data available for primary analysis at 2 years comprised of 443 and 413 testosterone and placebo-treated men, respectively. The primary and secondary outcomes are published (Wittert et al. 2021). Key findings and some implications relevant to translational science and future directions are detailed below.
In the testosterone compared to placebo-treated group, the proportion of participants with T2D at 2 years, based on a 2-h glucose ≤11.1mmol/L on the OGTT, was reduced by 40% (55/443, 12.4% vs 87/413, 21%). There was a 0.75 mmol/L greater decrease from baseline in the 2-h OGTT glucose concentration and a 0.17 mmol/L greater decrease in fasting glucose in the testosterone as compared to the placebo group.
Blood glucose normalised (2-h glucose <7.8 mmol/L) in 51.9% of testosterone and in 43.3% of placebo-treated men, respectively. While the difference is significant, the translational importance of over 40% of these high-risk men even if randomised to placebo treatment achieving normal glucose tolerance 2 years after the initiation of a relatively low-intensity lifestyle intervention, of proven cost-effectiveness, is a crucial finding.
Glucose response to oral challenge vs HbA1c: impact of red blood cell dynamics
In the T4DM study, the benefits of injectable testosterone undecanoate (TU) for glycaemia were shown in the 2-h GTT but were not accompanied by any change in HbA1C (Wittert et al. 2021). Previous small studies had previously reported little or no consistent benefit of TU treatment for HbA1C in studies of men without (Svartberg et al. 2008) or with T2D (Gianatti et al. 2014, Hackett et al. 2014, Ramachandran et al. 2020).
HbA1c is a function of the average blood glucose to which haemoglobin in the red blood cell (RBC) is exposed, and the lifespan of that RBC (Cohen et al. 2008). Improved glycaemia would be expected to produce a lower HbA1c so the unchanged HbA1c may reflect modest or minimal chronic glycaemic changes from testosterone treatment. However, the discordance between testosterone effects on acute (fasting glucose and 2-h OGTT) vs chronic (HbA1c) glycaemia may be due to testosterone’s erythropoietic effects influencing RBC lifespan; if testosterone increases RBC lifespan, this could increase HbA1c masking a glycaemic benefit of testosterone treatment. However, RBC lifespan does not differ significantly between males and females (Zhang et al. 2018) making changes in RBC lifespan due to ambient testosterone concentrations unlikely. Other possible explanations for the discrepancy is that testosterone treatment may abrogate the effects of endogenous inhibitors of deglycation such as fructosamine-3-kinase (Szwergold et al. 2001, Delpierre et al. 2002) or alternative deglycation mechanisms (Szwergold & Beisswenger 2003) operative in erythrocytes; however, their testosterone sensitivity has not been established. If testosterone inhibits deglycation enzymes, then HbA1c will increase. This area warrants further investigation.
Pharmacological not physiological effect of testosterone
For purposes of screening, and entry into the study, serum testosterone concentration was measured by platform chemiluminescent immunoassay (CLIA) (Wittert et al. 2019). Blood was subsequently sampled fasting in the morning prior to the first injection of TU or matched placebo and prior to each subsequent TU injection, serum testosterone concentration was measured by LC-MS/MS in batched samples at study end. Based on the CLIA data testosterone concentrations at screening were <8 nmol/L, 8–11 nmol/L and >11–14 nmol/L in approximately 20%, 40% and 40% of participants, respectively. By contrast, baseline analysis by LCMS showed that 67% of participants had serum testosterone concentrations >11 nmol/L and 41% >14 nmol/L. However, there were no significant relationships between serum testosterone concentration measured either by screening (CLIA) or baseline (LC-MS/MS) and glycaemic outcome variables. This suggests that pharmacological studies of testosterone effects on glycaemia in men without pathological hypogonadism should not employ baseline entry thresholds and should avoid the use of unreliable testosterone immunoassays.
Testosterone effects on body composition
Changes in fat and lean mass: translational considerations
In placebo-treated participants, fat mass, skeletal muscle mass and non-dominant hand grip strength decreased. In testosterone-treated participants, there was a greater decrease in fat mass and increase in skeletal muscle mass and non-dominant hand grip strength (Wittert et al. 2021). The magnitude of changes in lean and fat mass and hand grip strength are summarised in the Table 1 below.
Changes in fat mass, skeletal muscle mass and non-dominant hand grip strength after 2 years treatment with testosterone. Data from the T4DM Study (Wittert et al. 2021).
Placebo | Testosterone | Between group difference | P | |
---|---|---|---|---|
Fat mass | −1.9 kg | −4.6 kg | 2.7 | <0.0001 |
Skeletal muscle mass | −1.3 kg | +0.4 kg | 1.7 kg | <0.0001 |
Non-dominant hand grip strength | −1.3 kg | +1.7 kg | 2.19 kg | <0.0001 |
Because fat mass is positively, and skeletal muscle mass (Qadir et al. 2021) and grip strength (Li et al. 2016) are inversely, associated with the risk of T2D, we have sought to determine the extent to which changes in total fat mass (kg), abdominal fat mass (%), lean mass (kg) and non-dominant handgrip (kg) mediate the effect of testosterone treatment on glycaemia. Preliminary data (Wittert & Robledo 2022) show that some, but not all, of the testosterone effect (when coupled with lifestyle changes) is mediated via a decrease in fat mass and to some extent also by the increase in E2 concentration in accordance with the known effects of E2 on glucose metabolism in skeletal muscle documented in section 1.3. The observation that there is limited effect mediation by skeletal muscle mass and strength does not exclude non-trophic mechanisms other than E2, for example, increased GH secretion (Eakman et al. 1996), adiponectin production and GLUT-4 expression and translocation (Inada et al. 2016, Antinozzi et al. 2017) that may mediate the beneficial effects of testosterone.
Lifestyle modification programs are effective to reduce weight in obesity when adherence is high, but even under optimal circumstances, engagement is variable and adherence wanes with time (Fabricatore et al. 2009). Because sex steroid receptors are expressed in the brain, a motivational benefit of testosterone to increase adherence to the lifestyle program was proposed at the outset as a hypothetical mechanism (Wittert et al. 2019). However, the T4DM study provided no support for this motivational hypothesis as engagement in the lifestyle program during the study was not sensitive to testosterone treatment, while both groups achieved 70% sufficient physical activity at 2 years. Further we observed no mental or physical quality of life benefits and no improvement in quality of sleep (Wittert et al. 2021), findings that are consistent with data from the smaller and shorter T trials (Snyder et al. 2016)
Bone mineral density and ultrastructure: mechanistic considerations
Ageing is causally associated with decreasing bone mineral density (BMD), a process accelerated by diet-induced weight loss.
In men, testosterone both acting directly via the AR, as well as via its conversion by aromatisation to E2 to act on the ER, is required for the acquisition of peak bone mass attained at the completion of growth, as well as for the maintenance of bone mass during adulthood. Men with untreated congenital hypogonadism (e.g. congenital hypogonadotropic hypogonadism, or those with Klinefelter Syndrome) fail to achieve peak bone mass during puberty, and men with acquired pathological hypogonadism experience accelerated bone loss during adulthood (Vanderschueren et al. 2014). Small and ethically constrained, uncontrolled studies report that testosterone treatment of men with pathological hypogonadism reduces bone turnover markers and increases bone density (Katznelson et al. 1996, Snyder et al. 2000). The role of sex steroids in skeletal health in unselected older men without pathological hypogonadism is less clear. Population-based studies report that circulating sex steroids are inversely associated with accelerated bone loss, both if assessed by conventional DXA measuring areal bone mineral density (aBMD) and by high resolution–-peripheral quantitative computed tomography (HR-pqCT) measuring volumetric BMD (vBMD) and parameters of bone microarchitecture (David et al. 2022). Moreover, circulating sex steroids are inversely associated with fracture risk (Vanderschueren et al. 2014, David et al. 2022). However, such observational studies cannot ascribe causality.
