Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/316465852

Human Spermatogenesis and Its Regulation

Chapter · April 2017


DOI: 10.1007/978-3-319-53298-1_3

CITATIONS READS

34 6,963

4 authors, including:

Haiqi Chen Xiang Xiao


University of Texas Southwestern Medical Center The University of Hong Kong
46 PUBLICATIONS 980 CITATIONS 36 PUBLICATIONS 1,147 CITATIONS

SEE PROFILE SEE PROFILE

C. Yan Cheng
Population Council
585 PUBLICATIONS 28,834 CITATIONS

SEE PROFILE

All content following this page was uploaded by Haiqi Chen on 28 May 2019.

The user has requested enhancement of the downloaded file.


Human Spermatogenesis and Its
Regulation 3
Haiqi Chen PhD, Dolores Mruk PhD, Xiang Xiao PhD
and C. Yan Cheng PhD

Contents Introduction
Introduction ................................................................ 49
Structure and Composition of the Seminiferous In human spermatogenesis, undifferentiated
Epithelium .................................................................. 50 spermatogonia derived from spermatogonial
Sertoli Cells and the Blood–Testis Barrier (BTB).... 52 stem cells give rise to diploid spermatocytes
Cell–Cell Interactions in the Testis ........................... 53
Spermatogonia and Self-renewal ............................... 54 which undergo meiosis I/II to produce haploid
Spermatocytes and Meiosis ....................................... 56 spermatids [1–3]. Spermatids, in turn, undergo
Spermatids and Spermiogenesis ................................ 58 spermiogenesis to form functional spermatozoa
Cycle of the Seminiferous Epithelium ...................... 58 [3–5]. After puberty at *12 years of age, a
Factors that Regulate Spermatogenesis ..................... 59
Hormonal Regulation................................................. 59 healthy man produces upwards of 200 million
Small Non-coding RNAs (sncRNAs)........................ 62 sperm daily from his 20 s through 50 s [6, 7],
Obesity ....................................................................... 63 which declines gradually with a reduction of
Summary .................................................................... 64 *50% by his 80 s [8, 9]. Spermatogenesis is
References .................................................................. 64 composed of a series of cellular events which
includes (i) self-renewal of spermatogonial stem
cells and spermatogonia via mitosis, (ii) trans-
formation and differentiation of spermatocytes,
(iii) generation of haploid spermatids via meiosis
I/II, and (iv) final morphological maturation of
spermatids to become spermatozoa via spermio-
genesis. These processes thus involve multiple
H. Chen  D. Mruk  C.Y. Cheng (&) cellular events in the testis and are under com-
Center for Biomedical Research Population Council, plex controls by regulatory hormonal and sig-
1230 York Ave., New York, NY 10065, USA
e-mail: y-cheng@popcbr.rockefeller.edu
naling axes. The most important of these
regulatory pathways is the hypothalamic–pitu-
H. Chen
e-mail: hqchen1990@gmail.com
itary–testicular axis involving both FSH and LH,
and testosterone and estradiol-17ß as key regu-
D. Mruk
e-mail: mruk@popcbr.rockefeller.edu lators [10–14]. Since the subject of hormonal
regulation of spermatogenesis in men has been
X. Xiao
Department of Reproductive Physiology,
extensively reviewed [10–15], we focus our
Zhejiang Academy of Medical Sciences, Hangzhou, discussion herein on other topics in which
Zhejiang, China advances have been made in recent years.
e-mail: xxiao@zjams.cn

© Springer International Publishing AG 2017 49


S.J. Winters and I.T. Huhtaniemi (eds.), Male Hypogonadism,
Contemporary Endocrinology, DOI 10.1007/978-3-319-53298-1_3

sjwint01@louisville.edu
50 H. Chen et al.

A better understanding of human spermatogen- tubules combined per pair of testes is *400
esis is necessary since about 15% of married meters, and each tubule has a diameter of
couples are infertile, and half of these cases are *200 µm [3, 33]. Spermatogenesis occurs as
attributed to male factors, indicating that about shown in the cross section of a tubule (Fig. 3.1),
7% of men in the general population are infertile consisting of Sertoli cells and germ cells at dif-
[16]. In this context, it is of interest to note that ferent stages of development. These germ cells
male reproductive health can be considerably include undifferentiated spermatogonia, dark type
altered by the environment and life style factors A spermatogonia, pale type A spermatogonia,
[17–21]. Semen quality is believed to be type B spermatogonia; preleptotene, leptotene,
declining [22–25], illustrating the global trend of zygotene and pachytene primary spermatocytes,
impaired male reproductive health. While 6–18% and secondary spermatocytes; as well as Sa, Sb,
of non-obstructive azoospermia or severe oligo- Sc, Sd1, and Sd2 spermatids until Sd2 transform
zoospermia cases can be attributed to Y chro- into spermatozoa [34]. Sertoli and germ cells near
mosome microdeletions [26], as many as 25– the base of the seminiferous epithelium are in
40% of infertile men have no identifiable cause close contact with the basement membrane,
for hypospermatogenesis [27, 28]. In fact, infer- which is a modified form of extracellular matrix
tility is an emerging global public health issue and is composed of mostly collagen (type IV) and
right after cancer and cardiovascular diseases. laminins, heparin sulfate proteoglycan, and
Interestingly, experiments using mouse genetic nidogen/entactin [35, 36]. Behind the basement
models, including membrane is the type 1 collagen layer, to be
knockout/knockin/gene-trapped, transgenic, and followed by the myoid cell layer, lymph, and then
chemical-induced point mutant mice, have iden- lymphatic endothelium. Collectively, they con-
tified more than 400 genes pertinent to sper- stitute the tunica propria [35, 36] (Fig. 3.1).
matogenesis [28, 29], and the use of proteomics, During spermatogenesis, germ cells, in particular
epigenomics, and genomics have identified post-meiotic spermatids, rely almost solely on
>2300 genes pertinent to spermatogenesis and/or Sertoli cells for structural, nutritional, and para-
testis function. However, how this information crine support since haploid spermatids are
applies to humans remains largely unknown [30]. metabolically quiescent cells with relatively little
Future directions in “omics” research pertinent to cytosol to support their cellular functions [37,
human spermatogenesis have been recently 38]. Thus, the Sertoli cell is known as the
reviewed [30, 31]; herein, we focus on the local “mother” or “nurse” cell, with each Sertoli cell
regulation of spermatogenesis and highlight areas supporting *30–50 developing germ cells based
of research which can serve as helpful examples on morphometric analysis in the rat testis [39].
to guide future investigations. In humans, Sertoli cells undergo mitotic pro-
liferation in two separate phases. The first phase
takes place shortly after birth during the
Structure and Composition neonatal-infantile period when the Sertoli cell
of the Seminiferous Epithelium population increases via mitotic proliferation
[40]. During puberty, Sertoli cells again rapidly
In adult humans, as in rodents, the functional unit divide mitotically [40], giving rise to
that produces sperm via spermatogenesis is the *500  106 Sertoli cells per testis (each adult
seminiferous tubule. Spermatogenesis is sup- human testis weighs *18–19 gm). The Sertoli
ported by testosterone produced by Leydig cells cell number declines to *300  106 by
in the interstitium, and by estradiol-17ß produced 50–85 years of age, associated with a reduction
by Sertoli and germ cells [10–15], although there in daily sperm output [8]. Each testis has
is evidence that human Leydig cells also produce *4.5  106 Sertoli cells in mice [41] and 30
estradiol-17ß [32]. In men, each testis has *400– −40  106 Sertoli cells in rats [42–44] compared
600 seminiferous tubules, the length of all the to 500  106 in men [8], and a daily sperm

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 51

Adluminal (Apical) compartment F-actin bundle

Apical ES protein
Elongated complex
spermatid Endoplasmic
reticulum

Tight junction
Elongating protein complex
spermatid
Basal ES
protein complex
Sertoli cell
Pachytene Gap junction protein
spermatocyte
Sertoli cell Desmosomal protein
BTB

Intermediate filaments
compartment

Basement membrane
Basal

Sertoli cell Sertoli cell Type 1 collagen


Preleptotptene nucleus layer
nucleus
spermatocyte
Type A spermatogonium Myoid cell layer
Tunica
propria Type B spermatogonium
Lymph

Lymphatic
Blood Blood
endothelium
vessel vessel
Leydig Leydig
cell cell

Fig. 3.1 Structure and cellular composition of the sem- the apical compartment. The most notable junction in the
iniferous epithelium in the human testis. The seminiferous mammalian testis including humans is the ES, which is
epithelium, as seen in this schematic drawing of a cross typified by the presence of actin microfilament bundles
section of a seminiferous tubule, is composed of Sertoli sandwiched in-between cisternae of endoplasmic reticu-
and germ cells. The BTB is constituted by actin-based lum and the apposing Sertoli cell–cell plasma membranes.
tight junctions, basal ectoplasmic specialization (ES), and The ES at the BTB is called basal ES while the ES at the
gap junctions, as well as the intermediate filament-based Sertoli–spermatid interface is designated apical ES. Since
desmosome. As one of the tightest blood–tissue barriers in developing germ cells (e.g., preleptotene spermatocytes)
the human body, the BTB divides the seminiferous and developing spermatids are being transported across
epithelium into the adluminal (apical) and basal compart- the seminiferous epithelium, Sertoli cell–Sertoli cell and
ments. Spermatogonia (both type A, B and undifferenti- Sertoli–germ cell junctions are continuously remodeling
ated) and preleptotene spermatocytes reside in the basal throughout the 6 stages of the epithelial cycle of
compartment, whereas the other primary and secondary spermatogenesis in the human testis
spermatocytes and post-meiotic spermatids are found in

production of 8x [41], 70x [45], and 200x106 macrophages and microvessels (Fig. 3.1). Leydig
[46], respectively. The number of Sertoli cells is cells produce testosterone (T) under the influence
determined by FSH, thyroid hormones, growth of luteinizing hormone (LH) since LH receptors
hormone, and several paracrine growth factors are limited to Leydig cells in human and rodent
[40]. Outside the tubules in the interstitial space testes. Interestingly, the T level in the interstitial
lay the Leydig cells, fibroblasts, and some fluid and seminiferous tubule fluid is at least

sjwint01@louisville.edu
52 H. Chen et al.

70- and 50-fold higher than the T level in the divide and become fully differentiated, con-
systemic circulation in the rat [47]. In humans, comitant with the onset of meiosis I/II [40]. It is
the intratesticular T (e.g., interstitial fluid) level is of interest to note that human Sertoli cells, sim-
also 100-fold higher than the systemic circulation ilar to Sertoli cells in rodents, can be
[48–50], illustrating a considerably higher T de-differentiated, becoming mitotically active
level is maintained in the testis to support when they are exposed to fetal bovine serum
spermatogenesis. [40], such as cultured in media containing 5–
10% fetal bovine serum (v/v) [56]. Under such
conditions, Sertoli cells can be maintained
Sertoli Cells and the Blood–Testis through multiple passes in vitro without detect-
Barrier (BTB) able changes in their functional and physiologi-
cal properties [57].
The seminiferous epithelium of the mammalian BTB function in rodents is dependent on
testis is further divided into two functional testosterone [58–61]. However, treatment of nor-
compartments: (1) the basal, and (2) the adlu- mal men with testosterone plus levonorgestrel
minal (apical) compartments (Fig. 3.1), due to (LNG), which is capable of suppressing sper-
the presence of the Sertoli cell blood–testis bar- matogenesis and causing azoospermia or
rier (BTB) located near the basement membrane. oligospermia with suppression of intratesticular
The BTB is constituted by coexisting actin-based androgen levels, did not affect BTB function since
basal ectoplasmic specialization (basal ES) and the distribution of claudin-3 remained relatively
tight junction (TJ), as well as coexisting basal ES unaltered [62]. However, in a more recent study in
and gap junction, and intermediate which healthy men were treated with T plus LNG
filament-based desmosome [51, 52]. As such, together with the GnRH antagonist acyline or the
undifferentiated spermatogonia, both type A and 5a-reductase inhibitor dutasteride (in order to
B spermatogonia, and preleptotene spermato- provide added suppression of spermatogenesis),
cytes differentiated from type B spermatogonia there was considerable down-regulation of
reside in the basal compartment. More advanced claudin-11, connexin-43, and vinculin at the BTB
primary spermatocytes, secondary spermato- compared to men treated wth T + LNG alone [63].
cytes, and all haploid spermatids including Collectively, these findings suggest that human
spermatozoa reside in the adluminal compart- BTB is regulated in a manner similar to rodents.
ment (Fig. 3.1). Some species differences exist, however, such as
Briefly, the BTB provides a unique microen- the extent of androgen-dependency, since it
vironment in the adluminal compartment for appears that a considerable reduction in the
meiosis I/II and post-meiotic spermatid devel- intratesticular T level is necessary to suppress the
opment so that these events are segregated from BTB integrity in humans.
the systemic circulation, behind an immunolog- Due to the limited access to normal human
ical barrier. Studies have shown, however, that testes for functional analysis, it is difficult to
the BTB contributes minor significance to the compare the regulation of the human BTB
immune privilege of the testis since germ cells integrity with that of rodents. With advances in
residing in the basal compartment are equally cell and tissue culture techniques, however,
immunogenic, containing multiple testis-specific human Sertoli cells, using media containing fetal
antigens [53, 54]. Instead, the testis immune bovine serum, have been cultured and main-
privilege is maintained mostly by Sertoli cells tained successfully for several weeks in vitro [56,
through their secretory immunosuppressive bio- 57, 64–66], with a functional tight junction-
molecules: cytokines, bioactive lipids and pep- permeability barrier [56, 64]. Using this
tides, and androgens from Leydig cells [55]. approach, the organization of F-actin that sup-
The BTB in humans is established at puberty by ports BTB integrity has been investigated at the
*12–13 years of age when Sertoli cells cease to human Sertoli cell BTB. It is now known that

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 53

human Sertoli cell F-actin organization is main-


tained, similar to that of rodents, by actin binding
proteins, including Arp2/3 (actin related protein
2/3) complex and Eps8 (epidermal growth factor
receptor pathway substrate 8) that confers bran-
ched actin network and bundled actin microfila-
ments, respectively [57]. Furthermore, exposure
of human Sertoli cells to environmental toxi-
cants, such as CdCl2, or bisphenol A (BPA),
rapidly perturbs F-actin organization in these
cells. Changes in the distribution of cell adhesion
proteins at the BTB, including the TJ-protein
ZO-1, and the basal ES proteins N-cadherin and
ß-catenin, from the cell surface to the cell cytosol
[57], likely reflect degradation via an
endosome-dependent degradation pathway.
These changes disrupt integrity and function of
the BTB. It is expected that much information
will be obtained using this in vitro system to gain
insight into the biology of the human BTB.

