SlideShare a Scribd company logo
1 of 11
Download to read offline
See	discussions,	stats,	and	author	profiles	for	this	publication	at:	https://www.researchgate.net/publication/285619565
Causes	and	consequences	of	oxidative	stress	in
spermatozoa
Article		in		Reproduction	Fertility	and	Development	·	January	2016
DOI:	10.1071/RD15325
CITATIONS
7
READS
566
5	authors,	including:
Some	of	the	authors	of	this	publication	are	also	working	on	these	related	projects:
small	noncoding	RNAs	in	the	male	reproductive	system	View	project
Mammalian	Sperm	Membrane	Protein	Complexes	View	project
Zamira	Gibb
University	of	Newcastle
29	PUBLICATIONS			266	CITATIONS			
SEE	PROFILE
Mark	A	Baker
University	of	Newcastle
97	PUBLICATIONS			3,491	CITATIONS			
SEE	PROFILE
Joel	R	Drevet
Université	Clermont	Auvergne
115	PUBLICATIONS			2,621	CITATIONS			
SEE	PROFILE
Parviz	Gharagozloo
CellOxess	Biotechnology
27	PUBLICATIONS			682	CITATIONS			
SEE	PROFILE
All	content	following	this	page	was	uploaded	by	Parviz	Gharagozloo	on	07	December	2015.
The	user	has	requested	enhancement	of	the	downloaded	file.	All	in-text	references	underlined	in	blue	are	added	to	the	original	document
and	are	linked	to	publications	on	ResearchGate,	letting	you	access	and	read	them	immediately.
Causes and consequences of oxidative stress
in spermatozoa
Robert John AitkenA,D
, Zamira GibbA
, Mark A. BakerA
, Joel DrevetB
and Parviz GharagozlooC
A
Priority Research Centre in Reproductive Science and Hunter Medical Research Institute,
Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia.
B
GReD laboratory, CNRS UMR6293-INSERM U1103-Clermont Universite´, 63171 BP80006,
Aubie`re cedex, France.
C
CellOxess LLC, 15 Roszel Street, Princeton, NJ 08540, USA.
D
Corresponding author. Email: john.aitken@newcastle.edu.au
Abstract. Spermatozoa are highly vulnerable to oxidative attack because they lack significant antioxidant protection
due to the limited volume and restricted distribution of cytoplasmic space in which to house an appropriate armoury of
defensive enzymes. In particular, sperm membrane lipids are susceptible to oxidative stress because they abound
in significant amounts of polyunsaturated fatty acids. Susceptibility to oxidative attack is further exacerbated by the
fact that these cells actively generate reactive oxygen species (ROS) in order to drive the increase in tyrosine
phosphorylation associated with sperm capacitation. However, this positive role for ROS is reversed when spermatozoa
are stressed. Under these conditions, they default to an intrinsic apoptotic pathway characterised by mitochondrial ROS
generation, loss of mitochondrial membrane potential, caspase activation, phosphatidylserine exposure and oxidative
DNA damage. In responding to oxidative stress, spermatozoa only possess the first enzyme in the base excision repair
pathway, 8-oxoguanine DNA glycosylase. This enzyme catalyses the formation of abasic sites, thereby destabilising the
DNA backbone and generating strand breaks. Because oxidative damage to sperm DNA is associated with both
miscarriage and developmental abnormalities in the offspring, strategies for the amelioration of such stress, including
the development of effective antioxidant formulations, are becoming increasingly urgent.
Additional keywords: apoptosis, fertilizing potential, lipid peroxidation, male germ line, oxidative DNA damage,
ROS generation.
Introduction
Traditionally, spermatozoa are regarded as highly specialised
cells that have but one function in life: to achieve fertilisation
and deliver the paternal component of the embryonic genome
to an MII oocyte. Although defective sperm function has long
been recognised as a major cause of human infertility (Hull
et al. 1985), this condition has conventionally been equated
with the ability of spermatozoa to achieve fertilisation (Aitken
et al. 1987; Aitken 2006). With the passage of time, we have
come to understand that the functional competence of sper-
matozoa cannot be defined merely in terms of the ability of
these cells to fertilise an oocyte; it also needs to incorporate an
assessment of their ability to program a normal pattern of
embryonic development. Spermatozoa may affect embryonic
development via both genetic and a variety of epigenetic
mechanisms involving the methylation profile of the DNA,
the post-translational modification of nuclear histones and the
composition of a variety of coding and non-coding RNA
species that are integrated into these cells, to find ultimate
expression in the zygote and early embryo (Aitken 1999;
Ostermeier et al. 2005; Prescott et al. 2012; Hosken and
Hodgson 2014; Metzler-Guillemain et al. 2015; Soubry 2015).
These sperm-borne epigenetic marks are, in turn, affected
by a variety of paternal factors, including genotype, age,
obesity, smoking and exposure to environmental contaminants
(Aitken 2014).
The mechanisms by which such environmental and lifestyle
factors affect mammalian spermatozoa, to define both their
potential for fertilisation and the subsequent initiation of embry-
onic development, are poorly understood. The central hypothe-
sis outlined in this article is that all such factors ultimately
converge to induce a high level of oxidative stress in the male
germline. Oxidative stress is known to interfere with the
fertilising capacity of spermatozoa, to damage sperm nuclear
DNA and to affect the epigenetic profile of these cells (Aitken
et al. 2014b). Herein, we review the evidence relating to the
origins and consequences of such stress and consider potential
strategies for its remediation.
CSIRO PUBLISHING
Reproduction, Fertility and Development, 2016, 28, 1–10
http://dx.doi.org/10.1071/RD15325
Journal compilation Ó IETS 2016 www.publish.csiro.au/journals/rfd
Oxidative stress and fertilisation potential
The notion that sperm function may be compromised by the
onset of oxidative stress can be traced back to the early studies of
Evans (1947), who observed that heavily irradiated seawater
impaired the fertilising capacity of sea urchin spermatozoa. He
concluded that the irradiation process had generated hydrogen
peroxide (H2O2) and that this powerful oxidising agent was
damaging to spermatozoa. Although this conclusion was not
subsequently supported by Barron et al. (1949), these authors
did generate unequivocal data indicating that H2O2 is extremely
damaging to sperm function. Around the same time, Tosic and
Walton (1946) demonstrated that the metabolite generated by
bovine spermatozoa in the presence of egg yolk-based cryo-
preservatives was H2O2 and that this oxidant actively sup-
pressed their respiration. Furthermore, these authors identified
the source of the H2O2 to be an L-amino acid oxidase with an
affinity for aromatic amino acids, particularly phenylalanine,
which is abundant in egg yolk (Tosic and Walton 1950).
MacLeod (1943) also demonstrated that human spermatozoa
lost motility at high oxygen tensions via mechanisms that could
be reversed by catalase, again suggesting that H2O2 generation
was causally involved in the loss of sperm function. The par-
ticular destructive power of H2O2 relative to any other reactive
oxygen species (ROS) was later emphasised in studies revealing
that catalase, but not superoxide dismutase, was able to relieve
the detrimental effect of ROS generated by the xanthine oxidase
free radical-generating system on human sperm motility in vitro
(Aitken et al. 1993a).
The possibility that excess ROS generation may be associated
with defective sperm function in vivo was highlighted by two
papers that appeared in 1987 and demonstrated that the sperma-
tozoa of infertile males were characterised by high levels of ROS
generation and the induction of lipid peroxidation (Aitken and
Clarkson 1987; Alvarez et al. 1987). The susceptibility of human
spermatozoa to lipid peroxidation had previously been highlight-
ed by Thaddeus Mann (Jones et al. 1979), who pointed out that
these cells contain exceptionally high levels of polyunsaturated
fatty acids (PUFA; particularly docosahexanoic acid), which are
vulnerable to free radical attack, generating lipid peroxides and
aldehydes that have a direct inhibitory action on sperm move-
ment. These early studies have subsequently been confirmed
in many independent laboratories, all of which agree on the
fundamental tenet that defective sperm function is frequently
induced by oxidative stress, affecting the motility of these cells,
their DNA integrity and their competence for sperm–oocyte
fusion (e.g. Aitken et al. 1991, 2010; Zalata et al. 1995; Sanocka
et al. 1996; Sharma and Agarwal 1996; Nakamura et al. 2002;
Kao et al. 2008; Sakamoto et al. 2008; Bejarano et al. 2014;
Morielli and O’Flaherty 2015).
Oxidative stress and lipid peroxidation
The way in which oxidative stress suppresses sperm motility
appears to be directly related to the induction of lipid per-
oxidation. When ROS attack the PUFA that abound in human
spermatozoa, a variety of lipid metabolites is generated,
including lipid peroxyl radicals, alkoxyl radicals and various
aldehydes, such as malondialdehyde, 4-hydroxynonenal (4HNE)
and acrolein (Jones et al. 1978; Moazamian et al. 2015). The
addition of both lipid peroxides and lipid aldehydes to popula-
tions of human spermatozoa results in the rapid immobilisation
of these cells via different mechanisms. Lipid peroxyl radicals
destabilise the sperm plasma membrane by virtue of their ten-
dency to abstract hydrogen atoms from adjacent PUFA to
achieve a measure of stabilisation as the corresponding lipid
hydroperoxide. This process creates carbon-centred lipid radi-
cals that combine with oxygen to generate more peroxyl radi-
cals, which, in turn, abstract hydrogen from adjacent PUFA to
stabilise, generating additional lipid radicals and promoting the
propagation of the lipid peroxidation chain reaction (Fig. 1a).
The lipid peroxides generated in this process destabilise the
plasma membrane by becoming targets for phospholipase A2,
which moves into the plasma membranes to cleave out the lipid
peroxides so they can be further processed by glutathione per-
oxidase (van Kuijk et al. 1985). This process, in turn, generates
lysophospholipids that destabilise the sperm plasma membrane,
affecting the microarchitecture of this structure and changing
the functions of integral membrane proteins that are critical to
the maintenance of sperm motility, such as ATP-dependent ion
pumps and voltage-regulated ion channels (Nishikawa et al.
1989; Lundbaek and Andersen 1994). The disruptive effect of
peroxidative damage on lipid membrane architecture also
affects the ability of spermatozoa to participate in the membrane
fusion events associated with fertilisation (Aitken et al. 1989,
1993b, 1993c).
The lipid peroxidation chain reactions initiated in spermato-
zoa may also result in the formation of a cascade of aldehyde
by-products that include alkanals, such as malondialdehyde, and
alkenals, such as 4HNE and acrolein. These compounds, partic-
ularly 4HNE and acrolein, are powerful electrophiles that form
adducts with several proteins within the spermatozoa that, in
turn, affect sperm function. For example, the formation of
adducts with the flagellar axonemal protein, dynein heavy chain,
may explain the effect of these aldehydes on sperm movement
(Baker et al. 2015; Moazamian et al. 2015). In addition, 4HNE
has been shown to bind to mitochondrial proteins in human
spermatozoa, triggering electron leakage and the formation of
ROS (Fig. 1b). The oxidative stress associated with the latter
then forces the spermatozoa to enter the intrinsic apoptotic
cascade, beginning with a loss of mitochondrial membrane
potential and terminating in oxidative DNA adduct formation,
DNA strand breakage and cell death (Aitken et al. 2012).
Sources of ROS and oxidative stress in spermatozoa
With oxidative stress being such a major factor in the aetiology
of defective human sperm function, resolving the possible
causes of this condition is critical. In considering this matter, it is
important to emphasise that spermatozoa are not only vulnerable
to oxidative stress because of the targets they offer for free
radical attack in the form of PUFA, proteins and nucleic acids,
but they are also lacking significant intracellular antioxidant
protection, including ROS-metabolising enzymes, such as
catalase and glutathione peroxidase, by virtue of the limited
volume and restricted distribution of cytoplasmic space in which
to house such mediators of cell survival. Furthermore, these
2 Reproduction, Fertility and Development R. J. Aitken et al.
cells actively generate physiological levels of ROS in order to
drive the tyrosine phosphorylation events associated with sperm
capacitation (Aitken and Nixon 2013). The involvement of ROS
in the capacitation of mammalian spermatozoa has been
appreciated since the pioneering studies of Claude Gagnon in
the 1990s (de Lamirande and Gagnon 1993a). The ROS
responsible for sperm capacitation have been variously reported
as H2O2 (Bise et al. 1991; Aitken et al. 1995, 1996; Rivlin et al.
2004) superoxide anion (de Lamirande and Gagnon 1993b) and
the peroxynitrite radical generated by the reaction of superoxide
anion with another free radical species, namely nitric oxide
(Herrero et al. 2001; Rodriguez and Beconi 2009). In reality, the
interconversion of these various ROS and reactive nitrogen
species is very rapid and it is probable that several different
redox entities are involved in various aspects of the capacitation
process, including the suppression of tyrosine phosphatase
activity and the stimulation of cAMP generation (Aitken and
Nixon 2013). It has recently been hypothesised that the con-
tinued generation of ROS, particularly peroxynitrite, to achieve
capacitation ultimately overwhelms the limited antioxidant
defences of these cells and precipitates a state of apoptosis.
According to this concept, capacitation and the intrinsic apo-
ptotic cascade are the opposite ends of a metabolic continuum
driven by ROS (Aitken et al. 2015b).
If ROS are so important for sperm function, what is the
subcellular source of these molecules? In mammalian sperma-
tozoa there can be little doubt that the major sources of ROS are
the mitochondria. Human sperm mitochondria are particularly
active in the generation of ROS via mechanisms that are not
dependent on a loss of mitochondrial membrane potential
(Koppers et al. 2008). Activation of ROS generation at Complex
III was found to stimulate the rapid release of H2O2 into the
(a) PUFA
R
Free
radical
attack
H
O2
Initiation
OO •
R
Lipid radical
Mitochondrial ROS generation
Lipid radical
PUFA Peroxyl radical Lipid hydroperoxide
R
R
H
R
Hydrogen
abstraction
R
OOH
OO •
R
Peroxyl radical
Lipid aldehydes
4HNE/acrolein
(c)
H2O2
(b)
ϩ H2O
•
OH
•
•
Nucleus
Mitochondria
Adduction of
mitochondrial
proteins
Lipid
peroxidation
ϩ
Fig. 1. Oxidative stress in mammalian spermatozoa. (a) Spermatozoa are susceptible to oxidative stress because they contain high
concentrations of polyunsaturated fatty acids (PUFA). Free radical attack leads to the formation of lipid radicals that then combine with the
universal electron acceptor, oxygen, to generate a lipid peroxyl radical. In order to stabilise as a hydroperoxide, the latter extracts hydrogen
atoms from adjacent lipids, generating lipid radicals that then perpetuate the peroxidation cascade. (b) Lipid aldehydes generated as a
consequence of lipid peroxidation, such as 4-hydroxynonenal (4HNE), bind to mitochondrial proteins, including succinic acid
dehydrogenase and stimulate yet more free radical generation, further enhancing lipid peroxidation in a self-propagating cycle that
propels spermatozoa towards an apoptotic fate. (c) The unusual architecture of spermatozoa means that nucleases activated in the midpiece
cytoplasm, or released from the mitochondria, cannot enter the nuclear compartment. The only product of apoptosis that can pass from the
midpiece to the sperm head to damage the DNA is H2O2; this is why most DNA damage in spermatozoa is oxidative.
Oxidative stress in spermatozoa Reproduction, Fertility and Development 3
extracellular space, but no detectable peroxidative damage.
Conversely, the induction of ROS on the matrix side of
the inner mitochondrial membrane at Complex I resulted in
peroxidative damage to the midpiece and a loss of sperm
