ArticlePDF Available

The Role of Diet and Weight Loss in Improving Secondary Hypogonadism in Men with Obesity with or without Type 2 Diabetes Mellitus

MDPI
Nutrients
Authors:

Abstract

Despite growing recognition of the issue, obesity represents one of the most common public health problems, and its rates are still increasing globally. Among the number of comorbidities and complications associated with obesity, hypogonadism is listed, and this disorder, although frequently neglected, is characterized by a relevant impact on both quality of life and life expectancy. It is generally accepted that hypogonadism secondary to obesity is functional since it is reversible following weight loss. This review summarizes all current research examining the bidirectional relationship between excess body weight and low testosterone levels. Specifically, it evaluates the role that diet, with or without physical activity, plays in improving body weight and hypogonadism in adult and elderly men with obesity, with or without type 2 diabetes mellitus.
nutrients
Discussion
The Role of Diet and Weight Loss in Improving
Secondary Hypogonadism in Men with Obesity with
or without Type 2 Diabetes Mellitus
Vito Angelo Giagulli 1,2 ,* , Marco Castellana 3, Isanna Murro 4, Carla Pelusi 5,
Edoardo Guastamacchia 1, Vincenzo Triggiani 1and Giovanni De Pergola 4
1Section of Internal Medicine, Geriatrics, Endocrinology and Rare Disease, Interdisciplinary Department of
Medicine, University of Bari, School of Medicine, Policlinico, 70124 Bari, Italy;
edoardo.guastamacchia@uniba.it (E.G.); vincenzo.triggiani@uniba.it (V.T.)
2Outpatients Clinic of Endocrinology and Metabolic Disease, Conversano Hospital, 70014 Bari, Italy
3National Institute of Gastroenterology “S. De Bellis”, Castellana Grotte, 70013 Bari, Italy;
mcastellana01@yahoo.it
4Clinical Nutrition Unit, Medical Oncology, Department of Biomedical Science and Human Oncology,
University of Bari, School of Medicine, 70124 Bari, Italy; murro.isanna3@gmail.com (I.M.);
giovanni.depergola@uniba.it (G.D.P.)
5Division of Endocrinology and Center for Applied Biomedical Research, Department of Medical and
Surgical Sciences, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Hospital,
40138 Bologna, Italy; Carla.pelusi2@unibo.it
*Correspondence: vitogiagulli58@gmail.com; Tel.: +39-80-409-1368
Received: 27 October 2019; Accepted: 2 December 2019; Published: 5 December 2019


Abstract:
Despite growing recognition of the issue, obesity represents one of the most common
public health problems, and its rates are still increasing globally. Among the number of comorbidities
and complications associated with obesity, hypogonadism is listed, and this disorder, although
frequently neglected, is characterized by a relevant impact on both quality of life and life expectancy.
It is generally accepted that hypogonadism secondary to obesity is functional since it is reversible
following weight loss. This review summarizes all current research examining the bidirectional
relationship between excess body weight and low testosterone levels. Specifically, it evaluates the
role that diet, with or without physical activity, plays in improving body weight and hypogonadism
in adult and elderly men with obesity, with or without type 2 diabetes mellitus.
Keywords:
obesity; hypogonadism; testosterone; diet; physical activity; insulin resistance;
type 2 diabetes mellitus
1. Introduction
Obesity is one of the major public health issues globally, since it is associated with a higher morbidity [
1
]
and mortality [
2
], leading to a relevant economic impact on the affected people, their families, and the
health care system [
3
]. Subjects with obesity have a higher risk of a number of comorbidities, including
insulin resistance, type 2 diabetes mellitus (T2DM), dyslipidemia, hypertension, cardiovascular disease,
some types of cancer, hyperuricemia, deep vein thrombosis and pulmonary embolism, sleep-related
breathing disorders, osteoarthritis, and hypovitaminosis D [
4
6
]. In males, obesity represents the clinical
condition most strongly associated with hypogonadism, leading to erectile dysfunction and infertility
due to oligospermia/azoospermia [
7
9
]. In addition, in a vicious cycle, male hypogonadism impairs
body composition (reduced muscle mass, higher fat mass), fat metabolism, bone mineralization,
and cognitive function. Particularly, central fat accumulation has been linked with all the disorders
Nutrients 2019,11, 2975; doi:10.3390/nu11122975 www.mdpi.com/journal/nutrients
Nutrients 2019,11, 2975 2 of 16
above, and waist circumference has been found as the most reliable predictor of both lower serum
testosterone (T) and risk of developing metabolic and cardiovascular diseases [
10
12
]. The first-line
strategy for the management of overweight and obesity is represented by lifestyle interventions,
including diet and physical activity; in patients not achieving targets and meeting specific criteria,
pharmacological therapy and bariatric surgery should be considered [6,13].
In the present narrative review, the pathophysiology of functional secondary hypogonadism
is summarized, the impact of hypogonadism on body weight evaluated, and the role of lifestyle
interventions with a special focus on diet in improving this disorder in men with obesity with or
without type 2 diabetes mellitus discussed. Also, the possible eects of T supplementation in these
patients is reported.
2. Pathophysiology of Functional Secondary Hypogonadism
In the last few years, a new classification of hypogonadism has been proposed. Together with the
organ responsible for the disorder (either testis in primary forms or hypothalamus/pituitary gland
in secondary forms), the mechanism underlying hypogonadism should be considered as functional
and organic forms dierentiated. To be classified as functional, hypogonadism should be reversible
following the treatment of underlying causes. No structural, destructive, or congenital disease should
aect the hypothalamic–pituitary–testicular axis (HPT) [1416].
Among adult males, the most common form of hypogonadism is functional secondary
hypogonadism, also known as dysmetabolic hypogonadotropic hypogonadism or male obesity
secondary hypogonadism (MOSH). The pathogenesis of this disorder is multifactorial and generally
related to chronic diseases, including obesity, T2DM, and the metabolic syndrome [
15
,
16
]. Regardless of
the latter diseases, an inverse relationship between T and excess body weight has been reported in men
of all ages, with lower T levels found in subjects with higher body mass index (BMI) [
7
]. Indeed, low total
testosterone (TT) primarily reflects reduced concentrations of sex hormone-binding globulin (SHBG) in
mild obesity, with no clear evidence of clinical androgen deficiency or defective spermatogenesis [
17
].
Conversely, more severe forms of obesity lead to overt hypogonadotropic hypogonadism, due to the
suppression of the HPT axis, and lower levels of both TT and free testosterone (FT) and even of inhibin
B [
17
23
]. Interestingly, this mechanism of HPT axis damage seems to be already present at puberty in
boys with obesity [
24
]. Gonadotropins (follicle-stimulating hormone [FSH] and luteinizing hormone
FSH [LH]) may be inappropriately normal or low in mild obesity [
8
], and a specific decrease in pulse
amplitudes of LH has been reported in middle-aged men with severe obesity [
25
]. Several factors are
involved in the pathogenesis of this form of hypogonadism, in particular, a role of estradiol, insulin,
leptin, and other pro-inflammatory cytokines has been proposed. A brief summary of the contribution
of each hormone is reported in the following sections.
2.1. Estradiol
In men with obesity, a part of androgens is converted to estrogens through peripheral aromatization
in adipose tissue [
26
]. The most represented estrogen is estradiol, and this has been suggested to
induce functional secondary hypogonadism due to the suppression of the HPT axis [
27
,
28
]. Indeed,
early studies showed higher estradiol concentrations in young to middle-aged subjects with obesity
compared to lean controls [
29
], and following studies reported the treatment of hypogonadal subjects
with obesity with selective estrogen receptor modulators (SERM) or anti-aromatase inhibitors (AI) to
be successful in stimulating the HPT axis and in achieving physiological T levels [
30
,
31
]. Based on
these findings, it was hypothesized that low TT levels in subjects with obesity and hypogonadism are
due to the estradiol-related HPT axis suppression and low levels of SHBG.
In recent years, this hypothesis has been questioned. Dierent studies found low levels of both
estradiol and TT in men with obesity [
32
] as well as in hypogonadal men with T2DM [
33
]. Indeed,
given that up to 40% of circulating estradiol is synthesized from the peripheral conversion of T to
estradiol, it is much more likely that men with obesity and low serum T may have low estradiol
Nutrients 2019,11, 2975 3 of 16
levels too [
25
,
34
]. Accordingly, no changes in estradiol levels after weight loss were reported [
18
],
thus supporting the picture of hypogonadal men with obesity as not being characterized by a chronic
estrogen excess [
35
]. Given the findings above, one would raise the question whether a vicious
cycle is there for obesity and estrogens, as the one described for T. Indeed, in preclinical studies,
adipose tissue inflammation and insulin resistance were found to reduce aromatase expression,
which returned to normal levels with improvements in the factors above [
36
]. In addition, adequate
estradiol concentrations were linked with favorable metabolic eects, including reduced white adipose
tissue accumulation, preserved insulin sensitivity in skeletal muscles and liver, reduced apoptosis of
pancreatic
β
-cells driven by oxidative stress, amyloid polypeptide toxicity, or lipotoxicity, and enhance
glucose-stimulated insulin biosynthesis. Furthermore, estrogens act on the central nervous system,
particularly in the hypothalamus, leading to a decrease food intake [37].
In all, circulating estradiol is primarily dependent on T, and there is growing evidence that its levels
tend to decrease rather than to increase along with the progressive increase of fat mass [
35
]. To date,
conflicting data are reported in the literature on the contribution of estrogens to the pathogenesis of
functional secondary hypogonadism and the possible mechanisms not definitely explained.
2.2. Insulin
In vitro
studies showed that insulin stimulates the secretion of gonadotropin-releasing factors
(GnRH) from the hypothalamus, gonadotropin from the pituitary gland, and T from the Leydig
cells in the testis [
38
40
]. Obesity and T2DM are characterized by insulin resistance and, in those
patients in which this is not oset by the hyperinsulinemia, hypogonadism can be found due to
the lack of response of the HPT axis. It is worth noting that in patients with T2DM, a suppression
of testicular function rather than the HPT axis is possible, and 5% of men with T2DM present
with hypergonadotropic hypogonadism. However, the largest part is characterized by functional
hypogonadal hypogonadism mainly due to concomitant obesity. Other mechanisms have also been
proposed to describe the relationship between insulin and low T. Particularly, insulin resistance and
hyperinsulinemia are known to inhibit hepatic SHBG secretion
in vitro
and
in vivo
[
41
]. This protein
bounds 40–50% of total testosterone and plays a key role in the crosstalk between hypogonadism
and metabolic disorders. Low levels of SHBG are commonly found in patients with obesity or
type 2 diabetes mellitus. Since SHBG influences the bioavailability of testosterone, a reduction in
SHBG is associated with transient higher FT which is aromatized to estradiol, possibly inducing
negative feedback on the hypothalamic–pituitary–testicular axis [
42
]. Also, in patients with obesity,
metabolic syndrome, diabetes mellitus, dyslipidemia, and hypertension, a common comorbidity is
represented by non-alcoholic fatty liver disease (NAFLD), which includes non-alcoholic fatty liver
(NAFL) and non-alcoholic steatohepatitis (NASH). This is a strong predictor of progression to type
2 diabetes mellitus and is an independent risk factor irrespective of age and BMI [
43
]. Given that
testosterone undergoes hepatic metabolization and its levels are influenced by circulating SHBG which
is secreted by the liver, it has been hypothesized that NAFLD may play a key role in secondary
hypogonadism [
44
,
45
]. Accordingly, several observational studies and a meta-analysis demonstrated
a significant correlation between being diagnosed with NAFLD and low levels of both TT and SHBG.
There were not enough data to draw a definitive conclusion on FT [
45
,
46
]. However, a recent study
showed that mechanisms linking inflammation to hypothalamic–pituitary downregulation and then to
overt secondary hypogonadism described in animal models apply to humans too [
47
]. Eighty patients
with obesity were included, and sex hormone levels and liver disease were evaluated by the means of
gold standard techniques, represented by mass spectroscopy and liver biopsy, respectively. Compared
to patients with NAFL, those with NASH showed lower serum FT levels and higher levels of free
estradiol/FT and FT/LH ratios, indicating an overt condition of hypogonadism driven by an increase
in adipose tissue aromatization. Thus, while confirming this relationship between low testosterone
and NAFLD, this provided a new and interesting pathophysiogical mechanism. As a matter of fact,
it demonstrated that the grade of inflammation, as well as the severity of liver disease, influence
Nutrients 2019,11, 2975 4 of 16
androgen levels [
48
]. It is worth noting that, to date, there are not enough data in order to support
a bidirectional correlation between hypogonadism and NAFLD [45].
2.3. Leptin
An important role in the pathogenesis of functional secondary hypogonadism is played by the
visceral adipose tissue, in which a variety of circulating mediators (including leptin and pro-cytokines)
contributing to the suppression of the HPT axis at multiple levels is synthesized [
17
,
41
,
49
,
50
]. Leptin is
a major determinant of energy homeostasis and key in the regulation of male fertility. Concerning to
former aspect, it acts both on the central nervous system, controlling food intake, energy expenditure,
and fat distribution, and peripherally, favoring insulin sensitivity, lipolysis, and free fatty acids
(FFA) oxidation [
51
]. Concerning the latter aspect, it acts on the HPT axis at multiple levels. In the
hypothalamus, it stimulates the release of kisspeptin from forebrain neurons, which induces GnRH and
gonadotropin secretion [
52
,
53
]. In the testis, it inhibits basal and hCG-stimulated T production in adult
rat testis [
54
,
55
]. These mechanisms seem to be in conflict with each other; furthermore, considering
the former mechanism, hypogonadism would be hard to explain, given that that the adipose tissue is
the main source of leptin and subjects with obesity are characterized by high levels of leptin. However,
as for insulin and T2DM, the main link between leptin and hypogonadism could be represented by
leptin resistance, leading to either defects in the intracellular signaling of the leptin receptor or an
abnormal leptin transport across the blood–brain barrier [
56
]. In male obese hypogonadal animals,
leptin resistance was shown to reduce GnRH secretion through a downregulation of kisspeptin gene
expression and kisspeptin receptors [
57
]. On the contrary, it is believed that leptin sensitivity is
maintained in the testis, thus inhibiting steroidogenesis [
54
]. In conclusion, in male patients with
obesity, the combination of leptin resistance in the hypothalamus and a preserved leptin sensitivity in
the testis leads to hypogonadism [58].
