ArticlePDF Available

A sex-specific role for androgens in angiogenesis

Rockefeller University Press
Journal of Experimental Medicine (JEM)
Authors:

Abstract and Figures

Mounting evidence suggests that in men, serum levels of testosterone are negatively correlated to cardiovascular and all-cause mortality. We studied the role of androgens in angiogenesis, a process critical in cardiovascular repair/regeneration, in males and females. Androgen exposure augmented key angiogenic events in vitro. Strikingly, this occurred in male but not female endothelial cells (ECs). Androgen receptor (AR) antagonism or gene knockdown abrogated these effects in male ECs. Overexpression of AR in female ECs conferred androgen sensitivity with respect to angiogenesis. In vivo, castration dramatically reduced neovascularization of Matrigel plugs. Androgen treatment fully reversed this effect in male mice but had no effect in female mice. Furthermore, orchidectomy impaired blood-flow recovery from hindlimb ischemia, a finding rescued by androgen treatment. Our findings suggest that endogenous androgens modulate angiogenesis in a sex-dependent manner, with implications for the role of androgen replacement in men.
This content is subject to copyright.
The Rockefeller University Press $30.00
J. Exp. Med. Vol. 207 No. 2 345-352
www.jem.org/cgi/doi/10.1084/jem.20091924
345
Brief Definitive Report
The role of androgens in cardiovascular disease
(CVD) remains controversial. Although men de-
velop CVD earlier than women, consistent with
increased atherosclerosis (Wingard et al., 1983;
Shaw et al., 2006), repair/adaptation mecha-
nisms may be enhanced in males (Joseph and
Dyson, 1965). For example, male sex is associ-
ated with increased collateralization in patients
with severe coronary artery lesions (Abaci et al.,
1999). Androgens have been implicated in play-
ing a detrimental role in CVD by numerous in
vitro studies (McCrohon et al., 1999; Ng et al.,
2003a,b), but in vivo and clinical studies fail to
support this relationship (Liu et al., 2003; Wu
and von Eckardstein, 2003; Harman, 2005).
Indeed, some but not all cohort studies suggest
that subnormal blood levels of testosterone are
deleterious and correlate with increased cardio-
vascular risk factors and mortality (Araujo et al.,
2007; Khaw et al., 2007; Laughlin et al., 2008;
Tivesten et al., 2009).
Angiogenesis is a prerequisite for embryonic
development and plays a critical role in adult
physiological processes such as wound repair
and in tissue responses to ischemia (Carmeliet,
2005). The cardiovascular system is an important
target of androgen action (Liu et al., 2003), and
although the eect of estrogen in angiogenesis
has been studied extensively (Losordo and Isner,
2001), the role of androgens in this process
remains largely unexplored.
Androgens have been implicated in the re-
newal of various tissues, including muscle and
bone (Harman, 2005), but their potential role
in the maintenance of cardiovascular repair
mechanisms is unknown. As androgens have been
implicated in the regulation of angiogenesis-
related genes (Ng et al., 2003b; Death et al.,
2004), we reasoned that androgens may play a
role in angiogenesis, a process critical in the re-
generative response after ischemia. To examine
the role of androgens in angiogenesis, we used
dihydrotestosterone (DHT), the most potent
natural androgen that acts via the androgen
receptor (AR). As testosterone action partly
depends on its conversion to estradiol by the
unique enzyme CYP19 (aromatase), whereas
CORRESPONDENCE
Martin K.C. Ng:
mkcng@med.usyd.edu.au
OR
Daniel P. Sieveking:
sievekingd@hri.org.au
Abbreviations used: AcLDL,
Dil-acetylated low density
lipoprotein; ANOVA, analysis
of variance; AR, androgen
receptor; CVD, cardiovascular
disease; DHT, dihydrotestos-
terone; EC, endothelial cell;
ER, estrogen receptor; HF,
hydroxyutamide; HIF-1,
hypoxia-inducible factor 1;
LDPI, laser Doppler perfusion
index; MNC, mononuclear
cell; mRNA, messenger
RNA; PI3K, phosphoinositol
3-kinase; SDF-1, stromal
cell–derived factor 1;
siRNA, small interfering
RNA; VEGF, vascular endo-
thelial growth factor; vWF,
von Willebrand factor.
A sex-specic role for androgens
in angiogenesis
Daniel P. Sieveking,1,2 Patrick Lim,1 Renée W.Y. Chow,1 Louise L. Dunn,1
Shisan Bao,3 Kristine C.Y. McGrath,1 Alison K. Heather,1
David J. Handelsman,4,5 David S. Celermajer,1,2,6 and Martin K.C. Ng1,2,6
1Heart Research Institute, Sydney 2042, Australia
2Department of Medicine and 3Department of Pathology, University of Sydney, Sydney 2050, Australia
4ANZAC Research Institute, Sydney 2139, Australia
5Department of Andrology, Concord Hospital, Sydney 2139, Australia
6Department of Cardiology, Royal Prince Alfred Hospital, Sydney 2050, Australia
Mounting evidence suggests that in men, serum levels of testosterone are negatively cor-
related to cardiovascular and all-cause mortality. We studied the role of androgens in
angiogenesis, a process critical in cardiovascular repair/regeneration, in males and females.
Androgen exposure augmented key angiogenic events in vitro. Strikingly, this occurred in
male but not female endothelial cells (ECs). Androgen receptor (AR) antagonism or gene
knockdown abrogated these effects in male ECs. Overexpression of AR in female ECs con-
ferred androgen sensitivity with respect to angiogenesis. In vivo, castration dramatically
reduced neovascularization of Matrigel plugs. Androgen treatment fully reversed this effect
in male mice but had no effect in female mice. Furthermore, orchidectomy impaired blood-
ow recovery from hindlimb ischemia, a nding rescued by androgen treatment. Our nd-
ings suggest that endogenous androgens modulate angiogenesis in a sex-dependent
manner, with implications for the role of androgen replacement in men.
© 2010 Sieveking et al. This article is distributed under the terms of an Attribu-
tion–Noncommercial–Share Alike–No Mirror Sites license for the rst six months
after the publication date (see http://www.jem.org/misc/terms.shtml). After six
months it is available under a Creative Commons License (Attribution–Noncom-
mercial–Share Alike 3.0 Unported license, as described at http://creativecommons
.org/licenses/by-nc-sa/3.0/).
The Journal of Experimental Medicine
346 Androgens and angiogenesis | Sieveking et al.
Chronic exposure to DHT (72 h) induced a dose-dependent
increase in male EC tubulogenesis (Fig. 1, D and E), as did 24 h
of exposure (Fig. S1). Similarly, DHT augmented male EC
proliferation in a dose-dependent fashion after 48 h (Fig. 1 F).
Again, addition of HF abolished all androgen-mediated ef-
fects on these key angiogenic events, consistent with an AR-
dependent mechanism (Fig. 1, B, C, E, and F).
As an independent method to verify the pivotal involve-
ment of AR in DHT-mediated angiogenic eects, we also
assessed tubulogenesis in male ECs subjected to small inter-
fering RNA (siRNA)–mediated knockdown of AR. DHT
eects were also abolished in ECs exposed to AR siRNA (Fig.
1 G). DHT can also mediate eects via direct cross-reactivity
at the ER at suciently high concentrations. Therefore, to
further exclude any contribution to DHT eects via eects
on ERs, we assessed tubulogenesis in male ECs treated with
the highest concentration of DHT with or without the ER
antagonist ICI182780. The addition of ICI182780 did not
have any eect on DHT-mediated augmentation of tubulo-
genesis (Fig. 1 H). These ndings are consistent with an AR-
mediated angiogenic eect in male ECs.
Androgens do not augment angiogenic events in vitro
in female ECs
Mounting evidence indicates that sex steroids regulate vari-
ous vascular biological processes in a sex-dependent fashion
DHT cannot be aromatized, the use of DHT avoids the
potentially confounding estrogen receptor (ER)–mediated
eects of testosterone.
RESULTS AND DISCUSSION
Androgens augment angiogenic events in vitro
in male endothelial cells (ECs)
Male ECs treated with DHT demonstrated a dose-dependent
increase in key angiogenic processes in vitro (Fig. 1). EC mi-
gration was assessed by two dierent methods. Cells were pre-
treated with varying doses of DHT (0, 4, 40, and 400 nM) and
subsequently assessed using a Boyden chamber assay or a
scratch wounding assay. Using the Boyden chamber assay, 24 h
of DHT administration induced dose-dependent male EC
migration (220 ± 22, 262 ± 38, and 336 ± 39% vs. 100% con-
trol value for DHT at 4, 40, and 400 nM, respectively; P <
0.001 using analysis of variance [ANOVA]; Fig. 1 B). Similar
results were obtained using a scratch wounding assay. Migra-
tion by male ECs was augmented in a dose-dependent fashion
(138 ± 7, 280 ± 23, and 306 ± 32%; P < 0.01 using ANOVA;
Fig. 1 C). Interestingly, in both assays, addition of the AR
antagonist hydroxyutamide (HF) abrogated DHT-mediated
EC migration (96 ± 6 and 94 ± 6% vs. 100% control value for
40 and 400 nM DHT + HF, respectively; P > 0.05; Fig. 1,
B and C). DHT eects on tubulogenesis were assessed using
both Matrigel and co-culture methods (Sieveking et al., 2008).