movement that could be prevented by the concomitant presence
of a-tocopherol (Koppers et al. 2008). Defective human sper-
matozoa spontaneously generate mitochondrial ROS in a man-
ner that is negatively correlated with motility (Koppers et al.
2008). Indeed, simultaneous measurement of total cellular ROS
with dihydroethidium indicated that 68% of the variability in
such measurements could be explained by differences in mito-
chondrial ROS production (Koppers et al. 2008).
Another potential source of ROS are the NADPH oxidase
enzymes (NOX), including the calcium-dependent NOX5,
which are known to be present in the spermatozoa of certain
species, including human (Ba´nfi et al. 2001), although other
species, such as the mouse, do not possess this enzyme. Expos-
ing spermatozoa to NADPH can trigger a redox response that is
detectable with the redox probe lucigenin and inhibitable by
diphenylene iodonium (DPI), a flavoprotein inhibitor (Aitken
et al. 1997; Vernet et al. 2001). However, this lucigenin-
dependent activity was subsequently shown to be due to the
direct enzymatic reduction of the probe by cytochrome P450
reductase (Baker et al. 2004) and cytochrome b5 reductase
(Baker et al. 2005) when the electron donors were NADPH
and NADH, respectively. In contrast, using luminol as a
ROS probe, clear evidence has been obtained for a calcium-
dependent increase in ROS generation, which is particularly
marked in the spermatozoa of infertile patients and potentially
reflective of an involvement of NOX5 in the aetiology of
defective sperm function (Aitken and Clarkson 1987). There
is even some evidence to suggest that NOX5 may be overrepre-
sented in the defective spermatozoa recovered from patients
exhibiting teratozoospermia (Ghani et al. 2013). However,
definitive proof that NOX5 is the source of ROS under such
circumstances is currently lacking. There is a possibility that the
calcium-dependent signals observed with unfractionated sperm
suspensions are the result of low-level leucocyte contamination
(Aitken and Clarkson 1987; Aitken et al. 1992). The ability
of the NOX inhibitor apocynin to suppress the ROS signals
generated by human sperm suspensions (Dona` et al. 2011) could
also be accounted for by leucocyte contamination because this
reagent prevents assembly of the key cytosolic components
of the NADPH oxidase system (p40phox
, p47phox
and p67phox
),
which is not necessary for NOX5 to be active. A detailed study
of the NOX species present in human spermatozoa is currently
lacking and the role of these enzymes in the creation of oxidative
stress within the germline remains unresolved. One possibility
that cannot be excluded is that the NOX enzymes present in
mammalian spermatozoa play no role at all in the regulation of
sperm function, but rather function much earlier in germ cell
production, controlling spermatogonial stem cell proliferation
(Morimoto et al. 2013).
Finally, L-amino acid oxidases with a particular affinity for
phenylalanine have been identified in bull, horse, human and
ram spermatozoa (Tosic and Walton 1946; Aitken et al. 2015a;
Houston et al. 2015). In the case of equine spermatozoa, which
are heavily dependent on oxidative phosphorylation (Gibb et al.
2014), the primary role for this amino acid oxidase may be to
support the energy metabolism of these cells through the
oxidative deamination of aromatic amino acids, generating keto
acids that are then processed by the sperm mitochondria.
However, in the case of human spermatozoa, oxidative phos-
phorylation appears to play a minor role in sperm metabolism
because these cells are largely dependent of glycolysis to meet
their energy needs (du Plessis et al. 2015). In these cells, the
L-amino acid oxidase (interleukin 4 induced protein 1, IL4I1)
seems to have acquired a new biological function in supplying
the redox drive to sperm capacitation (Houston et al. 2015).
Role of oxidative stress in DNA damage
One of the major complications associated with male infertility
is the presence of high levels of DNA damage in the sperma-
tozoa. Such damage can arise as a consequence of infertility
(Irvine et al. 2000; Aitken and Curry 2011) age (Singh et al.
2003) smoking (Fraga et al. 1996) antioxidant deficiency (Fraga
et al. 1991, 1996), obesity (Fariello et al. 2012) exposure to
infection (Reichart et al. 2000; Burrello et al. 2004), heat
(De Iuliis et al. 2009a; Santiso et al. 2012) acidic pH (Santiso
et al. 2012), metals, particularly transition metals such as iron
and copper (Aitken et al. 2014a), radiofrequency electromag-
netic radiation (De Iuliis et al. 2009a), ionising radiation (Singh
and Stephens 1998), environmental toxicants such as acrylam-
ide (Katen and Roman 2015), chemotherapeutic agents (Delbe`s
et al. 2010), air pollution, plasticisers, pesticides (Evenson and
Wixon 2005; O’Flaherty 2014) and chloracetanilide herbicides
such as alachlor (Grizard et al. 2007).
There can be little doubt that most of these factors affect the
integrity of sperm chromatin through the induction of oxidative
stress. Such stress results in the generation of oxidised DNA
base adducts such as 8-hydroxy-20
-deoxyguanosine (8OHdG),
particularly in areas of the genome that are not heavily prota-
minated (De Iuliis et al. 2009b; Noblanc et al. 2013). Spermato-
zoa only possess one enzyme in the base excision repair (BER)
pathway, 8-oxoguanine DNA glycosylase (OGG1). This glyco-
sylase is associated with the sperm nucleus and mitochondria
and can actively excise 8OHdG, releasing this base adduct into
the extracellular space. Remarkably, spermatozoa do not pos-
sess the downstream components of the BER pathway, namely
apurinic endonuclease 1 (APE1) and X-ray repair complement-
ing defective repair in Chinese hamster cells 1 (XRCC1). The
net result of this truncated DNA repair capacity is to generate
abasic sites at locations that have been affected by 8OHdG
formation. Such abasic sites destabilise the ribose–phosphate
backbone, leading to a b-elimination or a ring opening reaction
of the ribose unit and a consequential strand break. This type of
DNA chemistry has been identified as being central to the
initiation of cancer in other cell types. Therefore, oxidative
DNA base lesions are not only potentially mutagenic but,
importantly, also contribute indirectly to the DNA fragmenta-
tion observed in the patient population (Ohno et al. 2014).
An oxidative involvement in DNA damage to mammalian
spermatozoa has been observed in relation to infertility (Shen
and Ong 2000; Aitken et al. 2010), heat (De Iuliis et al. 2009a),
antioxidant deficiency (Fraga et al. 1991), age (Weir and
4 Reproduction, Fertility and Development R. J. Aitken et al.
Robaire 2007; Smith et al. 2013a), smoking (Fraga et al. 1991),
obesity (Bakos et al. 2011), radiofrequency electromagnetic
radiation (De Iuliis et al. 2009a), herbicides (Grizard et al.
2007), plasticisers (Erkekoglu et al. 2010; Zhou et al. 2010) and
chemotherapeutic agents (Ghosh et al. 2002). Indeed, it would
appear that most DNA damage in mammalian spermatozoa
is the result of an oxidative insult generated as a result of either
impaired antioxidant protection because of endogenous (e.g. age)
or exogenous (e.g. phthalate esters) factors or changes in the
redox status of spermatozoa because of internal (e.g. mitochon-
drial electron leakage) or external (e.g. radiation or alachlor)
influences. However, it is also undeniable that not every
spermatozoon afflicted with DNA damage shows signs of oxida-
tive stress. Under these conditions, it has been suggested that
nuclease-mediated DNA fragmentation must occur as a result of
spermatozoa defaulting to an apoptotic state rather than oxidative
stress (Muratori et al. 2015). This interesting hypothesis is
difficult to reconcile with the fact that apoptosis invariably
involves the induction of oxidative stress (Koppers et al. 2011),
so it isdifficult to imagine how these phenomena can beseparated
in vivo. A possible resolution of this dilemma is set out below.
Role of apoptosis in DNA damage
It is well known that testicular precursor germ cells can undergo
apoptosis as part of a physiological process designed to optimise
germ cell : Sertoli cell ratios and to bring a measure of quality
control to the spermatogenic process, ensuring that no defective
germ cells are allowed to differentiate into spermatozoa (Shukla
et al. 2012). Apoptosis may also occur during spermatogenesis
in response to adverse circumstances, including heat shock,
ionising radiation, growth factor deprivation and chemothera-
peutic agents. The apoptotic process is largely, but not exclu-
sively, targeted to spermatocytes and both the intrinsic
mitochondrial pathway and the extrinsic p53/Fas system have
been implicated as key modulators of this process (Boekelheide
2005; Lagos-Cabre´ and Moreno 2012). However, the focus of
this discussion is the spermatozoa.
Spermatozoa are highly differentiated, transcriptionally
silent cells that, by virtue of their inert nuclear constitution
and highly specialised architecture, cannot undergo apoptosis in
the conventional sense. Nevertheless, they can undergo a
truncated version of this process. One of the key features of
sperm cell biology is that we do not have to expend energy
searching for factors that will induce these cells to undergo
apoptosis. Rather, these cells are designed to undergo apoptosis;
it is their default position. Spermatozoa are the ultimate symbol
of disposable cell types; indeed, all these cells are destined to die
a lonely apoptotic death in the male or female tract. The
fortunate exceptions to this rule are the handful of individual
gametes that manage to fertilise an oocyte and, in so doing,
achieve potential immortality for the genotype they carry. When
apoptosis does eventually occur, it is generally the intrinsic
apoptotic cascade that is induced, mediated by the sperm
mitochondria. Although receptor-mediated extrinsic apoptosis
remains a theoretical possibility in spermatozoa, no ligands have
been convincingly described to date that are capable of eliciting
such a response in the fully differentiated gamete. There has
been a claim that bacterial lipopolysaccharide (LPS) can elicit
apoptosis in spermatozoa by interacting with Toll-like receptor
(TLR) 2 and TLR4 on the sperm surface (Fujita et al. 2011).
However these data have not yet been independently validated
and our research group has not yet been able to achieve apoptosis
using commercially available LPS (R. J. Aitken, unpubl. obs.).
As a result, our view is that the mature gamete has very little
capacity to activate the extrinsic apoptotic cascade, but is
extremely vulnerable to its intrinsic counterpart.
Spermatozoa are normally prevented from entering the
intrinsic apoptotic pathway by virtue of the continuing activity
of phosphatidylinositol 3-kinase (PI3K; Koppers et al. 2011). If
PI3K is inhibited, then the spermatozoa default to an apoptotic
cascade characterised by rapid loss of motility, generation of
ROS, caspase activation in the cytosol, annexin V binding to the
cell surface, cytoplasmic vacuolisation and oxidative DNA
damage. The anti-apoptotic action of PI3K appears to depend
on its ability to promote the phosphorylation of another kinase,
AKT, which, in turn, is responsible for phosphorylating anti-
apoptotic effector proteins such as Bcl-2-associated death pro-
moter (BAD). Phosphorylation of the latter is essential for BAD
to remain associated with its cytoplasmic keeper protein, 14-3-3.
However, dephosphorylation allows BAD to orchestrate an
apoptotic process that has many similarities with the intrinsic
apoptotic cascade observed in somatic cells. There are two
major points of difference between apoptosis in spermatozoa
and somatic cells, as follows:
1. Mammalian spermatozoa are structurally different from
somatic cells in that all the mitochondria and most of the
cytoplasm are compartmentalised in the mid-piece of the
cell, physically separated from the DNA in the sperm
nucleus. As a result, even if apoptosis is activated in these
cells, the endonucleases released from the mitochondria
(e.g. endonuclease G) or activated in the cytoplasm
(e.g. caspase-activated DNAse) are physically impeded from
attacking the sperm nucleus (Koppers et al. 2011). The only
element of the apoptotic cascade that can exit from the sperm
midpiece and penetrate the nuclear compartment is H2O2. It
is for this reason that most of the DNA damage present in
human spermatozoa appears to be oxidatively induced
(Fig. 1c; Aitken et al. 2010).
2. Spermatozoa are also characterised by a severely truncated
BER pathway, as discussed above, that stalls after OGG1 has
removed the oxidised base to create abasic sites that have to
be further processed by the oocyte following fertilisation.
One consequence of spermatozoa lacking the next enzyme
in this pathway, namely APE1, is that these cells cannot
create the 30
-OH termini that are required by the terminal
deoxyribonucleotidyl transferase-mediated dUTP–digoxigenin
nick end-labelling (TUNEL) assay. As a result, TUNEL is a
very insensitive methodology for assessing DNA damage in
spermatozoa. Under circumstances where DNA damage is
induced by, for example, exposure to H2O2, intracellular and
extracellular 8OHdG can be clearly detected in the affected
sperm suspension and DNA strand breakage can be detected
with the sperm chromatin structure assay (SCSA); however,
TUNEL signals are not apparent (Smith et al. 2013b).
Oxidative stress in spermatozoa Reproduction, Fertility and Development 5
The later do eventually appear when the cells are close to
death. At this point, it is possible that a DNAse does become
activated in the spermatozoa (Sotolongo et al. 2005). For the
reasons given above, such a nuclease would have to be
incorporated into the sperm chromatin, not released from
the mitochondria or activated in the cytoplasm. The nature of
this DNAse (topoisomerase? DNAse 1?) and its mechanisms
of activation are not known. It is possible such nuclease
activity may be activated by a rise in intracellular calcium as
cells die, because calcium-dependent nuclease activity has
been described in this cell type on several independent
occasions by independent groups (Sotolongo et al. 2005;
Sibirtsev et al. 2011).
Apoptosis or oxidative stress causes DNA damage
We can conclude from the foregoing discussion that there are
two mechanisms for damaging DNA in mammalian spermato-
zoa. It is now generally acknowledged that a wide variety of
different intrinsic and extrinsic factors converge to generate a
state of oxidative stress in the germline. Once such stress has
been initiated, it tends to become accentuated because the lipid
aldehydes generated during the peroxidative process bind to
proteins in the mitochondrial electron transport chain, particu-
larly succinic acid dehydrogenase, stimulating the generation of
yet more free radicals, more DNA damage and more lipid per-
oxidation to continue the downward spiral towards apoptosis
(Fig. 1b; Aitken et al. 2012). There is also an association
between oxidative DNA damage in the male germline and poor
chromatin protamination during spermiogenesis (De Iuliis et al.
2009b). This relationship may reflect a certain vulnerability
towards oxidative stress as a consequence of the failure of sperm
nuclear DNA to adequately compact. However, it may also be a
consequence of inadequate protamination, because these small,
basic proteins are thought to protect the DNA by acting as
sacrificial antioxidants and by chelating redox active metals
such as copper (Liang et al. 1999).
The relationship between oxidative stress and apoptosis is
complex. Clearly, spermatozoa do express the classical markers
of apoptosis, such as ROS generation, phosphatidylserine expo-
sure, caspase activation and DNA fragmentation (Koppers et al.
2011). If PI3K activity is inhibited with wortmannin, then
human spermatozoa rapidly default to the intrinsic apoptotic
cascade, displaying all of the above features, including high
levels of mitochondrial ROS generation (Koppers et al. 2011).
Therefore, entry of spermatozoa into the senescence-driven
apoptotic pathway as a consequence of compromised PI3K
activity inevitably results in an apoptotic cascade involving
the stimulation of mitochondrial ROS generation and the induc-
tion of oxidative DNA damage. Under these circumstances,