Fig. 3.2 Cell–cell interaction within the testis and its


Cell–Cell Interactions in the Testis functional relationship to the hypothalamic–pituitary–
testicular axis. The classic hormonal regulatory axis that
Due to the presence of the BTB, germ cell exerts its regulatory effects on the testis through the
development, in particular post-meiotic spermatid release of gonadotropin-releasing hormone (GnRH) from
the hypothalamus, which in turn regulates the secretion of
development, relies almost exclusively on the
luteinizing hormone (LH) and follicle-stimulating hor-
structural, nutritional, and paracrine support of mone (FSH) from the pituitary gland. Testosterone pro-
Sertoli cells. This is mediated via cell junctions at duced by interstitial Leydig cells provides the feedback
the Sertoli–germ cell interface, involving multiple loop to modulate the production of GnRH from the
hypothalamus and thereby LH and FSH from the pituitary
genes and their proteins [37, 38, 67, 68]
gland. In humans and primates in contrast to rodents,
(Fig. 3.2). Even though undifferentiated sper- testosterone has no direct feedback effect on the pituitary
matogonia [e.g., spermatogonial stem cells [240]. Inhibin produced by Sertoli cells provides selective
(SSCs)], type A and type B spermatogonia, and feedback to the production of FSH. There is cross talk
among germ cells (including spermatogonial stem cells)
preleptotene spermatocytes reside outside the
and Sertoli cells in the seminiferous epithelium, as well as
BTB, they also rely on the Sertoli cell for func- Leydig cells and peritubular myoid cells in the intersti-
tional and structural support through the expres- tium. Studies have shown an array of growth factors,
sion of unique genes (Fig. 3.2). This is genes and hormones are involved. Abbreviations: CSF1,
colony stimulating factor 1; BMP4, bone morphogenetic
particularly true for SSCs which are located at the
protein 4; GDF9, growth differentiation factor-9; GnRH,
stem cell niches—usually at the base of the gonadotropin-releasing hormone; P-Mod-S, peritubular
seminiferous epithelium wherein three seminif- myoid cell modulates Sertoli cell factor; TGF-b1, trans-
erous tubules meet, adjacent to the microvessels forming growth factor-1; Wt1, Wilms’ tumor 1
in the interstitium. In short, SSCs rely on Sertoli
cells for structural and functional support, besides
biomolecules from the microvessels [69, 70]. Leydig cell steroidogenesis [71–73]. One of the
Sertoli cells also modulate spermatogenesis paracrine factors produced by Sertoli cells that
through cross talk with Leydig cells. Sertoli cells modulates Leydig cell function has been partially
stimulate Leydig cell differentiation but inhibit characterized [74]. Studies using genetic models

sjwint01@louisville.edu
54 H. Chen et al.

have shown that Sertoli cells maintain Leydig modulated by peritubular myoid cells, unequiv-
cell number (both fetal and adult Leydig cells) ocally demonstrating that Sertoli and peritubular
and support Leydig cell development [75, 76], as myoid cells are functionally connected. Interest-
well as determine Leydig cell differentiation ingly, studies using Amh-Cre to induce expres-
status and cell fate through the Wt1 gene [77]. On sion of the Diphtheria toxin receptor in Sertoli
the other hand, Sertoli cells also interact with cells to cause controlled, cell-specific, and acute
germ cells to support spermatogenesis through ablation of the Sertoli cell population in adult
multiple paracrine factors [37, 78, 79] (Fig. 3.2). mice have shown that Sertoli cells also control
There is also functional cross talk between peritubular myoid cell fate, including its differ-
Sertoli cells and the tunica propria, in particular entiation status and cell population [76]. Using
peritubular myoid cells (Fig. 3.2). For instance, this approach, Sertoli cells were shown to mod-
Sertoli cells work in concert with peritubular ulate testicular vascular network development,
myoid cells in the production and deposition of including modulating circulating testosterone
extracellular matrix (ECM) components [80] to levels in adult mice [86]. It should be noted that
produce and maintain the basement membrane, much of the information stated above and
which is a modified form of ECM [35, 36]. depicted in Fig. 3.2 is derived from studies in
Studies have also suggested that peritubular rodents. Clearly, further studies are needed to
myoid cells modulate Sertoli cell function understand the cell–cell interactions between
wherein myoid cells produce a paracrine factor Sertoli cells, Leydig cells, and peritubular myoid
known as P-Mod-S that regulates Sertoli cell cells in the human testis.
function [81]. However, the identity of this factor
remains to be clarified. Another report has shown
that while peritubular myoid cells do not secrete Spermatogonia and Self-renewal
clusterin or a2-macroglobulin (a wide-spectrum
protease inhibitor [82]), co-cultures of myoid Spermatogonia are the progenitor cells of all
cells with Sertoli cell promote Sertoli cell a2- germ cells and are designated type A and type B
macroglubulin and clusterin secretion [83]. It [87, 88]. In humans, type A spermatogonia are
must be noted, however, that unlike Sertoli cells, further categorized into progenitor dark type A
which can be maintained in culture in serum-free spermatogonia (Ad) and pale type A spermato-
medium (e.g., F12/DMEM with several growth gonia (Ap) which are capable of undergoing
factors [84]) for up to *2-wk, peritubular myoid mitotic proliferation for self-renewal [89]
cell cultures require the presence of serum pro- (Fig. 3.3). Ap are the predominant spermatogonia
teins, such as *5–10% fetal calf serum for their that survive radio- and chemotherapy while Ad
survival [83]. are largely eliminated during such treatment,
The most conclusive study thus far to illus- illustrating that Ap are the potential spermato-
trate the importance of peritubular myoid cells in gonial stem cells [90]. At present, it is generally
spermatogenesis is the generation of a peritubular accepted that Ad constitute a pool of reserve
myoid cell-specific androgen receptor spermatogonia which remain quiescent but can
(AR) knockout (PTM-ARKO) mouse model give rise to Ap when needed and are considered
[85]. PTM-ARKO male mice have reduced to be the ‘true’ spermatogonial stem cells in
seminiferous tubule fluid production and are humans [4, 91]. The molecular basis for the
azoospermic and infertile, with reduced expres- regulation of human spermatogonia remains lar-
sion of certain androgen-dependent Sertoli cell gely unknown. cKIT, a transmembrane protein
genes [85]. These findings illustrate that ARs in tyrosine kinase receptor (also known as stem cell
Sertoli cells fail to assume the function of ARs in factor receptor or CD117), and its ligand cKIT
myoid cells to sustain spermatogenesis. Further- ligand (also known as stem cell factor (SCF)) are
more, these findings demonstrate that Sertoli cell known to be involved in the differentiation of
androgen-dependent gene expression is spermatogonia in humans, rodents, and primates

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 55

sjwint01@louisville.edu
56 H. Chen et al.

b Fig. 3.3 Comparison of germ cell development in the stage III (concomitant with spermiation that takes place at
human versus the rat testis. While the development of late stage II) of the epithelial cycle are transported across
germ cells through spermatogenesis is similar between the the BTB to enter the adluminal compartment to prepare
two species, there are some notable differences such as for meiosis I/II which takes place at stage VI of the
during spermatogonial self-renewal and differentiation. In epithelial cycle, and Sa spermatids, the equivalent of
the rat, Asingle spermatogonia (As) divide and form round spermatids in the rat, are found in stage I. In the rat,
Apaired spermatogonia (Apr), which further divide to a single diploid As can give rise to 4096 haploid sperm;
form Aaligned spermatogonia (Aal). The differentiating whereas in the human, only 16 sperm are derived from
Aal undergo 5 cell divisions (A1-4 and In) to form type B one type A spermatogonium. The number of different
spermatogonia. In humans, both Adark spermatogonia types of germ cells that can be derived from a single
(Ad) and Apale spermatogonia (Ap) are capable of committed type A spermatogonium is indicated. This
undergoing self-renewal through mitosis, and Ap also figure was prepared based on the information of earlier
give rise to type B spermatogonia, the latter of which are reports [1, 51, 127] and in the model of rat spermatoge-
then differentiate to form preleptotene spermatocytes. In nesis it is noted that Apr and Aal spermatogonia have been
the human testis, preleptotene spermatocytes formed in shown to be able to revert to As [241]

[91]. cKIT is expressed in spermatogonia, round spermatogenesis in humans is based on findings


spermatids, and spermatozoa as well as in Leydig that bisdichloroacetyldiamine WIN 18,446, an
cells whereas its ligand or SCF is primarily inhibitor of retinoic acid synthesis, inhibits
expressed in Sertoli cells in humans [92]. How- ALDH1A2 and induces reversible infertility in
ever, cKIT is not expressed in spermatocytes or mice, rats, monkeys, and men [100–103]. More
elongating spermatids [93]. Expression of cKIT study is needed to define the role of retinoic acid
in type A spermatogonia varies across the semi- in human spermatogonial differentiation.
niferous tubules, being highest in proliferating Spermatogonial differentiation is a
Ap spermatogonia and type B spermatogonia gonadotropin-dependent event in primates [104]
[93–95], illustrating the involvement of since FSH treatment increases the number of Ap
cKIT/SCF in spermatogonial self-renewal and and type B spermatogonia in monkeys [105,
differentiation. The reduced staining of cKIT in 106]. Emerging evidence suggests that FSH
type A spermatogonia has been shown in men plays a role in spermatogonial differentiation in
with subfertility, and its reduced expression is men [107]. However, in normal adult men,
associated with an increase in apoptosis in type A gonadotropins (FSH and LH) serve as sper-
spermatogonia [96]. matogonial survival factors by regulating the
GFRA1, a6-INTEGRIN, PLZF, GPR125, intrinsic apoptotic pathway to promote germ cell
SALL4, and THY1 are undifferentiated sper- survival but have no effect on germ cell prolif-
matogonia markers in mouse, monkey, and man, eration [108].
whereas UTF1 (undifferentiated embryonic cell
transcription factor 1) and FGFR3 (fibroblast
growth factor receptor 3, also known as CD333) Spermatocytes and Meiosis
are molecular markers specific for human type A
spermatogonia [91]. The molecular basis for the Spermatocytes, namely leptotene, zygotene,
transition of undifferentiated spermatogonia to pachytene, and diplotene spermatocytes are
type A spermatogonia has been established in found in the human seminiferous epithelium
rodents and involves retinoic acid (RA), the [109] following the initiation of meiosis in
active metabolite of vitamin A, which is also humans at puberty [110] (Figs. 3.3 and 3.4).
involved in the initiation of meiosis [97]. Indeed, Diplotene spermatocytes undergo meiosis I to
treatment of mice with the pan-retinoic acid form secondary spermatocytes (with haploid
receptor antagonist, BMS-189453 that blocks the number of chromosomes but 2n content of
action of RA in the mouse testis, leads to DNA), which rapidly progress to meiosis II so
reversible infertility due to meiotic arrest [98, that a single secondary spermatocyte produces
99]. The notion that RA is involved in two haploid round Sa spermatids. In this context,

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 57

Fig. 3.4 A schematic drawing illustrating the six stages the entire epithelial cycle takes 16 days to complete. The
of the epithelial cycle (I-VI) in the human testis, and the duration of each stage, from I to VI, is also annotated.
associated germ cell types in the seminiferous epithelium This drawing was prepared and modified from earlier
in each of these stages. It is noted that spermiation takes reports [4, 34, 242], and recent advances in the staging of
place in late stage II, and preleptotene spermatocytes arise human spermatogenesis have recently been discussed
in stage III, which are being transported across the BTB in [243]
stage III. Meiosis takes place at stage VI of the cycle, and

it is of interest to note that, unlike other somatic Sertoli cells by a monocarboxylate transporter
cells, neither spermatocytes nor spermatids MCT1 and taken up by meiotic and post-meiotic
metabolize glucose; instead, they rely on lactate germ cells via their specific monocarboxylate
supplied by Sertoli cells as their energy source transporter, MCT2 [111]. Studies have shown
[111, 112]. In brief, glucose is taken up by Ser- that energy metabolism of Sertoli and germ cells
toli cells via the specific glucose transporter is regulated by FSH, steroids, insulin and para-
GLUT1 and is processed into lactate glycolyti- crine factors, and their disruption leads to male
cally [113] by testis-specific lactate dehydroge- infertility [112, 114]. It is also noted that defects
nase C4 (LDHC4). Lactate is transported out of in meiosis, such as meiotic maturation arrest

sjwint01@louisville.edu
58 H. Chen et al.