Testosterone effects on bone structure and strength
The effects of testosterone treatment on skeletal outcomes in men without pathological hypogonadism have only come into focus more recently. An early meta-analysis of RCTs enrolling 264 men aged 60–76 years with a baseline serum testosterone of 10–15 nmol/L receiving transdermal or intramuscular testosterone (short acting esters) reported that testosterone treatment, compared to placebo, increased aBMD at the lumbar spine by 3.7%, without significant effect on the femur (Isidori et al. 2005). A more recent large placebo-controlled study of men aged 60 years or older (n = 211) with a baseline testosterone of <9.54 nmol/L showed that daily transdermal testosterone treatment over one year increased vBMD (by quantitative CT (QCT)) predominantly at the trabecular spine (+6.8%) with lesser effects at peripheral spine (+2.9%) (Snyder et al. 2017). Studies using higher resolution HR-pqQCT (82 µm vs ~500 µm with standard QCT) allow assessment cortical and trabecular bone microarchitecture which predicts fracture risk independent of aBMD and the FRAX score, a validated aBMD-independent predictor of fracture risk incorporating clinical risk factors for fracture (Samelson et al. 2019). However, RCTs assessing the effects of testosterone treatment on bone microarchitecture defined by HR-pqQCT in men were lacking. Therefore, we conducted testosterone for bone (T4Bone), a planned sub-study of testosterone for diabetes mellitus (T4DM). In T4Bone over 24 months, testosterone treatment, compared to placebo, increased total vBMD both at the tibia and radius (Fui et al. 2021). vBMD increased predominantly in cortical bone at both sites with potent treatment effect sizes ranging from 2.9% to 3.1%. However, effects on trabecular microarchitecture were minor. Testosterone also significantly increased aBMD at the lumbar spine (+3.3%) and the total hip (+1.9%) (Fui et al. 2021). The predominant effects of testosterone treatment on cortical bone were consistent with observational HR-pqQCT studies in older men with lower sex steroid concentrations displaying deficits predominantly in cortical, rather than trabecular bone (Argoud et al. 2014). Moreover, men commencing androgen deprivation therapy for prostate cancer, which leads to severe sex steroid deficiency experience predominant loss of cortical, rather than trabecular bone (Hamilton et al. 2010).
Testosterone effects on cortical vs trabecular bone
None of the RCTs evaluating the effects of testosterone treatment in older men has been powered for fracture outcomes. Of note, however, the findings of T4DM that testosterone induced increases in cortical bone, which comprises 80% of the human skeleton, suggests that it might. Preclinical studies report that the loss of cortical bone compromises bone strength to a greater extent than the loss of trabecular bone (Vanderschueren et al. 2014), and in observational HR-pqQCT studies in men, deficits in cortical parameters were more consistently associated with fractures than trabecular parameters (Szulc et al. 2011, Fink et al. 2018). Moreover, the effect sizes, ranging from 1.3% to 3.1% for total and cortical vBMD in T4DM compare favourably with those reported for conventional osteoporotic drug treatments. In postmenopausal women with low BMD, antiresorptive drug therapy (using bisphosphonates or denosumab which have proven anti-fracture efficacy) over 12–24 months produced effect sizes on HR-pqQCT indices ranging from 0.3% to 3.8% (Burghardt et al. 2010). Of note men participating T4Bone had a mean baseline serum testosterone of 13.6 nmol/L; there was no evident relationship of baseline serum testosterone or E2 concentrations and bone microarchitecture.
Mechanistic aspects of testosterone action on bone
Testosterone treatment in T4DM was pharmacological and not designed to explore the mechanisms by which testosterone treatment increased vBMD. It remains unclear first how much testosterone effect is direct i.e. mediated by sex steroid receptors in bone as opposed to effects other tissues such as muscle or fat, and secondly, how much of the testosterone effect is mediated via the AR rather than via aromatisation to E2 and the ER.
Regarding the first question, both AR and ER are expressed in bone cells, and targeted deletions of both the AR and the ERs in bone cells (osteoclasts, osteoblasts and osteocytes) have reported skeletal deficits in mice (Vanderschueren et al. 2014) and congruent findings are reported in men with comparable genetic inactivation. Therefore, the actions of sex steroid on skeletal health are likely to be direct, supported by observations in T4Bone that testosterone treatment reduced bone remodelling. However, in addition, testosterone also increases muscle mass (Wittert et al. 2021), and it is possible that increased muscle mass, although modest, promotes skeletal health either by exerting loading on the skeleton and/or by secretion of bone-anabolic myokines. In addition, increased muscle mass may reduce falls risk, a common proximal cause of fragility fractures. Moreover, in T4DM, testosterone reduced fat mass and WC and fat mass (especially visceral fat), all of which are negatively associated with bone mass (Gilsanz et al. 2009). The mechanisms remain unclear but may be via secretion of osteo-catabolic adipo-cytokines and/or associated insulin resistance (Vanderschueren et al. 2014). These metabolically favourable changes in body composition are, at least in part, responsible for the reduced T2D risk observed in T4DM. Insulin resistance and T2D have been associated with increased fracture risk, although precise mechanisms are unclear (Khosla et al. 2021). Therefore, it is possible that testosterone treatment may improve skeletal health, at least in part indirectly via effects on muscle and fat including possibly glucose metabolism. However, future mechanistic and clinical studies are necessary to test these hypotheses.
Androgens promote periosteal apposition to increase bone size in men, explaining why men generally have wider long bones than women. However, regarding the second question, in older men, some observational studies suggest that skeletal outcomes (reduced bone density, impaired microarchitecture and increased fracture risk) are more closely related to reductions in circulating E2, rather than in testosterone (Russell & Grossmann 2019). Experimental studies among men receiving GnRH analogue therapy to suppress gonadal steroids with testosterone addback with or without an aromatase inhibitor (to suppress aromatisation of the administered testosterone to E2) have suggested indirectly that E2 is important for maintaining bone architecture (Finkelstein et al. 2016). However, these studies have inferred E2 effects by its drug-induced absence, rather that assessing its effect directly. To directly assess the effects of E2 on skeletal health in men, a more recent RCT enrolled older men receiving androgen deprivation therapy for prostate cancer, which reduces both circulating testosterone and E2 to castrate levels (Russell et al. 2022). In this RCT, E2 treatment, compared to placebo improved bone microarchitecture and bone density, thus providing direct evidence that E2 can improve skeletal health in men in the absence of testosterone (Russell et al. 2022). From a clinical perspective, this supports the role of testosterone treatment with its full spectrum of androgen actions (rather than non-aromatisable DHT or synthetic androgens) as the prime mode of testosterone replacement in men with organic hypogonadism. Of note, the findings from T4Bone do not endorse testosterone treatment to improve skeletal health in men without pathological hypogonadism. This is because whether testosterone treatment improves patient important health outcomes (such as fractures) remains unknown, and the long-term risks of testosterone treatment are insufficiently explored.
Questions about safety: erythrocytosis and cardiovascular effects
Erythrocytosis: mechanistic considerations
Testosterone has a direct and dose-related effect on bone marrow to increase erythropoiesis (Coviello et al. 2008). Mechanisms of testosterone-induced erythropoiesis include iron mobilisation and incorporation into RBCs, increased haemoglobin synthesis and RBC production (Bachman et al. 2014, Hennigar et al. 2020). Increased iron flux is mediated by changes in erythropoietin (EPO), hepcidin and ferroportin (Roth et al. 2019). The primary stimulus to renal production of the cytokine, EPO, is hypoxia (Bachman et al. 2014). Hepcidin inhibits absorption of iron in the gut by binding to the iron transporter ferroportin and resulting in its degradation thus preventing iron transport across the basolateral membrane of the enterocytes (Atanasiu et al. 2007). Testosterone induces a sustained suppression of hepcidin (Guo et al. 2013).
Murine data suggest that EPO upregulation and hepcidin suppression operate both jointly and independently (Guo et al. 2013, Guo et al. 2020). Although one clinical trial observed no significant increase in serum EPO during testosterone treatment (Maggio et al. 2013), testosterone produces a decrease in the threshold for EPO release (Bachman et al. 2014).
E2 increases haemopoietic stem cell proliferation and survival in vitro (Calado et al. 2009) and high endogenous E2 following IVF hyperstimulation is reported to inhibit hepcidin in women (Lehtihet et al. 2016). However, testosterone alone is sufficient to maintain red cell mass and to increase net erythropoiesis with supra physiological doses (Rochira et al. 2009).
Erythrocytosis: as an adverse event in clinical trials
Erythrocytosis (haematocrit >0.54) is one of the most common adverse effects in clinical trials of testosterone therapy. Advancing age is associated with a higher risk that cannot be explained by differential effects on EPO or soluble transferrin receptor (Coviello 2008). Erythrocytosis was a particularly prominent adverse event in T4DM and occurred in 22% (106) of men treated with testosterone but only 1% (6) of those on placebo. Once the effect of dehydration was eliminated by a second non-fasting test, only 23 men were withdrawn from treatment because of the finding of elevated haematocrit on two occasions. Nevertheless this rate was substantially higher than generally reported (Warren & Grossmann 2022) possibly because we did not exclude men with obstructive sleep apnoea (OSA). Severe OSA leads to a modest increase in haematocrit (Rha et al. 2022) and the best characterised risk factor for androgen-induced erythrocytosis is elevated baseline haemoglobin/haematocrit (Idan et al. 2010) or trough serum testosterone in testosterone-treated men (Ip et al. 2010). The prevalence of moderate or severe OSA in a randomly selected cohort of community-dwelling men aged 40 years and over is approximately 25%, with half having severe OSA (Adams et al. 2016). The prevalence is higher with increasing age and obesity and is over 50% in the presence of T2D (Andayeshgar et al. 2022). At enrolment, 14% of men were using CPAP and this remained relatively constant for the study duration (unpublished data). Therefore, it is possible that the relatively high prevalence of increased erythrocytosis in T4DM is attributable to the combined effects of testosterone and overnight hypoxia.