apoptosis and oxidative DNA damage are inextricably linked.
Similarly, when spermatozoa are exposed to xenobiotics such as
alachlor, the induction of apoptosis is inextricably linked with
the induction of oxidative stress (Grizard et al. 2007). Within the
infertile population, DNA damage is again associated with the
simultaneous appearance of oxidative stress and apoptosis
(Wang et al. 2003; Aitken et al. 2010) via mechanisms that
can be reversed by the sustained administration of antioxidants
such as melatonin (Bejarano et al. 2014). Similarly, cryostorage
leads to the induction of oxidatively driven DNA damage and
apoptosis that can be reversed by the presence of the antioxidant
quercetin (Zribi et al. 2012). However, there are occasional
circumstances where DNA damage can be visualised in the
absence of any evidence that the cells have been subjected to
oxidative stress (Muratori et al. 2015). Under these circum-
stances, it is possible that the apoptosis involves stimulation of a
DNAse that is integrated into the sperm chromatin and somehow
becomes activated when these cells are under stress.
Developmental consequences of oxidative damage
in the germ line
Whatever the causes of oxidative stress in the male germline,
there can be no doubt that this pathophysiological mechanism
leads to both impaired fertility and disrupted embryonic
development. At high levels of oxidative stress, fertilisation is
prevented because the damage to the sperm plasma membrane
impairs both the motility of these cells and their competence for
fusion with the oocyte. At lower levels of oxidative stress the
spermatozoa can retain their capacity for fertilisation while the
DNA in their nuclei is still oxidatively damaged (Aitken et al.
1998). The developmental consequences of fertilising eggs with
spermatozoa exhibiting oxidative DNA damage has been
explored using the glutathione peroxidase 5 (GPx5)-knockout
mouse. In this mouse model, the spermatozoa suffer from
oxidative areas as they descend the epididymis (Chabory et al.
2009). The level of stress experienced by these spermatozoa
does not impair their fertilising capacity, but does induce high
levels of oxidative DNA damage in the sperm nuclei. The
consequence of this damage can be seen in the developmental
status of the embryos when Gpx5-null males are mated with
wild-type females, because such unions are accompanied by a
significant increase in the incidence of miscarriage and devel-
opmental abnormalities (Chabory et al. 2009). In related studies
in which mouse spermatozoa were oxidatively damaged by
exposing them to H2O2, several developmental abnormalities
were observed in the offspring, including a delay in embryonic
development rates, a decrease in the ratio of inner cell mass cells
in the resulting blastocyst and a reduction in implantation rates
(Lane et al. 2014). Crown–rump length at Day 18 of gestation
was also reduced in offspring produced from H2O2-treated
spermatozoa. Female offspring from peroxide-treated sperma-
tozoa were smaller, became glucose intolerant and accumulated
increased levels of adipose tissue compared with control female
offspring. Interestingly, the male offspring phenotype was less
severe, with increases in fat depots only seen at 4 weeks of age,
which returned to control levels later in life (Lane et al. 2014).
Studies in primates (Burruel et al. 2013) and cattle (Simo˜es et al.
2013) have confirmed the effect of oxidative DNA damage in
spermatozoa on the developmental potential of fertilised ova.
Given the developmental significance of this oxidative sperm
DNA damage, it is important that strategies are developed to
reduce such pathological changes as a matter of good practice
in assisted conception programs and as a matter of good con-
science in couples contemplating parenthood by natural means.
The data accumulated to date are encouraging in that the
6 Reproduction, Fertility and Development R. J. Aitken et al.
oxidative damage in spermatozoa associated with obesity, for
example, can clearly be reversed by a combination of diet and
exercise (Palmer et al. 2012). The use of antioxidants has also
met with some success in treating idiopathic oxidative stress in
spermatozoa (Gharagozloo and Aitken 2011; Showell et al.
2014). The advantages of using an effective oral antioxidant
treatment are that, in cases where oxidative stress has been
diagnosed in the male germline, it should result in increases in
both the fertilising potential of human spermatozoa and their
genetic integrity. The disadvantage of using antioxidants, par-
ticularly as an empirical treatment for patients where there is no
evidence of oxidative stress, is that it may create a reductive
stress (Chen et al. 2013). We still await the results of randomised
double-blind cross-over trials to definitively establish the
therapeutic value of different antioxidant formulations in the
treatment of male patients exhibiting high levels of oxidative
DNA damage in their spermatozoa. Such studies are eagerly
anticipated.
References
Aitken, R. J. (1999). The Amoroso Lecture. The human spermatozoon: a cell
in crisis? J. Reprod. Fertil. 115, 1–7. doi:10.1530/JRF.0.1150001
Aitken, R. J. (2006). Sperm function tests and fertility. Int. J. Androl. 29,
69–75. doi:10.1111/J.1365-2605.2005.00630.X
Aitken, R. J. (2014). Age, the environment and our reproductive future:
bonking baby boomers and the future of sex. Reproduction 147, S1–S11.
doi:10.1530/REP-13-0399
Aitken, R. J., and Clarkson, J. S. (1987). Cellular basis of defective sperm
function and its association with the genesis of reactive oxygen species
by human spermatozoa. J. Reprod. Fertil. 81, 459–469. doi:10.1530/
JRF.0.0810459
Aitken, R. J., and Curry, B. J. (2011). Redox regulation of human sperm
function: from the physiological control of sperm capacitation to the
etiology of infertility and DNA damage in the germ line. Antioxid. Redox
Signal. 14, 367–381. doi:10.1089/ARS.2010.3186
Aitken, R. J., and Nixon, B. (2013). Sperm capacitation: a distant landscape
glimpsed but unexplored. Mol. Hum. Reprod. 19, 785–793. doi:10.1093/
MOLEHR/GAT067
Aitken, R. J., Thatcher, S., Glasier, A. F., Clarkson, J. S., Wu, F. C., and
Baird, D. T. (1987). Relative ability of modified versions of the hamster
oocyte penetration test, incorporating hyperosmotic medium or the
ionophore A23187, to predict IVF outcome. Hum. Reprod. 2, 227–231.
Aitken, R. J., Clarkson, J. S., and Fishel, S. (1989). Generation of reactive
oxygen species, lipid peroxidation, and human sperm function. Biol.
Reprod. 41, 183–197. doi:10.1095/BIOLREPROD41.1.183
Aitken, R. J., Irvine, D. S., and Wu, F. C. (1991). Prospective analysis of
sperm–oocyte fusion and reactive oxygen species generation as criteria
for the diagnosis of infertility. Am. J. Obstet. Gynecol. 164, 542–551.
doi:10.1016/S0002-9378(11)80017-7
Aitken, R. J., Buckingham, D., West, K., Wu, F. C., Zikopoulos, K., and
Richardson, D. W. (1992). Differential contribution of leucocytes and
spermatozoa to the generation of reactive oxygen species in the ejacu-
lates of oligozoospermic patients and fertile donors. J. Reprod. Fertil.
94, 451–462. doi:10.1530/JRF.0.0940451
Aitken, R. J., Buckingham, D., and Harkiss, D. (1993a). Use of a xanthine
oxidase free radical generating system to investigate the cytotoxic
effects of reactive oxygen species on human spermatozoa. J. Reprod.
Fertil. 97, 441–450. doi:10.1530/JRF.0.0970441
Aitken, R. J., Harkiss, D., and Buckingham, D. (1993b). Relationship
between iron-catalysed lipid peroxidation potential and human sperm
function. J. Reprod. Fertil. 98, 257–265. doi:10.1530/JRF.0.0980257
Aitken, R. J., Harkiss, D., and Buckingham, D. W. (1993c). Analysis of lipid
peroxidation mechanisms in human spermatozoa. Mol. Reprod. Dev. 35,
302–315. doi:10.1002/MRD.1080350313
Aitken, R. J., Paterson, M., Fisher, H., Buckingham, D. W., and van Duin, M.
(1995). Redox regulation of tyrosine phosphorylation in human sperma-
tozoa and its role in the control of human sperm function. J. Cell Sci. 108,
2017–2025.
Aitken, R. J., Buckingham, D. W., Harkiss, D., Paterson, M., Fisher, H., and
Irvine, D. S. (1996). The extragenomic action of progesterone on human
spermatozoa is influenced by redox regulated changes in tyrosine
phosphorylation during capacitation. Mol. Cell. Endocrinol. 117,
83–93. doi:10.1016/0303-7207(95)03733-0
Aitken, R. J., Fisher, H. M., Fulton, N., Gomez, E., Knox, W., Lewis, B., and
Irvine, S. (1997). Reactive oxygen species generation by human sper-
matozoa is induced by exogenous NADPH and inhibited by the flavo-
protein inhibitors diphenylene iodonium and quinacrine. Mol. Reprod.
Dev. 47, 468–482. doi:10.1002/(SICI)1098-2795(199708)47:4,468::
AID-MRD14.3.0.CO;2-S
Aitken, R. J., Gordon, E., Harkiss, D., Twigg, J. P., Milne, P., Jennings, Z.,
and Irvine, D. S. (1998). Relative impact of oxidative stress on the
functional competence and genomic integrity of human spermatozoa.
Biol. Reprod. 59, 1037–1046. doi:10.1095/BIOLREPROD59.5.1037
Aitken, R. J., De Iuliis, G. N., Finnie, J. M., Hedges, A., and McLachlan, R. I.
(2010). Analysis of the relationships between oxidative stress, DNA
damage and sperm vitality in a patient population: development of
diagnostic criteria. Hum. Reprod. 25, 2415–2426. doi:10.1093/
HUMREP/DEQ214
Aitken, R. J., Whiting, S., De Iuliis, G. N., McClymont, S., Mitchell, L. A.,
and Baker, M. A. (2012). Electrophilic aldehydes generated by sperm
metabolism activate mitochondrial reactive oxygen species generation
and apoptosis by targeting succinate dehydrogenase. J. Biol. Chem. 287,
33 048–33 060. doi:10.1074/JBC.M112.366690
Aitken, R. J., Finnie, J. M., Muscio, L., Whiting, S., Connaughton, H. S.,
Kuczera, L., Rothkirch, T. B., and De Iuliis, G. N. (2014a). Potential
importance of transition metals in the induction of DNA damage by
sperm preparation media. Hum. Reprod. 29, 2136–2147. doi:10.1093/
HUMREP/DEU204
Aitken, R. J., Smith, T. B., Jobling, M. S., Baker, M. A., and De Iuliis, G. N.
(2014b). Oxidative stress and male reproductive health. Asian J. Androl.
16, 31–38. doi:10.4103/1008-682X.122203
Aitken, J. B., Naumovski, N., Grupen, C. G., Gibb, Z., and Aitken, R. J.
(2015a). Characterization of an L-amino acid oxidase in equine sperma-
tozoa. Biol. Reprod. 92, 125. doi:10.1095/BIOLREPROD.114.126052
Aitken, R. J., Baker, M. A., and Nixon, B. (2015b). Are sperm capacitation
and apoptosis the opposite ends of a continuum driven by oxidative
stress? Asian J. Androl. 17, 633–639. doi:10.4103/1008-682X.153850
Alvarez, J. G., Touchstone, J. C., Blasco, L., and Storey, B. T. (1987).
Spontaneous lipid peroxidation and production of hydrogen peroxide
and superoxide in human spermatozoa. Superoxide dismutase as major
enzyme protectant against oxygen toxicity. J. Androl. 8, 338–348.
Baker, M. A., Krutskikh, A., Curry, B. J., McLaughlin, E. A., and Aitken,
R. J. (2004). Identification of cytochrome P450-reductase as the enzyme
responsible for NADPH-dependent lucigenin and tetrazolium salt reduc-
tion in rat epididymal sperm preparations. Biol. Reprod. 71, 307–318.
doi:10.1095/BIOLREPROD.104.027748
Baker, M. A., Krutskikh, A., Curry, B. J., Hetherington, L., and Aitken, R. J.
(2005). Identification of cytochrome-b5 reductase as the enzyme respon-
sible for NADH-dependent lucigenin chemiluminescence in human
spermatozoa. Biol. Reprod. 73, 334–342. doi:10.1095/BIOLREPROD.
104.037960
Baker, M. A., Weinberg, A., Hetherington, L., Villaverde, A. I., Velkov, T.,
Baell, J., and Gordon, C. P. (2015). Defining the mechanisms by which
the reactive oxygen species by-product, 4-hydroxynonenal, affects
Oxidative stress in spermatozoa Reproduction, Fertility and Development 7
human sperm cell function. Biol. Reprod. 92, 108. doi:10.1095/BIOL
REPROD.114.126680
Bakos, H. W., Mitchell, M., Setchell, B. P., and Lane, M. (2011). The effect
of paternal diet-induced obesity on sperm function and fertilization in a
mouse model. Int. J. Androl. 34, 402–410. doi:10.1111/J.1365-2605.
2010.01092.X
Ba´nfi, B., Molna´r, G., Maturana, A., Steger, K., Hegeduˆs, B., Demaurex, N.,
and Krause, K. H. (2001). A Ca(2þ)-activated NADPH oxidase in
testis, spleen, and lymph nodes. J. Biol. Chem. 276, 37 594–37 601.
doi:10.1074/JBC.M103034200
Barron, E. S. G., Flood, V., and Gasvoda, B. (1949). The effect of
hydrogen peroxide and of X-ray irradiated sea water on the respiration
of sea urchin sperm and eggs. Biol. Bull. 97, 51–56. doi:10.2307/
1538093
Bejarano, I., Monllor, F., Marchena, A. M., Ortiz, A., Lozano, G., Jime´nez,
M. I., Gaspar, P., Garcı´a, J. F., Pariente, J. A., Rodrı´guez, A. B., and
Espino, J. (2014). Exogenous melatonin supplementation prevents
oxidative stress-evoked DNA damage in human spermatozoa. J. Pineal
Res. 57, 333–339. doi:10.1111/JPI.12172
Bize, I., Santander, G., Cabello, P., Driscoll, D., and Sharpe, C. (1991).
Hydrogen peroxide is involved in hamster sperm capacitation in vitro.
Biol. Reprod. 44, 398–403. doi:10.1095/BIOLREPROD44.3.398
Boekelheide, K. (2005). Mechanisms of toxic damage to spermatogenesis.
J. Natl Cancer Inst. Monogr. 2005, 6–8. doi:10.1093/JNCIMONO
GRAPHS/LGI006
Burrello, N., Calogero, A. E., Perdichizzi, A., Salmeri, M., D’Agata, R., and
Vicari, E. (2004). Inhibition of oocyte fertilization by assisted reproduc-
tive techniques and increased sperm DNA fragmentation in the presence
of Candida albicans: a case report. Reprod. Biomed. Online 8, 569–573.
doi:10.1016/S1472-6483(10)61104-2
Burruel, V., Klooster, K. L., Chitwood, J., Ross, P. J., and Meyers, S. A.
(2013). Oxidative damage to rhesus macaque spermatozoa results in
mitotic arrest and transcript abundance changes in early embryos. Biol.
Reprod. 89, 72. doi:10.1095/BIOLREPROD.113.110981
Chabory, E., Damon, C., Lenoir, A., Kauselmann, G., Kern, H., Zevnik, B.,
Garrel, C., Saez, F., Cadet, R., Henry-Berger, J., Schoor, M., Gottwald,
U., Habenicht, U., Drevet, J. R., and Vernet, P. (2009). Epididymis
seleno-independent glutathione peroxidase 5 maintains sperm DNA
integrity in mice. J. Clin. Invest. 119, 2074–2085.
Chen, S. J., Allam, J. P., Duan, Y. G., and Haidl, G. (2013). Influence of
reactive oxygen species on human sperm functions and fertilizing
capacity including therapeutical approaches. Arch. Gynecol. Obstet.
288, 191–199. doi:10.1007/S00404-013-2801-4
De Iuliis, G. N., Newey, R. J., King, B. V., and Aitken, R. J. (2009a). Mobile
phone radiation induces reactive oxygen species production and
DNA damage in human spermatozoa in vitro. PLoS One 4, e6446.
doi:10.1371/JOURNAL.PONE.0006446
De Iuliis, G. N., Thomson, L. K., Mitchell, L. A., Finnie, J. M., Koppers, A.
J., Hedges, A., Nixon, B., and Aitken, R. J. (2009b). DNA damage in
human spermatozoa is highly correlated with the efficiency of chromatin
remodeling and the formation of 8-hydroxy-20
-deoxyguanosine, a marker
of oxidative stress. Biol. Reprod. 81, 517–524. doi:10.1095/BIOLRE
PROD.109.076836
de Lamirande, E., and Gagnon, C. (1993a). Human sperm hyperactivation
and capacitation as parts of an oxidative process. Free Radic. Biol. Med.
14, 157–166. doi:10.1016/0891-5849(93)90006-G
de Lamirande, E., and Gagnon, C. (1993b). A positive role for the superoxide
anion in triggering hyperactivation and capacitation of human sperma-
tozoa. Int. J. Androl. 16, 21–25. doi:10.1111/J.1365-2605.1993.
TB01148.X
Delbe`s, G., Hales, B. F., and Robaire, B. (2010). Toxicants and human sperm
chromatin integrity. Mol. Hum. Reprod. 16, 14–22. doi:10.1093/
MOLEHR/GAP087
Dona`, G., Fiore, C., Andrisani, A., Ambrosini, G., Brunati, A., Ragazzi, E.,
Armanini, D., Bordin, L., and Clari, G. (2011). Evaluation of correct
endogenous reactive oxygen species content for human sperm capacita-
tion and involvement of the NADPH oxidase system. Hum. Reprod. 26,
3264–3273. doi:10.1093/HUMREP/DER321
du Plessis, S. S., Agarwal, A., Mohanty, G., and van der Linde, M. (2015).
Oxidative phosphorylation versus glycolysis: what fuel do spermatozoa
use? Asian J. Androl. 17, 230–235. doi:10.4103/1008-682X.135123
Erkekoglu, P., Rachidi, W., Yuzugullu, O. G., Giray, B., Favier, A., Ozturk,
M., and Hincal, F. (2010). Evaluation of cytotoxicity and oxidative DNA
damaging effects of di(2-ethylhexyl)-phthalate (DEHP) and mono
(2-ethylhexyl)-phthalate (MEHP) on MA-10 Leydig cells and protection
by selenium. Toxicol. Appl. Pharmacol. 248, 52–62. doi:10.1016/