(or early maturation arrest) is found in *10% of molecules such as p-FAK-Tyr407, as well as actin
men with non-obstructive azoospermia (NOA), binding/regulatory proteins (e.g., the Arp2/3
with the histopathological features of reduced complex, Eps8), involving degeneration of api-
tubule diameter, reduce germ cell number, and cal ES and generation of laminin fragments [5,
degenerating spermatocytes [115]. 119]. However, this process is poorly understood
in humans except that it is likely dependent on
FSH and testosterone. So far, human male
Spermatids and Spermiogenesis infertility has not been attributed to defects of
spermiation alone [3].
The process during which spermatids undergo
extensive morphological transformation through
round Sa spermatids, Sb1, Sb2, Sc, Sd1, and Sd2, Cycle of the Seminiferous Epithelium
and eventually to spermatozoa is known as
spermiogenesis [116] (Figs. 3.3 and 3.4). During In human as well as rodent testes, the distinctive
this process, no further cellular divisions occur, cellular associations in the seminiferous epithe-
newly formed round spermatids from secondary lium along the tubule can be defined into different
spermatocytes are characterized by a small stages which appear cyclically throughout sper-
spherical nucleus with the usual array of cyto- matogenesis, known as the epithelial cycle of
plasmic organelles, including the Golgi appara- spermatogenesis [88, 120, 121]. In the testis of
tus, mitochondria, and centrioles [116]. Each rats, mice, and humans, an epithelial cycle is
spermatid undergoes considerable morphological composed of I-XIV, I-XII, and I-VI stages,
changes that begin with a large granule called the respectively [4, 122, 123]. It is of interest to note
acrosomic granule which is generated by several that spermiation and meiosis I/II take place at stage
small proacrosomic granules in the area of the VIII and XIV, VIII and XII and II and VI, in the rat,
Golgi apparatus that grows over the nucleus. As mouse, and human testis, respectively. It takes
spermiogenesis continues, the acrosome trans- about 16 days to complete an epithelial cycle in
forms further, and nuclear chromatin condensa- the human [124] versus 8.6 days in the mouse
tion begins [117]. At the final steps, the [125] and 12.8 days in the rat [126]. This is the
spermatid nucleus completes its chromatin con- time it takes for a given spot in a seminiferous
densation, and the majority of the cytoplasm tubule at a specific stage of the epithelial cycle,
eventually detaches from the spermatid to form such as at stage II, when observed under the
the residual body, which is engulfed by the stereomicroscope, to undergo cyclic staging and
Sertoli cell and processed into a phagosome. The become stage II again. However, the duration of
phagosome is then transported to the base of the spermatogenesis, i.e., from type A spermatogonia
Sertoli cell for lysosomal degradation [118]. In to spermatozoa, is *68 days (4.2 cycles) in
humans, spermiogenesis can be disrupted, lead- humans versus *35 days (4 cycles) and
ing to: (i) late maturation arrest, manifested by an *58 days (4.5 cycles) in mice and rats as noted in
arrest in development in early spermatids with Fig. 3.4 for humans (for reviews, see [1, 127,
dark round nuclei, and (ii) hypospermatogenesis, 128]). This is the time it takes for a single human
with an arrest in further development of con- diploid Ap spermatogonium (progenitor germ cell,
densed oval spermatids [115]. Mature sperma- wherein Ad spermatogonium is the regenerative
tozoa, once formed, are released into the tubule reserve stem cell) to develop into multiple haploid
lumen via the final cellular process of sper- spermatozoa which requires *4.2 cycles. In this
matogenesis at spermiation [5, 119]. While the context, it is noted that the staging of the epithelial
molecular mechanism underlying spermiation cycle is largely defined according to changes in the
remains relatively unexplored, studies in rodents Golgi region of developing spermatids, namely the
have shown that spermiation is tightly regulated acrosome, when it is visualized by the periodic
by the spatiotemporal expression of signaling acid Schiff’s reaction (PAS). In mouse, rat, and

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 59

human, spermatids can be divided into 16, 19, and panoply of Sertoli cell genes. FSH promotes
6 steps, respectively, throughout spermiogenesis Sertoli cell proliferation before puberty, while
before they develop and transform into sperma- after puberty, FSH activates Sertoli cell to sup-
tozoa. The classification of human spermatids into port germ cell development. Despite the impor-
6 steps of Sa, Sb1, Sb2, Sc, Sd1, and Sd2 was tant role of FSH in spermatogenesis, this
based on the use of osmium-dichromate as fixative hormone, unlike testosterone (see below), is not
for the human testis, which was then stained with considered essential to, but rather fine-tunes
osmium and examined by transmission electron spermatogenesis. For instance, deletion of the
microscopy [117]. FSH-ß subunit in mice led to infertility in
FSH-deficient females due to a block in follicu-
logenesis, but FSH-deficient males were fertile
Factors that Regulate even though the testes of these mice were
Spermatogenesis reduced in size [140, 141]. Furthermore, men
with inactivating mutations of the FSH-b gene or
Numerous factors are known to affect human the FSH receptor have impaired spermatogenesis
spermatogenesis. Below, we highlight regulators but generally remain fertile—an observation
that are well established or are rapidly develop- explained in part by the finding that the FSH
ing in the field. receptor possesses low-level constitutive activity
in the absence of FSH [129, 142–144] (see
Chap. 6). On the other hand, it is of interest to
Hormonal Regulation note that men with congenital complete hypog-
onadotropic hypogonadism (CHH) are usually
Hormonal regulation of spermatogenesis in treated with hCG (human chorionic gonado-
humans is a complex biological event involving trophin) together with FSH to induce testis
both testosterone and estradiol-17ß [10, 13, 14, development, spermatogenesis, and fertility (see
129, 130] and is tightly regulated by the Chap. 20). Also, men with idiopathic oligoas-
hypothalamic–pituitary–testicular axis by thenoteratozoospermia treated with recombinant
follicle-stimulating hormone (FSH) and luteiniz- FSH for 3 months were found to have consid-
ing hormone (LH) that exert their effects on Sertoli erable improvement in seminal parameters vs.
and Leydig cells, respectively. Besides these hor- untreated controls [145, 146].
mones, inhibins, activins, follistatin, and other
paracrine factors are also involved. Since the LH
hormonal regulation of spermatogenesis in The primary function of LH is to stimulate
humans has been eminently reviewed recently [11, the production of testosterone by Leydig cells
13, 131–136], interested readers are encouraged to [147]. LHCG-receptor (luteinizing hormone/
seek further information from these earlier choriogonadotropin receptor, also called
reviews. Below is a summary of recent findings LHCG-R or LHR) expression begins in fetal
pertinent to the hormonal regulation of human Leydig cells [148]. Human males with inacti-
spermatogenesis. vating mutations of the LHCG-R develop
ambiguous genitalia and are testosterone-
FSH deficient indicating that hCG/LH signaling is
FSH is an important regulator of spermatogenesis essential for testosterone production in the fetus
through effects on Sertoli cells where FSH and adult [149–151]. LH stimulates testosterone
receptors are expressed. FSH activates at least production by stimulating the expression of key
five signaling pathways in Sertoli cells: steroidogenic enzyme genes as well as tran-
cAMP-PKA, MAPK, PI3 K-AKT, intracellular scription factors that are required for testosterone
Ca, and phospholipase A2 [137–139]. Gene biosynthesis to support spermatogenesis and
expression profiles indicate that FSH regulates a other male reproductive function [152]. Ligand

sjwint01@louisville.edu
60 H. Chen et al.

binding to the LHCG-R stimulates adenylate (i.e., to obtain ITT) and serum of fertile men in
cyclase, increases cAMP production and phos- samples obtained simultaneously were strongly
phorylates target proteins through protein kinases and positively correlated (r = 0.67) [158]. The
A and C, activates ERK1 and 2, and increases different results in the two studies may be due to
calcium signaling [148]. Interestingly, accumu- different assay approaches. In this context, it is of
lating evidence supports the notion that interest to note that a recent report has shown that
steroidogenesis in human fetal testes is highly high ITT is not necessary to support spermatoge-
sensitive to environmental toxicants or elected nesis in humans [159], and proposed that the ITT
lifestyle (e.g., cigarette smoking) which are dis- level may be high only because T is synthesized
ruptive to LH-mediated testosterone production locally by Leydig cells in the interstitial space of
by Leydig cells [17, 153]. the testis. Thus, the physiological significance of
the ITT level to support spermatogenesis requires
Intratesticular Testosterone additional further studies.
(ITT) Microenvironment Importantly, the earlier report of Roth et al.
Testicular aspirations of normal men have shown also demonstrated considerable variations in ITT
that T is the predominant intratesticular sex steroid levels in fertile men which correlated very
[48, 49, 154]. ITT concentration in men averages strongly with serum LH levels (r = 0.87). It was
609 ± 50 ng/ml, which is much higher than the hypothesized that LH secretory pulsatility might
average serum T level of 3.7 ± 0.3 ng/ml [48– cause pulsatility in ITT concentrations [158]. In
50]. This gradient between the testis and serum is fact, ITT pulsatility was suggested by an earlier
similar to findings in rodents, wherein ITT in rat study that quantified T and estradiol concentra-
testes is *100-fold higher than the serum T level tions over 4 h in cannulated gonadal veins of
[47, 155]. Collectively, these findings support the men with varicocele [32]. While the importance
notion that a high ITT level is necessary to main- of ITT pulsatility to spermatogenesis remains to
tain spermatogenesis. For instance, the high level be established, it may influence the cyclic nature
of T in the testis is known to support spermatid of spermatogenesis during the epithelial cycle.
adhesion, spermiogenesis, and BTB function in For instance, the requirements of the seminifer-
rodent testes [5, 51, 156]. In this context, it is of ous epithelium to support androgen-dependent
interest to note that the ITT concentration at cellular events at late stage II during spermiation
*2000 nM vesurs *12 nM of T in serum in are quite different from stages VI when meiosis
normal men is considerably higher than that of takes place. As such, it is likely that the ITT level
SHBG/ABP (sex hormone binding globulin/ required at stage II is different from stage VI and
androgen binding protein, *50 nM; note: other stages to support spermatogenesis.
SHBG/ABP reduces androgen bioavailability), Nonetheless, the ITT concentration necessary to
suggesting that most ITT is bioavailable [154]. support spermatogenesis in humans has not been
Studies by Jarow et al. found that *70% of the established by quantifying T levels in intrates-
total ITT is bioactive based on a novel androgen ticular fluid obtained during micro-TESE (mi-
bioactivity assay [154]. Interestingly, neither ITT crodissection testicular sperm extraction) [160].
concentrations nor intratesticular bioactive Furthermore, studies in rodents have shown that
androgen levels were strongly correlated with T, besides playing a crucial role in maintaining
serum T concentration (correlation coefficients, germ cell adhesion in particular developing
r = 0.38) nor serum bioactive androgen level spermatids, modulates endocytic vesicle-mediated
(r = 0.46). Yet SHBG/ABP levels in the testis and protein trafficking [161]. Due to the high level of
serum in humans were strongly correlated [48, cellular activities in the epithelium to maintain
154]. These data thus fail to explain the disparity the daily sperm production rate, many cellular
between intratesticular and serum bioactive proteins, in particular those found at the Sertoli–
androgen levels [48, 154, 157]. In contrast, Roth Sertoli or Sertoli–germ cell interface, are being
et al. found that T levels in testicular aspirations re-cycled [162]. Studies have shown that the