2.4. Other Pro-Inflammatory Cytokines
As already stated, obesity and metabolic syndrome are characterized by chronic inflammation
and high levels of cytokines that are known to impact on Leydig cell steroidogenesis and T levels either
directly interfering with the HPT axis or indirectly altering the insulin transduction pathway [
59
61
].
Besides leptin, these include tumor necrosis factor-alpha (TNF-
α
), interleukin-1
β
(IL-1
β
), interleukin-6
(IL-6), and interleukin-8 (IL-8).
In vitro
studies showed TNF-
α
and IL-6 to be able to suppress
GnRH and LH secretion [
62
,
63
]. Also, in the testis, TNF-
α
, IL-1
β
, and IL-6 were associated with
a dose-dependent decline in steroidogenesis in TM3 Leydig cells [
64
]. Furthermore, in rat models
of diet-induced obesity, high levels of TNF-
α
, IL-1
β
, and IL-6 were associated with the disruption
of testicular morphology, with a reduction in seminiferous tubule diameter, epithelial and germ
cell atrophy and apoptosis, abnormal adhesions between Sertoli and spermatogenic cells and the
blood–testis barrier disassembly [
65
]. On the other hand, even if IL-8 exerts a stimulating eect on TM3
Leydig cell growth, it appears to minimally aect T production and blood concentrations [
64
]. Recently,
other cytokines, such as resistin and visfatin, have been assessed for their possible involvement in the
alteration of the HPT axis through inducing an insulin resistance state [66].
The clinical relevance of targeting cytokines for the management of functional secondary
hypogonadism was recently shown in a randomized controlled trial (RCT) conducted in hypogonadal
men with obesity: the administration of an anti-inflammatory treatment with anakinra (recombinant
human IL-1 receptor antagonist) led to an increase in endogenous T levels [
67
]. Overall, these data
support the connection among chronic inflammation, high levels of cytokines, and hypogonadism,
even if the exact mechanisms and site of action on the HPT axis are still a matter of debate.
3. Preclinical Studies On the Impact of Hypogonadism on Dierent Tissues
Animal models are useful in evaluating the impact of a specific molecule on a specific tissue.
To achieve this target, an interesting option is represented by the generation of knockout models.
Nutrients 2019,11, 2975 5 of 16
Particularly, in the setting of hypogonadism, this result can be achieved through the knockout of the
androgen receptor (AR). Preclinical studies linked sex hormones (especially T) with glucose homeostasis
and insulin sensitivity. Indeed, endocrine pancreas (
α
-cells and
β
-cells), liver, muscle, adipose tissue,
and neuronal tissue express the AR and the estrogen receptor (ER). Despite both
α
-cell and
β
-cell
presenting AR, the eects of androgens in
α
-cell is still unknown. Conversely, the selective knockout of
the AR in mice
β
-cells was associated with glucose intolerance due to a reduction in glucose-stimulated
insulin secretion, leading to hypoinsulinemia, glucose intolerance, and hyperglycemia. Moreover,
in these models, T treatment could not improve glucose-stimulated insulin secretion [
68
]. The selective
knockout of the AR in mice liver was associated with steatosis, insulin resistance, increased lipogenesis,
and reduction in FFA oxidation [
69
]. The male genomic AR knockout mice were associated with
reduced muscle mass, strength, and fatigue resistance [
70
]. The same animal model was characterized
by visceral obesity and both insulin and leptin resistance at an advanced age. Of note, AR and ER
density varies according to the type of adipose tissue more than sex, and visceral adipose tissue
presents with higher AR density than subcutaneous adipose tissue, highlighting the crucial role of T in
modulating body composition and metabolism [
71
]. Then, several selective adipose tissue-specific
AR knockout models were generated. A first model was generated using adipocyte-specific fatty
acid binding 4 protein (aP2) Cre mice and provided a phenotype of hyperleptinemia without leptin
resistance [
72
]. Another model was generated by crossing the same aP2-Cre with a dierent floxAR
line, and it was characterized by hyperinsulinemia in the absence of obesity. However, these mice
presented increased susceptibility to visceral obesity, hyperglycemia, and impaired insulin secretion
whenever they started a high-fat diet [
73
]. Finally, a selective knockout of the AR in the mice central
nervous system was generated using synapsin I Cre and showed reduced insulin sensitivity, increased
visceral obesity, serum triglycerides, and FFA [
74
]. Overall, the global AR knockout models suggest
that the AR regulates a number of pathways, and this regulation might occur through AR actions in
organs other than fat tissue, including the pancreas, muscle, and the central nervous system.
4. Lifestyle Interventions for the Management of Functional Secondary Hypogonadism
Although obesity is responsible of functional hypogonadotropic hypogonadism, a decrease of
serum T levels is predictive of metabolic syndrome, unfavorable changes of body composition (increase
of fat mass and decrease of fat free mass), lower insulin sensitivity, higher waist circumference, fasting
glucose, triglyceride, and LDL-cholesterol levels at all ages [
75
]. Accordingly, low SHBG, T, and other
sex steroids in community-dwelling middle-aged to elderly men and androgen deprivation therapy,
as in the case of prostate cancer treatment, were associated with a higher incidence of T2DM and
cardiovascular diseases [
76
78
]. On the other hand, testosterone replacement therapy (TRT) was
associated with reduced fat mass and increased lean mass in hypogonadal dysmetabolic patients [
79
].
In fact, T plays a critical role in the regulation of energy utilization, including adipogenesis, carbohydrate
and fat metabolism, and nitrogen retention [80].
Weight loss, however obtained (by diet, physical exercise, pharmacological therapy, or bariatric
surgery), reverses functional hypogonadotropic hypogonadism secondary to obesity to eugonadism
and induces an increase in both TT and FT [
17
,
22
,
81
]. In particular, at least 10% of weight loss is
required to achieve a significant increase in circulating T in most men with obesity and in improving
clinical features related to androgen deficiency [
17
]. In general terms, lifestyle measures are less
eective in inducing weight loss and are associated with less evident increases in circulating T [
17
].
However, any second-line intervention for obesity (either pharmacological therapy or bariatric surgery)
should always be considered as an add-on to lifestyle measures, including diet and physical activity.
In addition, one longitudinal study performed in men with obesity submitted to bariatric surgery
has shown that the degree of weight loss was more closely associated with improvements in sexual
function rather than with the increase in T levels [82].
Nutrients 2019,11, 2975 6 of 16
4.1. Eects of Lifestyle: Diet Only
An increase in serum androgens levels was reported following weight loss achieved through
very-low-calorie diets (VLCD) or low-calorie diets (LCD). Stanik et al. investigated the hormonal
profiles after a very restricted dietary protocol (supplemented fasting program with 320 kcal/day) for
8–20 weeks in 24 moderately men with obesity, and they observed that a mean weight loss of 19 kg
induced the normalization of TT and FT levels [
83
]. Another study evaluated 38 men with obesity
divided into two groups: 19 subjects were treated for 12 weeks with a VLCD, with total energy intake of
approximately 800 kcal/day, and subsequently for 10 weeks with a behavioral variation; the remaining
19 subjects were untreated and were considered as control subjects. The rapid weight loss induced by
VLCD resulted in an increase in SHBG and TT levels, and a decrease in serum insulin and leptin levels;
all these changes were maintained until the end of the follow-up period (8 months). Interestingly, when
the changes induced by maintained weight loss were evaluated in a backward regression analysis,
the decrease in insulin resistance was the most significantly parameter related to serum T levels [
84
].
These data are in line with the results of another study, evaluating serum sex hormones levels, sexual
function, and low urinary tract symptoms (LUTS) assessed through IPSS score before and after 8 weeks
in 51 men with visceral obesity with or without T2DM assigned to LCD (850–900 kcal/day) compared
to a control group of 25 men without T2DM but with similar BMI and waist circumference assigned to
their usual diet. Weight loss was significantly correlated with increased insulin sensitivity, TT, sexual
function and sexual desire item scores; moreover, the two latter endpoints were directly associated
with the level of waist circumference reduction [
85
]. The eects of the weight loss induced by diet on
hormonal, sexual, and urinary functions were also confirmed in a cohort of 31 men aected by obesity
and T2DM, showing improvements in all endpoints either in an initial 8-week LCD or in the following
44-week maintenance high protein, low-fat, and reduced-carbohydrate diet having a decrease in daily
intake of approximately 600 kcal/day. This study also showed a significant decrease in inflammatory
markers, such as C-reactive protein and IL-6, and an improvement in endothelial function evaluated
through brachial artery flow-mediated dilatation and soluble E-selectin levels. Thus, mechanisms
other than higher T are possibly involved in the improvements in sexual and urinary functions in men
with obesity [
86
]. Finally, Niskanen et al. experimented with the eect of rapid weight loss obtained by
a 9-week VLCD followed by a 12-month weight maintenance in a cohort of 58 men with visceral obesity.
An increase in serum SHBG levels during VLCD and a little decrease during weight maintenance,
staying however higher than baseline, were found; FT was significantly increased, both following
the rapid weight loss induced by VLCD and at the end of the 1-year weight maintenance period,
whereas TT showed intermediate variations between those of SHBG and FT [
87
]. In obese men, a rise
in SHBG (and therefore TT) is well known after weight loss as a result of insulin sensitization. FT or
bioavailable testosterone measurement is needed for the appropriate assessment of gonadal status.
Relevant data on the impact of diet on FT have been published in recent years. A first meta-analysis
found a low-calorie diet to be associated with an increase in TT, SHBG, and FT. It is worth noting that
in this meta-analysis, the results on the latter outcome were based on only two studies evaluating diet
as intervention [
81
,
84
,
87
]. The results were updated in a following meta-analysis published in 2019 by
the same group. In this study, the results of eight studies were pooled, and the significant increase in
FT was confirmed [
88
]. In all, a limited number of studies are reported in the literature on the impact of
diet on FT. Based on available data, a positive correlation should be expected between weight loss and
change in FT. These data strongly emphasize that diet is key as the first-line treatment for functional
hypogonadal hypogonadism secondary to obesity.
Since cholesterol is a precursor of T, an interesting topic is the possible eects of vegetarian diets
on the production of T. Surprisingly, very few not-recent studies investigated this aspect and conflicting
findings were reported. One study found lower levels of T and estradiol in vegetarian men compared
to omnivores men [
89
]. Other studies showed similar concentrations of TT, FT, and estradiol in the
same populations, despite a higher SHBG in the former [
90
,
91
]. In all, apart from the clear higher
SHBG levels in vegetarians, which are mainly due to high fiber intake, clear evidence on the eects of
Nutrients 2019,11, 2975 7 of 16
vegetarian diets on T is currently lacking. Western-style diets (WSDs) are characterized by a high intake
of cholesterol and protein; thus, it represents an interesting model to examine whether food habits may
change T levels in men. It is worth noting that WSD is highly heterogeneous in quantities and qualities
of fat (saturated vs. unsaturated), carbohydrates (high glycemic vs. low glycemic), and protein content,
and recent evidence suggests that imbalance in the levels of sex steroids is associated with WSD.
However, whether this alteration is mediated through obesity and insulin resistance at least in part,
there is no clear demonstration that WSD per se is responsible for lower T levels and infertility [92].
4.2. Eects of Lifestyle: Diet and Physical Activity
Several studies reported that physical activity is associated with an increased endogenous T in
men with obesity. Kumagai et al. evaluated the impact of aerobic exercise on T levels in 28 men
with overweight/obesity. After 12 weeks, an increase in TT and FT in the latter group was found,
with vigorous exercise independently associated with improvements in TT [
93
]. Given the results of
diet and physical activity when assessed as single interventions, a number of studies were performed to
assess their combined eect. Armamento-Villareal et al. evaluated the impact of lifestyle interventions
on hormone levels in frail older men with obesity. After 12 months, weight loss was not associated
with improvements in either TT or FT levels. However, the clinical relevance of these findings could
be limited, given the inclusion criteria of the study population. Indeed, in frail and older men with
obesity, weight loss is not the best approach for raising T levels because it can worsen age-related
muscle and bone loss [
94
]. Recently, De Lorenzo et al. evaluated the eects of a 10% weight loss
achieved through diet and physical activity on the hormonal profile of MOSH patients with total
testosterone below 12.1 nmol/L (349 ng/dL). The prescribed diet consisted of a “Mediterranean” style
diet, with all macronutrients present in each of the five meals. This was characterized by high
consumption of fresh vegetables, fruits, and extra virgin olive oil, and proteins were given through
legumes, cereals, and fish [
95
]. Overall, it was a hypocaloric (170–250 kcal/day reduction compared to
basal metabolism), high-protein (1.5 g/day of ideal body weight) diet; calories were given through
carbohydrates (45–50%), proteins (20–25%), and fat (30%; saturated fat <7%, polyunsaturated fatty
acids 10–20%, monounsaturated fatty acids 10–20%, cholesterol <300 mg/day). Fiber intake was
25–30 g/day, sodium intake was <5 g/day; no alcoholic beverages were allowed. Concerning physical
activity, subjects were prescribed with 150 min/week of mild intensity aerobic (50–70% of max heart
rate) activity and/or 90 min/week of high-intensity (>70% max heart rate) activity to be practiced at
least three days/week. After 3
±
1 months, a significant increase in TT was observed, together with
a reduction in fat mass percentage [
96
]. The largest study examining the eects of weight changes on T
concentrations was the Diabetes Prevention Program (DPP) trial. Eight-hundred-eighty-six adult men
with a mean BMI 32 kg/m
2
and a mean age 52 years were randomized to placebo twice daily (n=278),
850 mg metformin twice daily (n=315), or intensive lifestyle intervention (n=293). After 12 months,
the weight loss was
0.12 kg,
2.73 kg, and
7.87 kg, respectively. TT increased only in the latter
group by 1.15 nmol/L [
97
]. In synthesis, this demonstrates that lifestyle interventions are eective in
improving body composition, hormonal and metabolic profiles. Therefore, they should represent the
first choice in the management of obesity and functional secondary hypogonadism.