J.TAAP.2010.07.016
Evans, T. C. (1947). Effects of hydrogen peroxide produced in the medium
by radiation of spermatozoa of Arbacia punctulata. Biol. Bull. 92,
99–109. doi:10.2307/1538160
Evenson, D. P., and Wixon, R. (2005). Environmental toxicants cause sperm
DNA fragmentation as detected by the sperm chromatin structure assay
(SCSA). Toxicol. Appl. Pharmacol. 207(Suppl.), 532–537. doi:10.1016/
J.TAAP.2005.03.021
Fariello, R. M., Pariz, J. R., Spaine, D. M., Cedenho, A. P., Bertolla, R. P.,
and Fraietta, R. (2012). Association between obesity and alteration of
sperm DNA integrity and mitochondrial activity. BJU Int. 110, 863–867.
doi:10.1111/J.1464-410X.2011.10813.X
Fraga, C. G., Motchnik, P. A., Shigenaga, M. K., Helbock, H. J., Jacob, R. A.,
and Ames, B. N. (1991). Ascorbic acid protects against endogenous
oxidative DNA damage in human sperm. Proc. Natl Acad. Sci. USA
88, 11 003–11 006. doi:10.1073/PNAS.88.24.11003
Fraga, C. G., Motchnik, P. A., Wyrobek, A. J., Rempel, D. M., and Ames,
B. N. (1996). Smoking and low antioxidant levels increase oxidative
damage to sperm DNA. Mutat. Res. 351, 199–203. doi:10.1016/0027-
5107(95)00251-0
Fujita, Y., Mihara, T., Okazaki, T., Shitanaka, M., Kushino, R., Ikeda, C.,
Negishi, H., Liu, Z., Richards, J. S., and Shimada, M. (2011). Toll-like
receptors (TLR) 2 and 4 on human sperm recognize bacterial endotoxins
and mediate apoptosis. Hum. Reprod. 26, 2799–2806. doi:10.1093/
HUMREP/DER234
Ghani, E., Keshtgar, S., Habibagahi, M., Ghannadi, A., and Kazeroni, M.
(2013). Expression of NOX5 in human teratozoospermia compared to
normozoospermia. Andrologia 45, 351–356. doi:10.1111/AND.12023
Gharagozloo, P., and Aitken, R. J. (2011). The role of sperm oxidative stress
in male infertility and the significance of oral antioxidant therapy. Hum.
Reprod. 26, 1628–1640. doi:10.1093/HUMREP/DER132
Ghosh, D., Das, U. B., and Misro, M. (2002). Protective role of alpha-
tocopherol–succinate (provitamin-E) in cyclophosphamide induced tes-
ticular gametogenic and steroidogenic disorders: a correlative approach
to oxidative stress. Free Radic. Res. 36, 1209–1218. doi:10.1080/
1071576021000016472
Gibb, Z., Lambourne, S. R., and Aitken, R. J. (2014). The paradoxical
relationship between stallion fertility and oxidative stress. Biol. Reprod.
91, 77. doi:10.1095/BIOLREPROD.114.118539
Grizard, G., Ouchchane, L., Roddier, H., Artonne, C., Sion, B., Vasson, M. P.,
and Janny, L. (2007). In vitro alachlor effects on reactive oxygen species
generation, motility patterns and apoptosis markers in human spermato-
zoa.Reprod. Toxicol.23, 55–62.doi:10.1016/J.REPROTOX.2006.08.007
Herrero, M. B., de Lamirande, E., and Gagnon, C. (2001). Tyrosine nitration
in human spermatozoa: a physiological function of peroxynitrite, the
reaction product of nitric oxide and superoxide. Mol. Hum. Reprod. 7,
913–921. doi:10.1093/MOLEHR/7.10.913
Hosken, D. J., and Hodgson, D. J. (2014). Why do sperm carry RNA?
Relatedness, conflict, and control. Trends Ecol. Evol. 29, 451–455.
doi:10.1016/J.TREE.2014.05.006
8 Reproduction, Fertility and Development R. J. Aitken et al.
Houston, B., Curry, B., and Aitken, R. J. (2015). Human spermatozoa
possess an IL4I1 L-amino acid oxidase with a potential role in sperm
function. Reproduction 149, 587–596. doi:10.1530/REP-14-0621
Hull, M. G. R., Glazener, C. M. A., Kelly, N. J., Conway, D. I., Foster, P. A.,
Hunton, R. A., Coulson, C., Lambert, P. A., Watt, E. M., and Desai,
K. M. (1985). Population study of causes, treatment and outcome of
infertility. Br. Med. J. (Clin. Res. Ed.) 291, 1693–1697. doi:10.1136/
BMJ.291.6510.1693
Irvine, D. S., Twigg, J. P., Gordon, E. L., Fulton, N., Milne, P. A., and Aitken,
R. J. (2000). DNA integrity in human spermatozoa: relationships with
semen quality. J. Androl. 21, 33–44.
Jones, R., Mann, T., and Sherins, R. J. (1978). Adverse effects of peroxidized
lipid on human spermatozoa. Proc. R. Soc. Lond. B Biol. Sci. 201,
413–417. doi:10.1098/RSPB.1978.0053
Jones, R., Mann, T., and Sherins, R. J. (1979). Peroxidative breakdown of
phospholipids in human spermatozoa: spermicidal effects of fatty acid
peroxides and protective action of seminal plasma. Fertil. Steril. 31,
531–537.
Kao, S. H., Chao, H. T., Chen, H. W., Hwang, T. I., Liao, T. L., and Wei, Y. H.
(2008). Increase of oxidative stress in human sperm with lower motility.
Fertil. Steril. 89, 1183–1190. doi:10.1016/J.FERTNSTERT.2007.05.029
Katen, A. L., and Roman, S. D. (2015). The genetic consequences of paternal
acrylamide exposure and potential for amelioration. Mutat. Res. 777,
91–100. doi:10.1016/J.MRFMMM.2015.04.008
Koppers, A. J., De Iuliis, G. N., Finnie, J. M., McLaughlin, E. A., and Aitken,
R. J. (2008). Significance of mitochondrial reactive oxygen species in
the generation of oxidative stress in spermatozoa. J. Clin. Endocrinol.
Metab. 93, 3199–3207. doi:10.1210/JC.2007-2616
Koppers, A. J., Mitchell, L. A., Wang, P., Lin, M., and Aitken, R. J. (2011).
Phosphoinositide 3-kinase signalling pathway involvement in a
truncated apoptotic cascade associated with motility loss and oxidative
DNA damage in human spermatozoa. Biochem. J. 436, 687–698.
doi:10.1042/BJ20110114
Lagos-Cabre´, R., and Moreno, R. D. (2012). Contribution of environmental
pollutants to male infertility: a working model of germ cell apoptosis
induced by plasticizers. Biol. Res. 45, 5–14. doi:10.4067/S0716-
97602012000100001
Lane, M., McPherson, N. O., Fullston, T., Spillane, M., Sandeman, L.,
Kang, W. X., and Zander-Fox, D. L. (2014). Oxidative stress in mouse
sperm impairs embryo development, fetal growth and alters adiposity
and glucose regulation in female offspring. PLoS One 9, e100832.
doi:10.1371/JOURNAL.PONE.0100832
Liang, R., Senturker, S., Shi, X., Bal, W., Dizdaroglu, M., and Kasprzak,
K. S. (1999). Effects of Ni(II) and Cu(II) on DNA interaction with the
N-terminal sequence of human protamine P2: enhancement of binding
and mediation of oxidative DNA strand scission and base damage.
Carcinogenesis 20, 893–898. doi:10.1093/CARCIN/20.5.893
Lundbaek, J. A., and Andersen, O. S. (1994). Lysophospholipids modulate
channel function by altering the mechanical properties of lipid bilayers.
J. Gen. Physiol. 104, 645–673. doi:10.1085/JGP.104.4.645
MacLeod, J. (1943). The role of oxygen in the metabolism and motility of
human spermatozoa. Am. J. Physiol. 138, 512–518.
Metzler-Guillemain,C.,Victorero,G.,Lepoivre,C.,Bergon,A.,Yammine,M.,
Perrin, J., Sari-Minodier, I., Boulanger, N., Rihet, P., and Nguyen, C.
(2015). Sperm mRNAs and microRNAs as candidate markers for the
impact of toxicants on human spermatogenesis: an application to tobacco
smoking. Syst Biol Reprod Med 61, 139–149. doi:10.3109/19396368.
2015.1022835
Moazamian, R., Polhemus, A., Connaughton, H., Fraser, B., Whiting, S.,
Gharagozloo, P., and Aitken, R. J. (2015). Oxidative stress and human
spermatozoa: diagnostic and functional significance of aldehydes gen-
erated as a result of lipid peroxidation. Mol. Hum. Reprod. 21, 502–515.
doi:10.1093/MOLEHR/GAV014
Morielli, T., and O’Flaherty, C. (2015). Oxidative stress impairs function
and increases redox protein modifications in human spermatozoa.
Reproduction 149, 113–123. doi:10.1530/REP-14-0240
Morimoto, H., Iwata, K., Ogonuki, N., Inoue, K., Atsuo, O., Kanatsu-
Shinohara, M., Morimoto, T., Yabe-Nishimura, C., and Shinohara, T.
(2013). ROS are required for mouse spermatogonial stem cell self-
renewal. Cell Stem Cell 12, 774–786. doi:10.1016/J.STEM.2013.04.001
Muratori, M., Tamburrino, L., Marchiani, S., Cambi, M., Olivito, B.,
Azzari, C., Forti, G., and Baldi, E. (2015). Investigation on the origin
of sperm DNA fragmentation: role of apoptosis, immaturity and oxida-
tive stress. Mol. Med. 21, 109–122. doi:10.2119/MOLMED.2014.00158
Musset,B.,Clark,R.A.,DeCoursey,T.E.,Petheo,G.L.,Geiszt,M., Chen, Y.,
Cornell, J.E., Eddy, C. A., Brzyski,R. G., and El Jamali, A.(2012).NOX5
in human spermatozoa: expression, function, and regulation. J. Biol.
Chem. 287, 9376–9388. doi:10.1074/JBC.M111.314955
Nakamura, H., Kimura, T., Nakajima, A., Shimoya, K., Takemura, M.,
Hashimoto, K., Isaka, S., Azuma, C., Koyama, M., and Murata, Y.
(2002). Detection of oxidative stress in seminal plasma and fractionated
sperm from subfertile male patients. Eur. J. Obstet. Gynecol. Reprod.
Biol. 105, 155–160. doi:10.1016/S0301-2115(02)00194-X
Nishikawa, T., Tomori, Y., Yamashita, S., and Shimizu, S. (1989). Inhibition
of Naþ
,Kþ
-ATPase activity by phospholipase A2 and several lysopho-
spholipids: possible role of phospholipase A2 in noradrenaline release
from cerebral cortical synaptosomes. J. Pharm. Pharmacol. 41, 450–458.
doi:10.1111/J.2042-7158.1989.TB06499.X
Noblanc, A.,Damon-Soubeyrand,C., Karrich, B., Henry-Berger,J., Cadet,R.,
Saez, F., Guiton, R., Janny, L., Pons-Rejraji, H., Alvarez, J. G., Jr,
Drevet, J. R., and Kocer, A. (2013). DNA oxidative damage in mamma-
lian spermatozoa: where and why the male nucleus is impacted? Free
Radic. Biol. Med. 65, 719–723. doi:10.1016/J.FREERADBIOMED.
2013.07.044
O’Flaherty, C. (2014). Iatrogenic genetic damage of spermatozoa. Adv. Exp.
Med. Biol. 791, 117–135. doi:10.1007/978-1-4614-7783-9_8
Ohno, M.,Sakumi,K.,Fukumura, R.,Furuichi,M., Iwasaki,Y.,Hokama, M.,
Ikemura, T., Tsuzuki, T., Gondo, Y., and Nakabeppu, Y. (2014).
8-Oxoguanine causes spontaneous de novo germline mutations in mice.
Sci. Rep. 4, 4689. doi:10.1038/SREP04689
Ostermeier, G. C., Goodrich, R. J., Moldenhauer, J. S., Diamond, M. P., and
Krawetz, S. A. (2005). A suite of novel human spermatozoal RNAs.
J. Androl. 26, 70–74.
Palmer, N. O., Bakos, H. W., Owens, J. A., Setchell, B. P., and Lane, M.
(2012). Diet and exercise in an obese mouse fed a high-fat diet improve
metabolic health and reverse perturbed sperm function. Am. J. Physiol.
Endocrinol. Metab. 302, E768–E780. doi:10.1152/AJPENDO.00401.
2011
Prescott, J., Du, M., Wong, J. Y., Han, J., and De Vivo, I. (2012). Paternal age
at birth is associated with offspring leukocyte telomere length in the
Nurses’ Health Study. Hum. Reprod. 27, 3622–3631. doi:10.1093/
HUMREP/DES314
Reichart, M., Kahane, I., and Bartoov, B. (2000). In vivo and in vitro
impairment of human and ram sperm nuclear chromatin integrity by
sexually transmitted Ureaplasma urealyticum infection. Biol. Reprod.
63, 1041–1048. doi:10.1095/BIOLREPROD63.4.1041
Rivlin, J., Mendel, J., Rubinstein, S., Etkovitz, N., and Breitbart, H. (2004).
Role of hydrogen peroxide in sperm capacitation and acrosome reaction.
Biol. Reprod. 70, 518–522. doi:10.1095/BIOLREPROD.103.020487
Rodriguez, P. C., and Beconi, M. T. (2009). Peroxynitrite participates in
mechanisms involved in capacitation of cryopreserved cattle. Anim.
Reprod. Sci. 110, 96–107. doi:10.1016/J.ANIREPROSCI.2007.12.017
Sakamoto, Y., Ishikawa, T., Kondo, Y., Yamaguchi, K., and Fujisawa, M.
(2008). The assessment of oxidative stress in infertile patients with
varicocele. BJU Int. 101, 1547–1552. doi:10.1111/J.1464-410X.2008.
07517.X
Oxidative stress in spermatozoa Reproduction, Fertility and Development 9
Sanocka, D., Miesel, R., Jedrzejczak, P., and Kurpisz, M. K. (1996).
Oxidative stress and male infertility. J. Androl. 17, 449–454.
Santiso,R.,Tamayo,M.,Gosa´lvez,J.,Johnston,S.,Marin˜o,A.,Ferna´ndez,C.,
Losada, C., and Ferna´ndez, J. L. (2012). DNA fragmentation
dynamics allows the assessment of cryptic sperm damage in human:
evaluation of exposure to ionizing radiation, hyperthermia, acidic pH
and nitric oxide. Mutat. Res. 734, 41–49. doi:10.1016/J.MRFMMM.
2012.03.006
Sharma, R. K., and Agarwal, A. (1996). Role of reactive oxygen species in
male infertility. Urology 48, 835–850. doi:10.1016/S0090-4295(96)
00313-5
Shen, H., and Ong, C. (2000). Detection of oxidative DNA damage in
human sperm and its association with sperm function and male infertility.
Free Radic. Biol. Med. 28, 529–536. doi:10.1016/S0891-5849(99)
00234-8
Showell, M. G., Mackenzie-Proctor, R., Brown, J., Yazdani, A., Stankiewicz,
M. T., and Hart, R. J. (2014). Antioxidants for male subfertility. Cochrane
Database Syst. Rev. 12, CD007411.
Shukla, K. K., Mahdi, A. A., and Rajender, S. (2012). Apoptosis, spermato-
genesis and male infertility. Front. Biosci. (Elite Ed.) E4, 746–754.
doi:10.2741/E415
Sibirtsev, J. T., Shastina, V. V., Menzorova, N. I., Makarieva, T. N., and
Rasskazov, V. (2011). A Ca2þ
, Mg2þ
-dependent DNase involvement in
apoptotic effects in spermatozoa of sea urchin Strongylocentrotus
intermedius induced by two-headed sphingolipid, rhizochalin. Mar.
Biotechnol. (NY) 13, 536–543. doi:10.1007/S10126-010-9324-9
Simo˜es, R., Feitosa, W. B., Siqueira, A. F., Nichi, M., Paula-Lopes, F. F.,
Marques, M. G., Peres, M. A., Barnabe, V. H., Visintin, J. A., and
Assumpc¸a˜o, M. E. (2013). Influence of bovine sperm DNA fragmenta-
tion and oxidative stress on early embryo in vitro development outcome.
Reproduction 146, 433–441. doi:10.1530/REP-13-0123
Singh, N. P., and Stephens, R. E. (1998). X-Ray induced DNA double-
strand breaks in human sperm. Mutagenesis 13, 75–79. doi:10.1093/
MUTAGE/13.1.75
Singh, N. P., Muller, C. H., and Berger, R. E. (2003). Effects of age on DNA
double-strand breaks and apoptosis in human sperm. Fertil. Steril. 80,
1420–1430. doi:10.1016/J.FERTNSTERT.2003.04.002
Smith, T. B., De Iuliis, G. N., Lord, T., and Aitken, R. J. (2013a). The
senescence-accelerated mouse prone 8 as a model for oxidative stress
and impaired DNA repair in the male germ line. Reproduction 146,
253–262. doi:10.1530/REP-13-0186
Smith, T. B., Dun, M. D., Smith, N. D., Curry, B. J., Connaughton, H. S., and
Aitken, R. J. (2013b). The presence of a truncated base excision repair
pathway in human spermatozoa that is mediated by OGG1. J. Cell Sci.
126, 1488–1497. doi:10.1242/JCS.121657
Sotolongo, B., Huang, T. T., Isenberger, E., and Ward, W. S. (2005). An
endogenous nuclease in hamster, mouse, and human spermatozoa
cleaves DNA into loop-sized fragments. J. Androl. 26, 272–280.
Soubry, A. (2015). Epigenetic inheritance and evolution: a paternal per-
spective on dietary influences. Prog. Biophys. Mol. Biol. 118, 79–85.
doi:10.1016/J.PBIOMOLBIO.2015.02.008
Tosic, J., and Walton, A. (1946). Formation of hydrogen peroxide by
spermatozoa and its inhibitory effect on respiration. Nature 158, 485.
doi:10.1038/158485A0
Tosic, J., and Walton, A. (1950). Metabolism of spermatozoa. The formation
and elimination of hydrogen peroxide by spermatozoa and effects on
motility and survival. Biochem. J. 47, 199–212.
van Kuijk, F. J., Handelman, G. J., and Dratz, E. A. (1985). Consecutive
action of phospholipase A2 and glutathione peroxidase is required for
reduction of phospholipid hydroperoxides and provides a convenient
method to determine peroxide values in membranes. J. Free Radic. Biol.
Med. 1, 421–427. doi:10.1016/0748-5514(85)90156-4
Vernet, P., Fulton, N., Wallace, C., and Aitken, R. J. (2001). Analysis of
reactive oxygen species generating systems in rat epididymal spermato-
zoa. Biol. Reprod. 65, 1102–1113. doi:10.1095/BIOLREPROD65.4.1102
Wang, X., Sharma, R. K., Sikka, S. C., Thomas, A. J., Jr, Falcone, T., and
Agarwal, A. (2003). Oxidative stress is associated with increased
apoptosis leading to spermatozoa DNA damage in patients with male
factor infertility. Fertil. Steril. 80, 531–535. doi:10.1016/S0015-0282
(03)00756-8
Weir, C. P., and Robaire, B. (2007). Spermatozoa have decreased antioxi-
dant enzymatic capacity and increased reactive oxygen species produc-
tion during aging in the Brown Norway rat. J. Androl. 28, 229–240.
doi:10.2164/JANDROL.106.001362
Zalata, A., Hafez, T., Mahmoud, A., and Comhaire, F. (1995). Relationship
between resazurin reduction test, reactive oxygen species generation,
and gamma-glutamyltransferase. Hum. Reprod. 10, 1136–1140.
Zhou, D., Wang, H., Zhang, J., Gao, X., Zhao, W., and Zheng, Y. (2010).
Di-n-butyl phthalate (DBP) exposure induces oxidative damage in testes
of adult rats. Syst. Biol. Reprod. Med. 56, 413–419. doi:10.3109/
19396368.2010.509902
Zribi, N., Chakroun, N. F., Ben Abdallah, F., Elleuch, H., Sellami, A.,
Gargouri, J., Rebai, T., Fakhfakh, F., and Keskes, L. A. (2012). Effect of
freezing–thawing process and quercetin on human sperm survival and
DNA integrity. Cryobiology 65, 326–331. doi:10.1016/J.CRYOBIOL.
2012.09.003
www.publish.csiro.au/journals/rfd
10 Reproduction, Fertility and Development R. J. Aitken et al.
View publication statsView publication stats