Another intriguing possibility is an interaction between testosterone and somatic mutations, which occur with increasing frequency over the ageing process, in genes regulating erythrocytosis. For example, somatic mutations of the Janus kinase 2 (JAK2) gene and SH2B adaptor protein 3 (SH2B3) gene cause most cases of primary-acquired erythrocytosis (Kristan et al. 2021). Somatic mutations indicative of clonal haematopoiesis in individuals with primary erythrocytosis have also been found in two homologous genes BCOR and BCORL1, which play key roles in haematopoiesis (Wouters et al. 2020). Moreover, erythrocytosis with clonal haematopoiesis has an independent association with cardiovascular morbidity and mortality (Wouters et al. 2020). This is an area of active investigation we are currently pursuing.
Implications of a high haematocrit
The relationship between high haematocrit (49–70%) and cardiovascular risk remains unresolved, with some suggesting an increase in risk (Gagnon et al. 1994, Kunnas et al. 2009) and others not (Brown et al. 2001, Puddu et al. 2002). The data from these studies cannot necessarily be used to infer the risk of an increase in haematocrit induced by treatment with testosterone. In a recent study, 5842 men who developed polycythaemia (haematocrit ≥52%) after commencing treatment with testosterone had a higher risk of major adverse cardiovascular events/venous thromboembolism (MACE/VTE) (number of outcomes: 301, 5.15%) in 1 year than 5842 matched men who did not develop polycythaemia (number of outcomes 226, 3.87%) (Ory et al. 2022).
In men treated with testosterone, aggressive management of cardiometabolic co-morbidities and risk factors including OSA decreases the risk of erythrocytosis and improves overall symptoms (Farber et al. 2020).
Cardiovascular effects: mechanistic studies
There is ongoing debate over the cardiovascular effects of testosterone. Mechanistic studies have been conducted using in vitro and animal models of atherogenesis. In an experimental study using aortic ring segments cultured in vitro after endothelial denudation, testosterone treatment inhibited neointimal plaque development, and effect associated with increased expression of androgen receptor mRNA in treated segments (Hanke et al. 2001). This concept was supported by an in vivo study in male miniature pigs, where neointima formation in response to induced coronary artery injury was increased in castrated animals compared to intact controls, and effect abrogated by testosterone treatment (Tharp et al. 2009). Testosterone treatment induces apoptosis in vascular smooth muscle cells, another mechanism by which atherosclerotic plaque growth or stability may be affected (Lopes et al. 2014). Of note, testosterone-augmented angiogenesis, a process central to cardiovascular repair/regeneration, in male but not female endothelial cells (Sieveking et al. 2010). Androgen receptor gene knockdown abrogated these effects in male endothelial cells, whereas androgen receptor overexpression in female endothelial cells conferred androgen responsiveness for angiogenesis. Testosterone treatment also increased cholesterol efflux in a monocyte-macrophage cell line (Kilby et al. 2021). Thus, these experimental data suggest an effect of testosterone to modulate mechanisms relevant to atherogenesis.
In castrated male rabbits fed a cholesterol-rich diet, aortic atherosclerosis was highest in placebo-treated rabbits and lowest in testosterone-treated rabbits, as well as in untreated, sham-operated controls (Alexandersen et al. 1999). In cholesterol-fed ovariectomised female and castrated male rabbits, oestrogen in female rabbits and testosterone in male rabbits reduced aortic plaque formation (Bruck et al. 1997, Alexandersen et al. 1999). Similar results were reported in male miniature pigs fed a high-fat and high-cholesterol diet where castration resulted in increased atherosclerotic plaque and intima-media thickness (Deng et al. 2021). In castrated atherosclerosis-prone mice, the anti-atherosclerosis of the effect of testosterone treatment was dependent of aromatisation to E2 (Nathan et al. 2001). Testosterone treatment reduced atherosclerotic lesion area to a greater extent in castrated wild type compared to AR knockout mice (Bourghardt et al. 2010). Testosterone treatment also attenuated fatty streak formation in mice with a non-functioning androgen receptor, suggesting effects independent of the classical androgen receptor (Nettleship et al. 2007). Taken together, these studies are consistent with an effect of testosterone, either directly or via aromatisation to E2, to regulate atherosclerotic plaque formation. However, testosterone treatment increased arterial calcification in apolipoprotein E knockout mice, with a lesser effect of the non-aromatisable androgen dihydrotestosterone (McRobb et al. 2009). Reconciling these experimental findings with the earlier onset and greater severity of atherosclerosis in men than in women remains an ongoing challenge.
Cardiovascular effects: clinical studies
The associations of endogenous testosterone concentrations with the incidence of cardiovascular events in prospective cohort studies and the risk of cardiovascular adverse events in interventional trials of testosterone therapy have been previously reviewed (seeYeap et al. 2022). Several recent studies of note are highlighted as follows. In a large prospective observational cohort study of 210,700 men aged 40–69 years with 8790 cardiovascular events occurring over 9 years follow-up, baseline testosterone concentrations were not associated with the incidence of myocardial infarction, stroke, heart failure or major cardiovascular adverse events (Yeap et al. 2022). An association of lower testosterone concentrations with higher risk of stroke, but not risk of myocardial infarction, has been reported in men aged ≥70 years (Yeap et al. 2014). In a clinical study of overweight men, testosterone treatment did not improve vascular function assessed using flow-mediated dilation nor did testosterone add to the benefits of exercise on vascular health (Chasland et al. 2021). A sub-study from the Testosterone Trials (T-Trials) of 138 men reported an increase in coronary atheroma measured as non-calcified plaque volume using coronary computed tomography angiography in testosterone-treated men over 12 months, insufficient duration to gauge cardiovascular consequences (Budoff et al. 2017). The authors concluded that larger and longer studies were needed to understand the clinical implications of this finding.
An RCT of 6 months transdermal testosterone or placebo gel treatment to increase lower limb strength and physical function in 209 men aged ≥65 years with mobility limitations was stopped prematurely due to excess adverse cardiovascular events in testosterone-treated men (Basaria et al. 2010). However, another trial in a similar population of intermediate-frail or frail men aged ≥65 years randomised 274 men to testosterone gel or placebo treatment for 6 months, finding an improvement in lean mass, knee extension strength and physical function, with no signal for adverse cardiovascular events (Srinivas-Shankar et al. 2010). A registry study of older men treated with testosterone for up to 10 years reported improvements in cardiometabolic risk factors; however, the limitations of such uncontrolled studies need to be considered (Yassin et al. 2016).
Cardiovascular adverse events in T4DM
In T4DM, cardiovascular adverse events were comparably distributed between testosterone and placebo-treated men; 13/503 men in the placebo arm, and 7/504 in the testosterone arm experienced an ischemic heart disease adverse event over the two-year trial duration. When major adverse cardiovascular events (ischemic heart disease, cerebrovascular disease and cardiovascular deaths) were considered, there were 17/503 events in the placebo arm and 12/504 in the testosterone arm (Wittert et al. 2021). This is in keeping with the T-Trials where 788 men were randomly allocated testosterone or placebo for a year, and 7 men in each arm of the study died from cardiovascular causes or had a myocardial infarction or stroke (Snyder et al. 2016). However, none of these trials was long enough to definitively assess adverse cardiovascular events. A recent systematic review and meta-analysis of RCTs including individual patient data, mostly of 12-month duration found no evidence of excess cardiovascular or cerebrovascular adverse events in testosterone-treated men (Hudson et al. 2022). These and the 2-year T4DM data provide some assurance regarding short-term risk; however, much longer-term follow-up over decades is required for a full safety evaluation. Additional information will be forthcoming when the ‘Testosterone Replacement therapy for Assessment of long-term Vascular Events and efficacy ResponSE in hypogonadal men’ (TRAVERSE) study, a Federal Drug Agency-mandated 5-year cardiovascular safety trial of testosterone in middle- to older-aged men with multiple risk factors or pre-existing cardiovascular disease is completed (Bhasin et al. 2022).