Concerning the eects of lifestyle measures on semen quality, there is still very little information.
One study evaluated 43 men with obesity before and after 14 weeks of a weight loss program based on
a healthy diet and physical exercise. Weight loss was significantly associated with an increase in total
sperm count, semen volume, TT, and serum SHBG levels, with those men achieving the largest weight
loss showing normal sperm morphology [98].
4.3. Eects of Adding Testosterone Replacement Therapy to Lifestyle Interventions
As already stated, lifestyle interventions should be regarded as relevant tools for the management
of functional secondary hypogonadism. However, the question arises as to whether TRT can enhance
the benefits of lifestyle changes and prevent the diet-associated loss of muscle mass. One RCT
Nutrients 2019,11, 2975 8 of 16
performed by Hidreth et al. in healthy older men with low-normal T levels (200–350 ng/dL) showed
that T supplementation compared to placebo improved body composition in all patients; however,
no eect on functional performance or strength in those patients allocated to progressive resistance
training was found, while an improved upper body strength only in those patients allocated to no
exercise was reported [
99
]. In another RCT, no additional benefit on body weight, BMI, and waist
circumference was found when T treatment or placebo were given to men with obesity and obstructive
sleep apnea on VLCD. It is worth noting that the baseline T levels in each group were 13.2 and
13.4 nmol/L, respectively; whether these results could be considered applicable to all patients with
functional secondary hypogonadism is unclear [
100
]. In a recent 56-week RCT performed by Ng
Tang Fui et al., middle-aged (median age 53 years) men with obesity and low TT (mean 6.9 nmol/L)
were subjected to a rigorous 10-week VLCD phase followed by 46 weeks of weight maintenance
and were randomized at baseline to TRT or placebo. Both groups lost a similar amount of body
weight at the end of the VLCD phase. However, men allocated to T had greater reductions in fat
mass (between-group dierence
2.9 kg) and visceral fat (
2678 mm
2
), and preservation of lean mass
(+3.4 kg), compared to placebo. Thus, TRT prevented the diet-associated loss in lean mass without
increasing the diet-associated weight loss [
101
]. When the same patients were evaluated more than one
year after the RCT completion, in those patients in which TRT was withdrawn, T concentrations fell
back to baseline concentrations. Two-thirds of the weight loss was regained, the eect of TRT on body
composition was not apparent anymore, and dierences in fat and lean mass between groups were
no longer maintained [
102
]. This suggests that relatively short-term (56 weeks) TRT is not associated
with sustained benefits and that long-term therapy should be considered to maintain the achieved
results [
16
,
101
,
102
]. However, TRT can be considered to overcome fatigue and engage a healthy
lifestyle in some groups of patients, in line with results of other studies [103].
As far as the mechanisms involved in the eects of T on changes in body composition is concerned,
two options can be considered: the first is represented by its direct eects on adipose and skeletal
muscle cells; the second may well be the spontaneous increase of physical activity observed in men
on TRT [
29
,
101
]. This treatment was associated with lower leptin levels, suggesting a decrease in
adipose tissue mass and/or a lessening of leptin resistance [
22
]. On the other hand, it is to note that TRT
does not significantly influence circulating concentrations of ghrelin, glucagon-like peptide-1, gastric
inhibitory polypeptide, peptide YY, pancreatic polypeptide, and amylin, suggesting that its eects
on body composition are unlikely to be mediated by changes in modulators of appetite and energy
homeostasis [
29
]. Regarding sexual function, TRT improves this aspect independently of the eect on
weight loss [
104
]. In the large T trials, sexual function improved modestly with TRT, suggesting that
even older men with a relatively high comorbid burden (T2DM or previous myocardial infarction) can
respond to T [105].
Hypogonadism is associated with impaired physical activity. A recent systematic review found
TRT to be associated with increased physical function without any impact on muscle strength [
106
].
These findings are not in line with one reported by Storer et al. in which three years of TRT was
associated with an increase in stair-climbing power, muscle mass, and power [
107
]. Further studies to
address this issue are needed.
Lower TT and FT are generally found in patients with T2DM [
108
,
109
]. However, the presence
of this disorder does not aect the positive eects of TRT. A prospective study, performed for over
five years in 181 men with obesity, including 72 patients aected by T2DM, treated by parenteral
testosterone undecanoate 1000 mg/12 weeks, showed a significant and sustained reduction in body
weight, BMI and waist circumference. In addition, this study showed that restoring serum testosterone
to normal in hypogonadal men with obesity is beneficial with regards to the metabolic syndrome:
fasting glucose declined over the first 18 months and then stabilized, with similar trend for serum total
cholesterol, LDL cholesterol, and triglycerides; HDL increased slightly but significantly; diastolic and
systolic blood pressure decreased. This pattern was similar in patients with or without T2DM and is in
line with earlier studies [110,111].
Nutrients 2019,11, 2975 9 of 16
Although male hypogonadism can be considered an independent risk factor for the development
of metabolic syndrome, cardiovascular disease, and all-cause mortality, TRT presents some concerns,
especially whenever it is taken into consideration for the treatment of elderly men suering from
overt hypogonadism [
112
114
]. Indeed, elderly men can be aected by dierent metabolic disorders,
which favors the appearance of clinically important cardiovascular events, causing a state of fragility
and vulnerability [
115
]. However, several clinical studies and a recent meta-analysis of randomized
controlled studies proved that TRT improved waist circumference, body composition, some metabolic
parameters such as homeostatic model assessment (HOMA) and glycemic control, and reduced the
progression from prediabetes to overt diabetes mellitus type 2 in adult dysmetabolic men [
116
118
].
In addition, a meta-analysis of pharmaco-epidemiological studies demonstrated that patients treated
with T compared with controls showed a reduction of both mortality and mobility, especially due
to acute myocardial infarction, stroke, acute coronary syndromes, and heart failure. Conversely,
data from randomized controlled studies suggest that TRT is not able to reduce cardiovascular risk,
however, when the therapy with T is correctly applied, at least it is not associated with an increased
cardiovascular risk. All in all, there is a need for adequately powered placebo-controlled studies with
cardiovascular disease as the primary endpoint carried out in men with late-onset hypogonadism in
order to strengthen the conclusion that TRT is not associated with higher cardiovascular risk [
119
].
Another delicate issue when TRT is considered is represented by prostatic cancer. Cunningham et
al. reported that, in older men with symptomatic hypogonadism, normalizing T levels for one year
was associated with a small but statistically significant greater increase in Prostate-Specific Antigen
(PSA) levels compared to placebo [
120
]. This may lead to subsequent urological consultation, prostate
biopsy, and more frequent detection of prostate cancers, often represented by indolent lesions with
consequent overtreatment and possible complications. It is worth noting that nearly half of the PSA
elevations observed in this study were not confirmed when the analysis was repeated; furthermore,
when patients with a confirmed elevation of PSA were referred to a urologist and prescribed with PSA
repetition, an additional 50% of PSA elevations were not confirmed anymore, suggesting that PSA
elevations are often the result of laboratory variability. Also, out of 790 patients during the 12 months
of treatment and the six months of follow-up, only four prostate cancers were found, three of which in
the testosterone replacement therapy arm and one in the placebo. In any case, the potential benefits
and related risks of TRT should always be discussed with the patient, and elevated PSA values should
always be confirmed by repeating the test [
121
124
]. In line with this, the Endocrine Society Guidelines
for T treatment of hypogonadal men recommend a monitoring plan aiming at minimize the risk of
unnecessary prostate biopsies avoiding the detection of indolent small tumors and leading at the same
time to the identification of men who are at increased risk of having aggressive, potentially lethal
prostate cancer. Urologic referral, in particular, is recommended for men who, in the first year of T
treatment, have a confirmed increase in PSA values of 1.4 ng/mL or a confirmed absolute value of
4.0 ng/mL (or 3 ng/mL in men at high risk of prostate cancer) [16].
5. Conclusions
In patients with obesity and functional hypogonadotropic hypogonadism, weight loss has been
shown to be eective in restoring eugonadism. Available studies emphasize the key role of lifestyle
interventions as the first-line treatment to achieve improvements in body weight and secondary
hypogonadism. Among diets, hypocaloric and high-protein diets with Mediterranean style seem to
have the most favorable eect on T levels, T/estradiol ratio, sexual function, and sperm quality. Also,
physical activity should always be recommended, given the positive influence on androgenicity. Lastly,
TRT can be considered in specific groups of patients, in the knowledge that there is the lack of evidence
of sustained benefits following its withdrawal. Further studies comparing dierent diet protocols
and with a longer follow-up are needed. In particular, it would be of interest to focus on dietary
protocols introduced in recent years, including the very-low-calorie ketogenic diet, the Paleolithic diet,
and intermittent fasting for which we were not able to find a high level of evidence in the literature.
Nutrients 2019,11, 2975 10 of 16
The same holds true for physical activity, for which the definition of a dose-response correlation
between duration or type of exercise and eects on T would be relevant.
Author Contributions:
Conceptualization, V.A.G., C.P. and G.D.P.; resources, V.A.G., C.P. and G.D.P.;
writing—Original draft preparation, V.A.G., C.P. and G.D.P.; writing—Review and editing, M.C.; supervision, V.T.,
I.M. and E.G.
Funding: This research received no external funding.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Astrup, A.; Rossner, S.; Finer, N.; Van Gaal, L. Obesity in Europe—Does anybody care? Expert Opin.
Pharmacother. 2013,14, 971–973. [CrossRef] [PubMed]
2.
Flegal, K.M.; Kit, B.K.; Orpana, H.; Graubard, B.I. Association of all-cause mortality with overweight and
obesity using standard body mass index categories: A systematic review and meta-analysis. JAMA
2013
,
309, 71–82. [CrossRef] [PubMed]
3.
von Lengerke, T.; Krauth, C. Economic costs of adult obesity: A review of recent European studies with
a focus on subgroup-specific costs. Maturitas 2011,69, 220–229. [CrossRef] [PubMed]
4.
De Pergola, G.; Silvestris, F. Obesity as a major risk factor for cancer. J. Obes.
2013
,2013, 291546. [CrossRef]
5.
Resta, O.; Foschino-Barbaro, M.P.; Legari, G.; Talamo, S.; Bonfitto, P.; Palumbo, A.; Minenna, A.; Giorgino, R.;
De Pergola, G. Sleep-related breathing disorders, loud snoring and excessive daytime sleepiness in obese
subjects. Int. J. Obes. Relat. Metab. Disord. 2001,25, 669. [CrossRef]
6.
Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastrebo, A.M.; Nadolsky, K.;
Pessah-Pollack, R.; Plodkowski, R. Reviewers of the AACE/ACE obesity clinical practice guidelines. American
Association of Clinical Endocrinologists and American College of Endocrinology comprehensive clinical
practice guidelines for medical care of patients with obesity. Endocr. Pract. 2016,22, 1–203. [CrossRef]
7.
Chambers, T.J.; Richard, R.A. The impact of obesity on male fertility. Hormones
2015
,14, 563–568. [CrossRef]
8.
Davidson, L.M.; Millar, K.; Jones, C.; Fatum, M.; Coward, K. Deleterious eects of obesity upon the hormonal
and molecular mechanisms controlling spermatogenesis and male fertility. Hum. Fertil.
2015
,18, 184–193.
[CrossRef]
9.
Bieniek, J.M.; Kashanian, J.A.; Deibert, C.M.; Grober, E.D.; Lo, K.C.; Brannigan, R.E.; Sandlow, J.I.;
Jarvi, K.A. Influence of increasing body mass index on semen and reproductive hormonal parameters
in a multi-institutional cohort of subfertile men. Fertil. Steril. 2016,106, 1070–1075. [CrossRef]
10.
Svartberg, J.; von Muhlen, D.; Sundsfjord, J.; Jorde, R. Waist circumference and testosterone levels in
community dwelling men. The Tromso study. Eur. J. Epidemiol. 2004,19, 657–663. [CrossRef]
11.
Balkau, B.; Deanfield, J.E.; D
é
pres, J.P.; Bassand, J.P.; Fox, K.A.; Smith, S.C.; Barter, P.; Tan, C.E.; Van Gaal, L.;
Wittchen, H.U.; et al. International day for the evaluation of abdominal obesity (IDEA): A study of waist
circumference, cardiovascular disease, and diabetes mellitus in 168,000 primary care patients in 63 countries.
