You are on page 1of 13

Journal of Pathology

J Pathol 2007; 211: 206–218


Published online in Wiley InterScience
(www.interscience.wiley.com) DOI: 10.1002/path.2077

Review Article

The ageing male reproductive tract


N Sampson,1 G Untergasser,2 E Plas3 and P Berger1 *
1 Institute for Biomedical Ageing Research, Austrian Academy of Sciences, Innsbruck, Austria
2 Tumor Biology and Angiogenesis Laboratory, Division of Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
3 Ludwig Boltzmann Institute for Urology and Andrology, Lainz Hospital, Vienna, Austria

*Correspondence to: Abstract


P Berger, Institute for Biomedical
Ageing Research, Austrian Ageing of the male reproductive system is characterized by changes in the endocrine
Academy of Sciences, system, hypogonadism, erectile dysfunction and proliferative disorders of the prostate
Innsbruck, Austria. gland. Stochastic damage accumulating within ageing leads to progressive dysregulation at
E-mail: peter.berger@oeaw.ac.at each level of the hypothalamic–pituitary–gonadal (HPG) axis and in local auto/paracrine
interactions, thereby inducing morphological changes in reproductive target organs, such
No conflicts of interest were
as the prostate, testis and penis. Despite age-related changes in the HPG axis, endocrine
declared.
functions are generally sufficient to maintain fertility in elderly men. Ageing of the male
reproductive system can give rise to clinically relevant manifestations, such as benign
prostatic hyperplasia (BPH), prostate cancer (PCa) and erectile dysfunction (ED). In
this review, we discuss morphological/histological changes occurring in these organs and
current views and concepts of the underlying pathology. Moreover, we emphasize the
molecular/cellular pathways leading to reduced testicular/penile function and proliferative
disorders of the prostate gland.
Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John
Wiley & Sons, Ltd.
Keywords: prostate; hyperplasia; erectile dysfunction; fertility; testis; testosterone

Introduction Like other ageing processes in the human body,


ageing of the male reproductive tract is caused by
Ageing is defined by biological and demographic multi-factorial, stochastic changes at molecular, cel-
parameters characterized by an impairment of func- lular and regulatory levels. Ageing leads to an array
tion, decreasing environmental responsiveness and, of symptoms similar in particular to endocrine defi-
reciprocally, by an increased susceptibility to age- ciencies of young men, such as sexual dysfunction,
related diseases and mortality [1]. In contrast to the altered body composition, including increased abdom-
genetically programmed development of young indi- inal obesity, decreased muscle mass and strength
viduals, ageing is non-directed and non-programmed. and psychosocial alterations. These and other symp-
Thus, in the sense of a genetic programme, ‘ageing’ or toms of hypogonadism in young men due to known
‘anti-ageing’ genes do not exist and it is unlikely that
causes, such as pituitary disease, can be treated by
a single gene regulates this complex process. Rather,
androgen replacement (reviewed in [3,5,6]). This led
ageing may be considered to be an impairment of
to the hypothesis that decreasing androgen levels in
body functions over time caused by the accumula-
tion of molecular damage in DNA, proteins and lipids. elderly men may be responsible for similar symp-
Some of this damage may be repaired by energy- toms and could be treated accordingly. However, age-
intensive repair mechanisms. This energy expenditure ing in men is a multifactorial process and should
is evolutionarily selected to suit the ecological niche not simply be considered a result of declining hor-
in a non-protected environment. However, in an envi- monal serum levels, particularly of androgens such
ronment artificially protected (for example, by med- as testosterone or dehydroepiandrosterone (DHEA).
ical and technological intervention), life is extended While large, prospective and randomized trials in
beyond the warranty of the soma and damage accumu- elderly men are still lacking, the rather non-specific
lates [2]. The accumulating damage may be eventually symptoms of elderly men do not appear to be sig-
manifested in age-related health issues, such as uri- nificantly correlated with circulating androgen levels
nary tract impairment, prostate hyperplasia, erectile [7–9]. Thus, prevention, retardation or reversal of
dysfunction and decreased fertility, osteoporosis and age-related molecular, cellular and regulatory changes
general frailty, which have a significant impact on the through androgen supplementation remain speculative.
independence, general well-being and morbidity of the This review describes the age-related changes
elderly [3,4]. in endocrine signalling and cellular and structural

Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
www.pathsoc.org.uk
The ageing male reproductive tract 207

alterations with respect to the ageing male reproductive host of non-specific symptoms, including nervousness,
system, with emphasis on gondal function, erectile irritability, psychological depression, impaired mem-
dysfunction and the ageing prostate. In particular, we ory, fatigue, insomnia, hot flushes, periodic sweating
focus on current research into the molecular mech- and loss of sexual vigour. Partial hypergonadotropic
anisms that underlie the age-related morphological hypogonadism characterized by lowered androgen
changes associated with the pathogenesis of benign serum levels due to primary testicular failure [26] leads
prostatic hyperplasia and prostate cancer, two of to compensatory elevated levels of FSH and LH [5,27].
the most common proliferative diseases affecting the This is interwoven with central defects of the hypotha-
elderly male. lamus–pituitary regulatory unit, causing mild hypog-
onadotropic hypogonadism due to secondary gonadal
failure.
Hypothalamus–pituitary–gonadal axis: The causative mechanisms of the rather mod-
age-related histomorphological, functional est age-related decline in serum T levels may be
and regulatory impairment located at all three organ and regulatory levels of
the HPG axis and may involve testicular steroido-
Ageing male characteristics develop as a result of genesis, particularly diminished T secretory capac-
morphological changes in organs that are coupled to ity, as well as changes in synchronized hormone
changes in endocrine networks and testicular function production and altered feed-back mechanisms in the
[10]. These characteristics vary significantly between hypothalamic–pituitary unit (Figure 1). These age-
individuals and appear to be strongly influenced by related changes include: (a) an age-related decrease
environmental and lifestyle factors [11–16]. Clinical in the maximal hypothalamic secretion of GnRH
relevance for the age-related changes with respect to gonadotropes in the pituitary; (b) decreased LH-
to sex steroid hormone serum levels has not been stimulated testosterone secretion caused by persis-
unequivocally demonstrated (in part due to inter- tence of basal LH pulse frequency but an age-related
individual variation), although statistically signifi- decline in maximal and mean LH pulse amplitude
cant correlations have been reported for senile bone and area; and (c) a reduction in negative feedback
loss [17,18], age-related changes in body composi- of tT, bioT and fT on GnRH-driven LH secretion
tion [19,20], reduced cognitive function [21,22] and [28].
atherosclerosis [23,24]. Changes in primary testicular function and histo-
Ageing of the male reproductive system is dis- morphology lead to partial primary hypogonadism
tinct from the female menopause, which is char- in elderly men and are associated with age-related
acterized by the cessation of reproductive capac- decreases in Leydig cell number [29,30], impaired tes-
ity coupled with sudden loss of gonadal endocrine ticular perfusion due to atherosclerosis [31], thicken-
function. Endocrine dysfunctions in young men due ing, widening and hernia-like protrusions of the basal
to pituitary deficiency, Klinefelter’s syndrome or membrane of the tubuli seminiferi, increasing deposits
anorchia are attributable to defects at distinct lev- of lipofuscin in Leydig cells, a functional decline
els, either originating from primary testicular defi- in Leydig cell mitochondrial steroidogenesis due to
ciency or due to secondary causes at the regu- reduced supply of mitochondrial steroid hormone pre-
latory level in the hypothalamus or pituitary. In cursors [32], and a blunted rise in T, i.e. residual
contrast, late-onset hypogonadism (LOH) develops but decreased testicular T secretion capacity upon
slowly due to molecular and cellular ageing processes stimulation by human chorionic gonadotropin (hCG)
and involves each of the different regulatory levels [5,33] (reviewed in [10]). Moreover, an age-related
(hypothalamus, pituitary, testis) [10]. LOH is charac- cyclooxygenase-2 (COX2)-dependent tonic inhibition
terized by progressive dysregulation of the hypothala- of Leydig cell steroidogenesis and the expression of
mic–pituitary–gonadal (HPG) axis, associated with a the steroidogenic acute regulatory protein (StAR) may
decline in serum testosterone [total testosterone (tT), inhibit the rate-limiting step of testosterone biosynthe-
longitudinally: −1.6%/year) and a counter-regulatory sis [34].
rise in luteinizing hormone (LH) and follicle stimu- Functional testicular impairment and increasing
lating hormone (FSH) of +0.9%/year and 3.1%/year, SHBG levels lead to decreasing levels of bioT that in
respectively [25]. Serum levels for sex hormone bind- turn should evoke a full compensatory up-regulation
ing globulin (SHBG) conversely increase at a rate of the hypothalamic–pituitary unit (for review, see
of 1.2%/year, resulting in a more rapid decrease of [5]). Although modestly elevated LH serum levels
circulating free testosterone (FT) and bioavailable T are observed in elderly men, these levels are lower
(bioT, free plus albumin-bound T) levels than tT than expected from studies with primary hypogo-
(2–3%/year). This gradual decline in tT is accom- nadal young adults and seem to be the result of a
panied by the appearance of clinical symptoms that longer metabolic half-life, rather than due to increased
bear similarities to hypogonadism in young men, secretion [35]. This indicates that with increasing age
including decreased bone and muscle mass, abdom- GnRH stimulation of high-amplitude LH secretion is
inal obesity and decreased sexual body hair and insufficient, due to the failure of hypothalamic neurons
beard growth. In addition, hypogonadism results in a to generate synchronized pulsatile GnRH secretion