Recovery of the hypothalamo–pituitary–testicular axis
Another potentially detrimental outcome arising from prolonged injectable testosterone treatment even at standard testosterone replacement doses is that it creates sustained suppression of the HPT axis. The slow but eventually complete recovery takes up to 12 months after cessation of testosterone treatment (Handelsman et al. 2022) is comparable with the time course of recovery from androgen abuse-induced HPT axis suppression (Shankara-Narayana et al. 2020). The congruence of these findings suggests that the recovery from androgen-induced HPT axis suppression is primarily related to duration since ceasing androgen intake rather than the doses or patterns of androgens used. Prolonged, slow recovery of the HPT axis can cause symptomatic androgen deficiency withdrawal symptoms during recovery and potential androgen dependence. These pose a practical limitation on the wider application of such injectable testosterone treatment, especially in men without pathologic hypogonadism. Whether shorter treatment duration might facilitate faster HPT axis recovery remains an interesting question; however, the minimal duration of injectable testosterone treatment to maintain similar improvements in acute (if not chronic) glycaemia shown in T4DM remains unknown, and careful investigation would be warranted to determine the benefits and risks of shorter duration of testosterone treatment on iatrogenic HPT axis suppression and recovery. The intermediary molecular mechanism of the HPT axis suppression by exogenous testosterone, and whether it can be modified independent of systemic androgen effects, warrants further investigation.
Conclusions
The primary finding was that over and above the effects of a lifestyle intervention, men treated with testosterone had a 40% lower risk of T2D diagnosis (by OGTT) after 2 years relative to placebo-treated men. Further, at 2 years, 40% of men in the placebo group and 50% of men in the testosterone group normalised glycaemia. Yet HbA1c was not reduced so the magnitude of the chronic glycaemic changes requires further long-term evaluation before wider application of pharmacological testosterone treatment to prevent T2DM is justified.
Testosterone treatment appears to have primarily mediated its effect by inducing metabolically favourable changes in body composition. There were no motivational or quality of life benefits from testosterone treatment.
T4DM illustrates how concepts gained from basic and mechanistic studies are translatable via the platform of a placebo-controlled RCT leading to new information and ongoing studies to answer questions generated. These include the importance of central adiposity, and the role of testosterone and E2 in regulation of adipose and skeletal muscle biology. Similarly, the observed improvements in skeletal microarchitecture and BMD are grounded in the role of androgen and OR in bone biology. Whether the benefits on bone are durable and clinically important require further investigation.
While mechanistic studies have explored the potential role of testosterone in mechanisms relevant to atherogenesis, in T4DM there was no signal of either cardiovascular risk or benefit. The risk of an unacceptably high haematocrit may be preventable by screening for and treating sleep apnoea, but other mechanisms remain to be investigated. Suppression of the endogenous HPT axis may last for up to 12 months, and therefore ill-conceived or uncontrolled trials of testosterone treatment may do more harm than good.
From a translational perspective, building on the results of T4DM, data from trials that are adequately powered to examine durability of treatment effect and offset, prevention of diabetes-related complications, fragility fractures, cardiovascular events and mortality would be important. Further studies are also needed to better define mechanisms and magnitude of testosterone effects for glycaemic and skeletal benefit.
The most important, and readily translatable, outcome of T4DM is that, in line with prior observations, T2D is preventable. Lifestyle changes are a logical first step. The health of men is likely to benefit most not from the from the measurement of serum testosterone and prescription of testosterone treatment but from careful risk assessment and an overall holistic approach to care.
Declaration of interest
GW has received research funding from Lawley Pharmaceuticals, Bayer, Weight Watchers and Lilly, speaker’s honoraria from Bayer Pharma and Besins Healthcare, and consulting fees from Bayer Pharma. MG has received research funding from Bayer Pharma, Novartis, Weight Watchers, Otsuka and Lilly and speaker’s honoraria from Bayer Pharma and Besins Healthcare. BBY has received speaker honoraria and conference support from Bayer, Lilly and Besins, held advisory roles for Lilly, Besins, Ferring and Lawley Pharmaceuticals, and received research support from Bayer, Lilly and Lawley Pharmaceuticals. DJH has received institutional funding for investigator-initiated pharmacology research from Lawley Pharmaceuticals and Besins Healthcare and has served as an expert witness in professional standards and antidoping tribunals and testosterone litigation.
Funding
NHMRC Project grant 1030123, Bayer, WW, Eli Lilly.
References
Adams R, Appleton S, Taylor A, McEvoy D & & Wittert G 2016 Are the ICSD-3 criteria for sleep apnoea syndrome too inclusive? Lancet. Respiratory Medicine 4 e19–e20. (https://doi.org/10.1016/S2213-2600(1600109-0)
Alexandersen P, Haarbo J, Byrjalsen I, Lawaetz H & & Christiansen C 1999 Natural androgens inhibit male atherosclerosis: a study in castrated, cholesterol-fed rabbits. Circulation Research 84 813–819. (https://doi.org/10.1161/01.res.84.7.813)
Andayeshgar B, Janatolmakan M, Soroush A, Azizi SM & & Khatony A 2022 The prevalence of obstructive sleep apnea in patients with type 2 diabetes: a systematic review and meta-analysis. Sleep Science and Practice 6 6. (https://doi.org/10.1186/s41606-022-00074-w)
Antinozzi C, Marampon F, Corinaldesi C, Vicini E, Sgro P, Vannelli GB, Lenzi A, Crescioli C & & Di Luigi L 2017 Testosterone insulin-like effects: an in vitro study on the short-term metabolic effects of testosterone in human skeletal muscle cells. Journal of Endocrinological Investigation 40 1133–1143. (https://doi.org/10.1007/s40618-017-0686-y)
Argoud T, Boutroy S, Claustrat B, Chapurlat R & & Szulc P 2014 Association between sex steroid levels and bone microarchitecture in men: the STRAMBO study. Journal of Clinical Endocrinology and Metabolism 99 1400–1410. (https://doi.org/10.1210/jc.2013-3233)
Atanasiu V, Manolescu B & & Stoian I 2007 Hepcidin--central regulator of iron metabolism. European Journal of Haematology 78 1–10. (https://doi.org/10.1111/j.1600-0609.2006.00772.x)
Atlantis E, Martin SA, Haren MT, Taylor AW & & Wittert GA 2009 Members of the Florey Adelaide male ageing study. Inverse associations between muscle mass, strength, and the metabolic syndrome. Metabolism 58 1013–1022. (https://doi.org/10.1016/j.metabol.2009.02.027)
Atlantis E, Fahey P, Martin S, O'Loughlin P, Taylor AW, Adams RJ, Shi Z & & Wittert G 2016 Predictive value of serum testosterone for type 2 diabetes risk assessment in men. BMC Endocrine Disorders 16 26. (https://doi.org/10.1186/s12902-016-0109-7)
Bachman E, Travison TG, Basaria S, Davda MN, Guo W, Li M, Connor Westfall J, Bae H, Gordeuk V & & Bhasin S 2014 Testosterone induces erythrocytosis via increased erythropoietin and suppressed hepcidin: evidence for a new erythropoietin/hemoglobin set point. Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 69 725–735. (https://doi.org/10.1093/gerona/glt154)