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 61

relative ratio of ITT/cytokines within the neurological disorder spinal and bulbar muscular
micro-environment of the seminiferous epithe- atrophy in which affected men have small testes
lium may be crucial to govern these protein and gynecomastia (SBMA) [172]. Attempts to
recycling events during the epithelial cycle [163]. correlate the severity of spermatogenic dysfunc-
These studies, while recently performed in tion with CAG-encoded polyglutamine length
rodents, can now be extended to humans by polymorphism in the AR gene have, however,
using the human Sertoli cell in vitro system [57]. yielded inconsistent and conflicting results [173–
176]. Interestingly, an insertion mutation near the
Androgen Receptor (AR) beginning of the CAG repeat in exon 1 of the AR
T exerts its effects through AR signaling [156, gene was found in an azoospermic man [177].
164–166]. In humans, AR expression is restricted Treatment of an infertile man who had a point
to Sertoli cells [167, 168]. Studies using murine mutation of p.Val686Ala in the AR
models have shown that specific deletion of AR ligand-binding domain with prolonged high-dose
in Sertoli cells (SC-specific AR KO) leads to testosterone therapy was found to produce
meiotic arrest and early spermatogenic matura- marked improvement in sperm count and semen
tion arrest [169, 170], and also terminal differ- quality [178]. Combined with intracytoplasmic
entiation of haploid spermatids [171]. microinjection, testosterone treatment resulted in
Collectively, these findings illustrate the signifi- fertility. A point mutation in the transactivation
cance of AR-mediated action in spermatogenesis, domain (TAD) of the AR gene was found in an
in particular murine meiosis and spermiogenesis. infertile man with gynecomastia with a high FSH
In men, the AR expression level visualized by level, small testes, and Sertoli cell-only syn-
immunohistochemistry in Sertoli cells was higher drome [179]. However, a fertile man with
in those with NOA (non-obstructive azoosper- gynecomastia was found to have a p.Pro69Ser
mia) vs. OA (obstructive azoospermia), and a mutation within the AR ligand-binding domain
significant positive correlation was observed but to display no considerable defects in semen
between FSH levels and Sertoli cell AR expres- quality [180]. Taking these data collectively, it is
sion in OA patients [167]. However, efforts to anticipated that more AR mutation-associated
relate AR function or distribution to infertility in male infertility cases will be identified. As more
humans have been unsuccessful. For instance, no data are available, a systemic analysis is war-
correlation was found between AR expression in ranted to relate mutation(s) of the AR gene with
Sertoli cells with serum T levels, or serum LH, idiopathic male infertility. At this point, based on
and FSH levels in men with NOA [167]. Also, the available data, it is likely that FSH-dependent
exogenous FSH was shown to trigger an increase AR expression is crucial for spermatogenesis.
in AR expression in the human testis [167]. However, more accurate functional assays are
However, hCG therapy had no apparent effect, necessary to pin-point the importance of AR in
thereby underscoring the significance of male infertility.
FSH-dependent Sertoli cell AR expression [167].
Exon 1 of the AR contains a CAG trinu- Estrogen
cleotide repeat that encodes a polyglutamine tract Testes are known to produce a considerable
in the N-terminus of the AR. This region is amount of estradiol-17ß via aromatase, which is
required for AR interaction with transcriptional essential to maintain spermatogenesis [10, 12,
co-regulators, and variation in CAG repeat 181]. In humans, the role of estrogen in sper-
length, even within normal alleles, influences AR matogenesis remains a subject of debate. Emerg-
function in that shorter repeats are associated ing evidence, however, supports the notion that
with a more transcriptionally active receptor. estrogen is involved in testis development, fluid
Furthermore, an expanded polyglutamine resorption in the rete testis, maintenance of sper-
tract confers toxic properties responsible for matogenesis, and in the maturation of spermatozoa
neuronal and non-neuronal degeneration in the [10, 181, 182]. Most importantly, aromatization of

sjwint01@louisville.edu
62 H. Chen et al.

T to estradiol-17ß in the hypothalamus provides an illustrate the significance of estrogen in human


important negative feedback signal to gonado- spermatogenesis. Furthermore, genetic and
tropin secretion [10]. Aromatase is expressed by pharmacologically induced estrogen deficiency
human Leydig cells, Sertoli cells, spermatocytes, leads to reduced libido in men [191, 192]. In
spermatids, and spermatozoa (its presence in summary, estrogen is known to be involved in
spermatogonia remains unknown) [181] while the development of the testis, maintenance of
estrogen is produced mostly by Leydig cells in the male reproductive tract, fluid resorption in the
adult human testis [10]. rete testis, and in particular sperm maturation in
Estrogen exerts its effects via estrogen recep- humans [12, 181, 193, 194].
tors ERa (ESR1) and ERß (ESR2) [10, 12].
Studies in rodents have shown that ERa is
expressed by Leydig cells and peritubular myoid Small Non-coding RNAs (sncRNAs)
cells; whereas ERß is found in some Leydig
cells, but mostly in Sertoli cells and germ cells As noted above, spermatogenesis is a highly
[183, 184]. In short, ERa is predominant in the complicated process that requires the intriguing
interstitium and ERß in the seminiferous epithe- participation of multiple genes at the transcrip-
lium. Subsequent studies, however, have tional and post-transcriptional levels to produce
demonstrated that ERa is also expressed in Ser- spermatozoa. Studies have shown that germ cells,
toli and germ cells in the rat testis [185]. In including spermatogonia, spermatocytes, and
human testes, the binding of estradiol-17ß to post-meiotic spermatids all contain abundant
human sperm was first reported in 1981 [186]. In levels of non-coding RNAs, such as siRNA
1998, human sperm were shown to express both (small interfering RNA, 20–25 nucleotides),
the ERa mRNA and protein [187]. Subsequent miRNA (microRNA, 21–24 nucleotides), and
studies have confirmed the presence of ERß as piRNAs (Piwi-interacting RNA, 26–31 nucleo-
well as ERa in human sperm [12]. It is now tides) [195–197]. The most important sncRNAs
generally accepted that ERa is expressed in that are involved in spermatogenesis are miRNAs
spermatogonia, pachytene spermatocytes, and and piRNAs and the associated pathways based
early round spermatids; whereas ERß is expres- on studies of genetic models in rodents [198–
sed in pachytene spermatocytes, early round 201]. These small RNAs are short single-stranded
spermatids, Sertoli cells, and Leydig cells in non-coding nucleotides that are known to directly
human testes [188, 189]. A genetic analysis study disrupt target mRNAs through degradation,
in 300 infertile Indian men and 255 fertile nor- causing translation repression to block protein
mal subjects identified single nucleotide poly- synthesis [20, 202]. Since miRNA can bind to
morphisms (SNPs) (found in 4 subjects) and more than one mRNA, a specific miRNA can
mutations (found in 8 subjects) in the ERß gene, regulate the function of more than a single gene.
suggesting ERß gene mutations are a cause of Small RNAs including miRNAs and piRNA are
spermatogenesis failure in men. These infertile mostly stored in the chromatoid body which is a
men displayed normal reproductive tract and dense structure in the germ cell cytosol composed
serum hormone levels [190]. The few men of mainly RNAs and RNA-binding proteins and
identified with aromatase deficiency due to are involved in the regulation of germ cell apop-
autosomal recessive inheritance of mutations in tosis, proliferation, and differentiation, as well as
the CYP19A1 gene tend to have small testes and spermatogonial stem cell self-renewal [200, 203,
an abnormal semen analysis. Additionally, hor- 204]. Studies have shown an alteration in the
mone levels in men with mutation in ERa and expression patterns of small RNAs in infertile
deficient in aromatase have considerable changes men [205–207]. For instance, miR-141, miR-429,
in serum T, estradiol, LH, and/or FSH compared and miR-7-1-3p were up-regulated in men with
to normal subjects (see Chap. 1). While the idiopathic non-obstructive azoospermia (NOA)
number of patients is small, these findings [206] while miR-34c-5p, miR-122, miR-146b-5p,

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 63

miR-181a, miR-374b, miR-509-5p, and gonadotropin levels, or other sperm parameters


miR-513a-5p were markedly down-regulated in despite reduced testosterone levels [215, 216].
men with NOA. These miRNAs are possibly The reason(s) behind the different conclusions is
involved in regulating germ cell apoptosis [207]. not immediately known. Nonetheless, studies
In this context, it is of interest to note that have demonstrated possible mechanism(s) by
miRNAs and siRNAs are processed by an which obesity might lead to reduced spermato-
RNase III endonuclease Dicer, the deletion of genesis capacity and daily sperm output, as well
which in mice leads to infertility. Dicer is essen- as reduced sperm count and sperm quality. One
tial for haploid spermatid differentiation [208] proposed mechanism is hyperestrogenism in
and for the assembly and maintenance of cell which serum estrogens are considerably higher in
junctions in the seminiferous epithelium during obese men due to an increase in peripheral con-
spermatogenesis [209]. Specific deletion of Dicer version of testosterone to estrogens by aromatase
in Sertoli cells also leads to infertility with com- in adipose tissue [217–219], coupled with a
plete absence of spermatozoa in seminiferous reduced testosterone production [214]. The
tubules and progressive testicular degeneration excessive estrogens in the systemic circulation
[210]. While the role of Dicer in human fertility thus inhibit the release of LH and FSH from the
remains to be elucidated, it is obvious that small pituitary gland via the negative feedback on the
regulatory RNAs represent a tempting target for hypothalamus, thereby reducing T production,
non-hormonal male contraception. Also, much intratesticular T levels, and the testosterone/
work is needed to better understand the role of estrogen ratio. The reduced T and FSH levels
small RNAs in human spermatogenesis, and their result in suppressed spermatogenesis. However,
use as diagnostic markers to monitor infertility, the correlation between high levels of estradiol
for example, men who are at risk because of and obesity is controversial and is further affected
industrial exposure to toxicant-induced infertility. by low SHBG [220], suggesting the need for
additional studies. Nonetheless, this mechanism
is supported by findings that obese men have
Obesity considerably lower levels of inhibin B than
healthy men [221–223]. Elevated levels of
According to NIH guidelines (see http://www. cytokines are another potential cause of
nhlbi.nih.gov/health/health-topics/topics/obe/ hypospermatogenesis in obese men [224]. Other
diagnosis), adults with a body mass index mechanisms that might cause defects in human
(BMI) >30 (kg/m2) are obese. Based on the latest spermatogenesis in obesity include reduced levels
statistics in 2014, the obese adult population in the of SHBG, insulin and leptin resistance, sleep
U.S. is at 27.7% (http://www.gallup.com/poll/ apnea, and adiponectin deficiency [225–229].
181271/obesity-rate-inches-2014.aspx), which is Additionally, factors involved in the pathogenesis
double the world’s average of 13% (http://www. of obesity, such as high calorie diet, genetic, and
who.int/mediacentre/factsheets/fs311/en/), illus- epigenetic disorders, might also play a role in
trating an alarming trend given the health risks perturbing sperm production [230, 231].
associated with obesity. A lower sex hormone Besides the disruptive effects of obesity on
binding globulin (SHBG) level is found in men spermatogenesis, there is emerging evidence that
with obesity and/or diabetes, which in turn lowers obesity also affects the molecular structure of
the total testosterone level (see Chap. 16 and testicular germ cells and mature spermatozoa,
[211]). Emerging evidence suggests a negative such as an impairment of acrosome reaction
correlation between rising BMI and sperm count, [232], leading to altered growth in offspring,
sperm concentration, and motile sperm, which increased susceptibility to disease in adults [233],
impedes male fertility [212–214]. Interestingly, and erectile dysfunction [234]. Studies also
while other studies suggest that obesity has no suggest that high-fat diets can affect the epige-
major impact on fertility, semen quality, netic content of sperm or the endocrine content

sjwint01@louisville.edu
64 H. Chen et al.