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
208 N Sampson et al

Male reproductive functions, sperm


parameters and genetic risks

The second major testicular function apart from


steroidogenesis is the production of sperm. The
increase in male mean life expectancy accompanied
by a trend towards higher paternal age and develop-
ments in assisted reproduction have raised interest in
age-related aspects of male fertility. To what extent
ageing affects semen parameters and sperm function
in men is still debated, as no prospective longitudi-
nal studies are available. Age-related changes of the
testis influence sperm parameters, notably semen vol-
ume, sperm count, motility, morphology and fecun-
dity, and are reflected by a longitudinal increases
in serum FSH and LH (see above) [27,36]. Consid-
ering the multifactorial stochastic nature of ageing
processes, high intra-individual variations of sperm
parameters are not surprising. Several studies describe
age-related declines in semen volume, sperm count
and motility [36–38], indicating decreases in semen
Figure 1. Ageing male hypogonadism. In ageing men volume of 3–22%, sperm motility of 3–37% and
stochastic damage leads to gradual impairment of the
hypothalamus–pituitary–testicular axis, which is manifested as morphological spermatozoal abnormalities in 4–18%
an age-related decrease of tT, fT and bioT (A). Primary testicular of men aged >50 years compared with younger men
changes, including decreased numbers of Leydig cells, increased (<30 years). However, these results are influenced by
deposits of lipofuscin and disrupted steroidogenesis, reduce increased latency time between ejaculations, due to
T synthesis and T synthesis reserve capacity. Age-associated reduced sexual activity. Whether ageing has a nega-
increases in SHBG serum levels further aggravate decreases
in bioT and fT compared to tT. Although decreased, tive correlation with sperm count is not yet clear. Even
testicular functional reserve capacity is generally sufficient when adjusted for sexual abstinence, conflicting stud-
for adequate GnRH/LH feed-forward signals, which should ies report increased, decreased or unchanged sperm
result in fully compensatory T secretion. However, inadequate counts with ageing [39–42].
amplitude of GnRH and LH synchronous pulses (B and Ageing is associated with testicular histomorpholog-
C, respectively) due to lower numbers, and insufficient
synchronization of hypothalamic neurons with ageing lowers ical changes that develop gradually. In addition to the
testicular T output and availability. Thus, mild secondary above-mentioned changes, a reduction in the number
hypogonadism of the hypothalamic–pituitary unit in elderly of type A dark spermatogonia, an increasing number
men results in the inability to compensate for mostly of multinucleated spermatogonia and development of
mild primary testicular hypogonadism/hypoandrogenism. This megalospermatocytes, giant spermatids and multilay-
secondary hypogonadism is observed, although there may
be reduced T feedback inhibition, maintained LH secretory
ered spermatogonia has been described (for review,
capacity of gonadotropes, increased LH metabolic half-life see [10,37,38]). A decrease in Sertoli cell number and
and increased efficacy of suboptimal effective GnRH pulses function with ageing may correlate with a decreasing
[5,28]. Men >80 years exhibit increasing LH levels, which may sperm count.
also occur in middle-aged men due to the loss of elevated Whilst ageing clearly has a negative effect on fecun-
opioid tone (reviewed in [6,10]). The effects of regulatory and
histomorphological changes in endocrine organs and networks
dity in females, there are few studies on sperm function
on reproduction and fertility in elderly men remain to be related to ageing. Despite the use of different assay
fully elucidated. Solid arrows represent age-related changes in techniques, (including the hamster oocyte penetration
organ functions and hormone secretion; broken lines indicate assay, acrosome reaction and spermatozoal chromatin
regulatory pathways. LC, Leydig cells; SC, Sertoli cells condensation) no significant change in sperm function
has been determined [43,44]. However, it seems that
men contribute to the reduced fertility of couples at
bursts of appropriately high amplitudes [5,10]. This the end of the fourth decade of life and to diminished
is despite a diminished sensitivity to inhibitory T fecundity a few years later [37]. Recent investigations
feedback and a greater potency of suboptimal GnRH suggest a reduced fecundity in terms of time to preg-
pulses on LH secretion [28]. In contrast to previous nancy (TTP) not only with rising maternal but also
observations that ageing men have an increased T paternal age [45], although further studies are required
feedback response [5], a recent publication convinc- to clearly demonstrate a correlation [37].
ingly showed that T feedback inhibition is reduced Male ageing seems to be coupled to genetic muta-
[28]. The functional and histomorphological age- tions and risks that include numerical and structural
related changes seem to contribute to diminished T chromosomal aberrations (increased incidence of tri-
biosynthesis and further decrease levels of fT and somy 21, Down’s syndrome), diseases of complex
bioT. aetiology (schizophrenia) and inherited autosomal

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The ageing male reproductive tract 209

dominant diseases (Apert’s syndrome and achondro- cavernosum, thereby amplifying smooth muscle
plasia; reviewed in [37,38]). However, it should be relaxation and vasodilation. These agents demonstrate
kept in mind that the statistical power is low due to improvement in ED and are well tolerated [52].
the extremely low incidence of these genetic disor-
ders, especially when combined with the relatively rare
event of fathers of advanced age. The ageing prostate

Structural and cellular organization of the prostate


Erectile dysfunction: the ageing penis The prostate is a walnut-sized sex accessory gland sur-
rounding the urethra at the base of the bladder and
Erectile dysfunction (ED), the inability to achieve produces secretions that comprise a major fraction
or maintain an erection of sufficient rigidity for (∼30%) of seminal plasma in human ejaculate. The
completion of the sexual act, affects millions of men human prostate can be divided into four anatomically
worldwide. ED is strongly associated with ageing and distinct zones; peripheral, transition and central zones
is particularly common in men older than 50 years and the anterior fibromuscular stroma. At least five
[46,47]. ED is frequently associated with age-related cell types [stem cells, transit-amplifying cells, basal,
disorders, such as atherosclerosis, hypertension [48], secretory and neuroendocrine (NE) cells] comprise
diabetes [49] and after radical prostatectomy for the the stratified epithelial cell layer, which is thought to
treatment of age-related prostatic disease [50]. exist in a continuum of differentiation [57–59]. Sup-
Penile erection is a complex neurovascular function porting the overlying epithelium, the prostatic stroma
that is predominantly mediated by nitric oxide–cGMP consists of smooth muscle cells (SMCs), fibroblasts,
(NO–cGMP) signalling in smooth muscle cells (SMC) myofibroblasts, endothelial cells and components of
of helicine arteries and in the trabeculae [51]. The the extracellular matrix (ECM). Growth of the prostate
messenger NO is synthesized in cavernosal nerves and is regulated by systemic and locally-produced steroid
endothelial cells, which produce neuronal (nNOS) and hormones and growth factors. Differentiated secretory
endothelial (eNOS) nitric oxide synthase, respectively. epithelial cells express androgen receptor (AR) and
Soluble NO activates guanylyl cyclases in SMC that in oestrogen receptor beta (ERβ), which promote the
turn produce cGMP, which activates protein kinase G expression of sex steroid hormone-responsive genes.
(PKG) to induce smooth muscle relation in the corpora Epithelially-derived growth factors, such as insulin-
cavernosa and blood flow into cavernosal cisternae. like growth factor (IGF), transforming growth factor
cGMP levels are degraded by phosphodiesterase-5 (TGF) and epidermal growth factor (EGF), act in a
(PDE-5) to terminate smooth muscle relaxation [52]. paracrine manner on the underlying stroma. Stromal
Male ageing is characterized by slight but detectable fibroblasts are responsive to sex steroid hormones via
decreases in bioT and tissue remodelling of the corpora expression of AR and ERα, which promote cellular
cavernosa [53]. Corpora cavernosa development, SMC proliferation and stimulate the production of stromal-
homeostasis, eNOS and PDE-5 expression are strongly derived factors that act in an autocrine and paracrine
dependent on the presence of androgens as shown in fashion. In addition, stromal cells express aromatase
hypogonadal patients. However, although T is essen- and 5-α-reductase enzymes, which catalyse the local
tial for normal erection, the efficiency of T monother- synthesis of bioactive steroid hormones.
apy is limited to a small number of ED patients with
severe hypogonadism, due to the multifactorial nature
of ED [54]. SMC percentage and elastic fibres of their Proliferative diseases of the ageing prostate
ECM meshwork in the corpora cavernosa decrease Benign prostatic hyperplasia (BPH) is a classical age-
with ageing and provoke mechanical and size alter- related disease present in 20% of men at age 40 years,
ations of the penis that reduce penile extensibility and with progression to 70% at age 60 [60,61]. Of the
elasticity of the tunica albuginea [53]. A recent study many species with prostate glands, only man, chim-
confirmed these findings by demonstrating that in older panzee and dog are known to develop BPH [62,63].
healthy men mean flaccid penis size is greater and the BPH occurs in the transition zone of the prostate and
mean erected penis size is smaller [55]. In addition is characterized by progressive histological changes
to tissue remodelling of the corpora cavernosa, ED that arise initially in the stromal compartment, which
is commonly caused by atherosclerotic disease of the becomes enlarged and altered in its cellular com-
penile arteries, which leads to decreased oxygen ten- position [64] (Figure 2). Focal proliferation of SMC
sion and ischaemia-induced fibrosis. Additionally, ED leads to a characteristic nodular arrangement caused
may arise due to reduced NO production as a con- by budding and branching of epithelial glandular tis-
sequence of diabetes or ageing, due to lower penile sue and is later accompanied by basal cell hyperplasia
nNOS or eNOS activity via neuronal degradation or of the epithelium [65]. BPH is a multifactorial disease
endothelial damage, respectively [56]. involving environmental, endocrine and genetic fac-
Compounds, such as Sildenafil, Tadalafil and tors. Whilst no specific genetic mutations appear to
Vardenafil have been developed that potentiate the be associated with BPH, DNA methylation is globally
NO–cGMP cell-signalling system in the corpus reduced [66]. Consistently, expression of a significant