Basaria S, Coviello AD, Travison TG, Storer TW, Farwell WR, Jette AM, Eder R, Tennstedt S, Ulloor J, Zhang A, et al.2010 Adverse events associated with testosterone administration. New England Journal of Medicine 363 109–122. (https://doi.org/10.1056/NEJMoa1000485)
Bhasin S, Lincoff AM, Basaria S, Bauer DC, Boden WE, Cunningham GR, Davey D, Dubcenco E, Fukumoto S, Garcia M, et al.2022 Effects of long-term testosterone treatment on cardiovascular outcomes in men with hypogonadism: rationale and design of the TRAVERSE study. American Heart Journal 245 41–50. (https://doi.org/10.1016/j.ahj.2021.11.016)
Bhasin S, Woodhouse L, Casaburi R, Singh AB, Bhasin D, Berman N, Chen X, Yarasheski KE, Magliano L, Dzekov C, et al.2001 Testosterone dose-response relationships in healthy young men. American Journal of Physiology. Endocrinology and Metabolism 281 E1172–E1181. (https://doi.org/10.1152/ajpendo.2001.281.6.E1172)
Bourghardt J, Wilhelmson AS, Alexanderson C, De Gendt K, Verhoeven G, Krettek A, Ohlsson C & & Tivesten A 2010 Androgen receptor-dependent and independent atheroprotection by testosterone in male mice. Endocrinology 151 5428–5437. (https://doi.org/10.1210/en.2010-0663)
Brand JS, Rovers MM, Yeap BB, Schneider HJ, Tuomainen TP, Haring R, Corona G, Onat A, Maggio M, Bouchard C, et al.2014 Testosterone, sex hormone-binding globulin and the metabolic syndrome in men: an individual participant data meta-analysis of observational studies. PLoS One 9 e100409. (https://doi.org/10.1371/journal.pone.0100409)
Brown DW, Giles WH & & Croft JB 2001 Hematocrit and the risk of coronary heart disease mortality. American Heart Journal 142 657–663. (https://doi.org/10.1067/mhj.2001.118467)
Bruck B, Brehme U, Gugel N, Hanke S, Finking G, Lutz C, Benda N, Schmahl FW, Haasis R & & Hanke H 1997 Gender-specific differences in the effects of testosterone and estrogen on the development of atherosclerosis in rabbits. Arteriosclerosis, Thrombosis, and Vascular Biology 17 2192–2199. (https://doi.org/10.1161/01.atv.17.10.2192)
Budoff MJ, Ellenberg SS, Lewis CE, Mohler ER 3rd, Wenger NK, Bhasin S, Barrett-Connor E, Swerdloff RS, Stephens-Shields A, Cauley JA, et al.2017 Testosterone treatment and coronary artery plaque volume in older men with low testosterone. JAMA 317 708–716. (https://doi.org/10.1001/jama.2016.21043)
Burghardt AJ, Kazakia GJ, Sode M, de Papp AE, Link TM & & Majumdar S 2010 A longitudinal HR-pQCT study of alendronate treatment in postmenopausal women with low bone density: relations among density, cortical and trabecular microarchitecture, biomechanics, and bone turnover. Journal of Bone and Mineral Research 25 2558–2571. (https://doi.org/10.1002/jbmr.157)
Burnett-Bowie SA, Roupenian KC, Dere ME, Lee H & & Leder BZ 2009 Effects of aromatase inhibition in hypogonadal older men: a randomized, double-blind, placebo-controlled trial. Clinical Endocrinology 70 116–123. (https://doi.org/10.1111/j.1365-2265.2008.03327.x)
Calado RT, Yewdell WT, Wilkerson KL, Regal JA, Kajigaya S, Stratakis CA & & Young NS 2009 Sex hormones, acting on the tert gene, increase telomerase activity in human primary hematopoietic cells. Blood 114 2236–2243. (https://doi.org/10.1182/blood-2008-09-178871)
Chasland LC, Naylor LH, Yeap BB, Maiorana AJ & & Green DJ 2021 Testosterone and exercise in middle-to-older aged men: combined and independent effects on vascular function. Hypertension 77 1095–1105. (https://doi.org/10.1161/HYPERTENSIONAHA.120.16411)
Cohen RM, Franco RS, Khera PK, Smith EP, Lindsell CJ, Ciraolo PJ, Palascak MB & & Joiner CH 2008 Red cell life span heterogeneity in hematologically normal people is sufficient to alter HbA1c. Blood 112 4284–4291. (https://doi.org/10.1182/blood-2008-04-154112)
Collaboration NCDRF 2015 Effects of diabetes definition on global surveillance of diabetes prevalence and diagnosis: a pooled analysis of 96 population-based studies with 331,288 participants. Lancet. Diabetes and Endocrinology 3 624–637. (https://doi.org/10.1016/S2213-8587(1500129-1)
Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB & & Bhasin S 2008 Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. Journal of Clinical Endocrinology and Metabolism 93 914–919. (https://doi.org/10.1210/jc.2007-1692)
David K, Narinx N, Antonio L, Evenepoel P, Claessens F, Decallonne B & & Vanderschueren D 2022 Bone health in ageing men. Reviews in Endocrine and Metabolic Disorders 23 1173–1208. (https://doi.org/10.1007/s11154-022-09738-5)
Delpierre G, Collard F, Fortpied J & & Van Schaftingen E 2002 Fructosamine 3-kinase is involved in an intracellular deglycation pathway in human erythrocytes. Biochemical Journal 365 801–808. (https://doi.org/10.1042/BJ20020325)
Deng L, Fu D, Zhu L, Huang J, Ling Y & & Cai Z 2021 Testosterone deficiency accelerates early stage atherosclerosis in miniature pigs fed a high-fat and high-cholesterol diet: urine (1)H NMR metabolomics targeted analysis. Molecular and Cellular Biochemistry 476 1245–1255. (https://doi.org/10.1007/s11010-020-03987-1)
Eakman GD, Dallas JS, Ponder SW & & Keenan BS 1996 The effects of testosterone and dihydrotestosterone on hypothalamic regulation of growth hormone secretion. Journal of Clinical Endocrinology and Metabolism 81 1217–1223. (https://doi.org/10.1210/jcem.81.3.8772602)
Fabricatore AN, Wadden TA, Moore RH, Butryn ML, Gravallese EA, Erondu NE, Heymsfield SB & & Nguyen AM 2009 Attrition from randomized controlled trials of pharmacological weight loss agents: a systematic review and analysis. Obesity Reviews 10 333–341. (https://doi.org/10.1111/j.1467-789X.2009.00567.x)
Farber NJ, Vij SC & & Shoskes DA 2020 Failure of testosterone replacement therapy to improve symptoms correlates with burden of systemic conditions. Translational Andrology and Urology 9 1108–1112. (https://doi.org/10.21037/tau-19-848)
Ferrando AA, Sheffield-Moore M, Yeckel CW, Gilkison C, Jiang J, Achacosa A, Lieberman SA, Tipton K, Wolfe RR & & Urban RJ 2002 Testosterone administration to older men improves muscle function: molecular and physiological mechanisms. American Journal of Physiology. Endocrinology and Metabolism 282 E601–E607. (https://doi.org/10.1152/ajpendo.00362.2001)
Fink HA, Langsetmo L, Vo TN, Orwoll ES, Schousboe JT, & Ensrud KE for the Osteoporotic Fractures in Men (MrOS) Study Group 2018 Association of high-resolution peripheral quantitative computed tomography (HR-pQCT) bone microarchitectural parameters with previous clinical fracture in older men: the Osteoporotic Fractures in Men (MrOS) study. Bone 113 49–56. (https://doi.org/10.1016/j.bone.2018.05.005)
Finkelstein JS, Lee H, Burnett-Bowie SA, Pallais JC, Yu EW, Borges LF, Jones BF, Barry CV, Wulczyn KE, Thomas BJ, et al.2013 Gonadal steroids and body composition, strength, and sexual function in men. New England Journal of Medicine 369 1011–1022. (https://doi.org/10.1056/NEJMoa1206168)
Finkelstein JS, Lee H, Leder BZ, Burnett-Bowie SA, Goldstein DW, Hahn CW, Hirsch SC, Linker A, Perros N, Servais AB, et al.2016 Gonadal steroid-dependent effects on bone turnover and bone mineral density in men. Journal of Clinical Investigation 126 1114–1125. (https://doi.org/10.1172/JCI84137)
Fui NT, Hoermann R, Bracken K, Handelsman DJ, Inder WJ, Stuckey BGA, Yeap BB, Ghasem-Zadeh A, Robledo KP, Jesudason D, et al.2021 Effect of Testosterone Treatment on Bone Microarchitecture and Bone Mineral Density in Men: a 2-Year RCT. The Journal of Clinical Endocrinology & Metabolism 106 e3143–e3158.