of seminal fluid, which in turn affects early fetal U54 HD029990 Project 5 (to C.Y.C.); National Natural
development [213]. Science Foundation of China (NSFC) 31371176 (to X.
X.), China Qianjiang Talents Program QJD1502029 (to
Bariatric surgery [235], such as Roux-en-Y X.X.) and Zhejiang Province Department of Science
gastric bypass surgery [236], has been reported to Technology Funding 2016F10010 (to X.X.)
increase T levels and improve sexual function in
obese men. Much of the increase in T, however,
results from an increase in the level of SHBG References
[236]. Interestingly, there are case reports in
which bariatric surgery was followed by 1. Ehmcke J, Schlatt S. A revised model for spermato-
impaired semen parameters, possibly by per- gonial expansion in man: lessons from non-human
turbing absorption of vitamins and trace elements primates. Reproduction. 2006;132:673–80.
[237, 238]. However, one study of six men 2. Schlatt S, Ehmcke J. Regulation of spermatogene-
sis: an evolutionary biologist’s perspective. Semin
showed no disruptive changes of bariatric sur- Cell Dev Biol. 2014;29:2–16.
gery on semen parameters, coupled with an 3. Neto FT, Bach PV, Najari BB, Li PS, Goldstein M.
increase in urinary total T levels [239]. Spermatogenesis in humans and its affecting factors.
Semin Cell Dev Biol. 2016.
4. Amann RP. The cycle of the seminiferous epithe-
lium in humans: a need to revisit? J Androl.
Summary 2008;29:469–87.
5. O’Donnell L, Nicholls PK, O’Bryan MK, McLach-
Spermatogenesis is a series of cellular events that lan RI, Stanton PG. Spermiation: the process of
sperm release. Spermatogenesis. 2011;1:14–35.
take place in the seminiferous tubules of the testis. 6. Amann RP, Howards SS. Daily spermatozoal
In this review, we provide a brief overview of production and epididymal spermatozoal reserves
human spermatogenesis, from spermatogonial of the human male. J Urol. 1980;124(2):211–5.
7. Johnson L, Petty CS, Neaves WB. A comparative
self-renewal via mitosis, meiosis, post-meiotic
study of daily sperm production and testicular
spermatid development via spermiogenesis, to composition in humans and rats. Biol Reprod.
the release of sperm at spermiation. We discuss the 1980;22(5):1233–43.
role of Sertoli cell and especially the role of the 8. Johnson L, Zane RS, Petty CS, Neaves WB.
Quantification of the human Sertoli cell population:
BTB in spermatogenesis. We also highlight some
its distribution, relation to germ cell numbers, and
specific areas of research that deserve future age-related decline. Biol Reprod. 1984;31:785–95.
attention. Due to the lack of human testis samples 9. Zitzmann M. Effects of age on male fertility. Best
for analysis, in particular those from normal sub- Pract Res Clin Endocrinol Metab. 2013;27:617–28.
10. O’Donnell L, Robertson KM, Jones ME, Simp-
jects, the study of human spermatogenesis lags far
son ER. Estrogen and spermatogenesis. Endocr
behind studies in rodents. However, human Sertoli Rev. 2001;22:289–318.
cells obtained at biopsy can now be cultured 11. McLachlan R, O’Donnell L, Meachem S, Stanton P,
in vitro and maintained up to weeks and months de Kretser D, Pratis K, et al. Hormonal regulation of
spermatogenesis in primates and man: insights for
where they remain mitotically active. These cells
development of the male hormonal contraceptive.
can be subcultured for creative experiments. We J Androl. 2002;23:149–62.
also briefly discuss some emerging fields of 12. Carreau S, Hess RA. Oestrogens and spermatoge-
research that focus on factors affecting human nesis. Philos Trans R Soc Lond B Biol Sci.
2010;365:1517–35.
spermatogenesis. These factors may deserve more
13. Sharpe RM. Regulation of spermatogenesis. In: The
attention by investigators in future years. Physiology of Reproduction. Eds. Knobil, E., Neill,
J.D. New York, Raven Press. 1994. pp. 1363–434.
Acknowledgements The authors wish to thank Drs. 14. O’Donnell L, Meachem SJ, Stanton PG, McLach-
Stephen Winters and Ilpo Huhtaniemi for their helpful lan RI. Endocrine regulation of spermatogenesis. In:
remarks, comments, insightful thoughts, and suggestions Neill JD, editor. Physiology of Reproduction. 3rd
during the preparation of this manuscript for this book. ed. Amsterdam: Elsevier; 2006. p. 1017–69.
Without this valuable help, this Chapter could not have 15. O’Shaughnessy PJ. Hormonal control of germ cell
been written.This work was supported by grants from the development and spermatogenesis. Semin Cell Dev
National Institutes of Health, R01 HD056034 (to C.Y.C.), Biol. 2014;29:55–65.

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 65

16. Kilchevsky A, Honig S. Male factor infertility in examination of testicles of 50 boys who suffered
2011: semen quality, sperm selection and from sudden death. Int J Androl. 1983;6:143–56.
hematospermia. Nat Rev Urol. 2012;9:68–70. 34. Clermont Y. The cycle of the seminiferous epithe-
17. Sharpe RM. Environmental/lifestyle effects on lium in man. Am J Anat. 1963;112:35–51.
spermatogenesis. Phil Trans R Soc Lond B Biol 35. Dym M. Basement membrane regulation of Sertoli
Sci. 2010;365:1697–712. cells. Endocr Rev. 1994;15:102–15.
18. Cheng CY, Wong EWP, Lie PPY, Li MWM, Su L, 36. Siu MKY, Cheng CY. Dynamic cross-talk between
Siu ER, et al. Environmental toxicants and male cells and the extracellular matrix in the testis.
reproductive function. Spermatogenesis. 2011;1:2–13. BioEssays. 2004;26:978–92.
19. Wei Y, Schatten H, Sun QY. Environmental 37. Mruk DD, Cheng CY. Sertoli-Sertoli and
epigenetic inheritance through gametes and impli- Sertoli-germ cell interactions and their significance
cations for human reproduction. Hum Reprod in germ cell movement in the seminiferous epithe-
Update. 2015;21(2):194–208. lium during spermatogenesis. Endocr Rev.
20. Campion S, Catlin N, Heger N, McConndell EV, 2004;25:747–806.
Pacheco SE, Saffarini C, et al. Male reprotoxicty 38. Cheng CY, Mruk DD. Cell junction dynamics in the
and endocrine disruption. EXS. 2012;101:315–60. testis: Sertoli-germ cell interactions and male contra-
21. Wan HT, Mruk DD, Wong CKC, Cheng CY. ceptive development. Physiol Rev. 2002;82:825–74.
Targeting testis-specific proteins to inhibit spermato- 39. Weber JE, Russell LD, Wong V, Peterson RN.
genesis - lesson from endocrine disrupting chemi- Three dimensional reconstruction of a rat stage V
cals. Expert Opin Ther Targets. 2013;17:839–55. Sertoli cell: II. Morphometry of Sertoli-Sertoli and
22. Geoffroy-Siraudin C, Loundou Ad, Romain F, Sertoli-germ cell relationships. Am J Anat.
Achard V, Courbiere B, Perrard MH, et al. Decline 1983;167:163–79.
of semen quality among 10932 males consulting for 40. Sharpe RM, McKinnell C, Kivlin C, Fisher JS.
couple infertility over a 20-year period in Marseille. Proliferation and functional maturation of Sertoli cells,
France. Asian J Androl. 2012;14:584–90. and their relevance to disorders of testis function in
23. Jorgensen N, Vierula M, Jacobsen R, Pukkala E, adulthood. Reproduction. 2003;125:769–84.
Perheentupa A, Virtanen HE, et al. Recent adverse 41. Auharek SA, Avelar GF, Lara NLM, Sharpe RM,
trends in semen quality and testis cancer incidence Franca LR. Sertoli cell numbers and spermatogenic
among Finnish men. Int J Androl. 2011;34:e37–48. efficiency are increased in inducible nitric oxide
24. Virtanen HE, Sadov S, Vierula M, Toppari J. Finland synthase (iNOS) mutant-mice. Int J Androl.
is following the trend-sperm quality in Finnish men. 2011;34:e621–9.
Asian J Androl. 2013;15(2):162–4. 42. Berndtson WE, Thompson TL. Changing relation-
25. Handelsman DJ, Cooper TG. Falling sperm counts ships between testis size, Sertoli cell number and
and global estrogenic pollution: what have we spermatogenesis in Sprague-Dawley rats. J Androl.
learned over 20 years? Asian J Androl. 2013;15 1990;11:429–35.
(2):159–61. 43. Wing TY, Christensen AK. Morphometric studies
26. Foresta C, Moro E, Ferlin A. Y chromosome on rat seminiferous tubules. Am J Anat.
microdeletions and alterations of spermatogenesis. 1982;165:13–25.
Endocr Rev. 2010;22:226–39. 44. Wang ZX, Wreford NG, de Kretser DM. Determi-
27. Greenberg SH, Lipshultz LI, Wein AJ. Experience nation of Sertoli cell numbers in the developing rat
with 425 subfertile male patients. J Urol. testis by sterological methods. Int J Androl.
1978;119:507–10. 1989;12:58–64.
28. Krausz C, Escamilla AR, Chianese C. Genetics of 45. Johnson L, Petty CS, Neaves WB. A comparative
male infertility: from research to clinic. Reproduc- study of daily sperm production and testicular
tion. 2015;150(5):R159–74. composition in humans and rats. Biol Reprod.
29. Jamsai D, O’Bryan MK. Mouse models in male 1980;22:1233–43.
fertility research. Asian J Androl. 2011;13:139–51. 46. Amann RP, Howards SS. Daily spermatozoal
30. Carrell DT, Aston KI, Oliva R, Emery BR, De production and epididymal spermatozoal reserves
Jonge CJ. The “omics” of human male infertility: of the human male. J Urol. 1980;124:211–5.
integrating big data in a systems biology approach. 47. Turner TT, Jones CC, Howards SS, Ewing LL,
Cell Tissue Res. 2016;363(1):295–312. Zegeye B, Gunsalus GL. On the androgen microen-
31. Com E, Melaine N, Chalmel F, Pineau C. Pro- vironment of maturing spermatozoa. Endocrinol-
teomics and integrative genomics for unraveling the ogy. 1984;115:1925–32.
mysteries of spermatogenesis: the strategies of a 48. Jarow JP, Chen H, Rosner W, Trentacoste S,
team. J proteomics. 2014;107:128–43. Zirkin BR. Assessment of the androgen environ-
32. Winters SJ, Troen P. Testosterone and estradiol are ment within the human testis: Minimally invasive
co-secreted episodially by the human testis. J Clin method to obtain intratesticular fluid. J Androl.
Invest. 1986;78:870–3. 2001;22:640–5.
33. Muller J, Skakkebaek NE. Quantification of germ 49. Roth MY, Page ST, Lin K, Anawalt BD, Mat-
cells and seminiferous tubules by stereological sumoto AM, Synder CN, et al. Dose-dependent

sjwint01@louisville.edu
66 H. Chen et al.