More Related Content

What's hot

Reactive Oxygen Species in Signal Transduction and its applications
Reactive Oxygen Species in Signal Transduction and its applicationsReactive Oxygen Species in Signal Transduction and its applications
Reactive Oxygen Species in Signal Transduction and its applicationsMostafa Mohamed
 
Role of ascorbate peroxidase in the antioxidant protection
Role of ascorbate peroxidase in the antioxidant protectionRole of ascorbate peroxidase in the antioxidant protection
Role of ascorbate peroxidase in the antioxidant protectionBilal051
 
Role of Antioxidants in the Treatment of Male Infertility-Crimson Publishers
Role of Antioxidants in the Treatment of Male Infertility-Crimson PublishersRole of Antioxidants in the Treatment of Male Infertility-Crimson Publishers
Role of Antioxidants in the Treatment of Male Infertility-Crimson PublishersCrimsonpublisherssmoaj
 
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...Premier Publishers
 
Science_handout_GYV_BioGenesis
Science_handout_GYV_BioGenesisScience_handout_GYV_BioGenesis
Science_handout_GYV_BioGenesisTikiTsakiris
 
Nico wanandy unsw mechanism of antioxidant for the skin
Nico wanandy   unsw mechanism of antioxidant for the skinNico wanandy   unsw mechanism of antioxidant for the skin
Nico wanandy unsw mechanism of antioxidant for the skinNico Wanandy
 
ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake Jake Elwood
 
Ameliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedAmeliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedRam Sahu
 
Defence mechanism of antioxidant in Human Body
Defence mechanism of antioxidant in Human BodyDefence mechanism of antioxidant in Human Body
Defence mechanism of antioxidant in Human BodyImad Khan
 
ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake Jake Elwood
 
The elixir of life
The elixir of lifeThe elixir of life
The elixir of lifeRachel Jacob
 
PPT on Protective Activity Of Certain Important Antioxidant
PPT on Protective Activity Of Certain Important Antioxidant PPT on Protective Activity Of Certain Important Antioxidant
PPT on Protective Activity Of Certain Important Antioxidant Naveen K L
 
Antioxidants /certified fixed orthodontic courses by Indian dental academy
Antioxidants /certified fixed orthodontic courses by Indian dental academy Antioxidants /certified fixed orthodontic courses by Indian dental academy
Antioxidants /certified fixed orthodontic courses by Indian dental academy Indian dental academy
 
Peroxiredoxins guardians against oxidative stress
Peroxiredoxins guardians against oxidative stressPeroxiredoxins guardians against oxidative stress
Peroxiredoxins guardians against oxidative stressArya Prakash Panda
 

What's hot (20)

Reactive Oxygen Species in Signal Transduction and its applications
Reactive Oxygen Species in Signal Transduction and its applicationsReactive Oxygen Species in Signal Transduction and its applications
Reactive Oxygen Species in Signal Transduction and its applications
 
Role of ascorbate peroxidase in the antioxidant protection
Role of ascorbate peroxidase in the antioxidant protectionRole of ascorbate peroxidase in the antioxidant protection
Role of ascorbate peroxidase in the antioxidant protection
 
Superoxide Dismutase (Group 13)
Superoxide Dismutase (Group 13)Superoxide Dismutase (Group 13)
Superoxide Dismutase (Group 13)
 
Role of Antioxidants in the Treatment of Male Infertility-Crimson Publishers
Role of Antioxidants in the Treatment of Male Infertility-Crimson PublishersRole of Antioxidants in the Treatment of Male Infertility-Crimson Publishers
Role of Antioxidants in the Treatment of Male Infertility-Crimson Publishers
 
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...
Antioxidant potentials of tannic acid on lipid peroxidation induced by severa...
 
REJUVENATION
REJUVENATION REJUVENATION
REJUVENATION
 
pharmaceutics
pharmaceuticspharmaceutics
pharmaceutics
 
Science_handout_GYV_BioGenesis
Science_handout_GYV_BioGenesisScience_handout_GYV_BioGenesis
Science_handout_GYV_BioGenesis
 
Nico wanandy unsw mechanism of antioxidant for the skin
Nico wanandy   unsw mechanism of antioxidant for the skinNico wanandy   unsw mechanism of antioxidant for the skin
Nico wanandy unsw mechanism of antioxidant for the skin
 
ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake
 
Ameliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedAmeliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation induced
 
Defence mechanism of antioxidant in Human Body
Defence mechanism of antioxidant in Human BodyDefence mechanism of antioxidant in Human Body
Defence mechanism of antioxidant in Human Body
 
ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake ASBMB_Poster_3_25_2015_Jake
ASBMB_Poster_3_25_2015_Jake
 
Heat shock proteins
Heat shock proteinsHeat shock proteins
Heat shock proteins
 
Antioxidants
AntioxidantsAntioxidants
Antioxidants
 
The elixir of life
The elixir of lifeThe elixir of life
The elixir of life
 
PPT on Protective Activity Of Certain Important Antioxidant
PPT on Protective Activity Of Certain Important Antioxidant PPT on Protective Activity Of Certain Important Antioxidant
PPT on Protective Activity Of Certain Important Antioxidant
 
Antioxidants /certified fixed orthodontic courses by Indian dental academy
Antioxidants /certified fixed orthodontic courses by Indian dental academy Antioxidants /certified fixed orthodontic courses by Indian dental academy
Antioxidants /certified fixed orthodontic courses by Indian dental academy
 
Peroxiredoxins guardians against oxidative stress
Peroxiredoxins guardians against oxidative stressPeroxiredoxins guardians against oxidative stress
Peroxiredoxins guardians against oxidative stress
 
2012Brookins_TeQuion
2012Brookins_TeQuion2012Brookins_TeQuion
2012Brookins_TeQuion
 

Similar to Aitken 2016 causes and consequences of oxidative stress in spermatozoa

MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANI
MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANIMICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANI
MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANIDR SHASHWAT JANI
 
Boar semen cueva et al 2013
Boar semen cueva et al 2013Boar semen cueva et al 2013
Boar semen cueva et al 2013Jorge Parodi
 
Revision parodi 2013
Revision parodi 2013Revision parodi 2013
Revision parodi 2013Jorge Parodi
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdfHadgi1
 
Hallmarks of Aging summary
Hallmarks of Aging summaryHallmarks of Aging summary
Hallmarks of Aging summarySteve Cepa
 
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...Super Males: Antioxidants boost male fertility and sperm viability in fruit f...
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...Weily Lang
 
Revision parodi 2013
Revision parodi 2013Revision parodi 2013
Revision parodi 2013Jorge Parodi
 
2015_Muldoon_Brook Stickleback Androgens
2015_Muldoon_Brook Stickleback Androgens2015_Muldoon_Brook Stickleback Androgens
2015_Muldoon_Brook Stickleback AndrogensBreda Rahmanian
 
Nrf2: A Guardian of Healthspan and Gatekeeper of Species Longevity
Nrf2:  A Guardian of Healthspan and Gatekeeper of Species LongevityNrf2:  A Guardian of Healthspan and Gatekeeper of Species Longevity
Nrf2: A Guardian of Healthspan and Gatekeeper of Species LongevityLifeVantage
 
The developmental and physiological interactions between free radicals and an...
The developmental and physiological interactions between free radicals and an...The developmental and physiological interactions between free radicals and an...
The developmental and physiological interactions between free radicals and an...Alexander Decker
 
oxgen tension.pdf
oxgen tension.pdfoxgen tension.pdf
oxgen tension.pdftenawsisay
 
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...Premier Publishers
 
Oxidative Stress in Health & Disease: The therapeutic potential of Nrf2 acti...
Oxidative Stress in Health & Disease:  The therapeutic potential of Nrf2 acti...Oxidative Stress in Health & Disease:  The therapeutic potential of Nrf2 acti...
Oxidative Stress in Health & Disease: The therapeutic potential of Nrf2 acti...LifeVantage
 
Essay On Epigenetic Resetting In Plants
Essay On Epigenetic Resetting In PlantsEssay On Epigenetic Resetting In Plants
Essay On Epigenetic Resetting In PlantsLindsey Campbell
 
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...ijtsrd
 

Similar to Aitken 2016 causes and consequences of oxidative stress in spermatozoa (20)

MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANI
MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANIMICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANI
MICRONUTRIENTS IN MALE INFERTILITY BY DR SHASHWAT JANI
 
Boar semen cueva et al 2013
Boar semen cueva et al 2013Boar semen cueva et al 2013
Boar semen cueva et al 2013
 
Revision parodi 2013
Revision parodi 2013Revision parodi 2013
Revision parodi 2013
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdf
 
Hallmarks of Aging summary
Hallmarks of Aging summaryHallmarks of Aging summary
Hallmarks of Aging summary
 
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...Super Males: Antioxidants boost male fertility and sperm viability in fruit f...
Super Males: Antioxidants boost male fertility and sperm viability in fruit f...
 
Revision parodi 2013
Revision parodi 2013Revision parodi 2013
Revision parodi 2013
 
2015_Muldoon_Brook Stickleback Androgens
2015_Muldoon_Brook Stickleback Androgens2015_Muldoon_Brook Stickleback Androgens
2015_Muldoon_Brook Stickleback Androgens
 
Profert Male forte
Profert Male forteProfert Male forte
Profert Male forte
 
Nrf2: A Guardian of Healthspan and Gatekeeper of Species Longevity
Nrf2:  A Guardian of Healthspan and Gatekeeper of Species LongevityNrf2:  A Guardian of Healthspan and Gatekeeper of Species Longevity
Nrf2: A Guardian of Healthspan and Gatekeeper of Species Longevity
 
Sandlund
SandlundSandlund
Sandlund
 
The developmental and physiological interactions between free radicals and an...
The developmental and physiological interactions between free radicals and an...The developmental and physiological interactions between free radicals and an...
The developmental and physiological interactions between free radicals and an...
 
oxgen tension.pdf
oxgen tension.pdfoxgen tension.pdf
oxgen tension.pdf
 
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...
Developmental Anomalies and Oxidative Stress Responses in Zebrafish (Danio Re...
 
Maxoza - Neo DGM slides.pptx
Maxoza - Neo DGM slides.pptxMaxoza - Neo DGM slides.pptx
Maxoza - Neo DGM slides.pptx
 
Oxidative Stress in Health & Disease: The therapeutic potential of Nrf2 acti...
Oxidative Stress in Health & Disease:  The therapeutic potential of Nrf2 acti...Oxidative Stress in Health & Disease:  The therapeutic potential of Nrf2 acti...
Oxidative Stress in Health & Disease: The therapeutic potential of Nrf2 acti...
 
Essay On Epigenetic Resetting In Plants
Essay On Epigenetic Resetting In PlantsEssay On Epigenetic Resetting In Plants
Essay On Epigenetic Resetting In Plants
 
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...
The Impact of Annona Muricata on Semen Quality and Antioxidants Levels in Alc...
 
stat ppt article (1) (1).docx
stat ppt article (1) (1).docxstat ppt article (1) (1).docx
stat ppt article (1) (1).docx
 
بحث ديالى
بحث ديالىبحث ديالى
بحث ديالى
 

Recently uploaded

Human Factors of XR: Using Human Factors to Design XR Systems
Human Factors of XR: Using Human Factors to Design XR SystemsHuman Factors of XR: Using Human Factors to Design XR Systems
Human Factors of XR: Using Human Factors to Design XR SystemsMark Billinghurst
 
My Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationMy Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationRidwan Fadjar
 
"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii SoldatenkoFwdays
 
SIP trunking in Janus @ Kamailio World 2024
SIP trunking in Janus @ Kamailio World 2024SIP trunking in Janus @ Kamailio World 2024
SIP trunking in Janus @ Kamailio World 2024Lorenzo Miniero
 
WordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your BrandWordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your Brandgvaughan
 
Dev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebDev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebUiPathCommunity
 
Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Mattias Andersson
 
Connect Wave/ connectwave Pitch Deck Presentation
Connect Wave/ connectwave Pitch Deck PresentationConnect Wave/ connectwave Pitch Deck Presentation
Connect Wave/ connectwave Pitch Deck PresentationSlibray Presentation
 
Designing IA for AI - Information Architecture Conference 2024
Designing IA for AI - Information Architecture Conference 2024Designing IA for AI - Information Architecture Conference 2024
Designing IA for AI - Information Architecture Conference 2024Enterprise Knowledge
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsRizwan Syed
 
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationBeyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationSafe Software
 
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024BookNet Canada
 
AI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsAI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsMemoori
 
Commit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyCommit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyAlfredo García Lavilla
 
Unleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubUnleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubKalema Edgar
 
APIForce Zurich 5 April Automation LPDG
APIForce Zurich 5 April  Automation LPDGAPIForce Zurich 5 April  Automation LPDG
APIForce Zurich 5 April Automation LPDGMarianaLemus7
 
DevEX - reference for building teams, processes, and platforms
DevEX - reference for building teams, processes, and platformsDevEX - reference for building teams, processes, and platforms
DevEX - reference for building teams, processes, and platformsSergiu Bodiu
 
Understanding the Laravel MVC Architecture
Understanding the Laravel MVC ArchitectureUnderstanding the Laravel MVC Architecture
Understanding the Laravel MVC ArchitecturePixlogix Infotech
 
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 3652toLead Limited
 

Recently uploaded (20)

Human Factors of XR: Using Human Factors to Design XR Systems
Human Factors of XR: Using Human Factors to Design XR SystemsHuman Factors of XR: Using Human Factors to Design XR Systems
Human Factors of XR: Using Human Factors to Design XR Systems
 
My Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationMy Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 Presentation
 
"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko"Debugging python applications inside k8s environment", Andrii Soldatenko
"Debugging python applications inside k8s environment", Andrii Soldatenko
 
SIP trunking in Janus @ Kamailio World 2024
SIP trunking in Janus @ Kamailio World 2024SIP trunking in Janus @ Kamailio World 2024
SIP trunking in Janus @ Kamailio World 2024
 
WordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your BrandWordPress Websites for Engineers: Elevate Your Brand
WordPress Websites for Engineers: Elevate Your Brand
 
Dev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebDev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio Web
 
Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?
 