Figure 1. DHT augments key angiogenic events in vitro in male ECs via the AR. (A and B) Boyden chamber (cells were stained with Ulex lectin
[UEA-1] and DAPI) and (C) scratch wounding assays of migration by male ECs pretreated with DHT or vehicle (0.1% EtOH) for 24 h ± HF, assessed after 6
and 24 h, respectively. Bar, 50 µm. (D and E) Vascular network formation by male ECs exposed to DHT or vehicle ± HF for 72 h. Tubule area was quantied
using image analysis software (ImageJ; available at http://rsbweb.nih.gov/ij/). Bar, 100 µm. (F) Proliferation of male ECs exposed to DHT or vehicle ± HF for
24 h, assessed by direct cell counting (n = 4 independent experiments for A–F, respectively). (G and H) Matrigel assays of male ECs transfected with siRNA
targeted to the AR (G) or pretreated with the ER blocker ICI182780 and treated with DHT (H; n = 3 independent experiments). **, P < 0.01; and ***, P <
0.001 versus control using ANOVA. All data are expressed as means ± SEM. For each independent experiment, cells from a different donor were used.
JEM VOL. 207, February 15, 2010 347
Brief Definitive Report
for 0, 4, 40, and 400 nM DHT, respectively; P = 0.0024;
Fig. 3 A). Similarly, DHT induced dose-dependent increases
in the messenger RNA (mRNA) expression of VEGF re-
ceptors 1 and 2 (Flt-1 and KDR, respectively; Fig. 3, B and C).
However, ow cytometric assessment of surface VEGF recep-
tor levels revealed that DHT augmented KDR but not Flt-1
expression in a dose-dependent manner (Fig. 3, D and E). As
KDR is the chief mediator of the mitogenic/angiogenic action
of VEGF in ECs, whereas Flt-1 may function as a negative
regulator of VEGF action (Ferrara et al., 2003), our ndings
are consistent with a proangiogenic role for DHT through
VEGF signaling via KDR. Furthermore, the addition of anti-
VEGF antibody abrogated DHT-mediated increases in tubu-
logenesis (Fig. 3 F). Likewise, LY294002, an inhibitor of
phosphoinositol 3-kinase (PI3K), a key enzyme in the PI3K–
AKT pathway of VEGF signaling, inhibited DHT-mediated
tubulogenesis (Fig. 3 F). These ndings suggest that the proan-
giogenic eects of DHT in male ECs are VEGF dependent.
Endogenous androgens are necessary for angiogenesis
in vivo in males but not females
Having demonstrated a sex-specic role for androgens in
angiogenesis in vitro, we next assessed the role of androgens
in angiogenesis in vivo, rst using a Matrigel plug implanta-
tion model. Sexually mature male and female mice under-
went gonadectomy or a sham gonadectomy followed by
DHT administration. Orchidectomy markedly decreased
in vivo vascularization of Matrigel plugs (27.3 ± 2% mm2
von Willebrand factor [vWF]+ cells/mm2 Matrigel vs. con-
trol as 100%; P < 0.0001 using ANOVA; Fig. 4 A). As can
be observed in the representative image, these plugs were
virtually acellular. Androgen treatment fully reversed the cas-
tration-related impairment of angiogenesis in males, returning
(McCrohon et al., 1999; Ng et al., 2001; Ng et al., 2003b).
For androgens, this is related to an approximately two- to
fourfold increased AR expression in male vascular cells com-
pared with females (McCrohon et al., 2000; Death et al.,
2004). We therefore hypothesized that the proangiogenic
eects of DHT observed in ECs may also be sex dependent.
Unlike male ECs, exposure of female ECs to DHT had no
eect on migration (Fig. 2, A and B), tubulogenesis (Fig. 2 C),
or proliferation (P > 0.05; Fig. 2 D), consistent with andro-
gen-mediated sex-specic eects. To further examine the
role of the AR in mediating the sex-dependent eects of
DHT on angiogenesis, we overexpressed the AR in female
ECs (Fig. S2) and assessed tubulogenesis after exposure to
DHT. Female ECs overexpressing the AR became respon-
sive to DHT and displayed augmented tubulogenesis (114 ±
6% vs. 100% control value; P < 0.05 using the Student’s
t test), albeit to a lesser extent than in male cells (Fig. 2 E).
These ndings indicate that the sex-specic eects of DHT
on angiogenesis are at least in part mediated by sex dier-
ences in AR expression. As there are a variety of coregulators
involved in AR signaling (Liu et al., 2003), it is also possible
that many of these are also expressed in a sex-specic fashion
and are important for androgenic modulation of angiogenesis.
The proangiogenic effect of androgens in vitro are vascular
endothelial growth factor (VEGF) dependent
Next, to further elucidate the means by which DHT aug-
mented migration, proliferation, and tubulogenesis in male
ECs, cells exposed to DHT were assessed for the expression of
various proangiogenic factors. Exposure of male ECs to DHT
produced a dose-dependent increase in the production of
VEGF, a key angiogenic growth factor (102.8 ± 7, 107.8 ± 7,
118.9 ± 6, and 147.2 ± 15 pg VEGF/100 µg of cell protein
Figure 2. DHT does not augment key angiogenic events in vitro in female ECs. (A–D) Boyden chamber (A) and scratch wounding (B) assays of
migration assessed after 6 and 24 h, respectively, Matrigel assays (C), and proliferation by female ECs treated with DHT or vehicle (D; n = 4 indepen-
dent experiments). P > 0.05 using ANOVA. (E) Matrigel assays of female ECs transfected with an AR overexpression plasmid and treated with DHT (n =
3 independent experiments). *, P < 0.05 using the Student’s t test. All data are expressed as means ± SEM. For each independent experiment, cells from
a different donor were used.
348 Androgens and angiogenesis | Sieveking et al.
detectable as early as day 3 and signicant by day 5 after isch-
emia (LDPI at day 5 after ischemia: control, 0.28 ± 0.04; or-
chidectomized + DHT, 0.4 ± 0.05; P < 0.05 vs. orchidectomized
mice; Fig. 5 A). These ndings were also borne out in the
functional assessments of recovery (Fig. S3). The serial changes
in perfusion observed after femoral artery ligation have previ-
ously been shown to correlate with changes in vessel density
as determined by histological analysis (Counhal et al.,
1998). As early as day 7, immunohistochemical staining of
the adductor muscles of the ischemic hindlimbs revealed that
capillary density was impaired in orchidectomized mice and
augmented in DHT-treated orchidectomized mice (0.17 ±
0.01 and 0.27 ± 0.01 vs. 0.22 ± 0.01 capillaries/myober for
orchidectomized mice, orchidectomized + DHT mice, and
controls, respectively; P < 0.001 using ANOVA; Fig. 5, B
and C), consistent with the ndings observed for limb perfu-
sion. Mean vessel diameter was also signicantly smaller in
orchidectomized mice (P < 0.001; Fig. 5 D). Interestingly,
mean vessel diameter was also smaller in orchidectomized
mice receiving DHT, but DHT treatment appeared to miti-
gate the eects of castration. These data suggest that, in the
context of ischemia, endogenous androgens inuence the
rate and extent of ischemia-induced angiogenesis and play a
role in arteriogenesis.
To elucidate the mechanisms of androgen-induced angio-
genesis in the context of hindlimb ischemia, we assessed the
early expression of angiogenesis- and ischemia-related genes.
Notably, 3 d after ischemia, orchidectomy signicantly re-
duced the expression of hypoxia-inducible factor 1 (HIF-1;
0.5 ± 0.1 vs. 1.8 ± 0.2 relative mRNA expression ischemic/
nonischemic for orchidectomized mice vs. controls, respec-
tively; P < 0.05 using ANOVA) and stromal cell–derived
factor 1 (SDF-1; 0.7 ± 0.1 vs. 2.4 ± 0.1 relative mRNA
vascularization of Matrigel plugs to levels not signicantly
dierent to controls (91 ± 6% mm2 vWF+ cells/mm2 Matri-
gel vs. control as 100%; P < 0.001).
Consistent with previous ndings, ovariectomy attenuated
Matrigel plug vascularization in females (Morales et al., 1995),
albeit to levels more modest than in males (53 ± 8% mm2
vWF+ cells/mm2 Matrigel vs. control as 100%; P < 0.0001 us-
ing ANOVA; Fig. 4 B). In striking contrast to their male coun-
terparts, gonadectomy-related impairment of angiogenesis in
females was not reversed by DHT treatment (55 ± 6% vs. con-
trol as 100%; P < 0.0001 using ANOVA). In accordance with
our in vitro ndings, these data suggest that endogenous andro-
gens play a role in angiogenesis in males but not females.