Circulation 2007,116, 1942–1951. [CrossRef] [PubMed]
12.
Lopez, D.S.; Qiu, X.; Advani, S.; Tsilidis, K.K.; Khera, M.; Kim, J.; Morgentaler, A.; Wang, R.; Canfield, S.
Double trouble: Co-occurrence of testosterone deficiency and body fatness associated with all-cause mortality
in US men. Clin. Endocrinol. 2018,88, 58–65. [CrossRef] [PubMed]
13.
Durrer Schutz, D.; Busetto, L.; Dicker, D.; Farpour-Lambert, N.; Pryke, R.; Toplak, H.; Widmer, D.; Yumuk, V.;
Schutz, Y. European practical and patient-centred guidelines for adult obesity management in primary care.
Obes. Facts 2019,12, 40–66. [CrossRef] [PubMed]
14.
Tajar, A.; Forti, G.; O’Neill, T.W.; Lee, D.M.; Silman, A.J.; Finn, J.D.; Bartfai, G.; Boonen, S.; Casanueva, F.F.;
Giwercman, A.; et al. Characteristics of secondary, primary, and compensated hypogonadism in aging men:
Evidence from the European Male Ageing Study. J. Clin. Endocrinol. Metab.
2010
,95, 1810–1818. [CrossRef]
15.
Grossmann, M.; Matsumoto, A.M. A perspective on middle-aged and older men with functional
hypogonadism: Focus on holistic management. J. Clin. Endocrinol. Metab.
2017
,102, 1067–1075. [CrossRef]
16.
Bhasin, S.; Brito, J.P.; Cunningham, G.R.; Hayes, F.J.; Hodis, H.N.; Matsumoto, A.M.; Snyder, P.J.;
Swerdlo, R.S.; Wu, F.C.; Yialamas, M.A. Testosterone therapy in men with hypogonadism: An Endocrine
Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2018,103, 1715–1744. [CrossRef]
Nutrients 2019,11, 2975 11 of 16
17.
Grossmann, M. Hypogonadism and male obesity: Focus on unresolved questions. Clin. Endocrinol. (Oxf.)
2018,89, 11–21. [CrossRef]
18.
Wu, F.C.; Tajar, A.; Pye, S.R.; Silman, A.J.; Finn, J.D.; O’Neill, T.W.; Bartfai, G.; Casanueva, F.; Forti, G.;
Giwercman, A.; et al. Hypothalamic-pituitary-testicular axis disruptions in older men are dierentially
linked to age and modifiable and modifiable risk factors: The European Male Aging Study. J. Clin. Endocrinol.
Metab. 2008,93, 2737–2745. [CrossRef]
19.
Haring, R.; Volzke, H.; Felix, S.B.; Schipf, S.; Dorr, M.; Rosskopf, D.; Nauck, M.; Schofl, C.; Wallaschetofsky, H.
Prediction of metabolic syndrome by low serum testosterone levels in men: Results from the study of health
in Pomerania. Diabetes 2009,58, 2027–2031. [CrossRef]
20.
Zitzmann, M. Testosterone deficiency, insulin resistance and the metabolic syndrome. Nat. Rev. Endocrinol.
2009,5, 673–681. [CrossRef]
21.
Gates, M.A.; Mekary, R.A.; Chiu, G.R.; Ding, E.L.; Wittert, G.A.; Araujo, A.B. Sex steroid hormone levels and
body composition in men. J. Clin. Endocrinol. Metab. 2013,98, 2442–2450. [CrossRef]
22.
Camacho, E.M.; Huhtaniemi, I.T.; O’Neill, T.W.; Finn, J.D.; Pye, S.R.; Lee, D.M.; Tajar, A.; Bartfai, G.; Boonen, S.;
Casanueca, F.F.; et al. Age-associated changes in hypothalamic-pituitary-testicular function in middle-aged
and older men are modified by weight change and lifestyle factors: Longitudinal results from the European
Male Ageing Study. Eur. J. Epidemiol. 2013,168, 445–455. [CrossRef]
23.
Saboor Aftab, S.A.; Kumar, S.; Barber, T.M. The role of obesity and type 2 diabetes mellitus in the development
of male obesity-associated secondary hypogonadism. Clin. Endocrinol.
2013
,78, 330–337. [CrossRef]
[PubMed]
24.
Noko, N.; Thurston, J.; Hilkin, A.; Pyle, L.; Zeitler, P.S.; Nadeau, K.J.; Santoro, N.; Kelsey, M.M. Sex
Dierences in Eects of Obesity on Reproductive Hormones and Glucose Metabolism in Early Puberty.
J. Clin. Endocrinol. Metab. 2019,104, 4390–4397. [CrossRef] [PubMed]
25.
Giagulli, V.A.; Kaufman, J.M.; Vermeulen, A. Pathogenesis of the decreased androgen levels in obese men.
J. Clin. Endocrinol. Metab. 1994,79, 997–1000.
26. Bray, G.A. Obesity and reproduction. Hum. Reprod. 1997,12, 26–32. [CrossRef]
27.
Baker, H.W. Reproductive eects of nontesticular illness. Endocrinol. Metab. Clin. N. Am.
1998
,27, 831–850.
[CrossRef]
28.
T’Sjoen, G.G.; Giagulli, V.A.; Delva, H.; Crabbe, P.; De Bacquer, D.; Kaufman, J.M. Comparative assessment
in young and elderly men of the gonadotropin response to aromatase inhibition. J. Clin. Endocrinol. Metab.
2005,90, 5717–5722. [CrossRef]
29.
Schneider, G.; Kirschner, M.A.; Berkowitz, R.; Ertel, N.H. Increased estrogen production in obese men. J. Clin.
Endocrinol. Metab. 1979,48, 633–638. [CrossRef]
30.
Kovac, J.R. Reproductive endocrinology: Oral enclomiphene citrate in obese men with hypogonadism. Nat.
Rev. Urol. 2016,13, 133–134. [CrossRef]
31.
Loves, S.; Ruinemans-Koerts, J.; de Boer, H. Letrozole once a week normalizes serum testosterone in
obesity-related male hypogonadism. Eur. J. Endocrinol. 2008,158, 741–747. [CrossRef]
32.
Feldman, H.A.; Longcope, C.; Derby, C.A.; Johannes, C.B.; Araujo, A.B.; Coviello, A.D.; Bremner, W.J.;
McKinlay, J.B. Age trends in the level of serum testosterone and other hormones in middle-aged men:
Longitudinal results from the Massachusetts male aging study. J. Clin. Endocrinol. Metab.
2002
,87, 589–598.
[CrossRef]
33.
Dhindsa, S.; Furlanetto, R.; Vora, M.; Ghanim, H.; Chaudhuri, A.; Dandona, P. Low estradiol concentrations
in men with subnormal testosterone concentrations and type 2 diabetes. Diabetes Care
2011
,34, 1854–1859.
[CrossRef] [PubMed]
34.
MacDonald, P.C.; Madden, J.D.; Brenner, P.F.; Wilson, J.D.; Siiteri, P.K. Origin of estrogen in normal men and
in women with testicular feminization. J. Clin. Endocrinol. Metab. 1979,49, 905–916. [CrossRef]
35.
Wu, A.; Shi, Z.; Martin, S.; Vincent, A.; Heilbronn, L.; Wittert, G. Age-related changes in estradiol and
longitudinal associations with fat mass in men. PLoS ONE 2018,13, e0201912. [CrossRef]
36.
Swerdlo, R.S.; Dudley, R.E.; Page, S.T.; Wang, C.; Salameh, W.A. Dihydrotestosterone: Biochemistry,
physiology, and clinical implications of elevated blood levels. Endocr. Rev. 2017,38, 220–254. [CrossRef]
37.
Gambineri, A.; Pelusi, C. Sex hormones, obesity and type 2 diabetes: Is there a link? Endocr. Connect.
2019
,8,
R1–R9. [CrossRef]
Nutrients 2019,11, 2975 12 of 16
38.
Burcelin, R.; Thorens, B.; Glauser, M.; Gaillard, R.C.; Pralong, F.P. Gonadotropin-releasing hormone secretion
from hypothalamic neurons: Stimulation by insulin and potentiation by leptin. Endocrinology
2003
,144,
4484–4491. [CrossRef]
39.
Pitteloud, N.; Hardin, M.; Dwyer, A.A.; Valassi, E.; Yialamas, M.; Elahi, D.; Hayes, F.J. Increasing insulin
resistance is associated with a decrease in Leydig cell testosterone secretion in men. J. Clin. Endocrinol. Metab.
2005,90, 2636–2641. [CrossRef]
40.
Salvi, R.; Castillo, E.; Voirol, M.J.; Glauser, M.; Rey, J.P.; Gaillard, R.C.; Vollenweider, P.; Pralong, F.P.
Gonadotropin-releasing hormone-expressing neurons immortalized conditionally are activated by insulin:
Implication of the mitogen-activated protein kinase pathway. Endocrinology 2006,147, 816–826. [CrossRef]
41.
Dhindsa, S.; Ghanim, H.; Batra, M.; Dandona, P. Hypogonadotropic hypogonadism in men with diabesity.
Diabetes Care 2018,41, 1516–1525. [CrossRef] [PubMed]
42.
Pivonello, R.; Menafra, D.; Riccio, E.; Garifalos, F.; Mazzella, M.; de Angelis, C.; Colao, A. Metabolic disorders
and male hypogonadotropic hypogonadism. Front. Endocrinol. (Lausanne)
2019
,10, 345. [CrossRef] [PubMed]
43.
Bellentani, S.; Scaglioni, F.; Marino, M.; Bedogni, G. Epidemiology of non-alcoholic fatty liver disease. Dig.
Dis. 2010,28, 155–161. [CrossRef] [PubMed]
44.
Giagulli, V.A.; Verdonck, L.; Deslypere, J.P.; Giorgino, R.; Vermeulen, A. Comparison of plasma androgen
glucuronide levels after percutaneous or peroral androgen treatment in men: Evidence for important
splanchnic contribution to plasma 17 beta-hydroxyandrogen glucuronides. J. Clin. Endocrinol. Metab.
1993
,
76, 429–431. [PubMed]
45.
Grossmann, M.; Wierman, M.E.; Angus, P.; Handelsman, D.J. Reproductive endocrinology of nonalcoholic
fatty liver disease. Endocr. Rev. 2019,40, 417–446. [CrossRef] [PubMed]
46.
Jaruvongvanich, V.; Sanguankeo, A.; Riangwiwat, T.; Upala, S. Testosterone, sex hormone-binding globulin
and nonalcoholic fatty liver disease: A systematic review and meta-analysis. Ann. Hepatol.
2017
,16, 382–394.
[CrossRef] [PubMed]
47.
Sarchielli, E.; Comeglio, P.; Squecco, R.; Ballerini, L.; Mello, T.; Guarnieri, G.; Idrizaj, E.; Mazzanti, B.;
Vignozzi, L.; Gallina, P.; et al. Tumor Necrosis Factor-
α
impairs kisspeptin signaling in human
gonadotropin-releasing hormone primary neurons. J. Clin. Endocrinol. Metab. 2017,102, 46–56. [CrossRef]
48.
Van de Velde, F.; Bekaert, M.; Hoorens, A.; Geerts, A.; T’Sjoen, G.; Fiers, T.; Kaufman, J.M.; Van
Nieuwenhove, Y.; Lapauw, B. Histologically proven hepatic steatosis associates with lower testosterone
levels in men with obesity. Asian J. Androl. 2019. [CrossRef]
49.
Gianatti, E.J.; Grossmann, M. Testosterone deficiency in men with type 2 diabetes: Pathophysiology and
treatment. Diabet. Med. 2019. [CrossRef]
50.
Rastrelli, G.; Filippi, S.; Sforza, A.; Maggi, M.; Corona, G. Metabolic syndrome in male hypogonadism. Front.
Horm. Res. 2018,49, 131–155. [CrossRef]
51.
Kelesidis, T.; Kelesidis, I.; Chou, S.; Mantzoros, C.S. Narrative review: The role of leptin in human physiology:
Emerging clinical applications. Ann. Intern. Med. 2010,152, 93–100. [CrossRef]
52.
Quennell, J.H.; Mulligan, A.C.; Tups, A.; Liu, X.; Phipps, S.J.; Kemp, C.J.; Herbison, A.E.; Grattan, D.R.;
Anderson, G.M. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology
2009,150, 2805–2812. [CrossRef] [PubMed]
53.
George, J.T.; Veldhuis, J.D.; Tena-Sempere, M.; Millar, R.P.; Anderson, R.A. Exploring the pathophysiology of
hypogonadism in men with type 2 diabetes: Kisspeptin-10 stimulates serum testosterone and LH secretion
in men with type 2 diabetes and mild biochemical hypogonadism. Clin. Endocrinol. (Oxf.)
2013
,79, 100–104.
[CrossRef] [PubMed]
54.
Isidori, A.M.; Caprio, M.; Strollo, F.; Moretti, C.; Frajese, G.; Isidori, A.; Fabbri, A. Leptin and androgens in
male obesity: Evidence for leptin contribution to reduced androgen levels. J. Clin. Endocrinol. Metab.
1999
,
84, 3673–3680. [CrossRef] [PubMed]
55.
Tena-Sempere, M.; Pinilla, L.; Gonz
á
lez, L.C.; Di
é
guez, C.; Casanueva, F.F.; Aguilar, E. Leptin inhibits
testosterone secretion from adult rat testis in vitro. J. Endocrinol. 1999,161, 211–218. [CrossRef]
56.