Connect Wave/ connectwave Pitch Deck Presentation
Connect Wave/ connectwave Pitch Deck PresentationConnect Wave/ connectwave Pitch Deck Presentation
Connect Wave/ connectwave Pitch Deck Presentation
 
Designing IA for AI - Information Architecture Conference 2024
Designing IA for AI - Information Architecture Conference 2024Designing IA for AI - Information Architecture Conference 2024
Designing IA for AI - Information Architecture Conference 2024
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL Certs
 
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationBeyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
 
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC CataList - Tech Forum 2024
 
AI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsAI as an Interface for Commercial Buildings
AI as an Interface for Commercial Buildings
 
Commit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easyCommit 2024 - Secret Management made easy
Commit 2024 - Secret Management made easy
 
Unleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubUnleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding Club
 
APIForce Zurich 5 April Automation LPDG
APIForce Zurich 5 April  Automation LPDGAPIForce Zurich 5 April  Automation LPDG
APIForce Zurich 5 April Automation LPDG
 
DevEX - reference for building teams, processes, and platforms
DevEX - reference for building teams, processes, and platformsDevEX - reference for building teams, processes, and platforms
DevEX - reference for building teams, processes, and platforms
 
DMCC Future of Trade Web3 - Special Edition
DMCC Future of Trade Web3 - Special EditionDMCC Future of Trade Web3 - Special Edition
DMCC Future of Trade Web3 - Special Edition
 
Understanding the Laravel MVC Architecture
Understanding the Laravel MVC ArchitectureUnderstanding the Laravel MVC Architecture
Understanding the Laravel MVC Architecture
 