Endogenous androgens modulate
ischemia-induced angiogenesis
Hypoxia is one of the most potent angiogenic stimuli (Ceradini
and Gurtner, 2005). To investigate the role of androgens in
ischemia-induced angiogenesis, we used a mouse model of
hindlimb ischemia involving the unilateral ligation and re-
moval of the femoral artery (Counhal et al., 1998). Male
mice were randomized to orchidectomy with or without
DHT treatment or a sham orchidectomy. Laser Doppler per-
fusion imaging demonstrated that orchidectomy markedly
inhibited the rate of recovery from hindlimb ischemia (laser
Doppler perfusion index [LDPI] ischemic/nonischemic ratio
after 11 d: control, 0.54 ± 0.04; castrate, 0.39 ± 0.04; P <
0.05; Fig. 5 A). In functional assessments of the ischemic
hindlimbs, orchidectomized animals also displayed impaired
recovery in foot movement and ischemic damage (Fig. S3).
Interestingly, DHT not only rescued the orchidectomy-
induced impairment of blood ow recovery but also enhan-
ced recovery from ischemia, with improvements in flow
Figure 3. Proangiogenic effects of DHT in male ECs is VEGF dependent. (A) Production of VEGF by male ECs exposed to DHT for 48 h. Total cell
lysate protein was quantied and assayed via ELISA (n = 4 independent experiments). P = 0.002 using ANOVA for linear trend. (B) Expression of Flt-1
(P = 0.0128) and (C) KDR (P = 0.0295) mRNA and (D) Flt-1 and (E) KDR (P = 0.0005) protein in male ECs exposed to DHT for 48 h (using ANOVA for linear
trend). (F) Vascular network formation in male ECs treated with DHT with and without 1 µg/ml anti-VEGF antibody or 10 µM of the PI3K inhibitor
LY294002 (n = 3 independent experiments for B–F, respectively). ***, P < 0.001 compared with control using ANOVA. All data are expressed as means ±
SEM. For each independent experiment, cells from a different donor were used.
JEM VOL. 207, February 15, 2010 349
Brief Definitive Report
DHT, the increases observed for SDF-1 and KDR in DHT-
treated mice are also related to induction of HIF-1 (Ceradini
et al., 2004). There were trends toward a reduction in VEGF
expression in orchidectomized animals and an increase with
DHT treatment that were not statistically signicant. Never-
theless, given the critical role of HIF-1 in inducing VEGF-
related angiogenic events in ischemia and an increased expression
of KDR, our in vivo ndings are compatible with a role for
expression ischemic/nonischemic for orchidectomized mice
vs. controls, respectively; P < 0.05 using ANOVA), which are
key factors in the coordination of angiogenic gene expres-
sion and progenitor cell migration and homing, respectively
(Fig. 5, E and F, respectively; Ceradini et al., 2004). In stark
contrast, DHT treatment of orchidectomized mice augmented
expression of HIF-1 (3 ± 0.8; P < 0.01) and SDF-1 (3.6 ±
0.9; P < 0.01). These mice also demonstrated increased ex-
pression of KDR (3 ± 0.8 vs. 0.6 for orchidectomized + DHT
vs. controls, respectively; P < 0.01 using ANOVA; Fig. 5 G),
providing support for a role of DHT in VEGF signaling in
vivo. Furthermore, similar trends were observed for VEGF and
neuropillin-1 (Fig. S4). HIF-1 is the key subunit to HIF-1,
which is a critical, genome-wide transcription regulator re-
sponsible for oxygen homeostasis and responsive to hypoxic
stress (Semenza, 2002; Ceradini and Gurtner, 2005). It drives
the expression of >100 genes, including key genes associated
with angiogenesis such as VEGF and its receptors (Semenza,
2002). It is noteworthy that androgens have been associated
with HIF-1 expression in the prostate (Boddy et al., 2005).
Hence, it is possible that in addition to the direct eects of
Figure 4. Endogenous androgens play a role in angiogenesis in
males but not females. (A) Vascularization of Matrigel plugs harvested
from male mice after 14 d, with representative images (n = 8/group;
combined data from four independent experiments are shown). Sections
were immunohistochemically stained for vWF (3,3-diaminobenzidine
[DAB] + nickel, eosin). Bars, 100 µm. (B) Vascularization of Matrigel plugs
harvested from female mice, with representative images (n = 5–6/group;
combined data from three independent experiments are shown). Sections
were immunohistochemically stained for vWF (DAB, hematoxylin). Bars,
50 µm. ***, P < 0.001 versus control using ANOVA. All data are expressed
as means ± SEM.
Figure 5. Endogenous androgens modulate ischemia-induced an-
giogenesis. (A) LDPI. Limb perfusion ratio (ischemic/nonischemic) is pre-
sented with representative images (n = 8/group; combined data from four
independent experiments are shown). Differences between groups were
assessed by two-way ANOVA with Bonferroni correction (brackets at
right). *, P < 0.05; and **, P < 0.01 for comparisons between orchidecto-
mized and orchidectomized + DHT mice at individual time points. (B) Rep-
resentative images of neovascularization of ischemic hindlimbs at day 7.
Capillary density was measured in 8-µm frozen sections of the adductor
muscles via immunohistochemical staining for vWF (arrows). Bars, 100 µm.
(C) Capillary density expressed as the number of capillaries per myober
(eosin) and (D) mean vessel diameter (n = 6/group; combined data from
three independent experiments are shown). (E–G) Mechanisms of andro-
genic modulation of angiogenesis in hindlimb ischemia at day 3. Quantita-
tive RT-PCR for the expression of the angiogenesis- and ischemia-related
genes (E) HIF-1, (F) SDF-1, and (G) KDR (n = 6/group; combined data
from three independent experiments are shown). *, P < 0.05; **, P < 0.01;
and ***, P < 0.001 versus control using ANOVA. All data are expressed as
means ± SEM.
350 Androgens and angiogenesis | Sieveking et al.
castration-mediated decline in ulex+/AcLDL+ angiogenic cells
in both the spleen (17.8 ± 0.9 cells/200× eld; P > 0.05 using
ANOVA) and the bone marrow (31.7 ± 5 cells/200× eld;
P < 0.001 using ANOVA). SDF-1 is a key chemokine in the
migration and homing of bone marrow–derived progenitor
cells to sites of ischemia, and is itself a ligand for the receptor
CXCR4 (Jin et al., 2006); therefore, we also assessed the levels
of CXCR4+/Sca-1+ progenitor cells after induction of hindlimb
ischemia. Consistent with increased expression of SDF-1,
orchidectomized mice receiving DHT had signicantly ele-
vated numbers of CXCR4+/Sca-1+ cells present in the bone
marrow at day 3 after ischemia (0.8 ± 0.2% vs. 0.4 ± 0.07% of
MNCs for orchidectomized + DHT vs. controls, respectively;
P < 0.05 using ANOVA; Fig. 6 C). Trends similar to those
seen for ulex+/AcLDL+ cells were observed in the spleen for
CXCR4+/Sca-1+ cells (i.e., decrease with castration and increase
with DHT treatment), but these were not statistically signi-
cant (Fig. 6 D). The mobilization of cells from the bone mar-
row to the periphery is a highly coordinated, time-dependent
event. Therefore, because sampling took place at day 3, it is
possible that the signicantly elevated numbers of Sca-1+/
CXCR4+ cells in the bone marrow would not correspond to
higher levels in the periphery until a later time point. Overall,
our ndings suggest that androgens play a role in the prolifera-
tion and mobilization of circulating angiogenic cell populations
in the context of ischemia.
Finally, in light of the well-established eects of andro-
gens on erythropoiesis (Kennedy and Gilbertsen, 1957), we
assessed erythroid progenitor cell levels, as these cells have
been demonstrated to augment angiogenesis (Sasaki et al.,
2007). Gonadal status had no eect on the numbers of gran-
ulocyte/macrophage progenitors present in the bone mar-
row or the spleen (Fig. 6, E and F). However, consistent
VEGF in this context. Overall, these ndings suggest that
endogenous androgens play a role in the coordination of
ischemia-mediated angiogenesis by the regulation of key
angiogenesis-related genes, including HIF-1 and SDF-1.