Ahima, R.S. Revisiting leptin’s role in obesity and weight loss. J. Clin. Investig.
2008
,118, 2380–2383.
[CrossRef]
Nutrients 2019,11, 2975 13 of 16
57.
S
á
nchez-Garrido, M.A.; Ruiz-Pino, F.; Manfredi-Lozano, M.; Leon, S.; Garcia-Galiano, D.; Castaño, J.P.;
Luque, R.M.; Romero-Ruiz, A.; Castellano, J.M.; Di
é
guez, C.; et al. Obesity-induced hypogonadism in the
male: Premature reproductive neuroendocrine senescence and contribution of Kiss1-mediated mechanisms.
Endocrinology 2014,155, 1067–1079. [CrossRef]
58.
Behre, H.M.; Simoni, M.; Nieschlag, E. Strong association between serum levels of leptin and testosterone in
men. Clin. Endocrinol. (Oxf.) 1997,47, 237–240. [CrossRef]
59.
Hedger, M.P.; Meinhardt, A. Cytokines and the immune-testicular axis. J. Reprod. Immunol.
2003
,58, 1–26.
[CrossRef]
60.
Guazzone, V.A.; Jacobo, P.; Theas, M.S.; Lustig, L. Cytokines and chemokines in testicular inflammation: A
brief review. Microsc. Res. Tech. 2009,72, 620–628. [CrossRef]
61.
Morelli, A.; Sarchielli, E.; Comeglio, P.; Filippi, S.; Vignozzi, L.; Marini, M.; Rastrelli, G.; Maneschi, E.; Cellai, I.;
Persani, L.; et al. Metabolic syndrome induces inflammation and impairs gonadotropin-releasing hormone
neurons in the preoptic area of the hypothalamus in rabbits. Mol. Cell. Endocrinol.
2014
,382, 107–119.
[CrossRef] [PubMed]
62.
Watanobe, H.; Hayakawa, Y. Hypothalamic interleukin-1 beta and tumor necrosis factor-alpha, but not
interleukin-6, mediate the endotoxin-induced suppression of the reproductive axis in rats. Endocrinology
2003,144, 4868–7485. [CrossRef] [PubMed]
63.
Russell, S.H.; Small, C.J.; Stanley, S.A.; Franks, S.; Ghatei, M.A.; Bloom, S.R. The
in vitro
role of tumour
necrosis factor-alpha and interleukin-6 in the hypothalamic-pituitary gonadal axis. J. Neuroendocrinol.
2001
,
13, 296–301. [CrossRef] [PubMed]
64.
Leisegang, K.; Henkel, R. The
in vitro
modulation of steroidogenesis by inflammatory cytokines and insulin
in TM3 Leydig cells. Reprod. Biol. Endocrinol. 2018,16, 26. [CrossRef]
65.
Fan, W.; Xu, Y.; Liu, Y.; Zhang, Z.; Lu, L.; Ding, Z. Obesity or overweight, a chronic inflammatory status in
male reproductive system, leads to mice and human subfertility. Front. Physiol. 2018,8, 1117. [CrossRef]
66.
Syriou, V.; Papanikolaou, D.; Kozyraki, A.; Goulis, D.G. Cytokines and male infertility. Eur. Cytokine Netw.
2018,29, 73–82. [CrossRef]
67.
Ebrahimi, F.; Urwyler, S.A.; Straumann, S.; Doerpfeld, S.; Bernasconi, L.; Neyer, P.; Schuetz, P.; Mueller, B.;
Donath, M.Y.; Christ-Crain, M. IL-1 antagonism in men with metabolic syndrome and low testosterone: A
randomized clinical trial. J. Clin. Endocrinol. Metab. 2018,103, 3466–3476. [CrossRef]
68.
Navarro, G.; Xu, W.; Jacobson, D.A.; Wicksteed, B.; Allard, C.; Zhang, G.; De Gendt, K.; Kim, S.H.; Wu, H.;
Zhang, H.; et al. Extranuclear actions of the androgen receptor enhance glucose-stimulated insulin secretion
in the male. Cell Metab. 2016,23, 837–851. [CrossRef]
69.
Lin, H.Y.; Yu, I.C.; Wang, R.S.; Chen, Y.T.; Liu, N.C.; Altuwaijri, S.; Hsu, C.L.; Ma, W.L.; Jokinen, J.; Sparks, J.D.;
et al. Increased hepatic steatosis and insulin resistance in mice lacking hepatic androgen receptor. Hepatology
2008,47, 1924–1935. [CrossRef]
70.
MacLean, H.E.; Chiu, W.S.; Notini, A.J.; Axell, A.M.; Davey, R.A.; McManus, J.F.; Ma, C.; Plant, D.R.;
Lynch, G.S.; Zajac, J.D. Impaired skeletal muscle development and function in male, but not female, genomic
androgen receptor knockout mice. FASEB J. 2008,22, 2676–2689. [CrossRef]
71.
Rodriguez-Cuenca, S.; Monjo, M.; Proenza, A.M.; Roca, P. Depot dierences in steroid receptor expression
in adipose tissue: Possible role of the local steroid milieu. Am. J. Physiol. Endocrinol. Metab.
2005
,288,
E200–E207. [CrossRef] [PubMed]
72.
Yu, I.C.; Lin, H.Y.; Liu, N.C.; Wang, R.S.; Sparks, J.D.; Yeh, S.; Chang, C. Hyperleptinemia without obesity
in male mice lacking androgen receptor in adipose tissue. Endocrinology
2008
,149, 2361–2368. [CrossRef]
[PubMed]
73.
McInnes, K.J.; Smith, L.B.; Hunger, N.I.; Saunders, P.T.; Andrew, R.; Walker, B.R. Deletion of the androgen
receptor in adipose tissue in male mice elevates retinol binding protein 4 and reveals independent eects on
visceral fat mass and on glucose homeostasis. Diabetes 2012,61, 1072–1081. [CrossRef]
74.
Yu, I.C.; Lin, H.Y.; Liu, N.C.; Sparks, J.D.; Yeh, S.; Fang, L.Y.; Chen, L.; Chang, C. Neuronal androgen receptor
regulates insulin sensitivity via suppression of hypothalamic NF-
κ
B-mediated PTP1B expression. Diabetes
2013,62, 411–423. [CrossRef] [PubMed]
75.
Kelly, D.M.; Jones, T.H. Testosterone: A metabolic hormone in health and disease. J. Endocrinol.
2013
,217,
R25–R45. [CrossRef] [PubMed]
Nutrients 2019,11, 2975 14 of 16
76.
Gyawali, P.; Martin, S.A.; Heilbronn, L.K.; Vincent, A.D.; Taylor, A.W.; Adams, R.J.T.; O’Loughlin, P.D.;
Wittert, G.A. The role of sex hormone-binding globulin (SHBG), testosterone, and other sex steroids, on
the development of type 2 diabetes in a cohort of community-dwelling middle-aged to elderly men. Acta
Diabetol. 2018,55, 861–872. [CrossRef]
77.
Hamilton, E.J.; Gianatti, E.; Strauss, B.J.; Wentworth, J.; Lim-Joon, D.; Bolton, D.; Zajac, J.D.; Grossmann, M.
Increase in visceral and subcutaneous abdominal fat in men with prostate cancer treated with androgen
deprivation therapy. Clin. Endocrinol. 2011,74, 377–383. [CrossRef]
78.
Saylor, P.J.; Smith, M.R. Metabolic complications of androgen deprivation therapy for prostate cancer. J. Urol.
2013,189, S34–S42. [CrossRef]
79. Hackett, G. Type 2 diabetes and testosterone therapy. World J. Men’s Health 2019,37, 31–44. [CrossRef]
80.
Traish, A.M.; Kypreos, K.E. Testosterone and cardiovascular disease: An old idea with modern clinical
implications. Atherosclerosis 2011,214, 244–248. [CrossRef]
81.
Corona, G.; Rastrelli, G.; Monami, M.; Saad, F.; Luconi, M.; Lucchese, M.; Facchiano, E.; Sforza, A.; Forti, G.;
Mannucci, E.; et al. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: A
systematic review and meta-analysis. Eur. J. Endocrinol. 2013,168, 829–843. [CrossRef] [PubMed]
82.
Mora, M.; Aranda, G.B.; de Hollanda, A.; Flores, L.; Puig-Domingo, M.; Vidal, J. Weight loss is a major
contributor to improved sexual function after bariatric surgery. Surg. Endosc.
2013
,27, 3197–3204. [CrossRef]
[PubMed]
83.
Stanik, S.; Dornfeld, L.P.; Maxwell, M.H.; Viosca, S.P.; Korenman, S.G. The eect of weight loss on reproductive
hormones in obese men. J. Clin. Endocrinol. Metab. 1981,53, 828–832. [CrossRef] [PubMed]
84.
Kaukua, J.; Pekkarinen, T.; Sane, T.; Mustajoki, P. Sex hormones and sexual function in obese men losing
weight. Obes. Res. 2003,11, 689–694. [CrossRef]
85.
Khoo, J.; Piantadosi, C.; Worthley, S.; Wittert, G.A. Eects of a low energy diet on sexual function and lower
urinary tract symptoms in obese men. Int. J. Obes. 2010,34, 1396–1403. [CrossRef]
86.
Khoo, J.; Piantadosi, C.; Duncan, R.; Worthley, S.G.; Jenkins, A.; Noakes, M.; Worthley, M.I.; Lange, K.;
Wittert, G.A. Comparing eects of a low-energy diet and a high-protein low-fat diet on sexual and endothelial
function, urinary tract symptoms, and inflammation in obese diabetic men. J. Sex. Med.
2011
,8, 2868–2875.
[CrossRef]
87.
Niskanen, L.; Laaksonen, D.E.; Punnonen, K.; Mustajoki, P.; Kaukua, J.; Rissanen, A. Changes in sex
hormone-binding globulin and testosterone during weight loss and weight maintenance in abdominally
obese men with the metabolic syndrome. Diabetes Obes. Metab. 2004,6, 208–215. [CrossRef]
88.
Corona, G.; Rastrelli, G.; Morelli, A.; Sarchielli, E.; Cipriani, S.; Vignozzi, L.; Maggi, M. Treatment of functional
hypogonadism besides pharmacological substitution. World J. Men’s Health 2019. [CrossRef]
89.
Howie, B.J.; Shultz, T.D. Dietary and hormonal interrelationships among vegetarian Seventh-Day Adventists
and nonvegetarian men. Am. J. Clin. Nutr. 1985,42, 127–134. [CrossRef]
90.
B
é
langer, A.; Locong, A.; Noel, C.; Cusan, L.; Dupont, A.; Pr
é
vost, J.; Caron, S.; S
é
vigny, J. Influence of diet on
plasma steroids and sex hormone-binding globulin levels in adult men. J. Steroid. Biochem.
1989
,32, 829–833.
[CrossRef]
91.
Key, T.J.; Roe, L.; Thorogood, M.; Moore, J.W.; Clark, G.M.; Wang, D.Y. Testosterone, sex hormone-binding
globulin, calculated free testosterone, and oestradiol in male vegans and omnivores. Br. J. Nutr.
1990
,64,
111–119. [CrossRef] [PubMed]
92.
Varlamov, O. Western-style diet, sex steroids and metabolism. Biochim. Biophys. Acta Mol. Basis Dis.
2017
,
1863, 1147–1155. [CrossRef] [PubMed]
93.
Kumagai, H.; Yoshikawa, T.; Zempo-Miyaki, A.; Myoenzono, K.; Tsujimoto, T.; Tanaka, K.; Maeda, S. Vigorous
physical activity is associated with regular aerobic exercise-induced increased serum testosterone levels in
overweight/obese men. Horm. Metab. Res. 2018,50, 73–79. [CrossRef] [PubMed]
94.
Armamento-Villareal, R.; Aguirre, L.E.; Qualls, C.; Villareal, D.T. Eect of lifestyle intervention on the
hormonal profile of frail, obese older men. J. Nutr. Health Aging 2016,20, 334–340. [CrossRef] [PubMed]
95.
Di Daniele, N.; Noce, A.; Vidiri, M.F.; Moriconi, E.; Marrone, G.; Annicchiarico-Petruzzelli, M.; D’Urso, G.;
Tesauro, M.; Rovella, V.; De Lorenzo, A. Impact of mediterranean diet on metabolic syndrome, cancer and
longevity. Oncotarget 2017,8, 8947–8979. [CrossRef] [PubMed]
Nutrients 2019,11, 2975 15 of 16
96.
De Lorenzo, A.; Noce, A.; Moriconi, E.; Rampello, T.; Marrone, G.; Di Daniele, N.; Rovella, V. MOSH syndrome
(male obesity secondary hypogonadism): Clinical asssessment and possible therapeutic approaches. Nutrients
2018,10, 474. [CrossRef]
97.
Kim, C.; Barrett-Connor, E.; Aroda, V.R.; Mather, K.J.; Christophi, C.A.; Horton, E.S.; Pi-Sunyer, X.; Bray, G.A.;
Labrie, F.; Golden, S.H. Diabetes Prevention Program Research Group. Testosterone and depressive symptoms
among men in the Diabetes Prevention Program. Psychoneuroendocrinology 2016,72, 63–71. [CrossRef]
98.
Hakonsen, L.B.; Thulstrup, A.M.; Aggerholm, A.S.; Olsen, J.; Bonde, J.P.; Andersen, C.Y.; Bungum, M.;
Ernst, E.H.; Hansen, M.L.; Ernst, E.H.; et al. Does weight loss improve semen quality and reproductive
hormones? Results from a cohort of severely obese men. Reprod. Health 2011,8, 24. [CrossRef]
99.