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
210 N Sampson et al

Figure 2. Tissue remodelling in benign prostatic hyperplasia. Global reduction in chromatin methylation may lead to altered gene
expression, in particular over-expression of genes regulating cell proliferation and down-regulation of genes encoding apoptosis
mediators. Age-related changes in systemic sex steroid hormones, together with altered activity of hormone-metabolizing
enzymes lead to an increased intra-prostatic oestrogen : androgen ratio, which may subsequently alter the expression of
steroid hormone responsive genes. In addition, increased expression of androgen receptor (AR) is observed. Remodelling of
the stromal compartment occurs with proliferation of fibroblasts, which secrete growth factors that act on the overlying
epithelial compartment, inducing cell proliferation. Increased oxygen consumption of growing tissue may result in local hypoxia
with subsequent up-regulation of hypoxia-inducible factor 1 (HIF-1) and hypoxia response genes, including FGF-2 and FGF-7.
Hypertrophic basal cells actively secrete TGF-β, which induces transdifferentiation of stromal fibroblasts into SMCs and
myofibroblasts, which further produce mitogenic growth factors. Increased TGF-β also induces remodelling of the ECM, in
particular increased expression of matrix metalloproteinases (MMPs). Altered secretions of luminal cells lead to calcification,
clogged ducts and inflammation. Infiltrating lymphocytes produce inflammatory cytokines, which further promote cell proliferation
and differentiation. IFN-γ secreted by invading lymphocytes may induce NE cell differentiation from basal cells and may lead to
increased secretion of growth-inducing neuropeptides (NEPs)

number of genes is altered in BPH [67], in partic- Table 1. Distribution and frequency of cancer in different
ular those encoding epithelial/stromal-derived growth anatomical zones of the prostate [184]
factors (Figure 2). Percentage of total Frequency of
Prostate cancer (PCa) is the most common non- Zone glandular tissue (%) PCa (%)
cutaneous malignancy in men in Western countries
Transition 5–10 20
[68,69] and is strongly age-dependent. Pre-malignant Central 25 1–5
dysplastic lesions (prostatic intraepithelial neoplasia, Peripheral 70 70
PIN) are characterized by enlarged nuclei, attenuation
of basal epithelial layer, proliferation of secretory cells
and aberrant differentiation. PINs are regarded as the widely accepted [72–74]. Growing evidence supports
most likely pre-invasive stage of prostatic adenocarci- a PCa stem cell model in which genetic and/or epi-
noma [70,71]. PCa is a multifocal, non-clonal and het- genetic changes accumulate in ageing prostate stem
erogeneous tumour that is most commonly associated cells, associated factors or stem cell niches that pro-
with the peripheral zone (Table 1). Treatment using mote aberrant growth, cell survival and differentia-
androgen ablation strategies is initially successful in tion leading to tumourigenesis (Figure 3) [75–77].
most cases; however, highly aggressive and androgen- In support of this model, a potential PCa stem cell
independent (aiPCa) tumours may recur, for which population that forms tumours in vivo was recently
treatment is mainly palliative. The existence of pro- identified [78]. The most frequent tumour-associated
static stem cells located in the basal epithelium is now genetic event identified to date is fusion of the 5

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The ageing male reproductive tract 211

Figure 3. Schematic representation of differentiation processes in the development and progression of PCa. Androgen-independent
(AI) prostatic stem cells in the basal epithelial layer accumulate mutations in mediators of terminal growth arrest/differentiation
during ageing (red/green), leading to inappropriate proliferation and differentiation of transit-amplifying daughter cells (blue) into
intermediate luminal cells (white; bold black line). Transit-amplifying daughter cells may also differentiate into NE cells (grey;
thin black line). Changes in epithelial secreted proteins (e.g. tumour cells actively secrete TGF-β1) that act on the surrounding
stroma induce the formation of reactive stroma. Reactive stroma is associated with transdifferentiation of fibroblasts (yellow) into
SMC and myofibroblasts (pink), which in turn secrete growth factors that further stimulate stromal and epithelial proliferation
(broken line). Remodelling of the ECM by increased production of matrix metalloproteinases and structural components is
also apparent. Infiltrating lymphocytes (green stars) stimulate the production of inflammatory cytokines (such as IL-6 and
IL-8), which promote proliferation, angiogenesis and metastasis. Androgen ablation/acquirement of androgen independence
results in apoptosis of androgen-dependent (AD) secretory epithelial cells, leaving the ai stem and basal cells intact. Loss of
TGF-β-secreting AD differentiated cells releases stem/basal cells from the inhibitory effects of TGF-β, promoting further altered
proliferation/differentiation (red arrows), leading to the generation of an androgen-independent tumour [57,183] comprising
predominantly cells of an intermediate basal phenotype but also NE cells, which secrete neuropeptides (NEPs) that may contribute
to disease progression. The increased differentiation of clusters of NE cells is perhaps mediated by IFN-γ secreted by lymphocytes.
Increased levels of Her-2/neu enable basal cells to resume cell proliferation. Differentiation markers that classify each of the
different epithelial cell types are indicated. Prostate cancer stem cells have not been unequivocally isolated to date, thus this list of
markers (italicized) derives from studies of potential PCa stem cells [78], other cancer stem cells and non-tumourigenic prostatic
stem cells. Tumourigenic transit-amplifying cells lose expression of β1 integrin and differentiate into luminal cancer cells, which are
the predominant tumour cell type. The increased numbers of NE cells and their origin in tumours is unclear but may differentiate
from tumour luminal cells or from more differentiated AI intermediate basal cells

untranslated region of the androgen-regulated gene Local and systemic sex steroid hormones in
TMPRSS2 to the oncogenic ETS transcription factor prostatic disease
family members, ERG and ETV1, that are commonly
over-expressed in PCa [79]. However, although sev- Control of prostate growth and function is main-
eral genes with weak to moderate penetrance are asso- tained by a finely tuned balance between cellular
ciated with PCa, it is thought that multiple genetic concentrations of sex steroid hormones and locally
alterations lead to tumour development [80–83]. produced paracrine and autocrine growth factors.
The differences in zonal susceptibility to prostatic Intraprostatic levels of sex steroid hormones change
disease may result from different sensitivities to sex upon ageing, with the stromal oestrogen/androgen
steroid hormones and/or differences in expression of ratio increasing via enhanced aromatization of andro-
apoptosis-associated genes [84]. Although PCa and gens and decreased conversion of dihydrotestosterone
BPH are distinct pathologies arising from different (DHT) [90–92]. Despite numerous studies, there is no
cellular events, gene expression profiles of different conclusive evidence of a strong correlation between
stages of PCa [85–88] and BPH [67] reveal some elevated oestrogen or reduced androgen and increased
similarities, indicating a level of homogeneity [89]. incidence of BPH or PCa. However, at least permissive