Androgens modulate ischemia-induced
angiogenic/progenitor cell mobilization
It is now clear that the mobilization of various angiogenic/
progenitor cells from the bone marrow to sites of ischemia is
critical for neovascularization (Sieveking and Ng, 2009). Because
the spleen serves as a reservoir for peripheral stem/progenitor
cells (Heeschen et al., 2003), to further elucidate the mecha-
nisms of androgen-induced angiogenesis in hindlimb ischemia,
the early mobilization of angiogenic/progenitor cells was as-
sessed in both the bone marrow and the spleen. Mononuclear
cells (MNCs) staining positive for ulex lectin and uptake of Dil-
acetylated low density lipoprotein (AcLDL) represent a hetero-
geneous population of cells with angiogenic activity (Asahara
et al., 1997). Although the precise role of these cells in angiogen-
esis has been debated, these cells have been reported to aug-
ment angiogenesis in a paracrine fashion through the secretion
of various growth factors (Heil et al., 2004; Sieveking et al.,
2008). In females, estrogen has also been demonstrated to mo-
bilize bone marrow–derived angiogenic/progenitor cells that
participate in cardiovascular regeneration (Iwakura et al., 2006).
Similarly, the ndings of this study show that androgens modu-
late the mobilization of angiogenic cells. Orchidectomy mark-
edly decreased the numbers of ulex+/AcLDL+ cells in the bone
marrow (22.8 ± 1 vs. 13.4 ± 0.9 cells/200× eld for control vs.
orchidectomized mice, respectively; P < 0.001 using ANOVA;
Fig. 6 A) and the spleen (20.6 ± 0.8 vs. 15.2 ± 0.7 cells/200×
eld for control vs. orchidectomized mice, respectively; P <
0.001 using ANOVA; Fig. 6 B). DHT treatment rescued
Figure 6. Endogenous androgens modulate angiogenic/progenitor cell mobilization in response to ischemia. (A and B) Ulex+/AcLDL+ cells in the
bone marrow (A) and spleen (B). Cells were seeded at a density of 5 × 106 cells/mm2 and after 4 d were assessed for the ability to ingest 4 µg/ml AcLDL
(Invitrogen) and to bind 10 µg/ml FITC–UEA-1 (Sigma-Aldrich) via uorescence microscopy. (C and D) SCA-1+/CXCR4+ cells in the bone marrow (C) and
spleen (D). Hematopoietic progenitor cells were assessed using methylcellulose assays. (E and F) Granulocyte/macrophage CFUs in the bone marrow
(E) and spleen (F). (G and H) Erythroid CFUs in the bone marrow (G) and spleen (H; n = 6/group; combined data from three independent experiments
are shown). *, P < 0.05; **, P < 0.01; and ***, P < 0.001 using ANOVA. All data are expressed as means ± SEM.
JEM VOL. 207, February 15, 2010 351
Brief Definitive Report
Real-time RT-PCR. Real-time RT-PCR was performed in triplicate using
iQ SYBR Green Supermix and the iCycler Real-Time PCR Detection System
(Bio-Rad Laboratories). For the animal studies, the relative expression (R) of
each target mRNA was calculated based on the Pfa equation (Pfa, 2001),
as follows: R = (Etarget)∆Cttarget/(Ereference)∆Ctreference, where ∆Ct = Ctischemic
Ctnonischemic. The reference was -actin or 2-microglobuin mRNA.
Animal studies. All animal studies were performed under ethical approvals
of the Sydney South West Area Health Service Animal Welfare Commit-
tee. Male and female C57Bl6/J mice at 6 wk of age were randomized to or-
chidectomy, ovariectomy, or sham castration. 10 d later, castrated mice
received a 1-cm subdermal silastic implant lled with crystalline DHT.
Matrigel plug implantation model. Matrigel plug assays were performed
as described previously (Passaniti et al., 1992). Commercially available
Matrigel (BD) without phenol red was supplemented with 100 ng/ml of
basic broblast growth factor.
Hindlimb ischemia model. Hindlimb ischemia and subsequent perfusion
(Moor Instruments) and monitoring was performed as described previously
(Stabile et al., 2003).
Angiogenic/progenitor cell mobilization. For ow cytometric analysis
(FC500; Beckman Coulter), MNCs were stained with PE-conjugated anti-
bodies against SCA-1 (BioLegend) and FITC-conjugated antibodies against
CXCR4 (BD) with matched isotype controls. CFU assays for granulocyte/
macrophage and erythroid progenitors were set up in methylcellulose me-
dium (MethoCult; STEMCELL Technologies Inc.).
Statistical analysis. Data are expressed as means ± SEM. Comparisons be-
tween groups were performed using a two-tailed t test or by a one-way
ANOVA with post hoc analyses for pairwise comparisons (Newman-Keuls
multiple comparison). Prism software (version 4.00; GraphPad Software, Inc.)
for Windows was used for statistical analysis.
Online supplemental material. Fig. S1 depicts a dose-dependent aug-
mentation of Matrigel-based tubulogenesis in male ECs exposed to DHT for
24 h. In Fig. S2, we show dierential gene expression of the AR in male and
female ECs as well as female ECs transfected with an AR expression vector.
Fig. S3 demonstrates the eect of castration and DHT treatment on the
functional recovery of male mice subjected to femoral artery ligation. Fig. S4
shows the expression of angiogenesis-related genes in response to hindlimb
ischemia. Online supplemental material is available at http://www.jem
.org/cgi/content/full/jem.20091924/DC1.
We wish to thank Professor R.H. Karas for providing scientic advice during the
preparation of this paper.
This study was supported by a grant from the National Health and Medical
Research Council (457534). D.P. Sieveking was supported by a postgraduate support
grant from GlaxoSmithKline Australia.
The authors have no conicting nancial interests.
Submitted: 3 September 2009
Accepted: 21 December 2009
REFERENCES
Abaci, A., A. Ogˇ uzhan, S. Kahraman, N.K. Eryol, S. Unal, H. Arinç, and
A. Ergin. 1999. Eect of diabetes mellitus on formation of coronary
collateral vessels. Circulation. 99:2239–2242.
Akane, A. 1998. Sex determination by PCR analysis of the X-Y amelogenin
gene. Methods Mol. Biol. 98:245–249.
Araujo, A.B., V. Kupelian, S.T. Page, D.J. Handelsman, W.J. Bremner,
and J.B. McKinlay. 2007. Sex steroids and all-cause and cause-specic
mortality in men. Arch. Intern. Med. 167:1252–1260. doi:10.1001/
archinte.167.12.1252
with the enhanced recovery from ischemia observed in or-
chidectomized mice treated with DHT, erythroid progeni-
tors were signicantly elevated in these mice in both the
bone marrow (29.5 ± 2 vs. 20.5 ± 1 erythroid CFUs/105
MNCs for orchidectomized + DHT vs. control, respec-
tively; P < 0.01 using ANOVA; Fig. 6 G) and the spleen
(10.6 ± 1 vs. 5.5 ± 1 erythroid CFUs/105 MNCs for orchi-
dectomized + DHT vs. control, respectively; P < 0.01 using
ANOVA; Fig. 6 H). These data suggest that the enhanced
recovery from hindlimb ischemia seen in orchidectomized
males receiving DHT may be mediated in part by increased
mobilization of erythroid progenitors. In toto, the data pre-
sented in this report suggest that endogenous androgens are
necessary in the coordination of neovascularization in re-
sponse to critical ischemia and that consequent proangio-
genic eects of DHT in this context are mediated in part
through increased angiogenic/progenitor cell mobilization.
In summary, we report a sex-specic role for androgens
in angiogenesis. Androgens stimulate key angiogenic events
in male but not female cells in vitro, and these sex-specic
proangiogenic eects are mediated via the AR. In addition,
we report that endogenous androgens modulate angiogenesis
in males but not females in vivo. Moreover, in males, endog-
enous androgens are involved in the coordination and en-
hancement of neovascularization in the context of ischemic
injury. Our ndings suggest that androgens regulate vascular
regeneration in a sex-dependent manner. Given the age-
related decline of androgens, our ndings have implications
for the role of androgen replacement in men. Additionally,
these data may explain in part some of the observed sex dif-
ferences in the outcomes of CVD.
MATERIALS AND METHODS
Cell culture. Human umbilical vein ECs (HUVECs), freshly isolated as de-
scribed previously (Sieveking et al., 2008), were used in this study as they are
well validated in the study of angiogenesis and enabled the assessment of a
variety of male and female donors by primary cell harvest and culture. The
sex of HUVECs derived from male and female newborns was conrmed via
PCR analysis of the X-Y amelogenin gene (Akane, 1998). Cells (passages 2–4)
were grown in phenol red–free endothelial growth medium supplemented
with FBS that was stripped of endogenous steroids by 2% charcoal-dextran
treatment. Cells were treated with the nonaromatizable androgen DHT (0,
4, 40, or 400 nM) with and without the AR antagonist HF (40 µM), and as-
sayed for migration, proliferation, and vascular network formation as previ-
ously described (Sieveking et al., 2008), and a traditional Matrigel assay.
Umbilical cords were obtained under ethical approvals of the Sydney South
West Area Health Service Human Ethics Committee.