Hildreth, K.L.; Barry, D.W.; Moreau, K.L.; Vande Griend, J.; Meacham, R.B.; Nakamura, T.; Wolfe, P.;
Kohrt, W.M.; Ruscin, J.M.; Kittelson, J.; et al. Eects of testosterone and progressive resistance exercise in
healthy, highly functioning older men with low-normal testosterone levels. J. Clin. Endocrinol. Metab.
2013
,
98, 1891–1900. [CrossRef]
100.
Hoyos, C.M.; Killick, R.; Yee, B.J.; Grunstein, R.R.; Liu, P.Y. Eects of testosterone therapy on sleep and
breathing in obese men with severe obstructive sleep apnoea: A randomized placebo-controlled trial. Clin.
Endocrinol. 2012,77, 599–607. [CrossRef]
101.
Ng Tang Fui, M.; Hoermann, R.; Prendergast, L.A.; Zajac, J.D.; Grossmann, M. Symptomatic response to
testosterone treatment in dieting obese men with low testosterone levels in a randomized, placebo-controlled
clinical trial. Int. J. Obes. 2017,41, 420–426. [CrossRef] [PubMed]
102.
Ng Tang Fui, M.; Hoermann, R.; Zajac, J.D.; Grossmann, M. The eects of testosterone on body composition
in obese men are not sustained after cessation of testosterone treatment. Clin. Endocrinol.
2017
,87, 336–343.
[CrossRef] [PubMed]
103.
Yassin, A.; Almehmadi, Y.; Saad, F.; Doros, G.; Gooren, L. Eects of intermission and resumption of long-term
testosterone replacement therapy on body weight and metabolic parameters in hypogonadal in middle-aged
and elderly men. Clin. Endocrinol. 2016,84, 107–114. [CrossRef] [PubMed]
104.
Krasno, J.B.; Basaria, S.; Pencina, M.J.; Jasuja, G.K.; Vasan, R.S.; Ulloor, J.; Zhang, A.; Coviello, A.;
Kelly-Hayes, M.; D’Agostino, R.B.; et al. Free testosterone levels are associated with mobility limitation and
physical performance in community-dwelling men: The Framingham Ospring Study. J. Clin. Endocrinol.
Metab. 2010,95, 2790–2799. [CrossRef]
105.
Snyder, P.J.; Bhasin, S.; Cunningham, G.R.; Matsumoto, A.M.; Stephens-Shields, A.J.; Cauley, J.A.; Gill, T.M.;
Barrett-Connor, E.; Swerdlo, R.S.; Wang, C.; et al. Testosterone Trials Investigators. Eects of Testosterone
Treatment in Older Men. N. Engl. J. Med. 2016,374, 611–624. [CrossRef]
106.
Nam, Y.S.; Lee, G.; Yun, J.M.; Cho, B. Testosterone replacement, muscle strength, and physical function.
World J. Men’s Health 2018,36, 110–122. [CrossRef]
107.
Storer, T.W.; Basaria, S.; Traustadottir, T.; Harman, S.M.; Pencina, K.; Li, Z.; Travison, T.G.; Miciek, R.;
Tsitouras, P.; Hally, K.; et al. Eects of testosterone supplementation for 3 years on muscle performance and
physical function in older men. J. Clin. Endocrinol. Metab. 2017,102, 583–593. [CrossRef]
108.
Haider, A.; Saad, F.; Doros, G.; Louis Gooren, L. Hypogonadal obese men with and without diabetes mellitus
type 2 lose weight and show improvement in cardiovascular risk factors when treated with testosterone:
An observational study. Obes. Res. Clin. Pract. 2013,8, e339–e349. [CrossRef]
109.
Dhindsa, S.; Miller, M.G.; McWhirter, C.L.; Mager, D.E.; Ghanim, H.; Chaudhuri, A.; Dandona, P. Testosterone
concentrations in diabetic and nondiabetic obese men. Diabetes Care 2010,33, 1186–1192. [CrossRef]
110.
Kalinchenko, S.Y.; Tishova, Y.A.; Mskhalaya, G.J.; Gooren, L.J.; Giltay, E.J.; Saad, F. Eects of testosterone
supplementation on markers of the metabolic syndrome and inflammation in hypogonadal men with the
metabolic syndrome: The double-blinded placebo-controlled Moscow study. Clin. Endocrinol.
2010
,73,
602–612. [CrossRef]
111.
Aversa, A.; Bruzziches, R.; Francomano, D.; Rosano, G.; Isidori, A.M.; Lenzi, A.; Spera, G. Eects of
testosterone undecanoate on cardiovascular risk factors and atherosclerosis in middle-aged men with
late-onset hypogonadism and metabolic syndrome: Results from a 24-month, randomized, double-blind,
placebo-controlled study. J. Sex. Med. 2010,7, 3495–3503. [CrossRef]
112.
Haring, R.; Völzke, H.; Steveling, A.; Krebs, A.; Felix, S.B.; Schöfl, C.; Dörr, M.; Nauck, M.; Wallaschofski, H.
Low serum testosterone levels are associated with increased risk of mortality in a population-based cohort of
men aged 20–79. Eur. Heart J. 2010,31, 1494–1501. [CrossRef]
Nutrients 2019,11, 2975 16 of 16
113.
Laaksonen, D.E.; Niskanen, L.; Punnonen, K.; Nyyssönen, K.; Tuomainen, T.P.; Valkonen, V.P.; Salonen, R.;
Salonen, J.T. Testosterone and sex hormone-binding globulin predict the metabolic syndrome and diabetes
in middle-aged men. Diabetes Care 2004,27, 1036–1041. [CrossRef] [PubMed]
114.
Corona, G.; Rastrelli, G.; Monami, M.; Guay, A.; Buvat, J.; Sforza, A.; Forti, G.; Mannucci, E.; Maggi, M.
Hypogonadism as a risk factor for cardiovascular mortality in men: A meta-analytic study. Eur. J. Endocrinol.
2011,165, 687–701. [CrossRef] [PubMed]
115.
Mulligan, T.; Frick, M.F.; Zuraw, Q.C.; Stemhagen, A.; McWhirter, C. Prevalence of hypogonadism in males
aged at least 45 years: The HIM study. Int. J. Clin. Pract. 2006,60, 762–769. [CrossRef]
116.
Francomano, D.; Ilacqua, A.; Bruzziches, R.; Lenzi, A.; Aversa, A. Eects of 5-year treatment with testosterone
undecanoate on lower urinary tract symptoms in obese men with hypogonadism and metabolic syndrome.
Urology 2014,83, 167–173. [CrossRef]
117.
Corona, G.; Giagulli, V.A.; Maseroli, E.; Vignozzi, L.; Aversa, A.; Zitzmann, M.; Saad, F.; Mannucci, E.;
Maggi, M. Testosterone supplementation and body composition: Results from a meta-analysis study. Eur. J.
Endocrinol. 2016,174, R99–R116. [CrossRef]
118.
Yassin, A.; Haider, A.; Haider, K.S.; Caliber, M.; Doros, G.; Saad, F.; Garvey, W.T. Testosterone therapy in
men with hypogonadism prevents progression from prediabetes to type 2 diabetes: Eight-year data from
a registry study. Diabetes Care 2019,42, 1104–1111. [CrossRef]
119.
Corona, G.; Rastrelli, G.; Di Pasquale, G.; Sforza, A.; Mannucci, E.; Maggi, M. Testosterone and cardiovascular
risk: Meta-Analysis of interventional studies. J. Sex. Med. 2018,15, 820–838. [CrossRef]
120.
Cunningham, G.R.; Ellenberg, S.S.; Bhasin, S.; Matsumoto, A.M.; Parsons, J.K.; Preston, P.; Cauley, J.A.;
Gill, T.M.; Swerdlo, R.S.; Wang, C.; et al. Prostate-Specific Antigen levels during testosterone treatment
of hypogonadal older men: Data from a controlled trial. J. Clin. Endocrinol. Metab.
2019
,104, 6238–6246.
[CrossRef]
121.
Bhasin, S.; Singh, A.B.; Mac, R.P.; Carter, B.; Lee, M.I.; Cunningham, G.R. Managing the risks of prostate
disease during testosterone replacement therapy in older men: Recommendations for a standardized
monitoring plan. J. Androl. 2003,24, 299–311. [CrossRef] [PubMed]
122.
Jahn, J.L.; Giovannucci, E.L.; Stampfer, M.J. The high prevalence of undiagnosed prostate cancer at autopsy:
Implications for epidemiology and treatment of prostate cancer in the Prostate-specific Antigen-era. Int. J.
Cancer 2015,137, 2795–2802. [CrossRef] [PubMed]
123.
Fenton, J.J.; Weyrich, M.S.; Durbin, S.; Liu, Y.; Bang, H.; Melnikow, J. Prostate-Specific Antigen-based
screening for prostate cancer: Evidence report and systematic review for the US Preventive Services Task
Force. JAMA 2018,319, 1914–1931. [CrossRef] [PubMed]
124.
US Preventive Services Task Force; Grossman, D.C.; Curry, S.J.; Owens, D.K.; Bibbins-Domingo, K.;
Caughey, A.B.; Davidson, K.W.; Doubeni, C.A.; Ebell, M.; Epling, J.W., Jr.; et al. Screening for prostate cancer:
US Preventive Services Task Force Recommendation Statement. JAMA 2018,319, 1901–1913. [PubMed]
©
2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... A healthy diet and physical activity increase the concentration of sperms and their normal morphology and motility in healthy men 5 . Besides, diet and physical activity help to improve hypogonadism in men, irrespective of age 6 . In addition, various phytochemical compounds rich in the Mediterranean diet help counter environmental contaminants' adverse effects 7 . ...
Article
Full-text available
Age is a known determinant of reproductive health and fertility in both genders. The present work aims to assess the reproductive hormone profile of a middle-aged and elderly man. For this descriptive cross-sectional study, healthy male subjects (n=77) were recruited from the valley. Any individual suffering from any acute or chronic diseases and on drugs was ruled out from the study. Group A consisted of 40 elderly men between 60-70 years of age, and Group B comprised 37 men between 35-46 years of age. Blood samples were taken to estimate the reproductive hormone profile. Level of oxidant and antioxidant: Malondialdehyde and Glutathione. The demographic variables, which included retrospective and prospective questions, helped to assess the physical activity and diet intake behaviour of all inducted individuals. The analysis of the reproductive profile of both groups was similar and within the normal range of standards. However, the median level of LH was higher in group A than in group B: 6.7 mIU/ml versus 3.4 mIU/ml, respectively, and p<0.003. Both groups showed predominantly involvement in physical activity, >90%. The correlation of biochemical variables gives an insight into the fact that the Mediterranean diet and physical activity help to maintain a normal BMI. These implicate the normal secretion of various hormones, leading to intact spermatogenesis. We can safely deduce from this study that physically active lifestyles and a healthy diet are crucial factors in maintaining an endocrine profile. (Afr J Reprod Health 2024; 28 [4]: 22-29). Résumé L'âge est un déterminant connu de la santé reproductive et de la fécondité chez les deux sexes. Le présent travail vise à évaluer le profil hormonal de la reproduction d'un homme d'âge moyen et âgé. Pour cette étude transversale descriptive, des sujets masculins en bonne santé (n = 77) ont été recrutés dans la vallée. Toute personne souffrant de maladies aiguës ou chroniques et prenant des médicaments a été exclue de l'étude. Le groupe A était composé de 40 hommes âgés de 60 à 70 ans et le groupe B de 37 hommes âgés de 35 à 46 ans. Des échantillons de sang ont été prélevés pour estimer le profil des hormones reproductives. Niveau d'oxydant et d'antioxydant : Malondialdéhyde et Glutathion. Les variables démographiques, qui comprenaient des questions rétrospectives et prospectives, ont permis d'évaluer l'activité physique et le comportement alimentaire de tous les individus intronisés. L'analyse du profil reproducteur des deux groupes était similaire et se situait dans la fourchette normale des normes. Cependant, le taux médian de LH était plus élevé dans le groupe A que dans le groupe B : respectivement 6,7 mUI/ml versus 3,4 mUI/ml et p<0,003. Les deux groupes présentaient une participation prédominante à l'activité physique, > 90 %. La corrélation des variables biochimiques donne un aperçu du fait que le régime méditerranéen et l'activité physique contribuent à maintenir un IMC normal. Celles-ci impliquent la sécrétion normale de diverses hormones, conduisant à une spermatogenèse intacte. Nous pouvons déduire de cette étude qu'un mode de vie physiquement actif et une alimentation saine sont des facteurs cruciaux pour maintenir un profil endocrinien. (Afr J Reprod Health 2024; 28 [4]: 22-29).
... Numerous epidemiological studies have shown a negative correlation between obesity and T levels, and several meta-analyses have shown that weight loss produces a proportional increase in T concentrations. It is generally accepted that hypogonadism secondary to obesity is functional, as it is reversible after weight loss [41]. ...
... Lifestyle changes have been shown to reduce adipose tissue burden, decrease the in ammatory response 18 and suppress peripheral aromatization. 19 Controlling the metabolic status and body weight has been shown a proportional increase of serum testosterone. 20 Although many studies showed a relationship between obesity and ED, only a few studies have demonstrated that the VAI could homogeneously predict adipose tissue dysfunction than BMI and waist circumference. ...