increase in intratesticular testosterone by very claudin-11 at the Sertoli cell tight junction. Hum
low-dose human chorionic gonadotropin in normal Reprod. 2016;31(4):875–86.
men with experimental gonadotropin deficiency. 64. Robillard KR, Hoque MT, Bendayan R. Expression
J Clin Endocrinol Metab. 2010;95:3806–13. of ATP-binding cassette membrane transporters in
50. Takahashi J, Higashi Y, Lanasa JA, Winters SJ, rodent and human Sertoli cells: relevance to the
Oshima H, Troen P. Studies of the human testis. permeability of antiretroviral therapy at the
XVII. Gonadotropin regulation of intratesticular blood-testis barrier. J Pharmacol Exp Ther.
testosterone and estradiol in infertile men. J Clin 2012;340:96–108.
Endocrinol Metab. 1982;55:1073–80. 65. Hoque MT, Kis O, De Rosa MF, Bendayan R.
51. Cheng CY, Mruk DD. The blood-testis barrier and Raltegravir permeability across blood-tissue barriers
its implications for male contraception. Pharmacol and the potential role of drug efflux transporters.
Rev. 2012;64:16–64. Antimicrob Agents Chemother. 2015;59(5):2572–82.
52. Pelletier RM. The blood-testis barrier: the junctional 66. Jesus TT, Oliveira PF, Silva J, Barros A, Ferreira R,
permeability, the proteins and the lipids. Prog Sousa M, et al. Mammalian target of rapamycin
Histochem Cytochem. 2011;46:49–127. controls glucose consumption and redox balance in
53. Cheng YH, Wong EWP, Cheng CY. Cancer/testis human Sertoli cells. Fertil Steril. 2016;105(3):
(CT) antigens, carcinogenesis and spermatogenesis. 825–33 e3.
Spermatogenesis. 2011;1:209–20. 67. Bardin CW, Gunsalus GL, Cheng CY. The cell
54. Yule TD, Montoya GD, Russell LD, Williams TM, biology of the Sertoli cell. In: Desjardins C,
Tung KSK. Autoantigenic germ cells exist outside the Ewing L, editors. Cell and molecular biology of
blood testis barrier. J Immunol. 1988;141:1161–7. the testis. New York: Oxford University Press;
55. Meinhardt A, Hedger MP. Immunological, para- 1993. p. 189–219.
crine and endocrine aspects of testicular immune 68. Cheng CY, Mruk DD. Biochemistry of Sertoli
privilege. Mol Cell Endocrinol. 2011;335:60–8. cell/germ cell junctions, germ cell transport, and
56. Chui K, Trivedi A, Cheng CY, Cherbavaz DB, spermiation in the seminiferous epithelium. In:
Dazin PF, Huynh ALT, et al. Characterization and Griswold MD, editor. Sertoli Cell Biology. 2nd
functionality of proliferative human Sertoli cells. ed. Amsterdam: Elsevier; pp; 2015. p. 333–83.
Cell Transplant. 2011;20:619–35. 69. de Rooij DG. The spermatogonial stem cell niche.
57. Xiao X, Mruk DD, Tang EI, Wong CKC, Lee WM, Microsc Res Tech. 2009;72:580–5.
John CM, et al. Environmental toxicants perturb 70. Phillips BT, Gassei K, Orwig KE. Spermatogonial
human Serotli cell adhesive function via changes in stem cell regulation and spermatogenesis. Philos
F-actin organization medicated by actin regulatory Trans R Soc Lond B Biol Sci. 2010;365:1663–78.
proteins. Hum Reprod. 2014;29:1279–91. 71. Zwain I, Morris PL, Cheng CY. Identification of an
58. O’Donnell L, Stanton P, Bartles J, Robertson D. inhibitory factor from a Sertoli clonal cell line
Sertoli cell ectoplasmic specializations in the sem- (TM4) that modulates adult rat Leydig cell steroido-
iniferous epithelium of the testosterone-suppressed genesis. Mol Cell Endocrinol. 1991;80:115–26.
adult rat. Biol Reprod. 2000;63:99–108. 72. Zwain IH, Cheng CY. Rat seminiferous tubular
59. Meng J, Holdcraft RW, Shima JE, Griswold MD, culture medium contains a biological factor that
Braun RE. Androgens regulate the permeability of inhibits Leydig cell steroidogenesis: its purification
the blood-testis barrier. Proc Natl Acad Sci USA. and mechanism of action. Mol Cell Endocrinol.
2005;102:16696–700. 1994;104:213–27.
60. Chung NPY, Cheng CY. Is cadmium 73. Saez JM, Avallet O, Naville D, Perrard-Sapori MH,
chloride-induced inter-Sertoli tight junction perme- Chatelain PG. Sertoli-Leydig cell communications.
ability barrier disruption a suitable in vitro model to Ann N Y Acad Sci. 1989;564:210–31.
study the events of junction disassembly during 74. Tio S, Koppenaal D, Bardin C, Cheng C. Purifica-
spermatogenesis in the rat testis? Endocrinology. tion of gonadotropin surge inhibiting factor from
2001;142:1878–88. Sertoli cell-enriched culture medium. Biochem
61. Xiao X, Mruk DD, Lee WM, Cheng CY. c-Yes Biophys Res Commun. 1994;199:1229–36.
regulates cell adhesion at the blood-testis barrier 75. Rebourcet D, O’Shaughnessy PJ, Monteiro A,
and the apical ectoplasmic specialization in the Milne L, Cruickshanks L, Jeffrey N, et al. Sertoli
seminiferous epithelium of rat testes. Int J Biochem cells maintain Leydig cell number and peritubular
Cell Biol. 2011;43:651–65. myoid cell activity in the adult mouse testis.
62. Ilani N, Armanious N, Lue YH, Swerdloff RS, PLoS ONE. 2014;9(8):e105687.
Baravarian S, Adler A, et al. Integrity of the 76. Rebourcet D, O’Shaughnessy PJ, Pitetti JL, Mon-
blood-testis barrier in healthy men after suppression teiro A, O’Hara L, Milne L, et al. Sertoli cells
of spermatogenesis with testosterone and levonor- control peritubular myoid cell fate and support adult
gestrel. Hum Reprod. 2012;27(12):3403–11. Leydig cell development in the prepubertal testis.
63. McCabe MJ, Tarulli GA, Laven-Law G, Matthies- Development. 2014;141(10):2139–49.
son KL, Meachem SJ, McLachlan RI, et al. Gon- 77. Wen Q, Zheng QS, Li XX, Hu ZY, Gao F,
adotropin suppression in men leads to a reduction in Cheng CY, et al. Wt1 dictates the fate of fetal and

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 67

adult Leydig cells during development in the mouse 94. Medrano JV, Marques-Mari AI, Aguilar CE,
testis. Am J Physiol Endocrinol Metab. 2014;307 Riboldi M, Garrido N, Martinez-Romero A, et al.
(12):E1131–43. Comparative analysis of the germ cell markers
78. Jegou B. The Sertoli-germ cell communication c-KIT, SSEA-1 and VASA in testicular biopsies
network in mammals. Int Rev Cytol. 1993;147: from secretory and obstructive azoospermias. Mol
25–96. Hum Reprod. 2010;16(11):811–7.
79. Griswold M. Interactions between germ cells and 95. Gassei K, Ehmcke J, Dhir R, Schlatt S. Magnetic
Sertoli cells in the testis. Biol Reprod. 1995;52: activated cell sorting allows isolation of spermato-
211–6. gonia from adult primate testes and reveals distinct
80. Skinner M, Tung P, Fritz I. Cooperativity between CFRa1-positive subpopulations in men. J Med
Sertoli cells and testicular peritubular cells in the Primatol. 2010;39:83–91.
production and deposition of extracellular matrix 96. Feng HL, Sandlow JI, Sparks AE, Sandra A,
components. J Cell Biol. 1985;100:1941–7. Zheng LJ. Decreased expression of the c-kit recep-
81. Skinner MK, Fetterolf PM, Anthony CT. Purifica- tor is associated with increased apoptosis in
tion of a paracrine factor, P-Mod-S, produced by subfertile human testes. Fertil Steril. 1999;71
testicular peritubular cells that modulates Sertoli (1):85–9.
cell function. J Biol Chem. 1988;263:2884–90. 97. Hogarth CA, Griswold MD. The key role of vitamin
82. Cheng CY, Grima J, Stahler MS, Guglielmotti A, A in spermatogenesis. J Clin Invest. 2010;120:
Silvestrini B, Bardin CW. Sertoli cell synthesizes 956–62.
and secretes a protease inhibitor, a2-macroglobulin. 98. Chung SS, Wang X, Roberts SS, Griffey SM,
Biochemistry. 1990;29:1063–8. Reczek PR, Wolgemuth DJ. Oral administration of
83. Zwain IH, Grima J, Stahler MS, Saso L, Cailleau J, a retinoic acid receptor antagonist reversibly inhibits
Verhoeven G, et al. Regulation of Sertoli cell a2- spermatogenesis in mice. Endocrinology. 2011;152:
macroglobulin and clusterin (SGP-2) secretion by 2492–502.
peritubular myoid cells. Biol Reprod. 1993;48: 99. Chung SS, Wang X, Wolgemuth DJ. Prolonged
180–7. Oral Administration of a Pan-Retinoic Acid Recep-
84. Mruk DD, Cheng CY. An in vitro system to study tor Antagonist Inhibits Spermatogenesis in Mice
Sertoli cell blood-testis barrier dynamics. Methods With a Rapid Recovery and Changes in the
Mol Biol. 2011;763:237–52. Expression of Influx and Efflux Transporters.
85. Welsh M, Saunders PT, Atanassova N, Sharpe RM, Endocrinology. 2016;157(4):1601–12.
Smith LB. Androgen action via testicular peritubu- 100. Amory JK, Muller CH, Shimshoni JA, Isoherra-
lar myoid cells is essential for male fertility. nen N, Paik J, Moreb JS, et al. Suppression of
FASEB J. 2009;23:4218–30. spermatogenesis by bisdichloroacetyldiamines is
86. Rebourcet D, Wu J, Cruickshanks L, Smith SE, mediated by inhibition of testicular retinoic acid
Milne L, Fernando A, et al. Sertoli Cells Modulate biosynthesis. J Androl. 2011;32(1):111–9.
Testicular Vascular Network Development, Struc- 101. Paik J, Haenisch M, Muller CH, Goldstein AS,
ture, and Function to Influence Circulating Testos- Arnold S, Isoherranen N, et al. Inhibition of retinoic
terone Concentrations in Adult Male Mice. acid biosynthesis by the bisdichloroacetyldiamine
Endocrinology. 2016;157(6):2479–88. WIN 18,446 markedly suppresses spermatogenesis
87. Allen E. Studies on cell division in the albino rat. and alters retinoid metabolism in mice. J Biol
J Morphol. 1918;31:133–85. Chem. 2014;289(21):15104–17.
88. Clermont Y. Kinetics of spermatogenesis in mam- 102. Heller CG, Moore DJ, Paulsen CA. Suppression of
mals: seminiferous epithelium cycle and spermato- spermatogenesis and chronic toxicity in men by a
gonial renewal. Physiol Rev. 1972;52(1):198–236. new series of bis(dichloroacetyl) diamines. Toxicol
89. Clermont Y. Renewal of spermatogonia in man. Appl Pharmacol. 1961;3:1–11.
Am J Anat. 1966;118(2):509–24. 103. Drobeck HP, Coulston F. Inhibition and recovery of
90. Schulze C. Morphological characteristics of the spermatogenesis in rats, monkeys, and dogs med-
spermatogonial stem cells in man. Cell Tissue Res. icated with bis(dichloroacetyl) diamines. Exp Mol
1979;198(2):191–9. Pathol. 1962;1:251–74.
91. Boitani C, Di Persio S, Esposito V, Vicini E. 104. Plant TM. Undifferentiated primate spermatogonia
Spermatogonial cells: mouse, monkey and man and their endocrine control. Trends Endocrinol
comparison. Semin Cell Dev Biol. 2016. Metab. 2010;21:488–95.
92. Sandlow JI, Feng HL, Cohen MB, Sandra A. 105. van Alphen MM, van de Kant HJ, de Rooij DG.
Expression of c-KIT and its ligand, stem cell factor, Follicle-stimulating hormone stimulates spermato-
in normal and subfertile human testicular tissue. genesis in the adult monkey. Endocrinology.
J Androl. 1996;17(4):403–8. 1988;123:1449–55.
93. Unni SK, Modi DN, Pathak SG, Dhabalia JV, 106. Simorangkir DR, Ramaswarmy S, Marshall GR,
Bhartiya D. Stage-specific localization and expres- Pohl CR, Plant TM. A selective monotropic eleva-
sion of c-kit in the adult human testis. J Histochem tion of FSH but not that of LH, amplifies the
Cytochem. 2009;57(9):861–9. proliferation and differentiation of spermatogonia in

sjwint01@louisville.edu
68 H. Chen et al.