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
 

Aitken 2016 causes and consequences of oxidative stress in spermatozoa

  • 1. See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/285619565 Causes and consequences of oxidative stress in spermatozoa Article in Reproduction Fertility and Development · January 2016 DOI: 10.1071/RD15325 CITATIONS 7 READS 566 5 authors, including: Some of the authors of this publication are also working on these related projects: small noncoding RNAs in the male reproductive system View project Mammalian Sperm Membrane Protein Complexes View project Zamira Gibb University of Newcastle 29 PUBLICATIONS 266 CITATIONS SEE PROFILE Mark A Baker University of Newcastle 97 PUBLICATIONS 3,491 CITATIONS SEE PROFILE Joel R Drevet Université Clermont Auvergne 115 PUBLICATIONS 2,621 CITATIONS SEE PROFILE Parviz Gharagozloo CellOxess Biotechnology 27 PUBLICATIONS 682 CITATIONS SEE PROFILE All content following this page was uploaded by Parviz Gharagozloo on 07 December 2015. The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document and are linked to publications on ResearchGate, letting you access and read them immediately.
  • 2. Causes and consequences of oxidative stress in spermatozoa Robert John AitkenA,D , Zamira GibbA , Mark A. BakerA , Joel DrevetB and Parviz GharagozlooC A Priority Research Centre in Reproductive Science and Hunter Medical Research Institute, Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia. B GReD laboratory, CNRS UMR6293-INSERM U1103-Clermont Universite´, 63171 BP80006, Aubie`re cedex, France. C CellOxess LLC, 15 Roszel Street, Princeton, NJ 08540, USA. D Corresponding author. Email: john.aitken@newcastle.edu.au Abstract. Spermatozoa are highly vulnerable to oxidative attack because they lack significant antioxidant protection due to the limited volume and restricted distribution of cytoplasmic space in which to house an appropriate armoury of defensive enzymes. In particular, sperm membrane lipids are susceptible to oxidative stress because they abound in significant amounts of polyunsaturated fatty acids. Susceptibility to oxidative attack is further exacerbated by the fact that these cells actively generate reactive oxygen species (ROS) in order to drive the increase in tyrosine phosphorylation associated with sperm capacitation. However, this positive role for ROS is reversed when spermatozoa are stressed. Under these conditions, they default to an intrinsic apoptotic pathway characterised by mitochondrial ROS generation, loss of mitochondrial membrane potential, caspase activation, phosphatidylserine exposure and oxidative DNA damage. In responding to oxidative stress, spermatozoa only possess the first enzyme in the base excision repair pathway, 8-oxoguanine DNA glycosylase. This enzyme catalyses the formation of abasic sites, thereby destabilising the DNA backbone and generating strand breaks. Because oxidative damage to sperm DNA is associated with both miscarriage and developmental abnormalities in the offspring, strategies for the amelioration of such stress, including the development of effective antioxidant formulations, are becoming increasingly urgent. Additional keywords: apoptosis, fertilizing potential, lipid peroxidation, male germ line, oxidative DNA damage, ROS generation. Introduction Traditionally, spermatozoa are regarded as highly specialised cells that have but one function in life: to achieve fertilisation and deliver the paternal component of the embryonic genome to an MII oocyte. Although defective sperm function has long been recognised as a major cause of human infertility (Hull et al. 1985), this condition has conventionally been equated with the ability of spermatozoa to achieve fertilisation (Aitken et al. 1987; Aitken 2006). With the passage of time, we have come to understand that the functional competence of sper- matozoa cannot be defined merely in terms of the ability of these cells to fertilise an oocyte; it also needs to incorporate an assessment of their ability to program a normal pattern of embryonic development. Spermatozoa may affect embryonic development via both genetic and a variety of epigenetic mechanisms involving the methylation profile of the DNA, the post-translational modification of nuclear histones and the composition of a variety of coding and non-coding RNA species that are integrated into these cells, to find ultimate expression in the zygote and early embryo (Aitken 1999; Ostermeier et al. 2005; Prescott et al. 2012; Hosken and Hodgson 2014; Metzler-Guillemain et al. 2015; Soubry 2015). These sperm-borne epigenetic marks are, in turn, affected by a variety of paternal factors, including genotype, age, obesity, smoking and exposure to environmental contaminants (Aitken 2014). The mechanisms by which such environmental and lifestyle factors affect mammalian spermatozoa, to define both their potential for fertilisation and the subsequent initiation of embry- onic development, are poorly understood. The central hypothe- sis outlined in this article is that all such factors ultimately converge to induce a high level of oxidative stress in the male germline. Oxidative stress is known to interfere with the fertilising capacity of spermatozoa, to damage sperm nuclear DNA and to affect the epigenetic profile of these cells (Aitken et al. 2014b). Herein, we review the evidence relating to the origins and consequences of such stress and consider potential strategies for its remediation. CSIRO PUBLISHING Reproduction, Fertility and Development, 2016, 28, 1–10 http://dx.doi.org/10.1071/RD15325 Journal compilation Ó IETS 2016 www.publish.csiro.au/journals/rfd
  • 3. Oxidative stress and fertilisation potential The notion that sperm function may be compromised by the onset of oxidative stress can be traced back to the early studies of Evans (1947), who observed that heavily irradiated seawater impaired the fertilising capacity of sea urchin spermatozoa. He concluded that the irradiation process had generated hydrogen peroxide (H2O2) and that this powerful oxidising agent was damaging to spermatozoa. Although this conclusion was not subsequently supported by Barron et al. (1949), these authors did generate unequivocal data indicating that H2O2 is extremely damaging to sperm function. Around the same time, Tosic and Walton (1946) demonstrated that the metabolite generated by bovine spermatozoa in the presence of egg yolk-based cryo- preservatives was H2O2 and that this oxidant actively sup- pressed their respiration. Furthermore, these authors identified the source of the H2O2 to be an L-amino acid oxidase with an affinity for aromatic amino acids, particularly phenylalanine, which is abundant in egg yolk (Tosic and Walton 1950). MacLeod (1943) also demonstrated that human spermatozoa lost motility at high oxygen tensions via mechanisms that could be reversed by catalase, again suggesting that H2O2 generation was causally involved in the loss of sperm function. The par- ticular destructive power of H2O2 relative to any other reactive oxygen species (ROS) was later emphasised in studies revealing that catalase, but not superoxide dismutase, was able to relieve the detrimental effect of ROS generated by the xanthine oxidase free radical-generating system on human sperm motility in vitro (Aitken et al. 1993a). The possibility that excess ROS generation may be associated with defective sperm function in vivo was highlighted by two papers that appeared in 1987 and demonstrated that the sperma- tozoa of infertile males were characterised by high levels of ROS generation and the induction of lipid peroxidation (Aitken and Clarkson 1987; Alvarez et al. 1987). The susceptibility of human spermatozoa to lipid peroxidation had previously been highlight- ed by Thaddeus Mann (Jones et al. 1979), who pointed out that these cells contain exceptionally high levels of polyunsaturated fatty acids (PUFA; particularly docosahexanoic acid), which are vulnerable to free radical attack, generating lipid peroxides and aldehydes that have a direct inhibitory action on sperm move- ment. These early studies have subsequently been confirmed in many independent laboratories, all of which agree on the fundamental tenet that defective sperm function is frequently induced by oxidative stress, affecting the motility of these cells, their DNA integrity and their competence for sperm–oocyte fusion (e.g. Aitken et al. 1991, 2010; Zalata et al. 1995; Sanocka et al. 1996; Sharma and Agarwal 1996; Nakamura et al. 2002; Kao et al. 2008; Sakamoto et al. 2008; Bejarano et al. 2014; Morielli and O’Flaherty 2015). Oxidative stress and lipid peroxidation The way in which oxidative stress suppresses sperm motility appears to be directly related to the induction of lipid per- oxidation. When ROS attack the PUFA that abound in human spermatozoa, a variety of lipid metabolites is generated, including lipid peroxyl radicals, alkoxyl radicals and various aldehydes, such as malondialdehyde, 4-hydroxynonenal (4HNE) and acrolein (Jones et al. 1978; Moazamian et al. 2015). The addition of both lipid peroxides and lipid aldehydes to popula- tions of human spermatozoa results in the rapid immobilisation of these cells via different mechanisms. Lipid peroxyl radicals destabilise the sperm plasma membrane by virtue of their ten- dency to abstract hydrogen atoms from adjacent PUFA to achieve a measure of stabilisation as the corresponding lipid hydroperoxide. This process creates carbon-centred lipid radi- cals that combine with oxygen to generate more peroxyl radi- cals, which, in turn, abstract hydrogen from adjacent PUFA to stabilise, generating additional lipid radicals and promoting the propagation of the lipid peroxidation chain reaction (Fig. 1a). The lipid peroxides generated in this process destabilise the plasma membrane by becoming targets for phospholipase A2, which moves into the plasma membranes to cleave out the lipid peroxides so they can be further processed by glutathione per- oxidase (van Kuijk et al. 1985). This process, in turn, generates lysophospholipids that destabilise the sperm plasma membrane, affecting the microarchitecture of this structure and changing the functions of integral membrane proteins that are critical to the maintenance of sperm motility, such as ATP-dependent ion pumps and voltage-regulated ion channels (Nishikawa et al. 1989; Lundbaek and Andersen 1994). The disruptive effect of peroxidative damage on lipid membrane architecture also affects the ability of spermatozoa to participate in the membrane fusion events associated with fertilisation (Aitken et al. 1989, 1993b, 1993c). The lipid peroxidation chain reactions initiated in spermato- zoa may also result in the formation of a cascade of aldehyde by-products that include alkanals, such as malondialdehyde, and alkenals, such as 4HNE and acrolein. These compounds, partic- ularly 4HNE and acrolein, are powerful electrophiles that form adducts with several proteins within the spermatozoa that, in turn, affect sperm function. For example, the formation of adducts with the flagellar axonemal protein, dynein heavy chain, may explain the effect of these aldehydes on sperm movement (Baker et al. 2015; Moazamian et al. 2015). In addition, 4HNE has been shown to bind to mitochondrial proteins in human spermatozoa, triggering electron leakage and the formation of ROS (Fig. 1b). The oxidative stress associated with the latter then forces the spermatozoa to enter the intrinsic apoptotic cascade, beginning with a loss of mitochondrial membrane potential and terminating in oxidative DNA adduct formation, DNA strand breakage and cell death (Aitken et al. 2012). Sources of ROS and oxidative stress in spermatozoa With oxidative stress being such a major factor in the aetiology of defective human sperm function, resolving the possible causes of this condition is critical. In considering this matter, it is important to emphasise that spermatozoa are not only vulnerable to oxidative stress because of the targets they offer for free radical attack in the form of PUFA, proteins and nucleic acids, but they are also lacking significant intracellular antioxidant protection, including ROS-metabolising enzymes, such as catalase and glutathione peroxidase, by virtue of the limited volume and restricted distribution of cytoplasmic space in which to house such mediators of cell survival. Furthermore, these 2 Reproduction, Fertility and Development R. J. Aitken et al.
  • 4. cells actively generate physiological levels of ROS in order to drive the tyrosine phosphorylation events associated with sperm capacitation (Aitken and Nixon 2013). The involvement of ROS in the capacitation of mammalian spermatozoa has been appreciated since the pioneering studies of Claude Gagnon in the 1990s (de Lamirande and Gagnon 1993a). The ROS responsible for sperm capacitation have been variously reported as H2O2 (Bise et al. 1991; Aitken et al. 1995, 1996; Rivlin et al. 2004) superoxide anion (de Lamirande and Gagnon 1993b) and the peroxynitrite radical generated by the reaction of superoxide anion with another free radical species, namely nitric oxide (Herrero et al. 2001; Rodriguez and Beconi 2009). In reality, the interconversion of these various ROS and reactive nitrogen species is very rapid and it is probable that several different redox entities are involved in various aspects of the capacitation process, including the suppression of tyrosine phosphatase activity and the stimulation of cAMP generation (Aitken and Nixon 2013). It has recently been hypothesised that the con- tinued generation of ROS, particularly peroxynitrite, to achieve capacitation ultimately overwhelms the limited antioxidant defences of these cells and precipitates a state of apoptosis. According to this concept, capacitation and the intrinsic apo- ptotic cascade are the opposite ends of a metabolic continuum driven by ROS (Aitken et al. 2015b). If ROS are so important for sperm function, what is the subcellular source of these molecules? In mammalian sperma- tozoa there can be little doubt that the major sources of ROS are the mitochondria. Human sperm mitochondria are particularly active in the generation of ROS via mechanisms that are not dependent on a loss of mitochondrial membrane potential (Koppers et al. 2008). Activation of ROS generation at Complex III was found to stimulate the rapid release of H2O2 into the (a) PUFA R Free radical attack H O2 Initiation OO • R Lipid radical Mitochondrial ROS generation Lipid radical PUFA Peroxyl radical Lipid hydroperoxide R R H R Hydrogen abstraction R OOH OO • R Peroxyl radical Lipid aldehydes 4HNE/acrolein (c) H2O2 (b) ϩ H2O • OH • • Nucleus Mitochondria Adduction of mitochondrial proteins Lipid peroxidation ϩ Fig. 1. Oxidative stress in mammalian spermatozoa. (a) Spermatozoa are susceptible to oxidative stress because they contain high concentrations of polyunsaturated fatty acids (PUFA). Free radical attack leads to the formation of lipid radicals that then combine with the universal electron acceptor, oxygen, to generate a lipid peroxyl radical. In order to stabilise as a hydroperoxide, the latter extracts hydrogen atoms from adjacent lipids, generating lipid radicals that then perpetuate the peroxidation cascade. (b) Lipid aldehydes generated as a consequence of lipid peroxidation, such as 4-hydroxynonenal (4HNE), bind to mitochondrial proteins, including succinic acid dehydrogenase and stimulate yet more free radical generation, further enhancing lipid peroxidation in a self-propagating cycle that propels spermatozoa towards an apoptotic fate. (c) The unusual architecture of spermatozoa means that nucleases activated in the midpiece cytoplasm, or released from the mitochondria, cannot enter the nuclear compartment. The only product of apoptosis that can pass from the midpiece to the sperm head to damage the DNA is H2O2; this is why most DNA damage in spermatozoa is oxidative. Oxidative stress in spermatozoa Reproduction, Fertility and Development 3
  • 5. extracellular space, but no detectable peroxidative damage. Conversely, the induction of ROS on the matrix side of the inner mitochondrial membrane at Complex I resulted in peroxidative damage to the midpiece and a loss of sperm movement that could be prevented by the concomitant presence of a-tocopherol (Koppers et al. 2008). Defective human sper- matozoa spontaneously generate mitochondrial ROS in a man- ner that is negatively correlated with motility (Koppers et al. 2008). Indeed, simultaneous measurement of total cellular ROS with dihydroethidium indicated that 68% of the variability in such measurements could be explained by differences in mito- chondrial ROS production (Koppers et al. 2008). Another potential source of ROS are the NADPH oxidase enzymes (NOX), including the calcium-dependent NOX5, which are known to be present in the spermatozoa of certain species, including human (Ba´nfi et al. 2001), although other species, such as the mouse, do not possess this enzyme. Expos- ing spermatozoa to NADPH can trigger a redox response that is detectable with the redox probe lucigenin and inhibitable by diphenylene iodonium (DPI), a flavoprotein inhibitor (Aitken et al. 1997; Vernet et al. 2001). However, this lucigenin- dependent activity was subsequently shown to be due to the direct enzymatic reduction of the probe by cytochrome P450 reductase (Baker et al. 2004) and cytochrome b5 reductase (Baker et al. 2005) when the electron donors were NADPH and NADH, respectively. In contrast, using luminol as a ROS probe, clear evidence has been obtained for a calcium- dependent increase in ROS generation, which is particularly marked in the spermatozoa of infertile patients and potentially reflective of an involvement of NOX5 in the aetiology of defective sperm function (Aitken and Clarkson 1987). There is even some evidence to suggest that NOX5 may be overrepre- sented in the defective spermatozoa recovered from patients exhibiting teratozoospermia (Ghani et al. 2013). However, definitive proof that NOX5 is the source of ROS under such circumstances is currently lacking. There is a possibility that the calcium-dependent signals observed with unfractionated sperm suspensions are the result of low-level leucocyte contamination (Aitken and Clarkson 1987; Aitken et al. 1992). The ability of the NOX inhibitor apocynin to suppress the ROS signals generated by human sperm suspensions (Dona` et al. 2011) could also be accounted for by leucocyte contamination because this reagent prevents assembly of the key cytosolic components of the NADPH oxidase system (p40phox , p47phox and p67phox ), which is not necessary for NOX5 to be active. A detailed study of the NOX species present in human spermatozoa is currently lacking and the role of these enzymes in the creation of oxidative stress within the germline remains unresolved. One possibility that cannot be excluded is that the NOX enzymes present in mammalian spermatozoa play no role at all in the regulation of sperm function, but rather function much earlier in germ cell production, controlling spermatogonial stem cell proliferation (Morimoto et al. 2013). Finally, L-amino acid oxidases with a particular affinity for phenylalanine have been identified in bull, horse, human and ram spermatozoa (Tosic and Walton 1946; Aitken et al. 2015a; Houston et al. 2015). In the case of equine spermatozoa, which are heavily dependent on oxidative phosphorylation (Gibb et al. 2014), the primary role for this amino acid oxidase may be to support the energy metabolism of these cells through the oxidative deamination of aromatic amino acids, generating keto acids that are then processed by the sperm mitochondria. However, in the case of human spermatozoa, oxidative phos- phorylation appears to play a minor role in sperm metabolism because these cells are largely dependent of glycolysis to meet their energy needs (du Plessis et al. 2015). In these cells, the L-amino acid oxidase (interleukin 4 induced protein 1, IL4I1) seems to have acquired a new biological function in supplying the redox drive to sperm capacitation (Houston et al. 2015). Role of oxidative stress in DNA damage One of the major complications associated with male infertility is the presence of high levels of DNA damage in the sperma- tozoa. Such damage can arise as a consequence of infertility (Irvine et al. 2000; Aitken and Curry 2011) age (Singh et al. 2003) smoking (Fraga et al. 1996) antioxidant deficiency (Fraga et al. 1991, 1996), obesity (Fariello et al. 2012) exposure to infection (Reichart et al. 2000; Burrello et al. 2004), heat (De Iuliis et al. 2009a; Santiso et al. 2012) acidic pH (Santiso et al. 2012), metals, particularly transition metals such as iron and copper (Aitken et al. 2014a), radiofrequency electromag- netic radiation (De Iuliis et al. 2009a), ionising radiation (Singh and Stephens 1998), environmental toxicants such as acrylam- ide (Katen and Roman 2015), chemotherapeutic agents (Delbe`s et al. 2010), air pollution, plasticisers, pesticides (Evenson and Wixon 2005; O’Flaherty 2014) and chloracetanilide herbicides such as alachlor (Grizard et al. 2007). There can be little doubt that most of these factors affect the integrity of sperm chromatin through the induction of oxidative stress. Such stress results in the generation of oxidised DNA base adducts such as 8-hydroxy-20 -deoxyguanosine (8OHdG), particularly in areas of the genome that are not heavily prota- minated (De Iuliis et al. 2009b; Noblanc et al. 2013). Spermato- zoa only possess one enzyme in the base excision repair (BER) pathway, 8-oxoguanine DNA glycosylase (OGG1). This glyco- sylase is associated with the sperm nucleus and mitochondria and can actively excise 8OHdG, releasing this base adduct into the extracellular space. Remarkably, spermatozoa do not pos- sess the downstream components of the BER pathway, namely apurinic endonuclease 1 (APE1) and X-ray repair complement- ing defective repair in Chinese hamster cells 1 (XRCC1). The net result of this truncated DNA repair capacity is to generate abasic sites at locations that have been affected by 8OHdG formation. Such abasic sites destabilise the ribose–phosphate backbone, leading to a b-elimination or a ring opening reaction of the ribose unit and a consequential strand break. This type of DNA chemistry has been identified as being central to the initiation of cancer in other cell types. Therefore, oxidative DNA base lesions are not only potentially mutagenic but, importantly, also contribute indirectly to the DNA fragmenta- tion observed in the patient population (Ohno et al. 2014). An oxidative involvement in DNA damage to mammalian spermatozoa has been observed in relation to infertility (Shen and Ong 2000; Aitken et al. 2010), heat (De Iuliis et al. 2009a), antioxidant deficiency (Fraga et al. 1991), age (Weir and 4 Reproduction, Fertility and Development R. J. Aitken et al.