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
212 N Sampson et al

involvement is implicated by reports showing that factors that act on the prostatic epithelium, leading
men castrated before puberty develop neither PCa nor to reactivation of growth, hyperplasia and neoplas-
BPH, and prolonged or untimely treatment of rodents tic transformation. This so-called ‘reactive stroma’
with oestrogens leads to pre-malignant dysplasia and, exhibits numerous changes, including ECM deposi-
in combination with androgens, malignancy [93–95]. tion and fibroblast transdifferentiation to myofibrob-
Changes in androgen- and oestrogen-metabolizing lasts, which are highly abundant in BPH and PCa,
enzymes have also been reported in hyperplastic and and may be induced by oestrogens and epithelial-
malignant prostates [96–100] and may contribute to derived TGF-β1 [64,92,129]. Moreover, the subse-
aiPCa progression [101–103]. quent enhancement of fibroblast proliferation serves
Sex steroid hormones act via nuclear receptors that to regenerate the fibroblast pool, which, however, also
bind specific DNA motifs in the promoters of target undergoes transdifferentiation, generating a vicious
genes and via poorly understood genomic and non- circle whose consequence is manifested as the stro-
genomic alternative pathways [104–106]. Oestrogen mal expansion and prostate enlargement associated
receptors alpha and beta (ERα, ERβ) are found pre- with BPH. Significantly, the continual and irreversible
dominantly in the stroma and epithelium, respectively transdifferentiation into myofibroblasts becomes histo-
[102], although ERα is aberrantly expressed in pro- logically conserved and is thought to be one of the key
static epithelium in some PCa [100]. ERβ is reportedly mechanisms driving stromal expansion and prostate
initially up-regulated in early PCa but strongly down- enlargement in BPH [130]. The age-dependent loss of
regulated in high-grade PCa [107,108]. The androgen growth suppression was recently linked to increasing
receptor (AR) is expressed in both epithelial and stro- expression of stromal-derived factor 1 (SD-1) [131].
mal cells of the adult prostate. In contrast to rodents Several studies also implicate the pro-apoptotic gene
that do not develop BPH, AR is up-regulated in p53 in the development of reactive stroma. Initial up-
an age-associated manner in dog and man and pro- regulation of p53 in stromal fibroblasts by tumour cells
motes continued proliferation and differentiation of may subsequently select for an apoptosis-resistance
the prostate, perhaps contributing to BPH pathogenesis subpopulation of fibroblasts lacking p53 that then
[109–111]. AR is often increased in aiPCa and upon induce tumour progression in adjacent epithelial cell
androgen ablation in vitro, and continued expression populations [132,133].
of androgen-responsive genes has been shown [112]. The stromal insulin-like growth factor (IGF) axis,
In addition to changes in sex steroid hormone receptor which acts in a paracrine manner on prostatic epithe-
expression and function permitting aberrant activation lial cells, is modulated by the action of andro-
by weak steroid hormones, disease-associated alter- gens [134–136], locally produced proliferative IGFs
ations in steroid hormone intracellular signalling may and anti-proliferative IGF-binding proteins (IGFBPs),
also be mediated via changes in receptor co-regulators which play key roles in numerous aspects of nor-
[113–115] and/or by modulating the expression of mal and neoplastic prostate development [137]. Co-
hormone receptor target genes [116]. Given the large culture experiments demonstrate that IGF-I mediates
number of androgen- and oestrogen-responsive genes tumour-stromal cell interactions of PCa to accelerate
[117,118], age-associated changes in sex steroid hor- tumour growth [138]. IGFBP-3, the most abundant
mones potentially affect numerous cellular pathways. IGFBP, may promote the survival of aiPCa cells in
an androgen-depleted environment [139] and is also
Altered stromal–epithelial interactions in disease implicated in the development of BPH [140,141].
onset/progression Cellular senescence is proposed to be an ageing-
Prostatic stem cell homeostasis is maintained by associated process, with senescent cells accumulating
an androgen-controlled balance of the inhibitory in tissues with age [142,143]. Senescent fibroblasts
cytokine TGF-β and mitogenic cytokines, such as secrete factors that stimulate proliferation, differen-
EGF, FGF and SCF [119]. The TGF-β1 signalling tiation and invasiveness of pre-malignant epithelial
cascade appears to be down-regulated by AR, possi- cells directly or indirectly [144–146]. However, it
bly by direct interactions [120–123]. The age-related remains debatable whether senescence plays a role in
decrease in androgen levels may thus lead to increased the prostate in vivo.
TGF-β and perhaps sensitizes stem cells to stimula-
tory signals resulting in excessive cellular prolifera- Luminal–epithelial interactions/local
tion. Expression of the anti-apoptotic protein Bcl-2 is neuroendocrine system
often increased in BPH and PCa [124,125] and may
provide a mechanism by which cells overcome the Prostatic NE cells differentiate from prostatic epithe-
apoptotic effects of TGF-β in prostatic disease. Thus, lial progenitor cells [147] and secrete numerous pep-
an imbalance in the rate of proliferation and apoptosis tides, neuropeptides and hormones. Rather than exist-
may lead to enhanced cell survival and accumulation, ing as single isolated cells, NE cells appear to form
which perhaps contributes to prostate enlargement in networks of communicating cells with long neurite-
BPH [126–128]. like extensions, which may reach the lumen of the
Disruption of the androgen–oestrogen equilibrium prostatic acini [148]. Increased NE differentiation is
is thought to stimulate changes in stromal-derived associated with PCa and may contribute to disease

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The ageing male reproductive tract 213

progression via neuropeptide secretion [149–151]. In in angiogenesis [170,171] and autocrine regulation of
contrast, fewer NE cells are detected in large nod- differentiation and function of NE prostatic tumour
ules of BPH although they are present in smaller cells [172]. IL-8 is induced by TGF-β1 [173] and is
developing nodules, which may represent growing foci produced by senescent prostatic epithelial cells [174],
[152,153]. NE differentiation can be induced in cul- indicating a possible link between senescence of the
tured PCa cells under a variety of conditions, includ- ageing prostate and tissue growth in BPH/PCa.
ing serum starvation and androgen depletion, sug-
gesting the involvement of multiple pathways. IFN-γ Clinical aspects/future directions
induces NE transdifferentiation of primary prostatic
basal epithelial cells, raising the possibility that infil- Progress has been made in the treatment of BPH using
trating inflammatory T lymphocytes may contribute to combined therapy of α-adrenergic blocking agents and
NE differentiation [154]. Recent studies also impli- 5-α-reductase inhibitors [175]. However, due to treat-
cate the male-specific protocadherin-PC and EGF in ment side-effects and resistance to these therapies, sur-
NE differentiation [155,156]. NE phenotype increases gical intervention often remains the only effective but
during androgen-deprivation therapy and may even invasive and expensive treatment. A number of new
be directly induced by androgen-suppression treatment compounds are undergoing clinical trials for PCa and
regimes [157]. BPH [176,177]. Extensive cross-talk between numer-
ous signalling pathways and the dynamic molecular
changes during disease progression suggest that com-
Chronic inflammation bined or sequential targeting of several different sig-
Chronic intraprostatic inflammation is suspected to nalling pathways may be beneficial. There is an urgent
play a role in the pathogenesis of BPH and PCa need for the development of new molecular markers
[64,158–160]. Moreover, proliferative inflammatory for use as (a) early diagnostic tools, (b) prognostic
atrophy (PIA) shares molecular traits with PIN [70,71]. markers and (c) specific-therapeutic targets for BPH
Chemoattractants, such as GM-SCF produced by PCa and PCa. New biomarkers, such as autoantibody signa-
cells [161] and possibly BPH cells, lead to the initial tures, may improve early detection of PCa and report-
infiltration of cells, including chronically activated T edly show a greater specificity and sensitivity than
cells and macrophages into the glandular epithelium prostate-specific antigen (PSA) [178], which remains
and stroma [162,163]. These infiltrating cells are the most common, albeit limited, means of PCa diag-
responsible for increased production of inflammatory nosis and prognosis [179,180]. In addition, markers
cytokines (particularly IL-15 in stromal cells, IFN-γ are required that can reliably distinguish between clin-
in basal and stromal cells and IL-8 in epithelial cells) ically indolent prostate tumours and those with a high
and may support fibromuscular growth [162,164]. potential for recurrence. Large-scale gene profiling
Prostatic inflammation has been linked by several comparisons [181,182] are the most likely means to
studies to sex steroid hormones [165,166], although identify molecular components involved in PCa pro-
the mechanistic details remained unclear. An excel- gression and will facilitate the development of new
lent recent study showed that direct macrophage-PCa prognostic tools. In addition, such studies will provide
cell to cell interactions mediated by VCAM-1 stim- a much needed greater understanding of the molecu-
ulate macrophage production of cytokines, including lar mechanisms and in particular the role of steroid
IL-1β, which lead to removal of sex steroid hor- hormones in disease development.
mone receptor-bound co-repressor protein complexes
containing TAB 2, resulting in the transcriptional acti-
vation of a selective set of steroid hormone receptor Conclusion
downstream target genes [163]. The dismissal of the
holo-co-repressor complex appears to be mediated by Age-associated changes in the hypothalamus–pitui-
a phosphorylation-induced conformational change in tary–gonadal (HPG) axis play a pivotal role in the
TAB 2, which weakens its association with the lig- ageing male reproductive tract, resulting in altered
anded hormone receptor while enhancing its interac- testicular function and changes in semen output and
tion with other proteins in the holorepressor complex fecundity. These may be related to altered steroidoge-
[163]. This regulatory system may serve to integrate nesis of the ageing testis, ultrastructural histomorpho-
genome-wide responses to specific signalling path- logical changes at the testicular level and functional
ways, particularly in reversing negative gene regula- and morphological compromises of the ageing sperma-
tion by sex steroid hormones [163]. tozoa. The prostate is also sensitive to changes in the
The steroid hormone vitamin D is implicated in pre- HPG axis being regulated by a finely tuned balance of
venting PCa [167] and can suppress the high levels sex steroid hormones and locally produced paracrine
of IL-6 produced by SMCs in BPH and tumour cells and autocrine growth factors. Thus, the common anal-
via AR in a ligand-independent manner and can indi- ogy of male ageing with hypoandrogenism in young
rectly suppress IL-8 production [168,169]. IL-8 pro- adults is inappropriate, given the presumably small
motes stromal and epithelial cell proliferation and is contribution of hypoandrogenism to the non-specific
up-regulated in BPH and aiPCa. IL-8 is also implicated syndromal ensemble of widespread clinical signs of