AR silencing/overexpression. AR silencing was assessed using a com-
mercially available plasmid containing an oligonucleotide encoding an AR
siRNA (pKD-AR-v2; Millipore). To increase AR expression in female
cells, we used an AR expression plasmid (pSVARo; provided by A.O.
Brinkmann, Erasmus University Rotterdam, Rotterdam, Netherlands).
DHT and VEGF signaling in male ECs. After exposure to DHT for
48 h, cell lysates were assayed via ELISA (R&D Systems). Additionally, the
eect of DHT on male EC tubulogenesis was assessed in the presence of 1 µg/
ml anti-VEGF antibody (R&D Systems) or 10 µM LY294002 (Merck), an inhib-
itor of PI3K. Expression of Flt-1 and KDR was assessed via ow cytometry
using PE-conjugated antibodies against each antigen (R&D Systems).
352 Androgens and angiogenesis | Sieveking et al.
Losordo, D.W., and J.M. Isner. 2001. Estrogen and angiogenesis: A review.
Arterioscler. Thromb. Vasc. Biol. 21:6–12.
McCrohon, J.A., W. Jessup, D.J. Handelsman, and D.S. Celermajer. 1999.
Androgen exposure increases human monocyte adhesion to vascular
endothelium and endothelial cell expression of vascular cell adhesion
molecule-1. Circulation. 99:2317–2322.
McCrohon, J.A., A.K. Death, S. Nakhla, W. Jessup, D.J. Handelsman, K.K.
Stanley, and D.S. Celermajer. 2000. Androgen receptor expression is
greater in macrophages from male than from female donors. A sex dif-
ference with implications for atherogenesis. Circulation. 101:224–226.
Morales, D.E., K.A. McGowan, D.S. Grant, S. Maheshwari, D. Bhartiya,
M.C. Cid, H.K. Kleinman, and H.W. Schnaper. 1995. Estrogen pro-
motes angiogenic activity in human umbilical vein endothelial cells in
vitro and in a murine model. Circulation. 91:755–763.
Ng, M.K., W. Jessup, and D.S. Celermajer. 2001. Sex-related dierences
in the regulation of macrophage cholesterol metabolism. Curr. Opin.
Lipidol. 12:505–510. doi:10.1097/00041433-200110000-00005
Ng, M.K., S. Nakhla, A. Baoutina, W. Jessup, D.J. Handelsman, and D.S.
Celermajer. 2003a. Dehydroepiandrosterone, an adrenal androgen, in-
creases human foam cell formation: a potentially pro-atherogenic eect.
J. Am. Coll. Cardiol. 42:1967–1974. doi:10.1016/j.jacc.2003.07.024
Ng, M.K., C.M. Quinn, J.A. McCrohon, S. Nakhla, W. Jessup, D.J.
Handelsman, D.S. Celermajer, and A.K. Death. 2003b. Androgens up-
regulate atherosclerosis-related genes in macrophages from males but not
females: molecular insights into gender dierences in atherosclerosis.
J. Am. Coll. Cardiol. 42:1306–1313. doi:10.1016/j.jacc.2003.07.002
Passaniti, A., R.M. Taylor, R. Pili, Y. Guo, P.V. Long, J.A. Haney, R.R.
Pauly, D.S. Grant, and G.R. Martin. 1992. A simple, quantitative
method for assessing angiogenesis and antiangiogenic agents using re-
constituted basement membrane, heparin, and broblast growth factor.
Lab. Invest. 67:519–528.
Pfa, M.W. 2001. A new mathematical model for relative quantica-
tion in real-time RT-PCR. Nucleic Acids Res. 29:e45. doi:10.1093/
nar/29.9.e45
Sasaki, S., T. Inoguchi, K. Muta, Y. Abe, M. Zhang, K. Hiasa, K.
Egashira, N. Sonoda, K. Kobayashi, R. Takayanagi, and H. Nawata.
2007. Therapeutic angiogenesis by ex vivo expanded erythroid pro-
genitor cells. Am. J. Physiol. Heart Circ. Physiol. 292:H657–H665.
doi:10.1152/ajpheart.00343.2006
Semenza, G.L. 2002. HIF-1 and tumor progression: pathophysi-
ology and therapeutics. Trends Mol. Med. 8(Suppl.):S62–S67.
doi:10.1016/S1471-4914(02)02317-1
Shaw, L.J., C.N. Bairey Merz, C.J. Pepine, S.E. Reis, V. Bittner, S.F.
Kelsey, M. Olson, B.D. Johnson, S. Mankad, B.L. Sharaf, et al; WISE
Investigators. 2006. Insights from the NHLBI-sponsored Women’s
Ischemia Syndrome Evaluation (WISE) study: Part I: gender dierences
in traditional and novel risk factors, symptom evaluation, and gender-
optimized diagnostic strategies. J. Am. Coll. Cardiol. 47(Suppl. 1):S4–
S20. doi:10.1016/j.jacc.2005.01.072
Sieveking, D.P., and M.K. Ng. 2009. Cell therapies for therapeutic
angiogenesis: back to the bench. Vasc. Med. 14:153–166. doi:10.1177/
1358863X08098698
Sieveking, D.P., A. Buckle, D.S. Celermajer, and M.K. Ng. 2008. Strikingly
dierent angiogenic properties of endothelial progenitor cell subpopu-
lations: insights from a novel human angiogenesis assay. J. Am. Coll.
Cardiol. 51:660–668. doi:10.1016/j.jacc.2007.09.059
Stabile, E., M.S. Burnett, C. Watkins, T. Kinnaird, A. Bachis, A. la Sala,
J.M. Miller, M. Shou, S.E. Epstein, and S. Fuchs. 2003. Impaired arte-
riogenic response to acute hindlimb ischemia in CD4-knockout mice.
Circulation. 108:205–210. doi:10.1161/01.CIR.0000079225.50817.71
Tivesten, A., L. Vandenput, F. Labrie, M.K. Karlsson, O. Ljunggren, D.
Mellström, and C. Ohlsson. 2009. Low serum testosterone and es-
tradiol predict mortality in elderly men. J. Clin. Endocrinol. Metab.
94:2482–2488.
Wingard, D.L., L. Suarez, and E. Barrett-Connor. 1983. The sex dierential
in mortality from all causes and ischemic heart disease. Am. J. Epidemiol.
117:165–172.
Wu, F.C., and A. von Eckardstein. 2003. Androgens and coronary artery
disease. Endocr. Rev. 24:183–217. doi:10.1210/er.2001-0025
Asahara, T., T. Murohara, A. Sullivan, M. Silver, R. van der Zee, T. Li, B.
Witzenbichler, G. Schatteman, and J.M. Isner. 1997. Isolation of puta-
tive progenitor endothelial cells for angiogenesis. Science. 275:964–967.
doi:10.1126/science.275.5302.964
Boddy, J.L., S.B. Fox, C. Han, L. Campo, H. Turley, S. Kanga, P.R.
Malone, and A.L. Harris. 2005. The androgen receptor is signicantly
associated with vascular endothelial growth factor and hypoxia sens-
ing via hypoxia-inducible factors HIF-1a, HIF-2a, and the prolyl hy-
droxylases in human prostate cancer. Clin. Cancer Res. 11:7658–7663.
doi:10.1158/1078-0432.CCR-05-0460
Carmeliet, P. 2005. Angiogenesis in life, disease and medicine. Nature.
438:932–936. doi:10.1038/nature04478
Ceradini, D.J., and G.C. Gurtner. 2005. Homing to hypoxia: HIF-1 as a me-
diator of progenitor cell recruitment to injured tissue. Trends Cardiovasc.
Med. 15:57–63. doi:10.1016/j.tcm.2005.02.002
Ceradini, D.J., A.R. Kulkarni, M.J. Callaghan, O.M. Tepper, N. Bastidas,
M.E. Kleinman, J.M. Capla, R.D. Galiano, J.P. Levine, and G.C.
Gurtner. 2004. Progenitor cell tracking is regulated by hypoxic gra-
dients through HIF-1 induction of SDF-1. Nat. Med. 10:858–864.
doi:10.1038/nm1075
Counhal, T., M. Silver, L.P. Zheng, M. Kearney, B. Witzenbichler,
and J.M. Isner. 1998. Mouse model of angiogenesis. Am. J. Pathol.
152:1667–1679.
Death, A.K., K.C. McGrath, M.A. Sader, S. Nakhla, W. Jessup, D.J.
Handelsman, and D.S. Celermajer. 2004. Dihydrotestosterone promotes
vascular cell adhesion molecule-1 expression in male human endothe-
lial cells via a nuclear factor-kappaB-dependent pathway. Endocrinology.