Preprint
Full-text available
The metabolic syndrome (MeTS) negatively affects male sexual function. Lifestyle changes improve erectile function. However, the effects of lifestyle changes on sexual function in men with and without MeTS on the visceral adiposity index (VAI) have not been adequately investigated. Patients with MeTS (n = 48) and those with no MeTS (n = 31) were recruited. Sexual functions and psychogenic status were assessed using the International Index of Erectile Dysfunction (IIEF) questionnaire and Beck’s depression inventory (BDI), respectively. The VAI was calculated using the gender-specific formula. Demographic, clinical, biochemical and hormonal data were recorded. Participants were advised to 8500 steps per day and dietary control. At least three months later, demographic and biochemical tests were repeated, and the VAI was calculated. Association between sexual functions, presence of MeTS and lifestyle changes via the VAI were analyzed. Each integer increase of the VAI was related to a 1.74-fold increased risk of ED in the MeTS group. The MeTS was related to moderate to severe ED than those with no MeTS (p = 0.033). A 24% decrease of the VAI was recorded in the MeTS group at the end of the study (p = 0.014). The MeTS criteria were disappeared in eight patients at the end of the study, with a slight decrease of VAI (p = 0.07). In the beginning, patients with MeTS and no-MeTS showed similar sexual satisfaction, orgasm, desire, and general satisfaction scores. Sexual satisfaction, orgasm and general satisfaction were improved in the non-MeTS group (p < 0.05). At the end of the study, the MeTS group showed similar sexual satisfaction, desire, orgasm and general satisfaction rates (p > 0.05). Higher VAI levels were related to the lower erectile function scores, particularly in patients with MeTS. Lifestyle changes improved male sexual function scores only in the non-MeTS group. Therefore, fighting against MeTS should be adopted as a global healthcare policy.
... AT plays a critical role in the development of FHH via both direct and indirect mechanisms. Directly, it induces androgen deprivation both by sequestering testosterone from the systemic circulation, and through its conversion to estradiol (48)(49)(50). Obese subjects, in fact, have an increased aromatase activity, responsible for testosterone conversion into estrogens. On the other hand, increased estrogen levels reduce LH pulse amplitude and may directly drive adipogenesis and enhance visceral, subcutaneous and ectopic fat deposition (51). ...
Article
Full-text available
Functional hypogonadotropic hypogonadism (FHH) is an increasingly frequent condition, whose pathological mechanisms are not yet fully clarified. The concept of FHH has now completely replaced that of late onset hypogonadism, that only concerned the ageing man. FHH is the result of an impairment of the hypothalamic-pituitary gonadal axis (HPG-A) function, resulting in decreased testosterone concentrations associated with low or inappropriately normal gonadotropin levels and infertility; it can be diagnosed once organic causes of hypogonadism are excluded. The growing occurrence of FHH derives from its association with widespread conditions, such as obesity and diabetes mellitus, but also to the increasing ease and frequency of use of several drugs, such as opioids, glucocorticoids, and sex steroids. Moreover, given the tendency of many subjects to excessive physical activity and drastic reduction in caloric intake, FHH may also be secondary to low energy availability. Finally, the association with HIV infection should not be overlooked. Therefore, there is an important variability in the diseases that can lead to FHH. Despite the heterogeneity of the underlying pathologies, the mechanisms leading to FHH would seem quite similar, with the initial event represented by the impairment at the HPG-A level. Nevertheless, many different biological pathways are involved in the pathogenesis of FHH, therefore the aim of the current paper is to provide an overview of the main relevant mechanisms, through a detailed analysis of the literature, focusing specifically on pathogenesis and clinical, diagnostic and therapeutic aspects.
... Since obesity and diabetes mellitus are considered as relevant risk factors for poor prognosis once SARS-CoV-2 infection occurred, patients should be managed in order to obtain (and maintain) a significative weight loss and better glucose control also for restoring a normal gonadal function. Lifestyle modification, diet, physical activity and medications are essential for achieving these purposes [106][107][108][109][110] also considering that Governmental restrictive measures aimed to tackle the spread of COVID-19 could be a risk factor for sedentary and poorer quality of food intake, finally leading to weight gain and its related consequences [111]. ...
... Exercise and restricted-calorie diets can reduce body fat and weight and improve HG symptoms secondary to obesity (Giagulli et al., 2019). However, little is known about the effects of long-term physical exercise on ASP concentration in humans (Jahangiri et al., 2021;Kantorowicz et al., 2021). ...
Article
Full-text available
Background: Asprosin (ASP) is a recently discovered adipocyte factor that participates in glucose metabolism and inflammatory reactions. Recent findings suggest that it may be involved in the regulation of sex hormone secretion in the hypothalamic-pituitary-gonad (HPG) axis, but no studies have been reported in related populations. The purpose of this study was to evaluate the changes in serum ASP levels in healthy men and obese men, as well as before and after exercise weight loss, and to investigate male hypogonadism, insulin resistance, inflammatory response, and relationships induced by ASP and obesity. Methods: Thirty-eight young male volunteers were recruited and divided into a normal group ( n = 20) and an obese group ( n = 18) according to their body mass index. Fourteen of the obese men underwent a 14-week exercise and diet intervention (first 8 weeks of aerobic exercise at 60%–70% HR max for 30–50 min/4 days a week). Beginning at week 9, the intensity was increased to 75% HR max . Participants in the obese groups maintained a calorie-restricted diet throughout the study period. Results: Serum ASP levels in the obese group were significantly higher than those in the normal group, and serum gonadotropin-releasing hormone (GnRh), luteinizing hormone (LH), and testosterone (T) levels were decreased. After 14 weeks of exercise and diet intervention, serum ASP decreased significantly, the levels of body weight, lean body weight, body fat rate, fasting insulin (FINS), homeostatic model assessment for insulin resistance, TNF-α, IL-6, and IL-1β decreased significantly, and the serum GnRH, LH, and T levels increased significantly. ASP was positively correlated with body weight, body fat percentage, FINS, tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β and negatively correlated with relative lean body weight and serum LH and T levels. Conclusion: The serum ASP levels were increased in obese men compared with those of normal weight individuals, resulting in a chronic inflammatory reaction, high serum insulin, and HPG axis injury. Fourteen weeks of exercise and diet intervention effectively alleviated this phenomenon. It has been speculated that ASP might regulate male reproductive function by regulating the inflammatory response and insulin sensitivity.
... Lifestyle intervention, especially weight loss of at least 10% by any means, improves total and free testosterone levels [122][123][124] . Very low-calorie diets (VLCD) or low-calorie diets (LCD) with physical activity could reduce insulin resistance and leptin levels with improvements in SHBG and total testosterone levels 125 . A trial comparing the effects of LCD on insulin sensitivity, testosterone levels, and erectile function over 8-weeks in obese males who achieved a weight reduction (≥10% or more) found a significant increase in insulin sensitivity, testosterone levels (r = -0.34), ...
Article
Full-text available
Sexual dysfunction, which is defined as ‘difficulty during any stage of the sexual encounter that prevents or impairs the individual or couple from enjoying sexual activity’, is globally prevalent in males with prediabetes and diabetes. It is an early harbinger of cardiovascular diseases and has a profound impact on one's physical, mental, and social health. Among patients with either prediabetes or diabetes, the most common male sexual dysfunctions are hypogonadism, erectile dysfunction, and premature ejaculation. In Asia, although sexual health is an important factor of men's health, it is rarely discussed freely in real‐life practice. Addressing sexual health in Asian males has always been challenging with multiple barriers at the levels of patients and health care providers. Therefore, the assessment and management of sexual dysfunction in routine clinical practice should involve a holistic approach with effective patient–provider communication. In this review, we discuss the epidemiology, pathophysiology, and the management of hypogonadism, erectile dysfunction, and premature ejaculation among males with either prediabetes or diabetes (type 1 and type 2), as well as the evidence gaps across Asia. Sexual dysfunction is an early harbinger or cardiovascular disease. It also has a significant impact on an individual's physical, mental and social health. However, in Asia, addressing men's health is challenging with multiple barriers at the levels of health care providers and patients. Hence, in this review, we discuss the epidemiology, pathophysiological and management of hypogonadism, erectile dysfunction and premature ejaculation of men with either prediabetes or diabetes (type 1 and type 2), as well as the evidence gaps in Asia.
... There are contradictory results that do not definitively prove the use of this plant for patients with diabetes and hyperlipidemia, and according to local and local evidence about the effect of Carla on blood lipids [29,30]. in this study, the effect of Carla Existing on the level of blood lipids in laboratory rats, which if approved can be used as an available source with hyperlipidemic properties. ...
... Weight loss may improve T serum levels in obese men [82,83]. The same has been observed in obese and diabetic men, with a more relevant cumulative effect than glycemic control alone [84]. In diabetic and obese patients with functional hypogonadism, selective estrogen receptor modulators (e.g., clomiphene citrate) may reactivate the hypothalamic-pituitary-testis axis, and serum T can be restored [85][86][87]. ...
Article
Full-text available
Background Late-Onset Hypogonadism (LOH) is defined as a clinical and biochemical syndrome associated with advancing age. It is characterized by specific symptoms and less specific manifestations by deficiency of serum testosterone (T) levels. Aims This review aims to summarize the evidence related to LOH definition, diagnostic approach, and treatment to answer a clinical question: “Is Testosterone the fountain of youth for aging men?". Methodology. MEDLINE/PubMed and institutional websites were searched for original papers, guidelines, and position statements published in the last ten years. Results Observational and randomized controlled studies dealing with T replacement therapy in older men have been reported. Discussion and conclusion Despite some heterogeneities regarding diagnostic definition, therapeutic target, and testosterone prescription, all guidelines agreed that male hypogonadism should be diagnosed and managed in aged men as in adulthood. However, trials assessing the efficacy of T therapy conducted for male rejuvenating are lacking; thus, T prescription for this purpose is not recommended.
Book
Full-text available
Buku ini merupakan kolaborasi antar penulis yang berperan dalam dunia akademisi dan praktisi untuk menghasilkan referensi atau sumber bahan baca yang bisa dijadikan referensi dalam kehidupan sehari-hari. Membahas mengenai daur kehidupan manusia tidak pernah lepas dari peranan asupan gizi yang seimbang sesuai dengan tahap kehidupan. Bayi dalam kandungan, bayi yang sudah lahir dan tumbuh menjadi balita, anak-anak, remaja, dewasa dan lansia adalah proses kehidupan yang pasti dialami oleh setiap individu. Menghasilkan generasi yang unggul menjadi tantangan saat ini. Selain itu, upaya dalam menurunkan penyakit infeksi dan penyakit degeneratif yang berhubungan dengan asupan makanan dan gizi merupakan hal yang penting. Buku ini membahas secara rinci tentang peran dan kebutuhan gizi dalam setiap tahap perkembangan manusia yang tersaji dalam beberapa bab, diantaranya gizi ibu menyusui, gizi dalam daur kehidupan, ASI Eksklusif, MP-ASI, gizi bayi 0-12 bulan, gizi balita, gizi remaja, anemia pada remaja putri, gizi dewasa, gizi menopause dan andropause, hingga gizi lansia. Gizi Dalam Daur Kehidupan merupakan buku yang bisa menjadi panduan bagi tenaga kesehatan, bahan ajar, dan masyarakat luas untuk lebih bisa mengoptimalkan asupan gizi pada setiap fase atau tahap kehidupan manusia. Buku ini juga dilengkapi dengan kebutuhan gizi, cara pemberian makanan yang tepat hingga faktor-faktor yang berpengaruh terhadap asupan gizi individu serta cara pengoptimalannya.
Preprint
Full-text available
Male obesity secondary hypogonadism (MOSH) impairs fertility, sexual function, bone mineralization, fat metabolism, cognitive function, deteriorates muscle mass and alters body composition. The aim of this pilot study was to evaluate the effect of dietary intervention and physical activity on the MOSH patient’s hormonal profile after a 10% weight loss compared to baseline. Fourteen male patients were enrolled. Hormonal, lipid, glycemic profiles and body composition were determined at baseline and after a 10% weight loss. Aging Male Symptoms Scale (AMS) and Yale Food Addiction Scale (YFAS) were administered to patients in order to investigate hypogonadal symptoms and food addiction. Compared to baseline, a significant increase of Total Testosterone (TT) (300.2 ± 79.5 ng/dl vs 408.3 ± 125.9, p = 0.002, 95% CI 26.8; 167.7) and a reduction of 17-Beta Estradiol level (48.3 ± 14.9 pg/mL vs 39.2 ± 15.2, p = 0.049, 95% CI 3.1; 0.0) were observed. Total Fat Mass (FM) percentage, android and gynoid fat mass percentage (39.2 ± 6.4% vs 36.2 ± 5.8%, p = 0.0001, 95% CI 22.5; 62.3; 51.5 ± 6.8% vs 47.6 ± 6.8%, p = 0.001, 95% CI 0.6; 1.8, vs 39.2 ± 6.2% vs 36.5 ± 6.3% p= 0.0001, 95% CI 0.9; 2.0 respectively) were significantly decreased after nutritional intervention. In addition, total Fat Free Mass (FFM) in kg was significantly reduced after 10% weight loss (62.3± 2.8 kg vs 60.3± 7.7 kg, p = 0.002, 95% CI 45.0; 93.0). Lifestyle changes, specifically dietotherapy and physical activity, induce positive effects on hypogonadism due to obesity.