Fuller NR, Colagiuri S, Schofield D, Olson AD, Shrestha R, Holzapfel C, Wolfenstetter SB, Holle R, Ahern AL, Hauner H, et al.2013 A within-trial cost-effectiveness analysis of primary care referral to a commercial provider for weight loss treatment, relative to standard care--an international randomised controlled trial. International Journal of Obesity 37 828–834. (https://doi.org/10.1038/ijo.2012.139)
Gagnon DR, Zhang TJ, Brand FN & & Kannel WB 1994 Hematocrit and the risk of cardiovascular disease--the Framingham study: a 34-year follow-up. American Heart Journal 127 674–682. (https://doi.org/10.1016/0002-8703(9490679-3)
Gianatti EJ, Dupuis P, Hoermann R, Strauss BJ, Wentworth JM, Zajac JD & & Grossmann M 2014 Effect of testosterone treatment on glucose metabolism in men with type 2 diabetes: a randomized controlled trial. Diabetes Care 37 2098–2107. (https://doi.org/10.2337/dc13-2845)
Gilsanz V, Chalfant J, Mo AO, Lee DC, Dorey FJ & & Mittelman SD 2009 Reciprocal relations of subcutaneous and visceral fat to bone structure and strength. Journal of Clinical Endocrinology and Metabolism 94 3387–3393. (https://doi.org/10.1210/jc.2008-2422)
Gleicher S, Daugherty M, Ferry E & & Byler T 2020 Looking beyond hypogonadism: association between low testosterone and metabolic syndrome in men 20–59 years. International Urology and Nephrology 52 2237–2244. (https://doi.org/10.1007/s11255-020-02557-0)
Grossmann M 2018 Hypogonadism and male obesity: focus on unresolved questions. Clinical Endocrinology 89 11–21. (https://doi.org/10.1111/cen.13723)
Guo W, Bachman E, Li M, Roy CN, Blusztajn J, Wong S, Chan SY, Serra C, Jasuja R, Travison TG, et al.2013 Testosterone administration inhibits hepcidin transcription and is associated with increased iron incorporation into red blood cells. Aging Cell 12 280–291. (https://doi.org/10.1111/acel.12052)
Guo W, Schmidt PJ, Fleming MD & & Bhasin S 2020 Hepcidin is not essential for mediating testosterone's effects on erythropoiesis. Andrology 8 82–90. (https://doi.org/10.1111/andr.12622)
Hackett G, Cole N, Bhartia M, Kennedy D, Raju J, Wilkinson P & BLAST Study Group 2014 Testosterone replacement therapy improves metabolic parameters in hypogonadal men with type 2 diabetes but not in men with coexisting depression: the BLAST study. Journal of Sexual Medicine 11 840–856. (https://doi.org/10.1111/jsm.12404)
Hamilton EJ, Ghasem-Zadeh A, Gianatti E, Lim-Joon D, Bolton D, Zebaze R, Seeman E, Zajac JD & & Grossmann M 2010 Structural decay of bone microarchitecture in men with prostate cancer treated with androgen deprivation therapy. Journal of Clinical Endocrinology and Metabolism 95 E456–E463. (https://doi.org/10.1210/jc.2010-0902)
Handelsman DJ, Desai R, Conway AJ, Shankara-Narayana N, Stuckey BGA, Inder WJ, Grossmann M, Yeap BB, Jesudason D, Ly LP, et al.2022 Recovery of male reproductive endocrine function after ceasing prolonged testosterone undecanoate injections. European Journal of Endocrinology 186 307–318. (https://doi.org/10.1530/EJE-21-0608)
Handelsman DJ, Yeap B, Flicker L, Martin S, Wittert GA & & Ly LP 2015 Age-specific population centiles for androgen status in men. European Journal of Endocrinology 173 809–817. (https://doi.org/10.1530/EJE-15-0380)
Handelsman DJ 2000 Androgen physiology, pharmacology, use and misuse. In Endotext. Ed. Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, et al.South Dartmouth , MA: Endotext.
Hanke H, Lenz C, Hess B, Spindler KD & & Weidemann W 2001 Effect of testosterone on plaque development and androgen receptor expression in the arterial vessel wall. Circulation 103 1382–1385. (https://doi.org/10.1161/01.cir.103.10.1382)
Hennigar SR, Berryman CE, Harris MN, Karl JP, Lieberman HR, McClung JP, Rood JC & & Pasiakos SM 2020 Testosterone administration during energy deficit suppresses hepcidin and increases iron availability for erythropoiesis. Journal of Clinical Endocrinology and Metabolism 105. (https://doi.org/10.1210/clinem/dgz316)
Herbst KL & & Bhasin S 2004 Testosterone action on skeletal muscle. Current opinion in clinical nutrition and metabolic care7271–7. (https://doi.org/10.1097/00075197-200405000-00006)
Hudson J, Cruickshank M, Quinton R, Aucott L, Aceves-Martins M, Gillies K, Bhasin S, Snyder PJ, Ellenberg SS, Grossmann M, et al.2022 Adverse cardiovascular events and mortality in men during testosterone treatment: an individual patient and aggregate data meta-analysis. Lancet. Healthy Longevity 3 e381–e393. (https://doi.org/10.1016/S2666-7568(2200096-4)
Idan A, Griffiths KA, Harwood DT, Seibel MJ, Turner L, Conway AJ & & Handelsman DJ 2010 Long-term effects of dihydrotestosterone treatment on prostate growth in healthy, middle-aged men without prostate disease: a randomized, placebo-controlled trial. Annals of Internal Medicine 153 621–632. (https://doi.org/10.7326/0003-4819-153-10-201011160-00004)
Inada A, Fujii NL, Inada O, Higaki Y, Furuichi Y & & Nabeshima YI 2016 Effects of 17beta-estradiol and androgen on glucose metabolism in skeletal muscle. Endocrinology 157 4691–4705. (https://doi.org/10.1210/en.2016-1261)
Ip FF, di Pierro I, Brown R, Cunningham I, Handelsman DJ & & Liu PY 2010 Trough serum testosterone predicts the development of polycythemia in hypogonadal men treated for up to 21 years with subcutaneous testosterone pellets. European Journal of Endocrinology 162 385–390. (https://doi.org/10.1530/EJE-09-0717)
Isidori AM, Giannetta E, Greco EA, Gianfrilli D, Bonifacio V, Isidori A, Lenzi A & & Fabbri A 2005 Effects of testosterone on body composition, bone metabolism and serum lipid profile in middle-aged men: a meta-analysis. Clinical Endocrinology 63 280–293. (https://doi.org/10.1111/j.1365-2265.2005.02339.x)
Janiszewski PM, Janssen I & & Ross R 2007 Does waist circumference predict diabetes and cardiovascular disease beyond commonly evaluated cardiometabolic risk factors? Diabetes Care 30 3105–3109. (https://doi.org/10.2337/dc07-0945)
Jesudason DR, Dunstan K, Leong D & & Wittert GA 2003 Macrovascular risk and diagnostic criteria for type 2 diabetes: implications for the use of FPG and HbA(1c) for cost-effective screening. Diabetes Care 26 485–490. (https://doi.org/10.2337/diacare.26.2.485)
Katznelson L, Finkelstein JS, Schoenfeld DA, Rosenthal DI, Anderson EJ & & Klibanski A 1996 Increase in bone density and lean body mass during testosterone administration in men with acquired hypogonadism. Journal of Clinical Endocrinology and Metabolism 81 4358–4365. (https://doi.org/10.1210/jcem.81.12.8954042)
Khosla S, Samakkarnthai P, Monroe DG & & Farr JN 2021 Update on the pathogenesis and treatment of skeletal fragility in type 2 diabetes mellitus. Nature Reviews. Endocrinology 17 685–697. (https://doi.org/10.1038/s41574-021-00555-5)
Kilby EL, Kelly DM & & Jones TH 2021 Testosterone stimulates cholesterol clearance from human macrophages by stimulating LXRa. Life Sciences 269 119040. (https://doi.org/10.1016/j.lfs.2021.119040)
Kim M, Kyung YS & & Ahn TY 2020 Cross-sectional association of metabolic syndrome and its components with serum testosterone levels in a Korean-screened population. World Journal of Men’s Health 38 85–94. (https://doi.org/10.5534/wjmh.190030)
Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM & Diabetes Prevention Program Research Group 2002 Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. New England Journal of Medicine 346 393–403. (https://doi.org/10.1056/NEJMoa012512)
Kristan A, Pajic T, Maver A, Rezen T, Kunej T, Kolic R, Vuga A, Fink M, Zula Š, Podgornik H, et al.2021 Identification of variants associated with rare hematological disorder erythrocytosis using targeted next-generation sequencing analysis. Frontiers in Genetics 12 689868. (https://doi.org/10.3389/fgene.2021.689868)