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
214 N Sampson et al

ageing in the general male population. Rather, the 8. Spetz AC, Fredriksson MG, Hammar ML. Hot flushes in a male
complex ageing processes affecting testicular function population aged 55, 65, and 75 years, living in the community of
Linkoping, Sweden. Menopause 2003;10:81–87.
in combination with endocrine, cellular and structural 9. T’Sjoen G, Goemaere S, De Meyere M, Kaufman JM. Percep-
alterations of the prostate are determined by a com- tion of males’ aging symptoms, health and well-being in elderly
plex interaction of several systems affecting the male community-dwelling men is not related to circulating androgen
reproductive tract. levels. Psychoneuroendocrinology 2004;29:201–214.
Disruption of the androgen–oestrogen equilibrium 10. Hermann M, Untergasser G, Rumpold H, Berger P. Ageing of
the male reproductive system. Exp Gerontol 2000;35:1267–1279.
stimulates changes in stromal-derived factors that 11. Deslypere JP, Vermeulen A. Leydig cell function in normal men:
act on the prostatic epithelium leading to reactiva- effect of age, life-style, residence, diet, and activity. J Clin
tion of growth, hyperplasia and neoplastic transfor- Endocrinol Metab 1984;59:955–962.
mation. In particular, TGF-β1 is thought to provide 12. Vermeulen A, Kaufman JM, Giagulli VA. Influence of some
biological indexes on sex hormone-binding globulin and
the stimulus for the continual and irreversible his-
androgen levels in aging or obese males. J Clin Endocrinol Metab
tological reorganization of the prostatic stroma that 1996;81:1821–1826.
leads to prostate enlargement and BPH. Progression 13. Field AE, Colditz GA, Willett WC, Longcope C, McKinlay JB.
of PCa is also mediated by altered stromal–epithelial The relation of smoking, age, relative weight, and dietary intake
interactions, which are intimately regulated by sex to serum adrenal steroids, sex hormones, and sex hormone-
binding globulin in middle-aged men. J Clin Endocrinol Metab
steroid hormones. PCa is thought to arise due to
1994;79:1310–1316.
accumulating mutations in ageing stem cells that 14. Key TJ, Roe L, Thorogood M, Moore JW, Clark GM, Wang DY.
result in inappropriate proliferation and differentiation Testosterone, sex hormone-binding globulin, calculated free
of transit-amplifying daughter cells. Consequently, testosterone, and oestradiol in male vegans and omnivores. Br
altered growth factor secretion by inappropriately dif- J Nutr 1990;64:111–119.
15. Longcope C, Feldman HA, McKinlay JB, Araujo AB. Diet
ferentiated luminal cells contributes further to growth and sex hormone-binding globulin. J Clin Endocrinol Metab
dysregulation of both stromal and epithelial com- 2000;85:293–296.
partments, facilitating tumour progression. BPH- and 16. Pfeilschifter J, Scheidt-Nave C, Leidig-Bruckner G, Woitge HW,
PCa-associated cells secrete chemoattractants promot- Blum WF, Wuster C, et al. Relationship between circulating
ing infiltration of macrophages and cytokine secre- insulin-like growth factor components and sex hormones in a
population-based sample of 50–80 year-old men and women.
tion, which may induce further intra- and intercellular J Clin Endocrinol Metab 1996;81:2534–2540.
changes in molecular signalling. Due to their high 17. Stepan JJ, Lachman M, Zverina J, Pacovsky V, Baylink DJ.
frequency, BPH and PCa will become increasing clin- Castrated men exhibit bone loss: effect of calcitonin treatment
ical and socio-economic problems as the demographic on biochemical indices of bone remodeling. J Clin Endocrinol
trend of Western populations with higher percentages Metab 1989;69:523–527.
18. Stoch SA, Parker RA, Chen L, Bubley G, Ko YJ, Vincelette A,
of elderly people continues. Extended efforts in iden- et al. Bone loss in men with prostate cancer treated with
tifying factors involved in the mechanisms of organ- gonadotropin-releasing hormone agonists. J Clin Endocrinol
specific tissue remodelling are essential for the pre- Metab 2001;86:2787–2791.
vention and treatment of these age-related proliferative 19. Khaw KT, Barrett-Connor E. Lower endogenous androgens
prostatic diseases. predict central adiposity in men. Ann Epidemiol 1992;2:675–682.
20. Tenover JS. Androgen administration to aging men. Endocrinol
Metab Clin North Am 1994;23:877–892.
Acknowledgements 21. Yaffe K, Lui LY, Zmuda J, Cauley J. Sex hormones and cogni-
tive function in older men. J Am Geriatr Soc 2002;50:707–712.
This work was supported by the Austrian Science Fund 22. Moffat SD, Zonderman AB, Metter EJ, Blackman MR, Har-
(FWF; NRN S9307-B05). NS is a recipient of a Lise Meitner man SM, Resnick SM. Longitudinal assessment of serum free
testosterone concentration predicts memory performance and
Scholorship (FWF; M903-B05).
cognitive status in elderly men. J Clin Endocrinol Metab
2002;87:5001–5007.
23. Barrett-Connor E. Testosterone, HDL-cholesterol and cardiovas-
cular disease. In Pharmacology, Biology and Clinical Appli-
References cations of Androgens: Current Status and Future Prospects,
Bhasin S, Gabelnick HC, Spieler JM, Swerdloff RS, Wang C,
1. Kirkwood TB. Understanding the odd science of aging. Cell Kelly C (eds). Wiley-Liss: New York, 1996; 215–223.
2005;120:437–447. 24. English KM, Mandour O, Steeds RP, Diver MJ, Jones TH,
2. Kirkwood TB, Austad SN. Why do we age? Nature Channer KS. Men with coronary artery disease have lower levels
2000;408:233–238. of androgens than men with normal coronary angiograms. Eur
3. Hermann M, Berger P. Hormonal changes in aging men: a Heart J 2000;21:890–894.
therapeutic indication? Exp Gerontol 2001;36:1075–1082. 25. Feldman HA, Longcope C, Derby CA, Johannes CB, Araujo AB,
4. Bakshi S, Miller DK. Assessment of the aging man. Med Clin Coviello AD, et al. Age trends in the level of serum testosterone
North Am 1999;83:1131–1149. and other hormones in middle-aged men: longitudinal results
5. Kaufman JM, Vermeulen A. The decline of androgen levels in from the Massachusetts male aging study. J Clin Endocrinol
elderly men and its clinical and therapeutic implications. Endocr Metab 2002;87:589–598.
Rev 2005;26:833–876. 26. Schatzl G, Madersbacher S, Temml C, Krenn-Schinkel K, Nader
6. Hermann M, Berger P. Hormone replacement in the aging male? A, Sregi G, et al. Serum androgen levels in men: impact of health
Exp Gerontol 1999;34:923–933. status and age. Urology 2003;61:629–633.
7. Dunbar N, Gruman C, Reisine S, Kenny AM. Comparison 27. Madersbacher S, Stulnig T, Huber LA, Schonitzer D, Dirn-
of two health status measures and their associations with hofer S, Wick G, et al. Serum glycoprotein hormones and their
testosterone levels in older men. Aging Male 2001;4:1–7. free alpha-subunit in a healthy elderly population selected

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The ageing male reproductive tract 215