Article
Full-text available
A dichotomic distinction between “organic” and “functional” hypogonadism is emerging. The former is an irreversible condition due to congenital or “acquired” “organic” damage of the brain centers or of the testis. Conversely, the latter is a potentially reversible form, characterized by borderline low testosterone (T) levels mainly secondary to age-related comorbidities and metabolic derangements, including metabolic syndrome (MetS). Life-style modifications, – here reviewed and, when possible, meta-analyzed –, have documented that weight-loss and physical exercise are able to improve obesity-associated functional hypogonadism and its related sexual symptoms. A rabbit experimental model, of MetS originally obtained in our lab, showed that endurance training (PhyEx) completely reverted MetS-induced hypogonadotropic hypogonadism by reducing hypothalamus inflammation and testis fibrosis eventually allowing for a better corpora cavernosa relaxation and response to sildenafil. Physicians should strongly adapt all the reasonable strategies to remove/mitigate the known conditions underlying functional hypogonadism, including MetS and obesity. Physical limitations, including reduced muscle mass and increased fat mass, along with low self-confidence, also due to the sexual problems, might limit a subject’s propensity to increase physical activity and dieting. A short term T treatment trial, by improving muscle mass and sexual function, might help hypogonadal obese patients to overcome the overfed, inactive state and to become physically and psychologically ready for changing their lifestyle.
Article
Full-text available
Several studies highlight that testosterone deficiency is associated with, and predicts, an increased risk of developing metabolic disorders, and, on the other hand, is highly prevalent in obesity, metabolic syndrome and type-2 diabetes mellitus. Models of gonadotropin releasing hormone deficiency, and androgen deprivation therapy in patients with prostate cancer, suggest that hypogonadotropic hypogonadism might contribute to the onset or worsening of metabolic conditions, by increasing visceral adiposity and insulin resistance. Nevertheless, in functional hypogonadism, as well as in late onset hypogonadism, the relationship between hypogonadotropic hypogonadism and metabolic disorders is bidirectional, and a vicious circle between the two components has been documented. The mechanisms underlying the crosstalk between testosterone deficiency and metabolic disorders include increased visceral adipose tissue and insulin resistance, leading to development of metabolic disorders, which in turn contribute to a further reduction of testosterone levels. The decrease in testosterone levels might be determined by insulin resistance-mediated and, possibly, pro-inflammatory cytokine-mediated decrease of sex hormone binding globulin, resulting in a temporary increased free testosterone available for aromatization to estradiol in visceral adipose tissue, followed by a subsequent decrease in free testosterone levels, due to the excess of visceral adipose tissue and aromatization; by a direct inhibitory effect of increased leptin levels on Leydig cells; and by a reduced gonadotropin secretion induced by estradiol, inflammatory mediators, leptin resistance, and insulin resistance, with the ultimate determination of a substantial hypogonadotropic hypogonadism. The majority of studies focusing on the effects of testosterone replacement therapy on metabolic profile reported a beneficial effect of testosterone on body weight, waist circumference, body mass index, body composition, cholesterol levels, and glycemic control. Consistently, several interventional studies demonstrated that correction of metabolic disorders, in particular with compounds displaying a greater impact on body weight and insulin resistance, improved testosterone levels. The aim of the current review is to provide a comprehensive overview on the relationship between hypogonadotropic hypogonadism and metabolism, by clarifying the independent role of testosterone deficiency in the pathogenesis of metabolic disorders, and by describing the relative role of testosterone deficiency and metabolic impairment, in the context of the bidirectional relationship between hypogonadism and metabolic diseases documented in functional hypogonadotropic hypogonadism. These aspects will be assessed by describing metabolic profile in men with hypogonadotropic hypogonadism, and androgenic status in men with metabolic disorders; afterwards, the reciprocal effects of testosterone replacement therapy and corrective interventions on metabolic derangements will be reported.
Article
Full-text available
Men with obesity often present with low testosterone (T) and sex hormone-binding globulin (SHBG) levels. Several mechanisms for this have been proposed, but as SHBG is secreted by hepatocytes and sex steroids undergo hepatic metabolization, this study investigates whether severity and histological components of nonalcoholic fatty liver disease (NAFLD) are associated with sex steroid levels in obese men. This cross-sectional study included 80 obese men (age: 46 ± 11 years; body mass index: 42.2 ± 5.5 kg m-2). Serum levels of total T and estradiol (E2) were measured using liquid chromatography coupled with tandem mass spectroscopy (LC/MS-MS) and SHBG and gonadotropins by immunoassay. Liver biopsies were evaluated using Steatosis, Activity, and Fibrosis scoring. Participants with steatohepatitis had similar median (1stquartile-3rd quartile) total T levels (7.6 [5.0-11.0] nmol l-1 vs 8.2 [7.2-10.9] nmol l-1; P = 0.147), lower calculated free T (cFT) levels (148.9 [122.9-188.8] pmol l-1 vs 199.5 [157.3-237.6] pmol l-1; P = 0.006), and higher free E2/T ratios (10.0 [6.4-13.9] x10-3 vs 7.1 [5.7-10.7] x10-3.
Article
Full-text available
The first contact for patients with obesity for any medical treatment or other issues is generally with General Practitioners (GPs). Therefore, given the complexity of the disease, continuing GPs’ education on obesity management is essential. This article aims to provide obesity management guidelines specifically tailored to GPs, favouring a practical patient-centred approach. The focus is on GP communication and motivational interviewing as well as on therapeutic patient education. The new guidelines highlight the importance of avoiding stigmatization, something frequently seen in different health care settings. In addition, managing the psychological aspects of the disease, such as improving self-esteem, body image and quality of life must not be neglected. Finally, the report considers that achieving maximum weight loss in the shortest possible time is not the key to successful treatment. It suggests that 5–10% weight loss is sufficient to obtain substantial health benefits from decreasing comorbidities. Reducing waist circumference should be considered even more important than weight loss per se, as it is linked to a decrease in visceral fat and associated cardiometabolic risks. Finally, preventing weight regain is the cornerstone of lifelong treatment, for any weight loss techniques used (behavioural or pharmaceutical treatments or bariatric surgery).
Article
Context Prostate specific antigen (PSA) changes during testosterone treatment of older hypogonadal men have not been rigorously evaluated. The Endocrine Society Guidelines recommendation of urological referral for a confirmed increase in PSA >1.4 ng/mL is not based on PSA changes in testosterone-treated men. Design Double-blinded, placebo-controlled trial. Setting Twelve United States academic medical centers. Participants 790 hypogonadal men ≥65 years with average testosterone levels ≤275 ng/dL. Men at high risk for prostate cancer were excluded. Interventions Testosterone or placebo gel for 12 months. Main outcomes Percentile changes in PSA during testosterone treatment for 12 months. Results Testosterone treatment that increased testosterone levels from 232 ± 63 ng/dL to mid-normal was associated with a small but significantly greater increase (p<0.001) in PSA levels than placebo treatment. Serum PSA levels increased from 1.14±0.86 ng/mL (mean ±SD) at baseline by 0.47±1.1 ng/mL at 12 months in the testosterone group and from 1.25±0.86 ng/mL by 0.06±0.72 ng/mL in the placebo group. Five percent of men treated with testosterone had an increase ≥1.7 ng/mL and 2.5% of men had an increase of ≥3.4 ng/mL. A confirmed absolute PSA >4.0 ng/mL at 12 months was observed in 1.9% of men in the testosterone group and 0.3% in the placebo group. Four men were diagnosed with prostate cancer; two were Gleason 8. Conclusions When hypogonadal, older men with normal baseline PSA are treated with testosterone, 5% had an increase in PSA ≥1.7 ng/mL, and 2.5% had an increase ≥3.4 ng/mL.
Article
Epidemiological studies consistently demonstrate that lowered serum testosterone is not only common in men with established Type 2 diabetes, but also predicts future diabetic risks and increased mortality. Preclinical studies report plausible mechanisms by which low testosterone could mediate dysglycaemia. Exogenous testosterone treatment consistently reduces fat mass, increases muscle mass and improves insulin resistance in some studies, but the majority of currently available randomized controlled trials (RCTs) do not report a consistent glycaemic benefit. In men with diabetes, testosterone treatment effects on androgen deficiency‐like clinical features are inconsistent, and effects on sexual dysfunction may be attenuated compared with men without diabetes. The long‐term risks of testosterone treatment in older men without medical disease of the hypothalamic–pituitary–testicular axis are not known. Current RCTs are not definitive, owing to their small size, short duration and enrolment of men with mostly relatively good baseline glycaemic control not specifically selected for the presence of androgen deficiency symptoms. Although large, well‐designed clinical trials are needed, given the benefit–risk ratio of testosterone treatment is not well understood, routine serum testosterone testing or testosterone treatment of asymptomatic men with Type 2 diabetes is currently not recommended. Carefully selected, symptomatic men with low testosterone who are informed of the lack of high‐level evidence regarding the long‐term benefits and risks of this approach may be offered a trial of testosterone treatment in combination with lifestyle measures, weight loss and optimization of comorbidities. This article is protected by copyright. All rights reserved
Article
Context Obesity is known to impact reproductive function in adults, but little is known about its effects on reproductive hormones during puberty or sex differences in these effects. Objective To assess sex differences in effects of obesity on reproductive hormones and their relationship to insulin sensitivity and secretion. Design Cross-sectional study including anthropometrics, serum and urine reproductive hormone concentrations, and intravenous glucose tolerance testing (IVGTT) to assess acute insulin response to glucose (AIRg) and insulin sensitivity (Si) Setting Outpatient academic clinical research center Patients Girls (52%) and boys (48%) who were normal weight (NW, n=51, BMI-Z=-0.11 ± 0.77, age=11.5 ± 1.7 years) and obese (n=53, BMI-Z=2.22 ± 0.33, age=10.9 ± 1.5 years), Tanner stage 2-3 Results Boys with obesity had lower total testosterone (p<0.0001) and higher concentrations of the urinary estradiol metabolite, E1c, (p=0.046) than boys with NW. Girls with obesity had higher free androgen index (FAI, p=0.03) than NW girls. Both boys and girls with obesity had lower sex hormone-binding globulin (SHBG, p<0.0001) than NW. AIRg was inversely related to SHBG in boys (R²= 0.36, p<0.0001) and girls (R²=0.29, p=0.0001). Insulin resistance correlated with lower SHBG in boys (R²=0.45, p<0.0001) and girls (R²=0.24, p=0.0003), lower total testosterone for boys (R²=0.15, p=0.01), and higher FAI for girls (R²=0.08, p=0.04). Conclusion Youth with obesity have lower SHBG than youth with NW, but obesity has differential effects on reproductive hormones in girls vs. boys, which are apparent early in puberty. Ongoing longitudinal studies will evaluate the impact of obesity on reproductive hormones in girls and boys as puberty progresses.
Article
Objective: Type 2 diabetes (T2D) is a public health threat. Prediabetes represents a window of opportunity for intervention to prevent T2D. Men with T2D and prediabetes often have low testosterone. Since testosterone improves glycemic control in T2D, we investigated whether testosterone therapy (TTh) in men with hypogonadism and prediabetes prevents progression to T2D. Research design and methods: Three hundred and sixteen men with prediabetes (defined as HbA1c 5.7-6.4%) and total testosterone levels ≤12.1 nmol/L combined with symptoms of hypogonadism were analyzed. Two hundred and twenty-nine men received parenteral testosterone undecanoate (T-group), and 87 men with hypogonadism served as untreated control subjects. Metabolic and anthropometric parameters were measured twice yearly for 8 years. Results: HbA1c decreased by 0.39 ± 0.03% (P < 0.0001) in the T-group and increased by 0.63 ± 0.1% (P < 0.0001) in the untreated group. In the T-group, 90% achieved normal glucose regulation (HbA1c <5.7%). In the untreated group, 40.2% progressed to T2D (HbA1c >6.5%). TTh was also associated with significant improvements in fasting glucose, triglyceride:HDL ratio, triglyceride-glucose index, lipid accumulation product, total cholesterol, LDL, HDL, non-HDL, triglycerides, and Aging Males' Symptoms (AMS) scale. Significant deterioration in all these parameters was seen in the untreated group. Mortality was 7.4% in the T-group and 16.1% in the untreated group (P < 0.05). The incidence of nonfatal myocardial infarction was 0.4% in the T-group and 5.7% in the untreated group (P < 0.005). Conclusions: Long-term TTh completely prevents prediabetes progression to T2D in men with hypogonadism and improves glycemia, lipids, and AMS score. TTh holds tremendous potential for the large and growing population of men with prediabetes and hypogonadism.
Article
Many male infertility cases have no apparent cause, being characterized as idiopathic. Both inflammation and obesity have long been associated with infertility. On one hand, inflammation, such as orchitis and male accessory gland infections (MAGIs), are regulated by inflammatory cytokines. The latter are also produced in the testis by Leydig and Sertoli cells, being associated with gap junctional communication at the blood-testis barrier. Furthermore, they regulate spermatogenesis through cell interaction, Toll-like receptors and production of reactive oxygen species. Additionally, they affect testosterone production, acting at many levels of the pituitary - gonadal axis. Any imbalance in their production may result in infertility. On the other hand, obesity has also been associated with infertility. Adipokines, cytokines produced by white adipose tissue, regulate the lipid and glucose metabolism and the inflammatory system. Recent data on leptin show that it regulates reproduction by adjusting hypothalamus - pituitary - gonadal axis at both the central and peripheral levels. In this regard, resistin, visfatin and the GH secretagogue peptic hormone ghrelin affect spermatogenesis, whereas data on adiponectin are rather scarce. In conclusion, inflammatory cytokines and adipokines seem to have a pivotal role in the regulation of spermatogenesis; any imbalance in this stable environment may lead to infertility. Nevertheless, further studies are needed to clarify their exact role.