145:1889–1897. doi:10.1210/en.2003-0789
Ferrara, N., H.P. Gerber, and J. LeCouter. 2003. The biology of VEGF and
its receptors. Nat. Med. 9:669–676. doi:10.1038/nm0603-669
Harman, S.M. 2005. Testosterone in older men after the Institute of
Medicine Report: where do we go from here? Climacteric. 8:124–135.
doi:10.1080/13697130500042417
Heeschen, C., A. Aicher, R. Lehmann, S. Fichtlscherer, M. Vasa,
C. Urbich, C. Mildner-Rihm, H. Martin, A.M. Zeiher, and S.
Dimmeler. 2003. Erythropoietin is a potent physiologic stimulus
for endothelial progenitor cell mobilization. Blood. 102:1340–1346.
doi:10.1182/blood-2003-01-0223
Heil, M., T. Ziegelhoeer, B. Mees, and W. Schaper. 2004. A dierent
outlook on the role of bone marrow stem cells in vascular growth:
bone marrow delivers software not hardware. Circ. Res. 94:573–574.
doi:10.1161/01.RES.0000124603.46777.EB
Iwakura, A., S. Shastry, C. Luedemann, H. Hamada, A. Kawamoto, R.
Kishore, Y. Zhu, G. Qin, M. Silver, T. Thorne, et al. 2006. Estradiol
enhances recovery after myocardial infarction by augmenting incorpo-
ration of bone marrow-derived endothelial progenitor cells into sites
of ischemia-induced neovascularization via endothelial nitric oxide
synthase-mediated activation of matrix metalloproteinase-9. Circulation.
113:1605–1614. doi:10.1161/CIRCULATIONAHA.105.553925
Jin, D.K., K. Shido, H.G. Kopp, I. Petit, S.V. Shmelkov, L.M. Young, A.T.
Hooper, H. Amano, S.T. Avecilla, B. Heissig, et al. 2006. Cytokine-
mediated deployment of SDF-1 induces revascularization through
recruitment of CXCR4+ hemangiocytes. Nat. Med. 12:557–567.
doi:10.1038/nm1400
Joseph, J., and M. Dyson. 1965. Sex dierences in the rate of tissue regenera-
tion in the rabbit’s ear. Nature. 208:599–600. doi:10.1038/208599a0
Kennedy, B.J., and A.S. Gilbertsen. 1957. Increased erythropoiesis induced
by androgenic-hormone therapy. N. Engl. J. Med. 256:719–726.
Khaw, K.T., M. Dowsett, E. Folkerd, S. Bingham, N. Wareham, R.
Luben, A. Welch, and N. Day. 2007. Endogenous testosterone and
mortality due to all causes, cardiovascular disease, and cancer in men:
European prospective investigation into cancer in Norfolk (EPIC-
Norfolk) Prospective Population Study. Circulation. 116:2694–2701.
doi:10.1161/CIRCULATIONAHA.107.719005
Laughlin, G.A., E. Barrett-Connor, and J. Bergstrom. 2008. Low serum
testosterone and mortality in older men. J. Clin. Endocrinol. Metab.
93:68–75. doi:10.1210/jc.2007-1792
Liu, P.Y., A.K. Death, and D.J. Handelsman. 2003. Androgens and cardio-
vascular disease. Endocr. Rev. 24:313–340. doi:10.1210/er.2003-0005
... It is known; the estrogen increased the vascularization in some organs (uterus, heart, brain etc.) by expression and releasing of angiogenic factors in female [3][4][5] . There are few and limited studies about the effect of testosterone on angiogenesis [6,7] . In addition to the factors mentioned above angiogenesis are also affected by the age. ...
... Few studies have been made regarding the effects of androgens on angiogenesis [6,7] . It was determined that androgens stimulate erythropoietin production via VEGF in cell culture and endothelial stem cells are regulated hormonally. ...
... It was determined that androgens stimulate erythropoietin production via VEGF in cell culture and endothelial stem cells are regulated hormonally. In the reduced level of these hormones due to castration angiogenesis is also downward as it was shown previously [7] . The changes due to castration includes the decrease in endothelial function of vessel (Arteria femoralis) and deterioration in calcium channel activity as a result of decrease in testosterone level and the absence of androgen receptor in testicular feminized mice [11] . ...
Article
Full-text available
Citation of This Article Dabanoğlu İ, Bozdoğan B, Kum Ş, Ünübol Aypak S: The angiogenic effect of testosterone supplementation on brain of aged mice. Kafkas Univ Vet Fak Derg, 23 (2): 247-251, 2017. Abstract The aim of this study was to determine, by histological and molecular techniques, the effects of testosterone hormone treatment on angiogenesis in brain of aged mice. A total of 30 mice were used in 3 study groups: Sham operation group (Control), gonadectomy group (G) and gonadectomy and testosterone supplementation group (GTS). The capillary number and the inner diameter of larger capillary vessels in brain were measured by light microscope. The levels of VEGF (vascular endothelial growth factor) mRNA in brain tissues were determined by RT-PCR. After gonadectomy operation, capillary densities of brain decreased in male (P<0.05) but did not changed in female. Testosterone supplementation to gonadectomized aged mice caused a mild increase on capillary number in female brain but did not affect in male. Gonadectomy caused a decrease in VEGF mRNA levels of brain in male and female mice. Interestingly, testosterone replacement caused an important decrease (P<0.05) in the expression of VEGF of brain in male compared to control group whereas resulted with no change in female. The present results showed that testosterone hormone has different angiogenic effect on brain in male and female old mice. The gonadectomy operation in old male mice has a negative effect on angiogenic events. However, testosterone replacement in male was not sufficient to convert this change and did not increase angiogenesis.
... It is well known that estrogen increases the vascularization in some organs (e.g. the uterus, the heart, and the brain) by releasing angiogenic factors and increasing their expressions in female rats and women [4][5][6] . There are few and limited studies on the effect of testosterone on angiogenesis [7][8][9][10][11] . Testosterone has protective effects on formation of cells in the arterial vessels [12] . ...
... Limited studies on the eff ects of androgens on angiogenesis have been found [7,8] . Sieveking et al. [8] reported that androgens stimulate erythropoietin production via VEGF in cell culture and endothelial stem cells. ...
... Limited studies on the eff ects of androgens on angiogenesis have been found [7,8] . Sieveking et al. [8] reported that androgens stimulate erythropoietin production via VEGF in cell culture and endothelial stem cells. Angiogenesis is also down regulated when the levels of these hormones reduce after castration as stated previously [8] . ...
Article
Full-text available
How to Cite This Article Dabanoğlu İ, Bozdoğan B, Kum Ş: The effect of testosterone supplementation on capillar morphometry and VEGF expression level of the heart in aged mice. Kafkas Univ Vet Fak Derg, 25 (6): 801-806, 2019. Abstract The aim of this study was to determine the effect of testosterone hormone supplementation on the capillary morphometry and VEGF expression level of the heart of old mice using histological and molecular techniques. A total of 30 old mice were enrolled in 3 groups: sham operation group (Control), gonadectomy group (G) and gonadectomy and testosterone supplementation group (GTS). The capillary number and the inner diameter of larger capillary vessels in the heart were measured by light microscope in terms of angiogenic processes. The levels of angiogenic factor, VEGF (vascular endothelial growth factor), mRNA in the heart tissue were determined by RT-PCR. The capillary densities decreased in female mice, in which G and GTS (P<0.05) groups compared to the control group, whereas there was no alteration in male mice. Gonadectomy led to a reduction in VEGF expression. Interestingly, testosterone replacement (in GTS group) caused even more reduction in VEGF expression in male and female (P<0.05) compared to the control group. The present results showed that there was a variation affected by gender in the regulation of angiogenesis by testosterone in the heart. However, testosterone replacement was not sufficient to restore the effects of gonadectomy on the heart in old mice.
... There are publications in the literature demonstrating that androgens contribute to angiogenesis, which is considered one of the main mechanisms of CCC, at the microvascular level through the androgen receptor (17) . Moreover, it has been proposed that androgens can activate vascular repair and neovascularisation (18) . ...
... Moreover, it has been proposed that androgens can activate vascular repair and neovascularisation (18) . Although significant studies have been conducted on the relationship of androgens with CAD and neovascularisation, (11,17,18) studies on androgens and CCC are limited. In this study, we aimed to investigate the relationship between androgens and CCC in male patients with coronary artery occlusion, which plays a crucial role in the prevention of cardiovascular mortality and morbidity. ...
... Numerous in vivo and in vitro studies have shown that androgens contribute to angiogenesis by transcriptionally controlling the target genes through androgen receptors and by increasing the interaction of multiple signaling pathways (17,18) . Simultaneously, studies that have shown that androgens increase arterial blood flow causing vascular vasodilatation have proposed that androgens may contribute to angiogenesis, which is one of the fundamental mechanisms of coronary collateral circulation (CCC) (24,25) . ...