according to the SENIEUR protocol. Analyses with ultrasen- 52. Rosen RC, Kostis JB. Overview of phosphodiesterase 5 inhibi-
sitive time resolved fluoroimmunoassays. Mech Ageing Dev tion in erectile dysfunction. Am J Cardiol 2003;92:9M–18M.
1993;71:223–233. 53. Wespes E. Erectile dysfunction in the ageing man. Curr Opin
28. Keenan DM, Takahashi PY, Liu PY, Roebuck PD, Nehra AX, Urol 2000;10:625–628.
Iranmanesh A, et al. An ensemble model of the male gonadal 54. Mikhail N. Does testosterone have a role in erectile function? Am
axis: illustrative application in aging men. Endocrinology J Med 2006;119:373–382.
2006;147:2817–2828. 55. Schneider T, Sperling H, Lummen G, Syllwasschy J, Rubben H.
29. Neaves WB, Johnson L, Porter JC, Parker CR Jr, Petty CS. Does penile size in younger men cause problems in condom use?
Leydig cell numbers, daily sperm production, and serum A prospective measurement of penile dimensions in 111 young
gonadotropin levels in aging men. J Clin Endocrinol Metab and 32 older men. Urology 2001;57:314–318.
1984;59:756–763. 56. Gonzalez-Cadavid NF, Rajfer J. Molecular pathophysiology and
30. Johnson L. Spermatogenesis and aging in the human. J Androl gene therapy of aging-related erectile dysfunction. Exp Gerontol
1986;7:331–354. 2004;39:1705–1712.
31. Suoranta H. Changes in the small blood vessels of the 57. Tokar EJ, Ancrile BB, Cunha GR, Webber MM. Stem/progenitor
adult human testis in relation to age and to some and intermediate cell types and the origin of human prostate
pathological conditions. Virchows Arch A Pathol Pathol Anat cancer. Differentiation 2005;73:463–473.
1971;352:165–181. 58. Peehl DM. Primary cell cultures as models of prostate cancer
32. Takahashi J, Higashi Y, LaNasa JA, Yoshida K, Winters SJ, development. Endocr Relat Cancer 2005;12:19–47.
Oshima H, et al. Studies of the human testis. XVIII. 59. Long RM, Morrissey C, Fitzpatrick JM, Watson RW. Prostate
Simultaneous measurement of nine intratesticular steroids: epithelial cell differentiation and its relevance to the understand-
evidence for reduced mitochondrial function in testis of elderly ing of prostate cancer therapies. Clin Sci 2005;108:1–11.
men. J Clin Endocrinol Metab 1983;56:1178–1187. 60. Hermann M, Untergasser G, Rumpold H, Berger P. Ageing of
33. Harman SM, Tsitouras PD. Reproductive hormones in aging the male reproductive system. Exp Gerontol 2000;35:1267–1279.
men. I. Measurement of sex steroids, basal luteinizing hormone, 61. Isaacs JT. Etiology of benign prostatic hyperplasia. Eur Urol
and Leydig cell response to human chorionic gonadotropin. J Clin 1994;25:6–9.
Endocrinol Metab 1980;51:35–40. 62. De Marzo AM, Coffey DS, Nelson WG. New concepts in tissue
34. Wang X, Shen CL, Dyson MT, Eimerl S, Orly J, Hutson JC, specificity for prostate cancer and benign prostatic hyperplasia.
et al. Cyclooxygenase-2 regulation of the age-related decline in Urology 1999;53:29–39.
testosterone biosynthesis. Endocrinology 2005;146:4202–4208. 63. Steiner MS, Couch RC, Raghow S, Stauffer D. The chimpanzee
35. Bergendahl M, Aloi JA, Iranmanesh A, Mulligan TM, Veld- as a model of human benign prostatic hyperplasia. J Urol
huis JD. Fasting suppresses pulsatile luteinizing hormone (LH) 1999;162:1454–1461.
secretion and enhances orderliness of LH release in young but 64. Untergasser G, Madersbacher S, Berger P. Benign pro-
not older men. J Clin Endocrinol Metab 1998;83:1967–1975. static hyperplasia: age-related tissue-remodeling. Exp Gerontol
36. Kidd SA, Eskenazi B, Wyrobek AJ. Effects of male age on 2005;40:121–128.
semen quality and fertility: a review of the literature. Fertil Steril 65. Bonkhoff H, Remberger K. Morphogenetic concepts of normal
2001;75:237–248. and abnormal growth in the human prostate. Virchows Archiv
37. Kuhnert B, Nieschlag E. Reproductive functions of the ageing 1998;433:195–202.
male. Hum Reprod Update 2004;10:327–339. 66. Bedford MT, van Helden PD. Hypomethylation of DNA in
38. Plas E, Berger P, Hermann M, Pfluger H. Effects of aging on pathological conditions of the human prostate. Cancer Res
male fertility? Exp Gerontol 2000;35:543–551. 1987;47:5274–5276.
39. Auger J, Kunstmann JM, Czyglik F, Jouannet P. Decline in 67. Prakash K, Pirozzi G, Elashoff M, Munger W, Waga I, Dhir R,
semen quality among fertile men in Paris during the past 20 years. et al. Symptomatic and asymptomatic benign prostatic hyper-
N Engl J Med 1995;332:281–285. plasia molecular differentiation by using microarrays. Proc Natl
40. Eskenazi B, Wyrobek AJ, Sloter E, Kidd SA, Moore L, Young S, Acad Sci USA 2002;99:7598–7603.
et al. The association of age and semen quality in healthy men. 68. Howe HL, Wingo PA, Thun MJ, Ries LAG, Rosenberg HM,
Hum Reprod 2003;18:447–454. Feigal EG, et al. Annual report to the nation on the status
41. Rolf C, Kenkel S, Nieschlag E. Age-related disease pattern in of cancer (1973 through 1998), featuring cancers with recent
infertile men: increasing incidence of infections in older patients. increasing trends. J Natl Cancer Inst 2001;93:824–842.
Andrologia 2002;34:209–217. 69. Parkin DM, Bray FI, Devesa SS. Cancer burden in the year 2000.
42. Rolf C, Behre HM, Nieschlag E. Reproductive parameters of The global picture. Eur J Cancer 2001;37:S4–S66.
older compared to younger men of infertile couples. Int J Androl 70. Bostwick DG, Qian J. High-grade prostatic intraepithelial
1996;19:135–142. neoplasia. Mod Pathol 2004;17:360–379.
43. Nieschlag E, Lammers U, Freischem CW, Langer K, Wick- 71. Nelson WG, De Marzo AM, Isaacs WB. Prostate cancer. N Engl
ings EJ. Reproductive functions in young fathers and grandfa- J Med 2003;349:366–381.
thers. J Clin Endocrinol Metab 1982;55:676–681. 72. van Leenders G, Schalken JA. Stem cell differentiation within
44. Haidl G, Jung A, Schill WB. Ageing and sperm function. Hum the human prostate epithelium: implications for prostate
Reprod 1996;11:558–560. carcinogenesis. BJU Int 2001;88(suppl 2):35–42.
45. Hassan MA, Killick SR. Effect of male age on fertility: evidence 73. van Leenders G, Gage WR, Hicks JL, van Balken B, Aalders
for the decline in male fertility with increasing age. Fertil Steril TW, Schalken JA, et al. Intermediate cells in human prostate
2003;79(suppl 3):1520–1527. epithelium are enriched in proliferative inflammatory atrophy. Am
46. Seftel AD. Erectile dysfunction in the elderly: epidemiology, eti- J Pathol 2003;162:1529–1537.
ology and approaches to treatment. J Urol 2003;169:1999–2007. 74. Schalken JA, van Leenders G. Cellular and molecular biology of
47. O’Donnell AB, Araujo AB, McKinlay JB. The health of the prostate: stem cell biology. Urology 2003;62:11–20.
normally aging men: the Massachusetts Male Aging Study 75. Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea — a
(1987–2004). Exp Gerontol 2004;39:975–984. paradigm shift. Cancer Res 2006;66:1863–1890.
48. Kloner RA. Erectile dysfunction in the cardiac patient. Curr Urol 76. Hill RP. Identifying cancer stem cells in solid tumours: case not
Rep 2003;4:466–471. proven. Cancer Res 2006;66:1891–1896.
49. Richardson D, Vinik A. Etiology and treatment of erectile failure 77. Rizzo S, Attard G, Hudson DL. Prostate epithelial stem cells.
in diabetes mellitus. Curr Diab Rep 2002;2:501–509. Cell Prolif 2005;38:363–374.
50. Montorsi F, Burnett AL. Erectile dysfunction after radical 78. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ.
prostatectomy. BJU Int 2004;93:1–2. Prospective identification of tumourigenic prostate cancer stem
51. Wespes E. The ageing penis. World J Urol 2002;20:36–39. cells. Cancer Res 2005;65:10946–10951.

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
216 N Sampson et al

79. Tomlins SC, Rhodes DR, Perner S, Dhanasekaran SM, Mehra 99. Tang W, Norlin M. Regulation of steroid hydroxylase CYP7B1
R, Sun X-W, et al. Recurrent fusion of TMPRSS2 and by androgens and estrogens in prostate cancer LNCaP cells.
ETS transcription factor genes in prostate cancer. Science Biochem Biophys Res Commun 2006;344:540–546.
2005;310:644–648. 100. Ellem SJ, Schmitt JF, Pedersen JS, Frydenberg M, Risbridger
80. Hughes C, Murphy A, Martin C, Sheils O, O’Leary J. Molecular GP. Local aromatase expression in human prostate is altered in
pathology of prostate cancer. J Clin Pathol 2005;58:673–684. malignancy. J Clin Endocrinol Metab 2004;89:2434–2441.
81. Ribeiro FR, Diep CB, Jeronimo C, Henrique R, Lopes C, 101. Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA,
Eknaes M, et al. Statistical dissection of genetic pathways Penning TM, et al. Increased expression of genes converting
involved in prostate carcinogenesis. Genes Chromosomes Cancer adrenal androgens to testosterone in androgen-independent
2006;45:154–163. prostate cancer. Cancer Res 2006;66:2815–2825.
82. Schulz WA, Hatina J. Epigenetics of prostate cancer: beyond 102. Harkonen PL, Makela SI. Role of estrogens in development of
DNA methylation. J Cell Mol Med 2006;10:100–125. prostate cancer. J Steroid Biochem Mol Biol 2004;92:297–305.
83. Zhang Z, Karam J, Frenkel E, Sagalowsky A, Hsieh J-T. The 103. Harkonen P, Torn S, Kurkela R, Porvari K, Pulkka A, Lind-
application of epigenetic modifiers on the treatment of prostate fors A, et al. Sex hormone metabolism in prostate cancer cells
and bladder cancer. Urol Oncol 2006;24:152–160. during transition to an androgen-independent state. J Clin
84. Kirschenbaum A, Liu XH, Yao S, Narla G, Friedman SL, Endocrinol Metab 2003;88:705–712.
Martignetti JA, et al. Sex steroids have differential effects on 104. Hatzoglou A, Kampa M, Kogia C, Charalampopoulos I,
growth and gene expression in primary human prostatic epithelial Theodoropoulos PA, Anezinis P, et al. Membrane androgen
cell cultures derived from the peripheral versus transition zones. receptor activation induces apoptotic regression of human
Carcinogenesis 2006;27:216–224. prostate cancer cells in vitro and in vivo. J Clin Endocrinol Metab
85. Dhanasekaran SM, Barrette TR, Ghosh D, Shah R, Varam- 2005;90:893–903.
bally S, Kurachi K, et al. Delineation of prognostic biomarkers 105. Taylor RA, Cowin P, Couse JF, Korach KS, Risbridger GP.
in prostate cancer. Nature 2001;412:822–826. 17beta-Estradiol induces apoptosis in the developing rodent
86. Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, prostate independently of ERalpha or ERbeta. Endocrinology
White J, et al. The program of androgen-responsive genes 2006;147:191–200.
in neoplastic prostate epithelium. Proc Natl Acad Sci USA 106. Freeman MR, Cinar B, Lu ML. Membrane rafts as potential sites
2002;99:11890–11895. of nongenomic hormonal signaling in prostate cancer. Trends
87. DePrimo S, Diehn M, Nelson J, Reiter R, Matese J, Fero M, Endocrinol Metab 2005;16:273–279.
et al. Transcriptional programs activated by exposure of 107. Tsurusaki T, Aoki D, Kanetake H, Inoue S, Muramatsu M,
human prostate cancer cells to androgen. Genome Biol
Hishikawa Y, et al. Zone-dependent expression of estrogen
2002;3:research0032.0031–research0032.0012.
receptors alpha and beta in human benign prostatic hyperplasia.
88. Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, Mont-
J Clin Endocrinol Metab 2003;88:1333–1340.
gomery K, et al. Gene profiling identifies clinically rele-
108. Leav I, Lau KM, Adams JY, McNeal J, Taplin ME, Wang JP,
vant subtypes of prostate cancer. Proc Natl Acad Sci USA
et al. Comparative studies of the estrogen receptors beta and
2004;101:811–816.
alpha and the androgen receptor in normal human prostate glands,
89. Shah US, Getzenberg RH. Fingerprinting the diseased prostate:
dysplasia and in primary and metastatic carcinoma. Am J Pathol
associations between BPH and prostate cancer. J Cell Biochem
2001;159:79–92.
2004;91:161–169.
109. Prins GS, Jung MH, Vellanoweth RL, Chatterjee B, Roy AK.
90. Hiramatsu M, Maehara I, Ozaki M, Harada N, Orikasa S,
Age-dependent expression of the androgen receptor gene in
Sasano H. Aromatase in hyperplasia and carcinoma of the human
the prostate and its implication in glandular differentiation and
prostate. Prostate 1997;31:118–124.
hyperplasia. Dev Genet 1996;18:99–106.
91. Bonnet P, Reiter E, Bruyninx M, Sente B, Dombrowicz D,
de Leval J, et al. Benign prostatic hyperplasia and normal 110. Garraway LA, Lin D, Signoretti S, Waltregny D, Dilks J,
prostate ageing: differences in types I and II 5 alpha-reductase and Bhattacharya N, et al. Intermediate basal cells of the prostate:
steroid hormone receptor messenger ribonucleic acid (mRNA) in vitro and in vivo characterization. Prostate 2003;55:206–218.
levels, but not in insulin-like growth factor mRNA levels. J Clin 111. Banerjee PP, Banerjee S, Brown TR. Increased androgen recep-
Endocrinol Metab 1993;77:1203–1208. tor expression correlates with development of age-dependent,
92. Zhang J, Hess MW, Thurnher M, Hobisch A, Radmayr C, lobe-specific spontaneous hyperplasia of the brown Norway rat
Cronauer MV, et al. Human prostatic smooth muscle cells in prostate. Endocrinology 2001;142:4066–4075.
culture: estradiol enhances expression of smooth muscle cell- 112. Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA,
specific markers. Prostate 1997;30:117–129. Penning TM, et al. Increased expression of genes converting
93. Wang Y, Hayward SW, Donjacour AA, Young P, Jacks T, adrenal androgens to testosterone in androgen-independent
Sage J, et al. Sex hormone-induced carcinogenesis in Rb- prostate cancer. Cancer Res 2006;66:2815–2825.
deficient prostate tissue. Cancer Res 2000;60:6008–6017. 113. Culig Z, Comuzzi B, Steiner H, Bartsch G, Hobisch A. Expres-
94. Tam NN, Ghatak S, Ho SM. Sex hormone-induced alterations in sion and function of androgen receptor coactivators in prostate
the activities of antioxidant enzymes and lipid peroxidation status cancer. J Steroid Biochem Mol Biol 2004;92:265–271.
in the prostate of Noble rats. Prostate 2003;55:1–8. 114. Li W, Cavasotto CN, Cardozo T, Ha S, Dang T, Taneja SS,
95. Ricke WA, Ishii K, Ricke EA, Simko J, Wang Y, Hayward SW, et al. Androgen receptor mutations identified in prostate cancer
et al. Steroid hormones stimulate human prostate cancer and androgen insensitivity syndrome display aberrant ART-27
progression and metastasis. Int J Cancer 2006;118:2123–2131. coactivator function. Mol Endocrinol 2005;19:2273–2282.
96. Soronen P, Laiti M, Törn S, Härkonen P, Patrikainen L, Li Y, 115. Agoulnik IU, Vaid A, Bingman WEr, Erdeme H, Frolov A,
et al. Sex steroid hormone metabolism and prostate cancer. Smith CL, et al. Role of SRC-1 in the promotion of prostate
J Steroid Biochem Mol Biol 2004;92:281–286. cancer cell growth and tumor progression. Cancer Res
97. Pylkkanen L, Santti R, Salo L, Maentausta O, Vihko R, Nurmi M. 2005;65:7959–7967.
Immunohistochemical localisation of oestrogen-specific 17 beta- 116. Hendriksen PJM, Dits NFJ, Kokame K, Veldhoven A, van
hydroxysteroid oxidoreductase in the human and mouse prostate. Weerden WM, Bangma CH, et al. Evolution of the androgen
Prostate 1994;25:292–300. receptor pathway during progression of prostate cancer. Cancer
98. Fung KM, Samara EN, Wong C, Metwalli A, Krlin R, Bane B, Res 2006;66:5012–5020.
et al. Increased expression of type 2 3alpha-hydroxysteroid 117. Dehm SM, Tindall DJ. Molecular recognition of androgen action
dehydrogenase/type 5 17beta-hydroxysteroid dehydrogenase in prostate cancer. J Cell Biochem 2006;epub.
(AKR1C3) and its relationship with androgen receptor in prostate 118. Bektic J, Wrulich OA, Dobler G, Kofler K, Ueberall F, Culig Z,
carcinoma. Endocr Relat Cancer 2006;13:169–180. et al. Identification of genes involved in estrogenic action