Kunnas T, Solakivi T, Huuskonen K, Kalela A, Renko J & & Nikkari ST 2009 Hematocrit and the risk of coronary heart disease mortality in the TAMRISK study, a 28-year follow-up. Preventive Medicine 49 45–47. (https://doi.org/10.1016/j.ypmed.2009.04.015)
Lehtihet M, Bonde Y, Beckman L, Berinder K, Hoybye C, Rudling M, Sloan JH, Konrad RJ & & Angelin B 2016 Circulating Hepcidin-25 is reduced by endogenous estrogen in humans. PLoS One 11 e0148802. (https://doi.org/10.1371/journal.pone.0148802)
Li JJ, Wittert GA, Vincent A, Atlantis E, Shi Z, Appleton SL, Hill CL, Jenkins AJ, Januszewski AS & & Adams RJ 2016 Muscle grip strength predicts incident type 2 diabetes: population-based cohort study. Metabolism: Clinical and Experimental 65 883–892. (https://doi.org/10.1016/j.metabol.2016.03.011)
Lopes RA, Neves KB, Pestana CR, Queiroz AL, Zanotto CZ, Chignalia AZ, Valim YM, Silveira LR, Curti C & & Tostes RC 2014 Testosterone induces apoptosis in vascular smooth muscle cells via extrinsic apoptotic pathway with mitochondria-generated reactive oxygen species involvement. American Journal of Physiology. Heart and Circulatory Physiology 306 H1485–H1494. (https://doi.org/10.1152/ajpheart.00809.2013)
Maggio M, Snyder PJ, Ceda GP, Milaneschi Y, Luci M, Cattabiani C, Masoni S, Vignali A, Volpi R, Lauretani F, et al.2013 Is the haematopoietic effect of testosterone mediated by erythropoietin? The results of a clinical trial in older men. Andrology 1 24–28. (https://doi.org/10.1111/j.2047-2927.2012.00009.x)
Marriott RJ, Murray K, Hankey GJ, Manning L, Dwivedi G, Wu FCW & & Yeap BB 2022 Longitudinal changes in serum testosterone and sex hormone-binding globulin in men aged 40–69 years from the UK Biobank. Clinical Endocrinology 96 589–598. (https://doi.org/10.1111/cen.14648)
McMillin SL, Minchew EC, Lowe DA & & Spangenburg EE 2022 Skeletal muscle wasting: the estrogen side of sexual dimorphism. American Journal of Physiology. Cell Physiology 322 C24–C37. (https://doi.org/10.1152/ajpcell.00333.2021)
McRobb L, Handelsman DJ & & Heather AK 2009 Androgen-induced progression of arterial calcification in apolipoprotein E-null mice is uncoupled from plaque growth and lipid levels. Endocrinology 150 841–848. (https://doi.org/10.1210/en.2008-0760)
Nathan L, Shi W, Dinh H, Mukherjee TK, Wang X, Lusis AJ & & Chaudhuri G 2001 Testosterone inhibits early atherogenesis by conversion to estradiol: critical role of aromatase. Proceedings of the National Academy of Sciences of the United States of America 98 3589–3593. (https://doi.org/10.1073/pnas.051003698)
Nettleship JE, Jones TH, Channer KS & & Jones RD 2007 Physiological testosterone replacement therapy attenuates fatty streak formation and improves high-density lipoprotein cholesterol in the Tfm mouse: an effect that is independent of the classic androgen receptor. Circulation 116 2427–2434. (https://doi.org/10.1161/CIRCULATIONAHA.107.708768)
Ory J, Nackeeran S, Balaji NC, Hare JM & & Ramasamy AR 2022 Secondary polycythemia in men receiving testosterone therapy increases risk of major adverse cardiovascular events and venous thromboembolism in the first year of therapy. Journal of Urology 207 1295–1301. (https://doi.org/10.1097/JU.0000000000002437)
Pellegrino A, Tiidus PM & & Vandenboom R 2022 Mechanisms of estrogen influence on skeletal muscle: mass, regeneration, and mitochondrial function. Sports Medicine 52 2853–2869. (https://doi.org/10.1007/s40279-022-01733-9)
Puddu PE, Lanti M, Menotti A, Mancini M, Zanchetti A, Cirillo M, Angeletti M & Gubbio Study Research Group 2002 Red blood cell count in short-term prediction of cardiovascular disease incidence in the Gubbio population study. Acta Cardiologica 57 177–185. (https://doi.org/10.2143/AC.57.3.2005387)
Qadir R, Sculthorpe NF, Todd T & & Brown EC 2021 Effectiveness of resistance training and associated program characteristics in patients at risk for Type 2 diabetes: a systematic review and meta-analysis. Sports Medicine – Open 7 38. (https://doi.org/10.1186/s40798-021-00321-x)
Ramachandran S, Hackett GI & & Strange RC 2020 Testosterone replacement therapy: pre-treatment sex hormone-binding globulin levels and age may identify clinical subgroups. Andrology 8 1222–1232. (https://doi.org/10.1111/andr.12813)
Regadera J, Nistal M & & Paniagua R 1985 Testis, epididymis, and spermatic cord in elderly men. Correlation of angiographic and histologic studies with systemic arteriosclerosis. Archives of Pathology and Laboratory Medicine 109 663–667.
Rha MS, Jeong Y, Kim J, Kim CH, Yoon JH & & Cho HJ 2022 Is obstructive sleep apnea associated with erythrocytosis? A systematic review and meta-analysis. Laryngoscope Investigative Otolaryngology 7 627–635. (https://doi.org/10.1002/lio2.751)
Rochira V, Zirilli L, Madeo B, Maffei L & & Carani C 2009 Testosterone action on erythropoiesis does not require its aromatization to estrogen: insights from the testosterone and estrogen treatment of two aromatase-deficient men. Journal of Steroid Biochemistry and Molecular Biology 113 189–194. (https://doi.org/10.1016/j.jsbmb.2008.12.007)
Rosner W, Auchus RJ, Azziz R, Sluss PM & & Raff H 2007 Position statement: utility, limitations, and pitfalls in measuring testosterone: an Endocrine Society position statement. Journal of Clinical Endocrinology and Metabolism 92 405–413. (https://doi.org/10.1210/jc.2006-1864)
Ross R, Neeland IJ, Yamashita S, Shai I, Seidell J, Magni P, Santos RD, Arsenault B, Cuevas A, Hu FB, et al.2020 Waist circumference as a vital sign in clinical practice: a Consensus Statement from the IAS and ICCR Working Group on Visceral Obesity. Nature Reviews. Endocrinology 16 177–189. (https://doi.org/10.1038/s41574-019-0310-7)
Roth MP, Meynard D & & Coppin H 2019 Regulators of hepcidin expression. Vitamins and Hormones 110 101–129. (https://doi.org/10.1016/bs.vh.2019.01.005)
Russell N & & Grossmann M 2019 Mechanisms in endocrinology: estradiol as a male hormone. European Journal of Endocrinology 181 R23–R43. (https://doi.org/10.1530/EJE-18-1000)
Russell N, Ghasem-Zadeh A, Hoermann R, Cheung AS, Zajac JD, Shore-Lorenti C, Ebeling PR, Handelsman DJ & & Grossmann M 2022 Effects of estradiol on bone in men undergoing androgen deprivation therapy: a randomised placebo-controlled trial. European Journal of Endocrinology 187 241–256. (https://doi.org/10.1530/EJE-22-0227)
Samelson EJ, Broe KE, Xu H, Yang L, Boyd S, Biver E, Szulc P, Adachi J, Amin S, Atkinson E, et al.2019 Cortical and trabecular bone microarchitecture as an independent predictor of incident fracture risk in older women and men in the Bone Microarchitecture International Consortium (BoMIC): a prospective study. Lancet. Diabetes and Endocrinology 7 34–43. (https://doi.org/10.1016/S2213-8587(1830308-5)
Sartorius G, Spasevska S, Idan A, Turner L, Forbes E, Zamojska A, Allan CA, Ly LP, Conway AJ, McLachlan RI, et al.2012 Serum testosterone, dihydrotestosterone and estradiol concentrations in older men self-reporting very good health: the healthy man study. Clinical Endocrinology 77 755–763. (https://doi.org/10.1111/j.1365-2265.2012.04432.x)
Selva DM, Hogeveen KN, Innis SM & & Hammond GL 2007 Monosaccharide-induced lipogenesis regulates the human hepatic sex hormone-binding globulin gene. Journal of Clinical Investigation 117 3979–3987. (https://doi.org/10.1172/JCI32249)
Serra C, Bhasin S, Tangherlini F, Barton ER, Ganno M, Zhang A, Shansky J, Vandenburgh HH, Travison TG, Jasuja R, et al.2011 The role of GH and IGF-I in mediating anabolic effects of testosterone on androgen-responsive muscle. Endocrinology 152 193–206. (https://doi.org/10.1210/en.2010-0802)
Shankara-Narayana N, Yu C, Savkovic S, Desai R, Fennell C, Turner L, Jayadev V, Conway AJ, Kockx M, Ridley L, et al.2020 Rate and extent of recovery from reproductive and cardiac dysfunction due to androgen abuse in men. Journal of Clinical Endocrinology and Metabolism 105 1827–1839. (https://doi.org/10.1210/clinem/dgz324)