Article
Full-text available
Introduction: Coronary collateral circulation (CCC) is a natural bypass system for restoring blood flow, and a well-developed CCC is held to protect myocardial function and improve survival after coronary obstruction in patients with coronary artery disease (CAD). Sex steroids have been suggested as potentially hampering the course of CAD progression. We explored the relationship between the serum levels of sex steroids and CCC. Patients and Methods: A total of 115 males with chronic coronary artery disease who had at least one total coronary artery occlusion were included. Patients were divided into two groups: a well CCC group (Rentrop grades 2-3, n= 64) and a poorly developed CCC group (Rentrop grades 0-1, n= 51). Serum levels of total testosterone, free testosterone, sex hormone-binding globulin (SHBG), and dehydroepiandrosterone sulfate (DHEA-S) were recorded. A p-value below 0.05 was accepted as significant in all analyses. The confidence interval was accepted as 95%. Results: Serum total testosterone (ng/dL; 274.5 ± 57.7 vs. 329 ± 64.8, p< 0.001), free testosterone (pg/mL; 8.2 ± 2.4 vs. 12 ± 3.2, p< 0.001), DHEAS [ µg /dL; 111 (58) vs. 160 (85.5), p< 0.001] and SHBG concentrations (nmol/L; 29.3 ± 8.6 vs. 33.2 ± 10.2; p= 0.027) were significantly higher in the well coronary collateral group (WCG). According to the results of multiple regression analyses, diabetes [OR= 3.56, CI (1.26-3.5) p= 0.017], free testosterone level [OR= 1.57, CI (1.26-1.96), p< 0.001] and total testosterone level [OR= 1.01, CI (1.00-1.02), p= 0.009] were determined to be independent predictors. Conclusion: This study showed that a high level of sex steroids was a predictor of good collateral development in patients with chronic CAD.
... Testosterone's influence may also be mediated by sex, possibly due to hormonal history or other sex-associated differences in vascular physiology. Sieveking et al. (2010) found that androgen administration increased angiogenesis only in male mice models. Additionally, testosterone levels appear to have opposite actions on ET-1 (a vasoconstrictor) in cisgender vs. transgender men (Polderman et al., 1993;Kumanov et al., 2007;Abi-Ghanem et al., 2020). ...
Article
Full-text available
Gender-affirming hormone replacement therapy (gaHRT) is an important step for many in the gender diverse community, associated with increased quality-of-life and lower self-reported scores of depression and anxiety. However, considering the interactions that the involved sex hormones have on vasculature (with oestrogen and testosterone demonstrating vasodilatory and vasoconstricting properties, respectively), it is important for transgender healthcare research to examine how the manipulation of these hormones interact with cerebrovascular structure and functioning. There is a stark lack of research in this area. This mini-review outlines the research suggesting a vascular impact of these sex hormones using evidence from a range of cohorts (e.g., menopause, polycystic ovary syndrome) and discusses the work that has been done into cerebrovascular changes following gaHRT. Finally, recommendations for future research into cerebrovascular health in transgender cohorts following gaHRT are outlined.
... Specifically, similar to males, androgen receptors (ARs) for testosterone are expressed in vascular endothelial cells in females, although whereas activation of ARs in male endothelial cells results in angiogenesis and vascular repair, this effect is not demonstrated in females. 28,29 Although no independent relationship between testosterone level and vascular function was demonstrated in our study sample, an independent relationship between TE ratio and FMD was demonstrated in post hoc analyses. This finding suggests that higher levels of testosterone relative to estradiol are associated with improved endothelial function in reproductive-aged females living with CKD. ...
Article
Full-text available
Cardiovascular disease is the leading cause of death in women, and women with chronic kidney disease (CKD) experience especially elevated risk. This study examined the association between testosterone and vascular function in 61 reproductive-aged females with CKD. Testosterone levels and measures of vascular function were assessed, including pulse wave velocity, aortic augmentation, flow-mediated dilation (FMD), and velocity time integral. Multivariable linear regression analyses assessed the relationship between testosterone and each measure of vascular function. No associations were observed between testosterone and vascular function outcomes, although a significant positive association between testosterone-to-estradiol ratio and FMD was demonstrated. Although testosterone levels were not independently predictive of vascular function, the level of testosterone relative to estradiol was associated with FMD and may therefore influence endothelial function in the high-risk population of reproductive-aged female patients with CKD.
... One of the key regulators of SDF-1 and CXCR4 under hypoxic conditions is hypoxia-inducible factor-1 (HIF-1) [48][49][50]. Previously, it was reported that the administration of dihydrotestosterone (DHT) increased SDF-1 and CXCR4 levels in a mouse model of hindlimb ischemia [51]. As exposure to Cl 2 gas is known to cause hypoxia [52], it is possible that the increase in SDF-1 and CXCR4 after Cl 2 exposure is due to the upregulation of HIF-1. ...
Article
Full-text available
The aim of the study was to determine whether sex-related differences exist in immune response to inhalation lung injury. C57BL/6 mice were exposed to Cl2 gas (500 ppm for 15, 20, or 30 min). Results showed that male mice have higher rates of mortality and lung injury than females. The binding of the chemokine ligand C-X-C motif chemokine 12 (CXCL12), also called stromal-derived-factor-1 (SDF-1), to the C-X-C chemokine receptor type 4 (CXCR4) on lung cells promotes the migration of leukocytes from circulation to lungs. Therefore, the hypothesis was that elevated SDF-1/CXCR4 signaling mediates exaggerated immune response in males. Plasma, blood leukocytes, and lung cells were collected from mice post-Cl2 exposure. Plasma levels of SDF-1 and peripheral levels of CXCR4 in lung cells were higher in male vs. female mice post-Cl2 exposure. Myeloperoxidase (MPO) and elastase activity was significantly increased in leukocytes of male mice exposed to Cl2. Lung cells were then ex vivo treated with SDF-1 (100 ng/mL) in the presence or absence of the CXCR4 inhibitor, AMD3100 (100 nM). SDF-1 significantly increased migration, MPO, and elastase activity in cells obtained from male vs. female mice post-Cl2 exposure. AMD3100 attenuated these effects, suggesting that differential SDF-1/CXCR4 signaling may be responsible for sex-based disparities in the immune response to inhalation lung injury.
... MMP-2, MMP-7 and MMP-9), which control EC adhesion or detachment from the surrounding stroma; cyclooxygenase enzymes and their products (i.e. prostaglandins and thromboxanes), which can promote VEGF-A production, EC mobilisation and sprouting; specific microRNAs (miRNAs) like miR-296, which is generally overexpressed by PCa cells and regulates VEGF and PDGF receptors in ECs; tumour exosomes-derived sphingomyelin, which can promote EC migration, invasion and tube formation [101][102][103][104][105]. Of note, EC behaviour is influenced by androgens in a sex-specific manner [106]. Androgens increase angiogenic events in vitro and in vivo in male but not female ECs, and these sex-specific proangiogenic effects are VEGF-dependent. ...
Article
Full-text available
The genomic, epigenetic and metabolic determinants of prostate cancer pathobiology have been extensively studied in epithelial cancer cells. However, malignant cells constantly interact with the surrounding environment—the so-called tumour microenvironment (TME)—which may influence tumour cells to proliferate and invade or to starve and die. In that regard, stromal cells—including fibroblasts, smooth muscle cells and vasculature-associated cells—constitute an essential fraction of the prostate cancer TME. However, they have been largely overlooked compared to other cell types (i.e. immune cells). Indeed, their importance in prostate physiology starts at organogenesis, as the soon-to-be prostate stroma determines embryonal epithelial cells to commit toward prostatic differentiation. Later in life, the appearance of a reactive stroma is linked to the malignant transformation of epithelial cells and cancer progression. In this Review, we discuss the main mesenchymal cell populations of the prostate stroma, highlighting their dynamic role in the transition of the healthy prostate epithelium to cancer. A thorough understanding of those populations, their phenotypes and their transcriptional programs may improve our understanding of prostate cancer pathobiology and may help to exploit prostate stroma as a biomarker of patient stratification and as a therapeutic target.
... To our knowledge, it is unknown whether the hormonal status of the organism impacts functional activity in vitro at the tissue level. In HUVECs, androgen treatment augments proliferation and migration in male but not female cell lines and this effect is mediated by the androgen receptor, 27 which could explain sex-based differences in cardiovascular disease with aging. In murine lungs, there are age-and sex-based differences in endothelial cell function, angiogenesis, and gene transcription regulating these pathways, 28 which may also influence human cellular phenotypes. ...