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The ageing male reproductive tract 217

in the human prostate using microarray analysis. Genomics 138. Kawada M, Inoue H, Masuda T, Ikeda D. Insulin-like growth
2004;83:34–44. factor I secreted from prostate stromal cells mediates
119. Salm SN, Burger PE, Coetzee S, Goto K, Moscatelli D, Wil- tumor–stromal cell interactions of prostate cancer. Cancer Res
son EL. TGF-β maintains dormancy of prostatic stem cells in 2006;66:4419–4425.
the proximal region of ducts. J Cell Biol 2005;170:81–90. 139. Kojima S, Mulholland DJ, Ettinger S, Fazli L, Nelson CC,
120. Kang HY, Huang KE, Chang SY, Ma WL, Lin WJ, Chang C. Gleave ME. Differential regulation of IGFBP-3 by the androgen
Differential modulation of androgen receptor-mediated transac- receptor in the lineage-related androgen-dependent LNCaP and
tivation by Smad3 and tumor suppressor Smad4. J Biol Chem androgen-independent C4–2 prostate cancer models. Prostate
2002;277:43749–43756. 2006;66:971–986.
121. van der Poel HG. Androgen receptor and TGFbeta1/Smad 140. Untergasser G, Gander R, Lilg C, Lepperdinger G, Plas E,
signaling are mutually inhibitory in prostate cancer. Eur Urol Berger P. Profiling molecular targets of TGF-β1 in prostate
2005;48:1051–1058. fibroblast-to-myofibroblast transdifferentiation. Mech Age Dev
122. Carey JL, Sasur LM, Kawakubo H, Gupta V, Christian B, 2005;126:59–69.
Bailey PM, et al. Mutually antagonistic effects of androgen and 141. Cohen P, Nunn SE, Peehl DM. Transforming growth factor-
activin in the regulation of prostate cancer cell growth. Mol beta induces growth inhibition and IGF-binding protein-3
Endocrinol 2004;18:696–707. production in prostatic stromal cells: abnormalities in cells
123. Brodin G, ten Dijke P, Funa K, Heldin CH, Landstrom M. cultured from benign prostatic hyperplasia tissues. J Endocrinol
Increased smad expression and activation are associated with 2000;164:215–223.
apoptosis in normal and malignant prostate after castration. 142. Castro P, Giri D, Lamb DJ, Ittmann M. Cellular senescence
Cancer Res 1999;59:2731–2738. in the pathogenesis of benign prostatic hyperplasia. Prostate
124. Bruckheimer EM, Kyprianou N. Bcl-2 antagonises the com- 2003;55:30–38.
bined apoptotic effect of transforming growth factor beta 143. Dimri GP, Lee X, Basile G, et al. A biomarker that identifies
and dihydrotestosterone in prostate cancer cells. Prostate senescent human cells in culture and in aging skin in vivo. Proc
2002;53:133–142. Natl Acad Sci USA 1995;92:9363–9367.
125. Kyprianou N, Tu H, Jacobs SC. Apoptotic versus proliferative 144. Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J.
activities in human benign prostatic hyperplasia. Hum Pathol Senescent fibroblasts promote epithelial cell growth and
1996;27:668–675. tumourigenesis: a link between cancer and ageing. Proc Natl Acad
126. Shariat SF, Ashfaq R, Roehrborn C, Slawin KM, Lotan Y. Sci USA 2001;98:12072–12077.
Expression of survivin and apoptotic biomarkers in benign 145. Parrinello S, Coppe J-P, Krtolica A, Campisi J. Stro-
prostatic hyperplasia. J Urol 2005;174:2046–2050.
mal–epithelial interactions in aging and cancer: senescent
127. Royuela M, Arenas MI, Bethencourt FR, Sanchez-Chapado M,
fibroblasts alter epithelial cell differentiation. J Cell Sci
Fraile B, Paniagua R. Regulation of proliferation/apoptosis
2005;118:485–496.
equilibrium by mitogen-activated protein kinases in normal,
146. Bavik C, Coleman I, Dean JP, Knudsen BS, Plymate S, Nel-
hyperplastic, and carcinomatous human prostate. Hum Pathol
son PS. The gene expression program of prostate fibroblast senes-
2002;33:299–306.
cence modulates neoplastic epithelial cell proliferation through
128. Cordon-Cardo C, Koff A, Drobnjak M, Capodieci P, Osman I,
paracrine mechanisms. Cancer Res 2006;66:794–802.
Millard SS, et al. Distinct altered patterns of p27KIP1 gene
147. Rumpold H, Heinrich E, Untergasser G, Hermann M, Pfister G,
expression in benign prostatic hyperplasia and prostatic
Plas E, et al. Neuroendocrine differentiation of human prostatic
carcinoma. J Natl Cancer Inst 1998;90:1284–1291.
primary epithelial cells in vitro. Prostate 2002;53:101–108.
129. Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Row-
148. Berger P, Gruschwitz M, Spoettl G, Dirnhofer S, Maders-
ley DR. Reactive stroma in human prostate cancer: induction
bacher S, Gerth R, et al. Human chorionic gonadotropin
of myofibroblast phenotype and extracellular matrix remodelling.
Clin Cancer Res 2002;8:2912–2923. (hCG) in the male reproductive tract. Mol Cell Endocrinol
130. Rumpold H, Untergasser G, Madersbacher S, Berger P. The 2007;260–262:190–196.
development of benign prostatic hyperplasia by trans- 149. Jongsma J, Oomen MH, Noordzij MA, et al. Androgen-
differentiation of prostatic stromal cells. Exp Gerontol independent growth is induced by neuropeptides in human
2002;37:1001–1004. prostate cancer cell lines. Prostate 2000;42:34–44.
131. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delau- 150. Guate JL, Escaf S, Menendez CL, del Valle M, Vega JA.
nay T, Naeem R, et al. Stromal fibroblasts present in inva- Neuroendocrine cells in benign prostatic hyperplasia and
sive human breast carcinomas promote tumor growth and prostatic carcinoma: effect of hormonal treatment. Urol Int
angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 1997;59:149–153.
2005;121:335–348. 151. Uchida K, Masumori N, Takahashi A, Itoh N, Kato K,
132. Hill R, Song Y, Cardiff RD, Van Dyke T. Selective evolution Matusik RJ, et al. Murine androgen-independent neuroendocrine
of stromal mesenchyme with p53 loss in response to epithelial carcinoma promotes metastasis of human prostate cancer cell line
tumorigenesis. Cell 2005;123:1001–1011. LNCaP. Prostate 2006;66:536–545.
133. Kiaris H, Chatzistamou I, Trimis G, Frangous-Plemmenous M, 152. Xue Y, van der Laak J, Smedts F, Schoots C, Verhofstad A,
Pafiti-Kondi A, Kalofoutis A. Evidence for nonautonomous de la Rosette J, et al. Neuroendocrine cells during human
effect of p53 tumor suppressor in carcinogenesis. Cancer Res prostate development: does neuroendocrine cell density remain
2005;65:1627–1630. constant during fetal as well as postnatal life? Prostate
134. Pandini G, Mineo R, Frasca F, Roberts CT, Marcelli M, Vigneri 2000;42:116–123.
R, et al. Androgens up-regulate the insulin-like growth factor-I 153. Cockett AT, di Sant’Agnese PA, Gopinath P, Schoen SR,
receptor in prostate cancer cells. Cancer Res 2005;65:1849–1857. Abrahamsson PA. Relationship of neuroendocrine cells of
135. Le H, Arnold JT, McFann KK, Blackman MR. DHT and prostate and serotonin to benign prostatic hyperplasia. Urology
testosterone but not DHEA or E2 differentially modulate IGF- 1993;42:512–519.
I, IGFBP-2 and IGFBP-3 in human prostatic stromal cells. Am J 154. Untergasser G, Plas E, Pfister G, Heinrich E, Berger P. Interferon-
Physiol Endocrinol Metab 2006;290:E952–E960. gamma induces neuroendocrine-like differentiation of human
136. Wu JD, Haugk K, Woodke L, Nelson PS, Coleman I, Plymate S. prostate basal-epithelial cells. Prostate 2005;64:419–429.
Interaction of IGF signaling and the androgen receptor in prostate 155. Yang X, Chen MW, Terry S, Vacherot F, Chopin DK, Bemis DL,
cancer progression. J Cell Biochem 2006;25:epub. et al. A human- and male-specific protocadherin that acts
137. Gennigens C, Menetrier-Caux C, Droz JP. Insulin-like growth through the wnt signaling pathway to induce neuroen-
factor (IGF) family and prostate cancer. Crit Rev Oncol Hematol docrine transdifferentiation of prostate cancer cells. Cancer Res
2006;58:124–145. 2005;65:5263–5271.