Shi Z, Araujo AB, Martin S, O'Loughlin P & & Wittert GA 2013 Longitudinal changes in testosterone over five years in community-dwelling men. Journal of Clinical Endocrinology and Metabolism 98 3289–3297. (https://doi.org/10.1210/jc.2012-3842)
Sieveking DP, Lim P, Chow RW, Dunn LL, Bao S, McGrath KC, Heather AK, Handelsman DJ, Celermajer DS & & Ng MK 2010 A sex-specific role for androgens in angiogenesis. Journal of Experimental Medicine 207 345–352. (https://doi.org/10.1084/jem.20091924)
Sikaris K, McLachlan RI, Kazlauskas R, de Kretser D, Holden CA & & Handelsman DJ 2005 Reproductive hormone reference intervals for healthy fertile young men: evaluation of automated platform assays. Journal of Clinical Endocrinology and Metabolism 90 5928–5936. (https://doi.org/10.1210/jc.2005-0962)
Simpson E, Jones M, Misso M, Hewitt K, Hill R, Maffei L, Carani C & & Boon WC 2005 Estrogen, a fundamental player in energy homeostasis. Journal of Steroid Biochemistry and Molecular Biology 95 3–8. (https://doi.org/10.1016/j.jsbmb.2005.04.018)
Siren R, Eriksson JG & & Vanhanen H 2012 Waist circumference a good indicator of future risk for type 2 diabetes and cardiovascular disease. BMC Public Health 12 631. (https://doi.org/10.1186/1471-2458-12-631)
Snyder PJ, Bhasin S, Cunningham GR, Matsumoto AM, Stephens-Shields AJ, Cauley JA, Gill TM, Barrett-Connor E, Swerdloff RS, Wang C, et al.2016 Effects of testosterone treatment in older men. New England Journal of Medicine 374 611–624. (https://doi.org/10.1056/NEJMoa1506119)
Snyder PJ, Kopperdahl DL, Stephens-Shields AJ, Ellenberg SS, Cauley JA, Ensrud KE, Lewis CE, Barrett-Connor E, Schwartz AV, Lee DC, et al.2017 Effect of testosterone treatment on volumetric bone density and strength in older men with low testosterone: a controlled clinical trial. JAMA Internal Medicine 177 471–479. (https://doi.org/10.1001/jamainternmed.2016.9539)
Snyder PJ, Peachey H, Berlin JA, Hannoush P, Haddad G, Dlewati A, Santanna J, Loh L, Lenrow DA, Holmes JH, et al.2000 Effects of testosterone replacement in hypogonadal men. Journal of Clinical Endocrinology and Metabolism 85 2670–2677. (https://doi.org/10.1210/jcem.85.8.6731)
Srinivas-Shankar U, Roberts SA, Connolly MJ, O'Connell MD, Adams JE, Oldham JA & & Wu FC 2010 Effects of testosterone on muscle strength, physical function, body composition, and quality of life in intermediate-frail and frail elderly men: a randomized, double-blind, placebo-controlled study. Journal of Clinical Endocrinology and Metabolism 95 639–650. (https://doi.org/10.1210/jc.2009-1251)
Svartberg J, Agledahl I, Figenschau Y, Sildnes T, Waterloo K & & Jorde R 2008 Testosterone treatment in elderly men with subnormal testosterone levels improves body composition and BMD in the hip. International Journal of Impotence Research 20 378–387. (https://doi.org/10.1038/ijir.2008.19)
Szulc P, Boutroy S, Vilayphiou N, Chaitou A, Delmas PD & & Chapurlat R 2011 Cross-sectional analysis of the association between fragility fractures and bone microarchitecture in older men: the STRAMBO study. Journal of Bone and Mineral Reseach: the Official Journal for the American Society of Bone and Mineral Research 26 1358–1367. (https://doi.org/10.1002/jbmr.319)
Szwergold BS & & Beisswenger PJ 2003 Enzymatic deglycation--a new paradigm or an epiphenomenon? Biochemical Society Transactions 31 1428–1432. (https://doi.org/10.1042/bst0311428)
Szwergold BS, Howell S & & Beisswenger PJ 2001 Human fructosamine-3-kinase: purification, sequencing, substrate specificity, and evidence of activity in vivo. Diabetes 50 2139–2147. (https://doi.org/10.2337/diabetes.50.9.2139)
Tharp DL, Masseau I, Ivey J, Ganjam VK & & Bowles DK 2009 Endogenous testosterone attenuates neointima formation after moderate coronary balloon injury in male swine. Cardiovascular Research 82 152–160. (https://doi.org/10.1093/cvr/cvp038)
Umapathysivam M, Grossmann M & & Wittert GA 2022 Effects of androgens on glucose metabolism. Best Practice and Research. Clinical Endocrinology and Metabolism 36 101654. (https://doi.org/10.1016/j.beem.2022.101654)
Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Börjesson AE & & Ohlsson C 2014 Sex steroid actions in male bone. Endocrine Reviews 35 906–960. (https://doi.org/10.1210/er.2014-1024)
Warren AM & & Grossmann M 2022 Haematological actions of androgens. Best Practice and Research. Clinical Endocrinology and Metabolism 36 101653. (https://doi.org/10.1016/j.beem.2022.101653)
Wittert G, Atlantis E, Allan C, Bracken K, Conway A, Daniel M, Gebski V, Grossmann M, Hague W, Handelsman DJ, et al.2019 Testosterone therapy to prevent type 2 diabetes mellitus in at-risk men (T4DM): design and implementation of a double-blind randomized controlled trial. Diabetes, Obesity and Metabolism 21 772–780. (https://doi.org/10.1111/dom.13601)
Wittert G, Bracken K, Robledo KP, Grossmann M, Yeap BB, Handelsman DJ, Stuckey B, Conway A, Inder W, McLachlan R, et al.2021 Testosterone treatment to prevent or revert type 2 diabetes in men enrolled in a lifestyle programme (T4DM): a randomised, double-blind, placebo-controlled, 2-year, phase 3b trial. Lancet. Diabetes and Endocrinology 9 32–45. (https://doi.org/10.1016/S2213-8587(2030367-3)
Wittert G & & Grossmann M 2022 Obesity, type 2 diabetes, and testosterone in ageing men. Reviews in Endocrine and Metabolic Disorders 23 1233–1242. (https://doi.org/10.1007/s11154-022-09746-5)
Wittert G & & Robledo K 2022 A mediation analysis of the effect of testosterone treatment in the T4DM trial. ESA-SRB-APEG-NZSE Annual Scientific Meeting, Te Pae, Christchurch, New Zealand, Nov 12-16, Abstract #172.
Wouters HJCM, Mulder R, van Zeventer IA, Schuringa JJ, van der Klauw MM, van der Harst P, Diepstra A, Mulder AB & & Huls G 2020 Erythrocytosis in the general population: clinical characteristics and association with clonal hematopoiesis. Blood Advances 4 6353–6363. (https://doi.org/10.1182/bloodadvances.2020003323)
Yassin AA, Nettleship J, Almehmadi Y, Salman M & & Saad F 2016 Effects of continuous long-term testosterone therapy (TTh) on anthropometric, endocrine and metabolic parameters for up to 10 years in 115 hypogonadadal elderly men: real-life experience from an observational registry study. Andrologia 48 793–799. (https://doi.org/10.1111/and.12514)
Yeap BB, Alfonso H, Chubb SA, Hankey GJ, Handelsman DJ, Golledge J, Almeida OP, Flicker L & & Norman PE 2014 In older men, higher plasma testosterone or dihydrotestosterone is an independent predictor for reduced incidence of stroke but not myocardial infarction. Journal of Clinical Endocrinology and Metabolism 99 4565–4573. (https://doi.org/10.1210/jc.2014-2664)
Yeap BB, Manning L, Chubb SAP, Handelsman DJ, Almeida OP, Hankey GJ & & Flicker L 2018 Progressive impairment of testicular endocrine function in ageing men: testosterone and dihydrotestosterone decrease, and luteinizing hormone increases, in men transitioning from the 8th to 9th decades of life. Clinical Endocrinology 88 88–95. (https://doi.org/10.1111/cen.13484)
Yeap BB, Marriott RJ, Antonio L, Raj S, Dwivedi G, Reid CM, Anawalt BD, Bhasin S, Dobs AS, Handelsman DJ, et al.2022 Associations of serum testosterone and sex hormone-binding globulin with incident cardiovascular events in middle-aged to older men. Annals of Internal Medicine 175 159–170. (https://doi.org/10.7326/M21-0551)
Zhang HD, Ma YJ, Liu QF, Ye TZ, Meng FY, Zhou YW, Yu GP, Yang JP, Jiang H, Wang QS, et al.2018 Human erythrocyte lifespan measured by Levitt's CO breath test with newly developed automatic instrument. Journal of Breath Research 12 036003. (https://doi.org/10.1088/1752-7163/aaacf1)
Zhou L, Han L, Liu M, Lu J & & Pan S 2020 Impact of metabolic syndrome on sex hormones and reproductive function: a meta-analysis of 2923 cases and 14062 controls. Aging (Albany NY) 13 1962–1971. (https://doi.org/10.18632/aging.202160)