the adult rhesus monkey (Macaca mulatta). Hum 123. Xiao X, Mruk DD, Wong CK, Cheng CY. Germ
Reprod. 2009;24:1584–95. cell transport across the seminiferous epithelium
107. Selice R, Ferlin A, Garolla A, Caretta N, Foresta C. during spermatogenesis. Physiology (Bethesda).
Effects of endogenous FSH on normal human 2014;29(4):286–98.
spermatogenesis in adults. Int J Androl. 2011;34: 124. Heller CG, Clermont Y. Spermatogenesis in man:
e511–7. an estimate of its duration. Science. 1963;140
108. Ruwanpura SM, McLachlan RI, Matthiesson KL, (3563):184–6.
Meachem SJ. Gonadotrophins regulate germ cell 125. Oakberg EF. Duration of spermatogenesis in the
survival, not proliferation, in normal adult men. mouse and timing of stages of the cycle of the
Hum Reprod. 2008;23:403–11. seminiferous epithelium. Am J Anat. 1956;99
109. Schulze W, Rehder U. Organization and morpho- (3):507–16.
genesis of the human seminiferous epithelium. Cell 126. Clermont Y, Leblond CP, Messier B. Duration of
Tissue Res. 1984;237(3):395–407. the cycle of the seminal epithelium of the rat. Arch
110. Feng CW, Bowles J, Koopman P. Control of Anat Microsc Morphol Exp. 1959;48(Suppl):37–55.
mammalian germ cell entry into meiosis. Mol Cell 127. Ehmcke J, Wistuba J, Schlatt S. Spermatogonial
Endocrinol. 2014;382(1):488–97. stem cells: questions, models and perspectives.
111. Boussouar F, Benahmed M. Lactate and energy Human Reprod Update. 2006;12:275–82.
metabolism in male germ cells. Trends Endocrinol 128. de Kretser DM, Kerr JB. The cytology of the testis.
Metab. 2004;15:345–50. In: Knobil E, Neill JB, Ewing LL, Greenwald GS,
112. Dias TR, Alves MG, Silva BM, Oliveira BF. Sperm Markert CL, Pfaff DW, editors. The Physiology of
glucose transport and metabolism in diabetic indi- Reproduction, vol. 1. New York: Raven Press;
viduals. Mol Cell Endocrinol. 2014;396:37–45. 1988. p. 837–932.
113. Oliveira PF, Martins AD, Moreira AC, Cheng CY, 129. McLachlan RI, O’Donnell L, Meachem SJ, Stan-
Alves MG. The Warburg effect revisited - lession ton PG, De Kretser DM, Pratis K, et al. Identifica-
from the Sertoli cell. Med Res Rev. 2015;35: tion of specific sites of hormonal regulation in
126–51. spermatogenesis in rats, monkeys, and man. Recent
114. Oliveira BF, Alves MG, Rato L, Silva J, Sa R, Prog Horm Res. 2002;57:149–79.
Barros A, et al. Influence of 5a-dihydrotestosterone 130. Handelsman DJ. Hormonal regulation of spermato-
and 17b-estradiol on human Sertoli cells metabo- genesis: insights from constructing genetic models.
lism. Int J Androl. 2011;34:e612–20. Reprod Fertil Dev. 2011;23:507–19.
115. McLachlan RI, Rajpert-De Meyts E, Hoei-Hansen 131. Itman CM, Mendis SH, Barakat B, Loveland KL.
CE, de Kretser DM, Skakkebaek NE. Histological All in the family: TGF-b family action in testis
evaluation of the human testis—Approaches to development. Reproduction. 2006;132:233–46.
optimizing the clinical value of the assessment: 132. Foresta C, Selice R, Garolla A, Ferlin A.
Mini Review. Hum Reprod. 2007;22:2–16. Follicle-stimulating hormone treatment of male
116. Clermont Y, Leblond CP. Spermiogenesis of man, infertility. Curr Opin Urol. 2008;18:602–7.
monkey, ram and other mammals as shown by the 133. De Kretser DM, Hedger MP, Loveland KL,
periodic acid-Schiff technique. Am J Anat. 1955;96 Phillips DJ. Inhibins, activins and follistatin in
(2):229–53. reproduction. Hum Reprod Update. 2002;8:529–41.
117. de Kretser DM. Ultrastructural features of human 134. Winters SJ, Moore JP. Paracrine control of gonado-
spermiogenesis. Z Zellforsch Mikrosk Anat. trophs. Semin Reprod Med. 2007;25:379–87.
1969;98(4):477–505. 135. Winters SJ, Moore JP. Intra-pituitary regulation of
118. Clermont Y, Morales C, Hermo L. Endocytic gonadotrophs in male rodents and primates. Repro-
activities of Sertoli cells in the rat. Ann NY Acad duction. 2004;128:13–23.
Sci. 1987;513:1–15. 136. McLachlan R, Wreford N, O’Donnell L, de Kret-
119. Cheng CY, Mruk DD. Biochemistry of Sertoli ser D, Robertson D. The endocrine regulation of
cell/germ cell junctions, germ cell transport, and spermatogenesis: independent roles for testosterone
spermiation in the seminiferous epithelium. In: and FSH. J Endocr. 1996;148:1–9.
Sertoli Cell Biology, 2nd Edition. Ed. Griswold, 137. Huhtaniemi I. A short evolutionary history of
M.D., Amsterdam, Elsevier; pp. 333–383. doi: 10. FSH- = stimulated spermatogenesis. Hormones
1016/B978-0-12-417047-6.00012.0. 2015. (Athens). 2015;14:468–78.
120. Roosen-Runge EC. Kinetics of spermatogenesis in 138. Stanton PG. Regulation of the blood-testis barrier.
mammals. Ann N Y Acad Sci. 1952;55(4):574–84. Semin Cell Dev Biol (in press; PMID:27353840;
121. Leblond CP, Clermont Y. Definition of the stages of DOI:101016/jsemcdb201606018). 2016.
the cycle of the seminiferous epithelium in the rat. 139. Griswold MD, Heckert L, Linder C. The molecular
Ann N Y Acad Sci. 1952;55(4):548–73. biology of the FSH receptor. J Steroid Biochem
122. Hess RA, de Franca LR. Spermatogenesis and cycle Mol Biol. 1995;53:215–8.
of the seminiferous epithelium. Adv Exp Med Biol. 140. Kumar TR, Wang Y, Lu N, Matzuk MM. Follicle
2008;636:1–15. stimulating hormone is required for ovarian follicle

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 69

maturation but not male fertility. Nat Genet. 156. Wang RS, Yeh S, Tzeng CR, Chang C. Androgen
1997;15:201–4. receptor roles in spermatogenesis and fertility:
141. Kumar TR. FSHbeta knockout mouse model: a lessons from testicular cell-specific androgen recep-
decade ago and into the future. Endocrine. 2009;36 tor knockout mice. Endocr Rev. 2009;30:119–32.
(1):1–5. 157. Nieschlag E, Wickings EJ, Mauss J. Endocrine
142. Plant TM, Marshall GR. The functional significance testicular function in vivo and in vitro in infertile
of SH in spermatogenesis and the control of its men. Acta Endocrinol. 1979;90:544–51.
secretion in male primates. Endocr Rev. 158. Roth MY, Lin K, Amory JK, Matsumoto AM,
2001;22:764–86. Anawalt BD, Snyder CN, et al. Serum LH correlates
143. Walker WH, Cheng J. FSH and testosterone highly with intratesticular steroid levels in normal
signaling in Sertoli cells. Reproduction. men. J Androl. 2009;31:138–45.
2005;130:15–28. 159. Oduwole OO, Vydra N, Wood NE, Samanta L,
144. O’Shaughnessy PJ, Johnston H, Willerton L, Owen L, Keevil B, et al. Overlapping dose
Baker PJ. Failure of normal adult Leydig cell responses of spermatogenic and extragonadal
development in androgen-receptor-deficient mice. testosterone actions jeopardize the principle of
J Cell Sci. 2002;115:3491–6. hormonal male contraception.
145. Caroppo E, Niederberger C, Vizziello GM, D’Amato FASEB J. 2014;28:2566–76.
G. Recombinant human follicle-stimulating hormone 160. Shinjo E, Shiraishi K, Matsuyama H. The effect of
as a pretreatment for iodiopathic oligoasthenoterato- human chorionic gonadotropin-based hormonal
zoospermic patients undergoing intracytoplasmic therapy on intratesticular testosterone levels and
sperm injection. Fertil Sertil. 2003;80:1398–403. spermatogonial DNA synthesis in men with
146. Santi D, Granata AR, Simoni M. Follicle- non-obstructive azoospermia. Andrology. 2013;1
stimulating hormone treatement of male idiopathic (6):929–35.
infertility improves pregancy rate: a meta-analysis. 161. Yan HHN, Mruk DD, Lee WM, Cheng CY.
Endocr Connect. 2015;4:46–58. Blood-testis barrier dynamics are regulated by
147. Saez JM. Leydig cells: endocrine, paracrine, and testosterone and cytokines via their differential
autocrine regulation. Endocr Rev. 1994;15:574–626. effects on the kinetics of protein endocytosis and
148. Ascoli M, Fanelli F, Segaloff DL. The lutropin/ recycling in Sertoli cells. FASEB J. 2008;22:
choriogonadotropin receptor, a 2002 perspective. 1945–59.
Endocr Rev. 2002;23:141–74. 162. Xiao X, Wong EWP, Lie PPY, Mruk DD,
149. Themmen APN, Huhtaniemi IT. Mutations of Wong CKC, Cheng CY. Cytokines, polarity pro-
gonadotropins and gonadotropin receptors: Eluci- teins and endosomal protein trafficking and signal-
dating the physiology and pathophysiology of ing—the Sertoli cell blood-testis barrier in vitro as a
pituitary-gonadal function. Endocr Rev. 2000;21: study model. Methods Enzymol. 2014;534:181–94.
551–83. 163. Li MWM, Mruk DD, Lee WM, Cheng CY.
150. Latronico AC, Segaloff DL. Naturally occurring Cytokines and junction restructuring events during
mutations of the luteinizing-hormone receptor: spermatogenesis in the testis: An emerging concept
Lessons learned about rerproductive physiology of regulation. Cytokine Growth Factor Rev.
and G protein-coupled receptors. Am J Human 2009;20:329–38.
Genet. 1999;65:949–58. 164. Smith LB, Walker WH. The regulation of sper-
151. Latronico AC, Arnhold IJ. Inactivating mutations of matogenesis by androgens. Semin Cell Dev Biol.
the human luteinizing hormone receptor in both 2014;30:2–13.
sexes. Semin Reprod Med. 2012;30:382–6. 165. Pihlajamaa P, Sahu B, Janne OA. Determinants of
152. Winters SJ. Endocrine evaluatiojn of testicular receptor- and tissue-specific actions in androgen
function. Endocrinol Metab Clin North Am. signaling. Endocr Rev. 2015;36(4):357–84.
1994;23:709–23. 166. Walker WH. Non-classical actions of testosterone
153. Scott HM, Mason JI, Sharpe RM. Steroidogenesis and spermatogenesis. Philos Trans R Soc Lond B
in the fetal testis and its susceptibility to disruption Biol Sci. 2010;365:1557–69.
by exogenous compounds. Endocr Rev. 2009;30 167. Kato Y, Shiraishi K, Matsuyama H. Expression of
(7):883–925. testicular androgen receptor in non-obstructive
154. Jarow JP, Wright WW, Brown TR, Yan X, azoospermia and its change after hormonal therapy.
Zirkin BR. Bioactivity of androgens within the Andrology. 2014;2:734–40.
testes and serum of normal men. J Androl. 2005;26 168. Griswold M. The central role of Sertoli cells in
(3):343–8. spermatogenesis. Semin Cell Dev Biol.
155. Cheng CY, Gunsalus GL, Morris ID, Turner TT, 1998;9:411–6.
Bardin CW. The heterogeneity of rat androgen 169. De Gendt K, Swinnen J, Saunders P, Schoonjans L,
binding protein (rABP) in the vascular compartment Dewerchin M, Devos A, et al. A Sertoli
differs from that in the testicular tubular lumen: cell-selective knockout of the androgen receptor
further evidence for bidirectional secretion of causes spermatogenic arrest in meiosis. Proc Natl
rABP. J Androl. 1986;7:175–9. Acad Sci USA. 2004;101:1327–32.

sjwint01@louisville.edu
70 H. Chen et al.

170. Chang C, Chen Y, Yeh S, Xu D, Wang R, Guillou F, and estrogen receptors in adult male mouse repro-
et al. Infertility with defective spermatogenesis and ductive tract. J Androl. 2002;23:870–81.
hypotestosteronemia in male mice lacking the 184. Kotula-Balak M, Gancarczyk M, Sadowska J,
androgen receptor in Sertoli cells. Proc Natl Acad Bilinska B. The expression of aromatase, estrogen
Sci USA. 2004;101:6876–81. receptor a and estrogen receptor b in mouse Leydig
171. Holdcraft RW, Braun RE. Androgen receptor cells in vitro that derived from crytorchid males.
function is required in Sertoli cells for the terminal Eur J Histochem. 2005;49:59–62.
differentiation of haploid spermatids. Development. 185. Lucas TF, Siu ER, Esteves CA, Monteiro HP,
2004;131:459–67. Oliverira CA, Porto CS, et al. 17b-estradiol induces
172. La Spada AR, Wilson EM, Lubahn DB, Hard- the translocation of the estrogen receptors ESR1 and
ing AE, Fischbeck KH. Androgen receptor gene ESR2 to the cell membrane, MAPK3/1 phosphory-
mutations in X-linked spinal and bulbar muscular lation and proliferation of cultured immature rat
atrophy. Nature. 1991;352:77–9. Sertoli cells. Biol Reprod. 2008;78:101–14.
173. Delli Muti N, Agarwal A, Buldreghini E, Gioia A, 186. Cheng CY, Boettcher B, Rose RJ, Kay DJ, Tin-
Lenzi A, Boscaro M, et al. Have androgen receptor neberg HR. The binding of sex steroids to human
gene CAG and GGC repeat polymorphisms an spermatozoa. An autoradiographic study. Int J
effect on sperm motility in infertile men? Androlo- Androl. 1981;4:1–17.
gia. 2014;46(5):564–9. 187. Durkee TJ, Mueller M, Zinaman M. Identification
174. Fietz D, Geyer J, Kliesch S, Gromoll J, of estrogen receptor protein and messenger ribonu-
Bergmann M. Evaluation of CAG repeat length of celic acid in human spermatozoa. Am J Obstet
androgen receptor expressing cells in human testes Gynecol. 1998;178:1288–97.
showing different pictures of spermatogenic impair- 188. Fietz D, Bergmann M, Hartmann K. In situ
ment. Histochem Cell Biol. 2011;136:689–97. hybridization of estrogen receptors a and b and
175. Giagulli VA, Carbone MD, De Pergola G, Guasta- GPER in the human testis. Methods Mol Biol.
macchia E, Resta F, Licchelli B, et al. Could 2016;1366:189–205.
androgen receptor gene CAG tract polymorphism 189. Pelletier G, El-Alfy M. Immunocytochemical local-
affect spermatogenesis in men with idiopathic ization of estrogen receptors a and b in the human
infertility? J Assit Reprod Genet. 2014;31:689–97. reproductive organs. J Clin Endocrinol Metab.
176. Casella R, Maduro MR, Misfud A, Lipshultz LI, 2000;85:4835–40.
Yong EL, Lamb DJ. Androgen receptor gene 190. Khattri A, Pandey RK, Gupta NJ, Chakravarty B,
polyglutamine length is associated with testicular Deenadayal M, Singh L, et al. Estrogen receptor
histology in infertile patients. J Urol. beta gene mutations in Indian infertile men. Mol
2003;169:224–7. Hum Reprod. 2009;15(8):513–20.
177. Chen YH, Xu HY, Wang ZY, Zhu ZH, Li CD, 191. Carani C, Rochira V, Faustini-Fustini M, Balestri-
Wu ZG, et al. An insertion mutation in the androgen eri A, Granata AR. Roel of oestrogen in male sexual
receptor gene in a patient with azoospermia. Asian J behaviour: Insights from the natural model of
Androl. 2015;17(5):857–8. aromatase deficiency. Clin Endocrinol. 1999;51:
178. Tordjman KM, Yaron M, Berkovitz A, Botchan A, 517–24.
Sultan C, Lumbroso S. Fertility after high-dose 192. Finkelstein JS, Yu EW, Burnett-Bowie SA. Gon-
testosterone and intracytoplasmic sperm injection in adal steroids and body composition, strength, and
a patient with androgen insensitivity syndrome with sexual function in men. N Engl J Med. 2013;369:
a previously unreported androgen receptor muta- 2457.
tion. Andrologia. 2014;46:703–6. 193. Sharpe RM. Hormones and testis development and
179. Xu HY, Li CD, Tang LL, Wang LL, Yu X, Gu XM, the possible adverse effects of environmental
et al. An infertile man with gynecomastia caused by chemicals. Toxicol Lett. 2001;120:221–32.
a novel mutation of the androgen receptor gene. 194. Stahl PJ, Stember DS, Goldstein M. Contemporary
Asian J Androl. 2015;17(3):509–10. management of male infertility. Annu Rev Med.
180. Petroli RJ, Hiort O, Struve D, Maciel-Guerra AT, 2012;63:525–40.
Guerra-Junior G, Palandi de Mello M, et al. 195. Song R, Hennig GW, Wu Q, Jose C, Zheng H,
Preserved fertility in a patient with gyknecomastia Yan W. Male germ cells express abundant endoge-
associated with the p.Pro695Ser mutation in the nous siRNAs. Proc Natl Acad Sci USA. 2011;108:
androgen receptor. Sex Dev. 2014;8:350–5. 13159–131564.
181. Carreau S, Wolczynski S, Galeraud-Denis I. Aro- 196. Song R, Ro S, Michaels JD, Park C, McCarrey JR,
matase, estrogens and human male reproduction. Phil Yan W. Many X-linked microRNAs escape meiotic
Trans R Soc Lond B Biol Sci. 2010;365:1571–9. sex chromosome inactivation. Nat Genet.
182. Hess RA. Estrogen in the adult male reproductive 2009;41:488–93.
tract: A review. Reprod Biol Endocrinol. 2003;1 197. Holt JE, Stanger SJ, Nixon B, McLaughlin EA.
(52):1–14. Non-coding RNA in spermatogenesis and epididy-
183. Zhou Q, Nie R, Prins GS, Saunders PTK, Katzenel- mal maturation. Adv Exp Med Biol. 2016;886:95–
lenbogen BS, Hess RA. Localization of androgen 120.