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
218 N Sampson et al

156. Humez S, Monet M, Legrand G, Lepage G, Delcourt P, 170. Zhang F, Lee J, Lu S, Pettaway CA, Dong Z. Blockade of
Prevarskaya N. Epidermal growth factor-induced neuroen- transforming growth factor-beta signaling suppresses progression
docrine differentiation and apoptotic resistance of androgen- of androgen-independent human prostate cancer in nude mice.
independent human prostate cancer cells. Endocr Relat Cancer Clin Cancer Res 2005;11:4512–4520.
2006;14:181–195. 171. Liu J, Schuff-Werner P, Steiner M. Thrombin/thrombin receptor
157. Mosca A, Berruti A, Russo L, Torta M, Dogliotti L. The (PAR-1)-mediated induction of IL-8 and VEGF expression
neuroendocrine phenotype in prostate cancer: basic and clinical in prostate cancer cells. Biochem Biophys Res Commun
aspects. J Endocrinol Invest 2005;28:141–145. 2006;343:183–189.
158. Nelson WG, De Marzo AM, DeWeese TL, Isaacs WB. The role 172. Huang J, Yao JL, Zhang L, Bourne PA, Quinn AM, di
of inflammation in the pathogenesis of prostate cancer. J Urol Sant’Agnese PA, et al. Differential expression of interleukin-
2004;172:S6–11; discussion S11–12. 8 and its receptors in the neuroendocrine and non-
159. Palapattu GS, Sutcliffe S, Bastian PJ, Platz EA, De Marzo AM, neuroendocrine compartments of prostate cancer. Am J Pathol
Isaacs WB, et al. Prostate carcinogenesis and inflammation: 2005;166:1807–1815.
emerging insights. Carcinogenesis 2005;26:1170–1181. 173. Lu S, Dong Z. Characterization of TGF-beta-regulated
160. Kramer G, Marberger M. Could inflammation be a key interleukin-8 expression in human prostate cancer cells. Prostate
component in the progression of benign prostatic hyperplasia? 2006;66:996–1004.
Curr Opin Urol 2006;16:25–29. 174. Castro P, Xia C, Gomez L, Lamb DJ, Ittmann M. Interleukin-
161. Chung TD, Yu JJ, Spiotto MT, Bartkowski M, Simons JW. 8 expression is increased in senescent prostatic epithelial cells
Characterization of the role of IL-6 in the progression of prostate and promotes the development of benign prostatic hyperplasia.
cancer. Prostate 1999;38:199–207. Prostate 2004;60:153–159.
162. Kramer G, Steiner GE, Handisurya A, Stix U, Haitel A, Knerer 175. Tarter TH, Vaughan EDJ. Inhibitors of 5alpha-reductase in the
B, et al. Increased expression of lymphocyte-derived cytokines in treatment of benign prostatic hyperplasia. Curr Pharm Des
benign hyperplastic prostate tissue, identification of the producing 2006;12:775–783.
cell types, and effect of differentially expressed cytokines on 176. Armstrong AJ, Carducci MA. New drugs in prostate cancer. Curr
stromal cell proliferation. Prostate 2002;52:43–58. Opin Urol 2006;16:138–145.
163. Zhu P, Baek SH, Bourk EM, Ohgi KA, Garcia-Bassets I, 177. Bullock TL, Andriole GLJ. Emerging drug therapies for
Sanjo H, et al. Macrophage/cancer cell interactions mediate benign prostatic hyperplasia. Expert Opin Emerging Drugs
hormone resistance by a nuclear receptor derepression pathway. 2006;11:111–123.
Cell 2006;124:615–29. 178. Bradford TJ, Xiaoju W, Chinnaiyan AM. Cancer immunomics:
164. Giri D, Ittmann M. Interleukin-8 is a paracrine inducer using autoantibody signatures in the early detection of prostate
of fibroblast growth factor 2, a stromal and epithelial cancer. Urol Oncol 2006;24:237–242.
growth factor in benign prostatic hyperplasia. Am J Pathol 179. Small EJ, Roach MR. Prostate-specific antigen in prostate cancer:
2001;159:139–147. a case study in the development of a tumour marker to monitor
165. Robinette CL. Sex-hormone induced inflammation and fibro- recurrence and assess response. Sem Oncol 2002;29:264–273.
muscular proliferation in the rat lateral prostate. Prostate 180. Kirby RS, Christmas TJ, Brawer MK. In Prostate Cancer.
1988;12:271–286. Mosby: London, 2001.
166. Asirvatham AJ, Schmidt M, Gao B, Chaudhary J. Androgens 181. Nanni S, Priolo C, Grasselli A, D’Eletto M, Merola R, Moretti R,
regulate the immune/inflammatory response and cell survival et al. Epithelial-restricted gene profile of primary cultures from
pathways in rat ventral prostate epithelial cells. Endocrinology human prostate tumours: a molecular approach to predict clinical
2006;147:257–271. behaviour of prostate cancer. Mol Cancer Res 2006;4:79–92.
167. Schwartz GG. Vitamin D and the epidemiology of prostate 182. True L, Coleman I, Hawley S, Huang CY, Gifford D, Cole-
cancer. Semin Dial 2005;18:276–289. man R, et al. A molecular correlate to the Gleason grading
168. Nonn L, Peng L, Feldman D, Peehl DM. Inhibition of p38 system for prostate adenocarcinoma. Proc Natl Acad Sci USA
by vitamin D reduces interleukin-6 production in normal 2006;103:10991–10996.
prostate cells via mitogen-activated protein kinase phosphatase 5: 183. Craft N, Chhor C, Tran C, Belldegrun A, DeKernion J, Witte ON,
implications for prostate cancer prevention by vitamin D. Cancer et al. Evidence for clonal outgrowth of androgen-independent
Res 2006;66:4516–4524. prostate cancer cells from androgen-dependent tumours through
169. Bao BY, Yao J, Lee YF. 1α,25-dihydroxyvitamin D3 suppresses a two-step process. Cancer Res 1999;59:5030–5036.
interleukin-8-mediated prostate cancer cell angiogenesis. Car- 184. Che M, Grignon D. Pathology of prostate cancer. Cancer Metast
cinogenesis 2006;27:1883–1893. Rev 2002;21:381–395.

J Pathol 2007; 211: 206–218 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

You might also like