Article
Full-text available
It is unknown whether biological sex influences phenotypes of commercially available human pulmonary artery endothelial cells (HPAECs). Ten lots of commercial HPAECs were used (Lonza Biologics; PromoCell). Five (50%) were confirmed to be genotypically male (SRY+) and five (50%) were confirmed to be female (SRY ‐). Experiments were conducted between passages five – eight. HPAEC phenotype was confirmed with a panel of cell expression markers. Standard assays for proliferation, migration and tube formation were performed in triplicate with technical replicates, under three treatment conditions (EndoGRO, Sigma‐Aldrich). Apoptosis was assessed by exposing cells treated with complete media or low serum media to hypoxic (1% oxygen) or normoxic (20% oxygen) conditions. Laboratory staff was blinded. The median (range) age of male and female donors from whom the HPAECs were derived was 58 (48 – 60) and 56 (33 – 67), respectively. Our results suggest decreased proliferation in genotypically female cells compared with male cells (p=0.09). With increasing donor age, female cells were less proliferative and male cells were more proliferative (p=0.001). Female cells were significantly more apoptotic than male cells by condition (p=0.001). Female cells were significantly more migratory than male cells in complete media but less migratory than male cells under VEGF enriched conditions (p=0.001). There are subtle sex‐based differences in the behavior of HPAECs that depend on donor sex and, less so, age. These differences may undermine rigor and reproducibility. Future studies should define whether biological sex is an important regulator of HPAEC function in health and disease. This article is protected by copyright. All rights reserved.
Article
Testosterone acting via the androgen receptor, and via aromatisation to oestradiol, an activator of the oestrogen receptor, plays key roles in adipose tissue, bone, and skeletal muscle biology. This is reflected in epidemiological studies associating obesity and disordered glucose metabolism with lower serum testosterone concentrations and an increased risk of type 2 diabetes (T2D) in men. Testosterone also modulates erythrocytosis and vascular endothelial and smooth muscle cell function, with potential impacts on haematocrit and the cardiovascular system. The Testosterone for the Prevention of Type 2 Diabetes (T4DM) study enrolled men aged 50 years and over with a waist circumference of 95cm or over, impaired glucose tolerance or newly diagnosed T2D, and a serum testosterone concentration (as measured by chemiluminescence immunoassay) <14.0 nmol/L. The study reported that 2 years treatment with testosterone undecanoate 1000mg, administered 3-monthly intramuscularly, on the background of a lifestyle program, reduced the likelihood of T2D diagnosis by 40% compared to placebo. This effect was accompanied by a decrease in fasting serum glucose, and associated with favourable changes in body composition, hand grip strength, bone mineral density and skeletal microarchitecture, but not in HbA1c, a red blood cell-dependent measure of glycaemic control. There was no signal for cardiovascular adverse events. With the objective of informing translational science and future directions, this commentary discusses mechanistic studies underpinning the rationale for T4DM, and translational implications of the key outcomes relating to glycaemia, and body composition, together with effects on erythrocytosis, and cardiovascular risk and slow recovery of the hypothalamo-pituitary-testicular axis.
Article
Use of the real-time polymerase chain reaction (PCR) to amplify cDNA products reverse transcribed from mRNA is on the way to becoming a routine tool in molecular biology to study low abundance gene expression. Real-time PCR is easy to perform, provides the necessary accuracy and produces reliable as well as rapid quantification results. But accurate quantification of nucleic acids requires a reproducible methodology and an adequate mathematical model for data analysis. This study enters into the particular topics of the relative quantification in real-time RT-PCR of a target gene transcript in comparison to a reference gene transcript. Therefore, a new mathematical model is presented. The relative expression ratio is calculated only from the real-time PCR efficiencies and the crossing point deviation of an unknown sample versus a control. This model needs no calibration curve. Control levels were included in the model to standardise each reaction run with respect to RNA integrity, sample loading and inter-PCR variations. High accuracy and reproducibility (<2.5% variation) were reached in LightCycler PCR using the established mathematical model.
Article
IT has been found that when wounds are made through the full-thickness of the pinna of the rabbit's ear, and tissue of area 1 cm2 is excised (Fig. 1), the tissue deficit is almost completely made good within 98 days, though a small hole always remains (Fig. 2). This entails the regeneration of relatively mobile dorsal skin, elastic cartilage, connective tissue, nerve fibres and immobile vental skin. Furthermore, the process takes place significantly faster in males than in females. It has been reported elsewhere that if either skin1 or cartilage2 is removed from the rabbit's ear it regenerates, the skin completely and the cartilage partially. The replacement of full-thickness deficits has not, however, been reported.
Article
Age-related reduction of serum testosterone may contribute to the signs and symptoms of aging, but previous studies report conflicting evidence about testosterone levels and male mortality. No large prospective cohort study has determined a possible association between serum estradiol and mortality in men. The main objective was to examine the association between serum testosterone and estradiol and all-cause mortality in elderly men. We used specific gas chromatography-mass spectrometry to analyze serum sex steroids at baseline in older men who participated in the prospective population-based MrOS Sweden cohort (n = 3014; mean age, 75 yr; range, 69-80 yr). All-cause mortality by serum testosterone and estradiol levels. During a mean follow-up period of 4.5 yr, 383 deaths occurred. In multivariate hazards regression models, low levels (within quartile 1 vs. quartiles 2-4) of both testosterone [hazard ratio (HR), 1.65; 95% confidence interval (CI), 1.29-2.12] and estradiol (HR, 1.54; 95% CI, 1.22-1.95) associated with mortality. A model including both hormones showed that both low testosterone (HR, 1.46; 95% CI, 1.11-1.92) and estradiol (HR, 1.33; 95% CI, 1.02-1.73) predicted mortality. Risk of death nearly doubled (HR, 1.96; 95% CI, 1.46-2.62) in subjects with low levels of both testosterone and estradiol compared with subjects within quartiles 2-4 of both hormones. Elderly men with low serum testosterone and estradiol have increased risk of mortality, and subjects with low values of both testosterone and estradiol have the highest risk of mortality.
Article
The discovery, over a decade ago, of endothelial progenitor cells that are able to participate in neovascularization of adult tissue has been greeted enthusiastically because of the potential for new cell-based therapies for therapeutic angiogenesis. Since that time, an ever-growing list of candidate cells has been proposed for cardiovascular regeneration. However, to date, pre-clinical and clinical studies evaluating the therapeutic potential of various cell therapies have reported conflicting results, generating controversy. Key issues within the field of cell therapy research include a lack of uniform cellular definitions, as well as inadequate functional characterization of the role of putative stem/progenitor cells in angiogenesis. Given the mixed results of initial clinical studies, there is now a scientific imperative to understand better the vascular biology of candidate cells in order to better translate cell therapy to the bedside. This review will provide a translationally relevant overview of the biology of candidate stem/progenitor cells for therapeutic angiogenesis.
Article
Blood vessel growth is necessary for normal tissue homeostatis and contributes to solid tumor growth. Methods to quantitate neovascularization should be useful in testing biological factors and drugs that regulate angiogenesis or to induce a vascular supply to promote wound healing. An extract of basement membrane proteins (Matrigel) was found to reconstitute into a gel when injected subcutaneously into C57/BL mice and to support an intense vascular response when supplemented with angiogenic factors. New vessels and von Willebrand factor antigen staining were apparent in the gel 2-3 days after injection, reaching a maximum after 3-5 days. Hemoglobin content of the gels was found to parallel the increase in vessels in the gel allowing ready quantitation. Angiogenesis was obtained with both acidic and basic fibroblast growth factors and was enhanced by heparin. Several substances were tested for angiostatic activity in this assay by coinjection in Matrigel with fibroblast growth factor and heparin. Platelet-derived growth factor BB, interleukin 1-beta, interleukin-6, and transforming growth factor-beta were potent inhibitors of neovascularization induced by fibroblast growth factor. Tumor necrosis factor-alpha did not alter the response but was alone a potent inducer of neovascularization when coinjected with Matrigel and heparin. Consistent with the previously demonstrated importance of collagenase in mediating endothelial cell invasion, a tissue inhibitor of metalloproteinases that also inhibits collagenases was found to be a potent inhibitor of fibroblast growth factor-induced angiogenesis. Our assay allows the ready quantitative assessment of angiogenic and anti-angiogenic factors and should be useful in the isolation of endothelial cells from the capillaries that penetrate into the gel.
Article
Wingard, D. L. (Dept. of Community and Family Medicine, U. of California, San Diego, La Jolla, CA 92093), L. Suarez and E. Barrett-Connor. The sex differential in mortality from all causes and ischemic heart disease. Am J Epidemiol 1983; 117: 165–72. The sex differential in mortality from all causes and ischemic heart disease is examined in an upper-middle class Caucasian community of 3516 adults in southern California, who were followed for a minimum of seven years. The influence of several demographic, behavioral, and biologic risk factors is simultaneously controlled for by means of a multiple logistic analysis. Risk factors include age, marital status, education, cigarette smoking, cholesterol, systolic blood pressure, fasting plasma glucose, and obesity. Both the prevalence and relative mortality risk associated with several risk factors differ by sex. Adjustment decreases the sex differential for mortality from 1.7 to 1.3 for all causes and from 4.8 to 2.4 for ischemic heart disease. When analysis is limited to healthy men and women, the adjusted sex differential in mortality is 1.2 for all causes and 2.0 for ischemic heart disease. Findings of this study are compared with two other population-based studies.