sjwint01@louisville.edu
3 Human Spermatogenesis and Its Regulation 71

198. de Mateo S, Sassone-Corsi P. Regulation of sper- from humans to animal models. Hum Reprod
matogenesis by small non-coding RNAs: role of the Update. 2011;17(5):667–83.
germ granule. Semin Cell Dev Biol. 2014;29:84– 216. Ramlau-Hansen CH, Hansen M, Jensen CR,
92. Olsen JC, Bonde JP, Thulstrup AM. Semen quality
199. Inui M, Martello G, Piccolo S. MicroRNA control and reproductive hormones according to birth-
of signal transduction. Nat Rev Mol Cell Biol. weight and body mass index in childhood and adult
2010;11:252–63. lief: two decades of follow-up. Fertil Sertil.
200. Eisenberg I, Kotaja N, Goldman-Wohl D, Imbar T. 2010;94:610–8.
microRNA in Human Reproduction. Adv Exp Med 217. de Boer H, Verschoor L, Ruinemans-Koerts J,
Biol. 2015;888:353–87. Jansen M. Letrozole normalizes serum testosterone
201. Bartel DP. MicroRNAs: genomics, biogenesis, in severely obese men with hypogonadotropic
mechanism, and function. Cell. 2004;116:281–97. hypogonadism. Diabetes Obesity Metabol.
202. Brodersen P, Voinnet O. Revisiting the principles of 2005;7:211–5.
microRNA target recognition and mode of action. 218. Schneider G, Kirschner MA, Berkowitz R,
Nat Rev Mol Cell Biol. 2009;10:141–8. Ertel NH. Increased estrogen production in obese
203. Meikar O, Da Ros M, Korhonen H, Kataja N. men. J Clin Endocrinol Metab. 1979;48:633–8.
Chromatoid body and small RNAs in male germ 219. Ghosh S, Ashcraft K, Jahid MJ, April C, Gha-
cells. Reproduction. 2011;142:195–209. jar CM, Ruan J, et al. Regulation of adipose
204. Yadav RP, Kotaja N. Small RNAs in spermatoge- oestrogen output by mechanical stress. Nat Com-
nesis. Mol Cell Endocrinol. 2014;382(1):498–508. mun. 2013;4:1821.
205. Lian J, Zhang X, Tian H, Liang N, Wang Y, 220. Dhindsa S, Batra M, Kuhadiya N, Dan-
Liang C, et al. Altered microRNA expression in dona P. Oestradiol concentrations are not elevated
patients with non-obstructive azoospermia. Reprod in obesity-associated hypogonadotrophic hypogo-
Biol Endocrinol. 2009;7:13. nadism. Clin Endocrinol. 2014;80(3):464.
206. Wu W, Qin Y, Li Z, Dong J, Dai J, Lu C, et al. 221. Burger HG. Physiological principles of endocrine
Genome-wide microRNA expression profiling in replacement: estrogen. Horm Res. 2001;56(Suppl
idiopathic non-obstructive azoospermia: significant 1):82–5.
up-regulation of miR-141, miR-429 and 222. Gutorova NV, Kleshchyov MA, Tipisova EV,
miR-7-1-3p. Hum Reprod. 2013;28(7):1827–36. Osadchuk LV. Effects of overweight and obesity
207. Wang C, Yang C, Chen X, Yao B, Yang C, Zhu C, on the spermogram values and levels of reproduc-
et al. Altered profile of seminal plasma microRNAs tive hormones in the male population of the
in the molecular diagnosis of male infertility. Clin European north of Russia. Bull Exp Biol Med.
Chem. 2011;57(12):1722–31. 2014;157:95–8.
208. Korhonen HM, Meikar O, Yadav RP, Papaioan- 223. Giagulli VA, Kaufman JM, Vermeulen A. Patho-
nou MD, Romero Y, Da Ros M, et al. Dicer is genesis of the decreased androgen levels in obese
required for haploid male germ cell differentiation men. J Clin Endocrinol Metab. 1994;79:997–1000.
in mice. PLoS ONE. 2011;6(9):e24821. 224. Veldhuis J, Yang R, Roelfsema F, Taka-
209. Korhonen HM, Yadav RP, Da Ros M, Chalmel F, hashi P. Proinflammatory Cytokine Infusion Atten-
Zimmermann C, Toppari J, et al. DICER regulates uates LH’s Feedforward on Testosterone Secretion:
the formation and maintenance of cell-cell Junctions Modulation by Age. J Clin Endocrinol Metab.
in the mouse seminiferous epithelium. Biol Reprod. 2016;101(2):539–49.
2015;93(6):139. 225. Mah PM, Wittert GA. Obesity and testicular
210. Papaioannou MD, Pitetti JL, Ro S, Park C, Aubry F, function. Mol Cell Endocrinol. 2010;316:180–6.
Schaad O, et al. Sertoli cell Dicer is essential for 226. Nielsen TL, Hagen C, Wraae K, Brixen K,
spermatogenesis in mice. Dev Biol. 2009;326 Petersen PH, Haug E, et al. Visceral and subcuta-
(1):250–9. neous adipose tissue assessed by magnetic reso-
211. Winters SJ. Monitoring testosterone levels in nance imaging in relation to circulating androgens,
testosterone-treated men. Curr Med Res Opin. sex hormone-binding globulin, and luteinizing hor-
2016;32(2):271–2. mone in young men. The Journal of clinical
212. Kiess W, Wagner IV, Kratzsch J, Korner A. Male endocrinology and metabolism. 2007;92(7):2696–
obesity. Endocrinol Metab Clin North Am. 705.
2015;44:761–72. 227. Landry D, Cloutier F, Martin LJ. Implications of
213. Chambers TJG, Anderson RA. The impact of leptin in neuroendocrine regulation of male repro-
obesity on male fertility. Hormones. 2015;14:563–8. duction. Reprod Biol Endocrinol. 2013;13:1–14.
214. Stokes VJ, Anderson RA, George JT. How does 228. Luboshitzky R, Lavie L, Shen-Orr Z, Herer O.
obesity affect fertility in men—and what are the Altered luteinizing hormone and testosterone secre-
treatment options? Clin Endocrinol. 2015;82 tion in middle-aged obese men with obstructive
(5):633–8. sleep apnea. Obes Res. 2005;13:780–6.
215. Teerds KJ, de Rooij DG, Keijer J. Functional 229. Ec Tsai. Matsumoto AM, Fujimoto WY,
relationship between obesity and male reproduction: Boyko EJ. Association of bioavailable, free, and

sjwint01@louisville.edu
72 H. Chen et al.

total testosterone with insulin resistance: Influence 237. di Frega AS, Dale B, Di Matteo L, Wilding M.
of sex hormone-bindign globulin and body fat. Secondary male factor infertility after Roux-en-Y
Diabetes Care. 2004;27:861–8. gastric bypass for morbid obesity: case report.
230. Hammoud AO, Gibson M, Peterson CM, Hamil- Human Reprod. 2005;20:997–8.
ton BD, Carrell DT. Obesity and male reproductive 238. Lazaros L, Hatzi E, Markoula S, Takenaka A,
potential. J Androl. 2006;27:619–26. sofikitis N, Zikopoulos K, et al. Dramatic reduction
231. Rato L, Alves MG, Cavaco JE, Oliveeira PF. in sperm parameters following bariatric surgery:
High-energy diets: a threat for male fertility? report of two cases. Andrologia. 2012;44:428–32.
Obesity Rev. 2014;15:996–1007. 239. Legro RS, Kunselman AR, Meadows JW, Kes-
232. Samavat J, Natali I, Degl’Innocenti S, filimberti E, ner JS, Krieg EFJ, Rogers AM, et al. Time-related
Cantini G, Di Franco A, et al. Acrosome reaction is increase in urinary testosterone levels and stable
impaired in spermatozoa of obese men: a prelimi- semen analysis parameters after bariatric surgery in
nary study. Fertil Steril. 2014;102:1274–81. men. Reprod Biomed Online. 2015;30:150–6.
233. Palmer NO, Bakos HW, Fullston T, Lane M. Impact 240. Kawakami S, Winters SJ. Regulation of leutinizing
of obesity on male fertility, sperm function and hormone secretion and subunit messenger ribonu-
molecular composition. Spermatogenesis. 2012;2 cleic acid expression by gonadal steroids in peri-
(4):253–63. fused pituitary cells from male monkeys and rats.
234. Feldman HA, Johannes CB, Derby CA, Klein- Endocrinology. 1999;140:3587–93.
man KP, Mohr BA, Araujo AB, et al. Erectile 241. Nakagawa T, Sharma M, Nabeshima Y, Braun RE,
dysfunction and coronary risk factors: prospective Yoshida S. Functional hierarchy and reversibility
results from the Massachusetts male aging study. within the murine spermatogenic stem cell com-
Prev Med. 2000;30:328–38. partment. Science. 2010;328(5974):62–7.
235. Lucchese M, Maggi M. Hypogonadism as a new 242. Heller CG, Clermont Y. Kinetics of the germinal
comorbidity in male patient’s selection for bariatric epithelium in man. Recent Prog Horm Res.
surgery: towards an extended concept of metabolic 1964;20:545–75.
surgery? Obes Surg. 2013;23:2018–9. 243. Muciaccia B, Boitani C, Berloco BP, Nudo F,
236. Hammoud A, Gibson M, Hunt SC, Adams TD, Spadetta G, Stefanini M, et al. Novel stage classi-
Carrell DT, Kolotkin RL, et al. Effect of Roux-en-Y fication of human spermatogenesis based on acro-
gastric bypass surgery on the sex steroids and some development. Biol Reprod. 2013;89(3):60.
quality of life in obese men. J Clin Endocrinol
Metab. 2009;94(4):1329–32.

sjwint01@louisville.edu
View publication stats

You might also like