  • 6. Robaire 2007; Smith et al. 2013a), smoking (Fraga et al. 1991), obesity (Bakos et al. 2011), radiofrequency electromagnetic radiation (De Iuliis et al. 2009a), herbicides (Grizard et al. 2007), plasticisers (Erkekoglu et al. 2010; Zhou et al. 2010) and chemotherapeutic agents (Ghosh et al. 2002). Indeed, it would appear that most DNA damage in mammalian spermatozoa is the result of an oxidative insult generated as a result of either impaired antioxidant protection because of endogenous (e.g. age) or exogenous (e.g. phthalate esters) factors or changes in the redox status of spermatozoa because of internal (e.g. mitochon- drial electron leakage) or external (e.g. radiation or alachlor) influences. However, it is also undeniable that not every spermatozoon afflicted with DNA damage shows signs of oxida- tive stress. Under these conditions, it has been suggested that nuclease-mediated DNA fragmentation must occur as a result of spermatozoa defaulting to an apoptotic state rather than oxidative stress (Muratori et al. 2015). This interesting hypothesis is difficult to reconcile with the fact that apoptosis invariably involves the induction of oxidative stress (Koppers et al. 2011), so it isdifficult to imagine how these phenomena can beseparated in vivo. A possible resolution of this dilemma is set out below. Role of apoptosis in DNA damage It is well known that testicular precursor germ cells can undergo apoptosis as part of a physiological process designed to optimise germ cell : Sertoli cell ratios and to bring a measure of quality control to the spermatogenic process, ensuring that no defective germ cells are allowed to differentiate into spermatozoa (Shukla et al. 2012). Apoptosis may also occur during spermatogenesis in response to adverse circumstances, including heat shock, ionising radiation, growth factor deprivation and chemothera- peutic agents. The apoptotic process is largely, but not exclu- sively, targeted to spermatocytes and both the intrinsic mitochondrial pathway and the extrinsic p53/Fas system have been implicated as key modulators of this process (Boekelheide 2005; Lagos-Cabre´ and Moreno 2012). However, the focus of this discussion is the spermatozoa. Spermatozoa are highly differentiated, transcriptionally silent cells that, by virtue of their inert nuclear constitution and highly specialised architecture, cannot undergo apoptosis in the conventional sense. Nevertheless, they can undergo a truncated version of this process. One of the key features of sperm cell biology is that we do not have to expend energy searching for factors that will induce these cells to undergo apoptosis. Rather, these cells are designed to undergo apoptosis; it is their default position. Spermatozoa are the ultimate symbol of disposable cell types; indeed, all these cells are destined to die a lonely apoptotic death in the male or female tract. The fortunate exceptions to this rule are the handful of individual gametes that manage to fertilise an oocyte and, in so doing, achieve potential immortality for the genotype they carry. When apoptosis does eventually occur, it is generally the intrinsic apoptotic cascade that is induced, mediated by the sperm mitochondria. Although receptor-mediated extrinsic apoptosis remains a theoretical possibility in spermatozoa, no ligands have been convincingly described to date that are capable of eliciting such a response in the fully differentiated gamete. There has been a claim that bacterial lipopolysaccharide (LPS) can elicit apoptosis in spermatozoa by interacting with Toll-like receptor (TLR) 2 and TLR4 on the sperm surface (Fujita et al. 2011). However these data have not yet been independently validated and our research group has not yet been able to achieve apoptosis using commercially available LPS (R. J. Aitken, unpubl. obs.). As a result, our view is that the mature gamete has very little capacity to activate the extrinsic apoptotic cascade, but is extremely vulnerable to its intrinsic counterpart. Spermatozoa are normally prevented from entering the intrinsic apoptotic pathway by virtue of the continuing activity of phosphatidylinositol 3-kinase (PI3K; Koppers et al. 2011). If PI3K is inhibited, then the spermatozoa default to an apoptotic cascade characterised by rapid loss of motility, generation of ROS, caspase activation in the cytosol, annexin V binding to the cell surface, cytoplasmic vacuolisation and oxidative DNA damage. The anti-apoptotic action of PI3K appears to depend on its ability to promote the phosphorylation of another kinase, AKT, which, in turn, is responsible for phosphorylating anti- apoptotic effector proteins such as Bcl-2-associated death pro- moter (BAD). Phosphorylation of the latter is essential for BAD to remain associated with its cytoplasmic keeper protein, 14-3-3. However, dephosphorylation allows BAD to orchestrate an apoptotic process that has many similarities with the intrinsic apoptotic cascade observed in somatic cells. There are two major points of difference between apoptosis in spermatozoa and somatic cells, as follows: 1. Mammalian spermatozoa are structurally different from somatic cells in that all the mitochondria and most of the cytoplasm are compartmentalised in the mid-piece of the cell, physically separated from the DNA in the sperm nucleus. As a result, even if apoptosis is activated in these cells, the endonucleases released from the mitochondria (e.g. endonuclease G) or activated in the cytoplasm (e.g. caspase-activated DNAse) are physically impeded from attacking the sperm nucleus (Koppers et al. 2011). The only element of the apoptotic cascade that can exit from the sperm midpiece and penetrate the nuclear compartment is H2O2. It is for this reason that most of the DNA damage present in human spermatozoa appears to be oxidatively induced (Fig. 1c; Aitken et al. 2010). 2. Spermatozoa are also characterised by a severely truncated BER pathway, as discussed above, that stalls after OGG1 has removed the oxidised base to create abasic sites that have to be further processed by the oocyte following fertilisation. One consequence of spermatozoa lacking the next enzyme in this pathway, namely APE1, is that these cells cannot create the 30 -OH termini that are required by the terminal deoxyribonucleotidyl transferase-mediated dUTP–digoxigenin nick end-labelling (TUNEL) assay. As a result, TUNEL is a very insensitive methodology for assessing DNA damage in spermatozoa. Under circumstances where DNA damage is induced by, for example, exposure to H2O2, intracellular and extracellular 8OHdG can be clearly detected in the affected sperm suspension and DNA strand breakage can be detected with the sperm chromatin structure assay (SCSA); however, TUNEL signals are not apparent (Smith et al. 2013b). Oxidative stress in spermatozoa Reproduction, Fertility and Development 5
  • 7. The later do eventually appear when the cells are close to death. At this point, it is possible that a DNAse does become activated in the spermatozoa (Sotolongo et al. 2005). For the reasons given above, such a nuclease would have to be incorporated into the sperm chromatin, not released from the mitochondria or activated in the cytoplasm. The nature of this DNAse (topoisomerase? DNAse 1?) and its mechanisms of activation are not known. It is possible such nuclease activity may be activated by a rise in intracellular calcium as cells die, because calcium-dependent nuclease activity has been described in this cell type on several independent occasions by independent groups (Sotolongo et al. 2005; Sibirtsev et al. 2011). Apoptosis or oxidative stress causes DNA damage We can conclude from the foregoing discussion that there are two mechanisms for damaging DNA in mammalian spermato- zoa. It is now generally acknowledged that a wide variety of different intrinsic and extrinsic factors converge to generate a state of oxidative stress in the germline. Once such stress has been initiated, it tends to become accentuated because the lipid aldehydes generated during the peroxidative process bind to proteins in the mitochondrial electron transport chain, particu- larly succinic acid dehydrogenase, stimulating the generation of yet more free radicals, more DNA damage and more lipid per- oxidation to continue the downward spiral towards apoptosis (Fig. 1b; Aitken et al. 2012). There is also an association between oxidative DNA damage in the male germline and poor chromatin protamination during spermiogenesis (De Iuliis et al. 2009b). This relationship may reflect a certain vulnerability towards oxidative stress as a consequence of the failure of sperm nuclear DNA to adequately compact. However, it may also be a consequence of inadequate protamination, because these small, basic proteins are thought to protect the DNA by acting as sacrificial antioxidants and by chelating redox active metals such as copper (Liang et al. 1999). The relationship between oxidative stress and apoptosis is complex. Clearly, spermatozoa do express the classical markers of apoptosis, such as ROS generation, phosphatidylserine expo- sure, caspase activation and DNA fragmentation (Koppers et al. 2011). If PI3K activity is inhibited with wortmannin, then human spermatozoa rapidly default to the intrinsic apoptotic cascade, displaying all of the above features, including high levels of mitochondrial ROS generation (Koppers et al. 2011). Therefore, entry of spermatozoa into the senescence-driven apoptotic pathway as a consequence of compromised PI3K activity inevitably results in an apoptotic cascade involving the stimulation of mitochondrial ROS generation and the induc- tion of oxidative DNA damage. Under these circumstances, apoptosis and oxidative DNA damage are inextricably linked. Similarly, when spermatozoa are exposed to xenobiotics such as alachlor, the induction of apoptosis is inextricably linked with the induction of oxidative stress (Grizard et al. 2007). Within the infertile population, DNA damage is again associated with the simultaneous appearance of oxidative stress and apoptosis (Wang et al. 2003; Aitken et al. 2010) via mechanisms that can be reversed by the sustained administration of antioxidants such as melatonin (Bejarano et al. 2014). Similarly, cryostorage leads to the induction of oxidatively driven DNA damage and apoptosis that can be reversed by the presence of the antioxidant quercetin (Zribi et al. 2012). However, there are occasional circumstances where DNA damage can be visualised in the absence of any evidence that the cells have been subjected to oxidative stress (Muratori et al. 2015). Under these circum- stances, it is possible that the apoptosis involves stimulation of a DNAse that is integrated into the sperm chromatin and somehow becomes activated when these cells are under stress. Developmental consequences of oxidative damage in the germ line Whatever the causes of oxidative stress in the male germline, there can be no doubt that this pathophysiological mechanism leads to both impaired fertility and disrupted embryonic development. At high levels of oxidative stress, fertilisation is prevented because the damage to the sperm plasma membrane impairs both the motility of these cells and their competence for fusion with the oocyte. At lower levels of oxidative stress the spermatozoa can retain their capacity for fertilisation while the DNA in their nuclei is still oxidatively damaged (Aitken et al. 1998). The developmental consequences of fertilising eggs with spermatozoa exhibiting oxidative DNA damage has been explored using the glutathione peroxidase 5 (GPx5)-knockout mouse. In this mouse model, the spermatozoa suffer from oxidative areas as they descend the epididymis (Chabory et al. 2009). The level of stress experienced by these spermatozoa does not impair their fertilising capacity, but does induce high levels of oxidative DNA damage in the sperm nuclei. The consequence of this damage can be seen in the developmental status of the embryos when Gpx5-null males are mated with wild-type females, because such unions are accompanied by a significant increase in the incidence of miscarriage and devel- opmental abnormalities (Chabory et al. 2009). In related studies in which mouse spermatozoa were oxidatively damaged by exposing them to H2O2, several developmental abnormalities were observed in the offspring, including a delay in embryonic development rates, a decrease in the ratio of inner cell mass cells in the resulting blastocyst and a reduction in implantation rates (Lane et al. 2014). Crown–rump length at Day 18 of gestation was also reduced in offspring produced from H2O2-treated spermatozoa. Female offspring from peroxide-treated sperma- tozoa were smaller, became glucose intolerant and accumulated increased levels of adipose tissue compared with control female offspring. Interestingly, the male offspring phenotype was less severe, with increases in fat depots only seen at 4 weeks of age, which returned to control levels later in life (Lane et al. 2014). Studies in primates (Burruel et al. 2013) and cattle (Simo˜es et al. 2013) have confirmed the effect of oxidative DNA damage in spermatozoa on the developmental potential of fertilised ova. Given the developmental significance of this oxidative sperm DNA damage, it is important that strategies are developed to reduce such pathological changes as a matter of good practice in assisted conception programs and as a matter of good con- science in couples contemplating parenthood by natural means. The data accumulated to date are encouraging in that the 6 Reproduction, Fertility and Development R. J. Aitken et al.
  • 8. oxidative damage in spermatozoa associated with obesity, for example, can clearly be reversed by a combination of diet and exercise (Palmer et al. 2012). The use of antioxidants has also met with some success in treating idiopathic oxidative stress in spermatozoa (Gharagozloo and Aitken 2011; Showell et al. 2014). The advantages of using an effective oral antioxidant treatment are that, in cases where oxidative stress has been diagnosed in the male germline, it should result in increases in both the fertilising potential of human spermatozoa and their genetic integrity. The disadvantage of using antioxidants, par- ticularly as an empirical treatment for patients where there is no evidence of oxidative stress, is that it may create a reductive stress (Chen et al. 2013). We still await the results of randomised double-blind cross-over trials to definitively establish the therapeutic value of different antioxidant formulations in the treatment of male patients exhibiting high levels of oxidative DNA damage in their spermatozoa. Such studies are eagerly anticipated. References Aitken, R. J. (1999). The Amoroso Lecture. The human spermatozoon: a cell in crisis? J. Reprod. Fertil. 115, 1–7. doi:10.1530/JRF.0.1150001 Aitken, R. J. (2006). Sperm function tests and fertility. Int. J. Androl. 29, 69–75. doi:10.1111/J.1365-2605.2005.00630.X Aitken, R. J. (2014). Age, the environment and our reproductive future: bonking baby boomers and the future of sex. Reproduction 147, S1–S11. doi:10.1530/REP-13-0399 Aitken, R. J., and Clarkson, J. S. (1987). Cellular basis of defective sperm function and its association with the genesis of reactive oxygen species by human spermatozoa. J. Reprod. Fertil. 81, 459–469. doi:10.1530/ JRF.0.0810459 Aitken, R. J., and Curry, B. J. (2011). Redox regulation of human sperm function: from the physiological control of sperm capacitation to the etiology of infertility and DNA damage in the germ line. Antioxid. Redox Signal. 14, 367–381. doi:10.1089/ARS.2010.3186 Aitken, R. J., and Nixon, B. (2013). Sperm capacitation: a distant landscape glimpsed but unexplored. Mol. Hum. Reprod. 19, 785–793. doi:10.1093/ MOLEHR/GAT067 Aitken, R. J., Thatcher, S., Glasier, A. F., Clarkson, J. S., Wu, F. C., and Baird, D. T. (1987). Relative ability of modified versions of the hamster oocyte penetration test, incorporating hyperosmotic medium or the ionophore A23187, to predict IVF outcome. Hum. Reprod. 2, 227–231. Aitken, R. J., Clarkson, J. S., and Fishel, S. (1989). Generation of reactive oxygen species, lipid peroxidation, and human sperm function. Biol. Reprod. 41, 183–197. doi:10.1095/BIOLREPROD41.1.183 Aitken, R. J., Irvine, D. S., and Wu, F. C. (1991). Prospective analysis of sperm–oocyte fusion and reactive oxygen species generation as criteria for the diagnosis of infertility. Am. J. Obstet. Gynecol. 164, 542–551. doi:10.1016/S0002-9378(11)80017-7 Aitken, R. J., Buckingham, D., West, K., Wu, F. C., Zikopoulos, K., and Richardson, D. W. (1992). Differential contribution of leucocytes and spermatozoa to the generation of reactive oxygen species in the ejacu- lates of oligozoospermic patients and fertile donors. J. Reprod. Fertil. 94, 451–462. doi:10.1530/JRF.0.0940451 Aitken, R. J., Buckingham, D., and Harkiss, D. (1993a). Use of a xanthine oxidase free radical generating system to investigate the cytotoxic effects of reactive oxygen species on human spermatozoa. J. Reprod. Fertil. 97, 441–450. doi:10.1530/JRF.0.0970441 Aitken, R. J., Harkiss, D., and Buckingham, D. (1993b). Relationship between iron-catalysed lipid peroxidation potential and human sperm function. J. Reprod. Fertil. 98, 257–265. doi:10.1530/JRF.0.0980257 Aitken, R. J., Harkiss, D., and Buckingham, D. W. (1993c). Analysis of lipid peroxidation mechanisms in human spermatozoa. Mol. Reprod. Dev. 35, 302–315. doi:10.1002/MRD.1080350313 Aitken, R. J., Paterson, M., Fisher, H., Buckingham, D. W., and van Duin, M. (1995). Redox regulation of tyrosine phosphorylation in human sperma- tozoa and its role in the control of human sperm function. J. Cell Sci. 108, 2017–2025. Aitken, R. J., Buckingham, D. W., Harkiss, D., Paterson, M., Fisher, H., and Irvine, D. S. (1996). The extragenomic action of progesterone on human spermatozoa is influenced by redox regulated changes in tyrosine phosphorylation during capacitation. Mol. Cell. Endocrinol. 117, 83–93. doi:10.1016/0303-7207(95)03733-0 Aitken, R. J., Fisher, H. M., Fulton, N., Gomez, E., Knox, W., Lewis, B., and Irvine, S. (1997). Reactive oxygen species generation by human sper- matozoa is induced by exogenous NADPH and inhibited by the flavo- protein inhibitors diphenylene iodonium and quinacrine. Mol. Reprod. Dev. 47, 468–482. doi:10.1002/(SICI)1098-2795(199708)47:4,468:: AID-MRD14.3.0.CO;2-S Aitken, R. J., Gordon, E., Harkiss, D., Twigg, J. P., Milne, P., Jennings, Z., and Irvine, D. S. (1998). Relative impact of oxidative stress on the functional competence and genomic integrity of human spermatozoa. Biol. Reprod. 59, 1037–1046. doi:10.1095/BIOLREPROD59.5.1037 Aitken, R. J., De Iuliis, G. N., Finnie, J. M., Hedges, A., and McLachlan, R. I. (2010). Analysis of the relationships between oxidative stress, DNA damage and sperm vitality in a patient population: development of diagnostic criteria. Hum. Reprod. 25, 2415–2426. doi:10.1093/ HUMREP/DEQ214 Aitken, R. J., Whiting, S., De Iuliis, G. N., McClymont, S., Mitchell, L. A., and Baker, M. A. (2012). Electrophilic aldehydes generated by sperm metabolism activate mitochondrial reactive oxygen species generation and apoptosis by targeting succinate dehydrogenase. J. Biol. Chem. 287, 33 048–33 060. doi:10.1074/JBC.M112.366690 Aitken, R. J., Finnie, J. M., Muscio, L., Whiting, S., Connaughton, H. S., Kuczera, L., Rothkirch, T. B., and De Iuliis, G. N. (2014a). Potential importance of transition metals in the induction of DNA damage by sperm preparation media. Hum. Reprod. 29, 2136–2147. doi:10.1093/ HUMREP/DEU204 Aitken, R. J., Smith, T. B., Jobling, M. S., Baker, M. A., and De Iuliis, G. N. (2014b). Oxidative stress and male reproductive health. Asian J. Androl. 16, 31–38. doi:10.4103/1008-682X.122203 Aitken, J. B., Naumovski, N., Grupen, C. G., Gibb, Z., and Aitken, R. J. (2015a). Characterization of an L-amino acid oxidase in equine sperma- tozoa. Biol. Reprod. 92, 125. doi:10.1095/BIOLREPROD.114.126052 Aitken, R. J., Baker, M. A., and Nixon, B. (2015b). Are sperm capacitation and apoptosis the opposite ends of a continuum driven by oxidative stress? Asian J. Androl. 17, 633–639. doi:10.4103/1008-682X.153850 Alvarez, J. G., Touchstone, J. C., Blasco, L., and Storey, B. T. (1987). Spontaneous lipid peroxidation and production of hydrogen peroxide and superoxide in human spermatozoa. Superoxide dismutase as major enzyme protectant against oxygen toxicity. J. Androl. 8, 338–348. Baker, M. A., Krutskikh, A., Curry, B. J., McLaughlin, E. A., and Aitken, R. J. (2004). Identification of cytochrome P450-reductase as the enzyme responsible for NADPH-dependent lucigenin and tetrazolium salt reduc- tion in rat epididymal sperm preparations. Biol. Reprod. 71, 307–318. doi:10.1095/BIOLREPROD.104.027748 Baker, M. A., Krutskikh, A., Curry, B. J., Hetherington, L., and Aitken, R. J. (2005). Identification of cytochrome-b5 reductase as the enzyme respon- sible for NADH-dependent lucigenin chemiluminescence in human spermatozoa. Biol. Reprod. 73, 334–342. doi:10.1095/BIOLREPROD. 104.037960 Baker, M. A., Weinberg, A., Hetherington, L., Villaverde, A. I., Velkov, T., Baell, J., and Gordon, C. P. (2015). Defining the mechanisms by which the reactive oxygen species by-product, 4-hydroxynonenal, affects Oxidative stress in spermatozoa Reproduction, Fertility and Development 7
  • 9. human sperm cell function. Biol. Reprod. 92, 108. doi:10.1095/BIOL REPROD.114.126680 Bakos, H. W., Mitchell, M., Setchell, B. P., and Lane, M. (2011). The effect of paternal diet-induced obesity on sperm function and fertilization in a mouse model. Int. J. Androl. 34, 402–410. doi:10.1111/J.1365-2605. 2010.01092.X Ba´nfi, B., Molna´r, G., Maturana, A., Steger, K., Hegeduˆs, B., Demaurex, N., and Krause, K. H. (2001). A Ca(2þ)-activated NADPH oxidase in testis, spleen, and lymph nodes. J. Biol. Chem. 276, 37 594–37 601. doi:10.1074/JBC.M103034200 Barron, E. S. G., Flood, V., and Gasvoda, B. (1949). The effect of hydrogen peroxide and of X-ray irradiated sea water on the respiration of sea urchin sperm and eggs. Biol. Bull. 97, 51–56. doi:10.2307/ 1538093 Bejarano, I., Monllor, F., Marchena, A. M., Ortiz, A., Lozano, G., Jime´nez, M. I., Gaspar, P., Garcı´a, J. F., Pariente, J. A., Rodrı´guez, A. B., and Espino, J. (2014). Exogenous melatonin supplementation prevents oxidative stress-evoked DNA damage in human spermatozoa. J. Pineal Res. 57, 333–339. doi:10.1111/JPI.12172 Bize, I., Santander, G., Cabello, P., Driscoll, D., and Sharpe, C. (1991). Hydrogen peroxide is involved in hamster sperm capacitation in vitro. Biol. Reprod. 44, 398–403. doi:10.1095/BIOLREPROD44.3.398 Boekelheide, K. (2005). Mechanisms of toxic damage to spermatogenesis. J. Natl Cancer Inst. Monogr. 2005, 6–8. doi:10.1093/JNCIMONO GRAPHS/LGI006 Burrello, N., Calogero, A. E., Perdichizzi, A., Salmeri, M., D’Agata, R., and Vicari, E. (2004). Inhibition of oocyte fertilization by assisted reproduc- tive techniques and increased sperm DNA fragmentation in the presence of Candida albicans: a case report. Reprod. Biomed. Online 8, 569–573. doi:10.1016/S1472-6483(10)61104-2 Burruel, V., Klooster, K. L., Chitwood, J., Ross, P. J., and Meyers, S. A. (2013). Oxidative damage to rhesus macaque spermatozoa results in mitotic arrest and transcript abundance changes in early embryos. Biol. Reprod. 89, 72. doi:10.1095/BIOLREPROD.113.110981 Chabory, E., Damon, C., Lenoir, A., Kauselmann, G., Kern, H., Zevnik, B., Garrel, C., Saez, F., Cadet, R., Henry-Berger, J., Schoor, M., Gottwald, U., Habenicht, U., Drevet, J. R., and Vernet, P. (2009). Epididymis seleno-independent glutathione peroxidase 5 maintains sperm DNA integrity in mice. J. Clin. Invest. 119, 2074–2085. Chen, S. J., Allam, J. P., Duan, Y. G., and Haidl, G. (2013). Influence of reactive oxygen species on human sperm functions and fertilizing capacity including therapeutical approaches. Arch. Gynecol. Obstet. 288, 191–199. doi:10.1007/S00404-013-2801-4 De Iuliis, G. N., Newey, R. J., King, B. V., and Aitken, R. J. (2009a). Mobile phone radiation induces reactive oxygen species production and DNA damage in human spermatozoa in vitro. PLoS One 4, e6446. doi:10.1371/JOURNAL.PONE.0006446 De Iuliis, G. N., Thomson, L. K., Mitchell, L. A., Finnie, J. M., Koppers, A. J., Hedges, A., Nixon, B., and Aitken, R. J. (2009b). DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-20 -deoxyguanosine, a marker of oxidative stress. Biol. Reprod. 81, 517–524. doi:10.1095/BIOLRE PROD.109.076836 de Lamirande, E., and Gagnon, C. (1993a). Human sperm hyperactivation and capacitation as parts of an oxidative process. Free Radic. Biol. Med. 14, 157–166. doi:10.1016/0891-5849(93)90006-G de Lamirande, E., and Gagnon, C. (1993b). A positive role for the superoxide anion in triggering hyperactivation and capacitation of human sperma- tozoa. Int. J. Androl. 16, 21–25. doi:10.1111/J.1365-2605.1993. TB01148.X Delbe`s, G., Hales, B. F., and Robaire, B. (2010). Toxicants and human sperm chromatin integrity. Mol. Hum. Reprod. 16, 14–22. doi:10.1093/ MOLEHR/GAP087 Dona`, G., Fiore, C., Andrisani, A., Ambrosini, G., Brunati, A., Ragazzi, E., Armanini, D., Bordin, L., and Clari, G. (2011). Evaluation of correct endogenous reactive oxygen species content for human sperm capacita- tion and involvement of the NADPH oxidase system. Hum. Reprod. 26, 3264–3273. doi:10.1093/HUMREP/DER321 du Plessis, S. S., Agarwal, A., Mohanty, G., and van der Linde, M. (2015). Oxidative phosphorylation versus glycolysis: what fuel do spermatozoa use? Asian J. Androl. 17, 230–235. doi:10.4103/1008-682X.135123 Erkekoglu, P., Rachidi, W., Yuzugullu, O. G., Giray, B., Favier, A., Ozturk, M., and Hincal, F. (2010). Evaluation of cytotoxicity and oxidative DNA damaging effects of di(2-ethylhexyl)-phthalate (DEHP) and mono (2-ethylhexyl)-phthalate (MEHP) on MA-10 Leydig cells and protection by selenium. Toxicol. Appl. Pharmacol. 248, 52–62. doi:10.1016/ J.TAAP.2010.07.016 Evans, T. C. (1947). Effects of hydrogen peroxide produced in the medium by radiation of spermatozoa of Arbacia punctulata. Biol. Bull. 92, 99–109. doi:10.2307/1538160 Evenson, D. P., and Wixon, R. (2005). Environmental toxicants cause sperm DNA fragmentation as detected by the sperm chromatin structure assay (SCSA). Toxicol. Appl. Pharmacol. 207(Suppl.), 532–537. doi:10.1016/ J.TAAP.2005.03.021 Fariello, R. M., Pariz, J. R., Spaine, D. M., Cedenho, A. P., Bertolla, R. P., and Fraietta, R. (2012). Association between obesity and alteration of sperm DNA integrity and mitochondrial activity. BJU Int. 110, 863–867. doi:10.1111/J.1464-410X.2011.10813.X Fraga, C. G., Motchnik, P. A., Shigenaga, M. K., Helbock, H. J., Jacob, R. A., and Ames, B. N. (1991). Ascorbic acid protects against endogenous oxidative DNA damage in human sperm. Proc. Natl Acad. Sci. USA 88, 11 003–11 006. doi:10.1073/PNAS.88.24.11003 Fraga, C. G., Motchnik, P. A., Wyrobek, A. J., Rempel, D. M., and Ames, B. N. (1996). Smoking and low antioxidant levels increase oxidative damage to sperm DNA. Mutat. Res. 351, 199–203. doi:10.1016/0027- 5107(95)00251-0 Fujita, Y., Mihara, T., Okazaki, T., Shitanaka, M., Kushino, R., Ikeda, C., Negishi, H., Liu, Z., Richards, J. S., and Shimada, M. (2011). Toll-like receptors (TLR) 2 and 4 on human sperm recognize bacterial endotoxins and mediate apoptosis. Hum. Reprod. 26, 2799–2806. doi:10.1093/ HUMREP/DER234 Ghani, E., Keshtgar, S., Habibagahi, M., Ghannadi, A., and Kazeroni, M. (2013). Expression of NOX5 in human teratozoospermia compared to normozoospermia. Andrologia 45, 351–356. doi:10.1111/AND.12023 Gharagozloo, P., and Aitken, R. J. (2011). The role of sperm oxidative stress in male infertility and the significance of oral antioxidant therapy. Hum. Reprod. 26, 1628–1640. doi:10.1093/HUMREP/DER132 Ghosh, D., Das, U. B., and Misro, M. (2002). Protective role of alpha- tocopherol–succinate (provitamin-E) in cyclophosphamide induced tes- ticular gametogenic and steroidogenic disorders: a correlative approach to oxidative stress. Free Radic. Res. 36, 1209–1218. doi:10.1080/ 1071576021000016472 Gibb, Z., Lambourne, S. R., and Aitken, R. J. (2014). The paradoxical relationship between stallion fertility and oxidative stress. Biol. Reprod. 91, 77. doi:10.1095/BIOLREPROD.114.118539 Grizard, G., Ouchchane, L., Roddier, H., Artonne, C., Sion, B., Vasson, M. P., and Janny, L. (2007). In vitro alachlor effects on reactive oxygen species generation, motility patterns and apoptosis markers in human spermato- zoa.Reprod. Toxicol.23, 55–62.doi:10.1016/J.REPROTOX.2006.08.007 Herrero, M. B., de Lamirande, E., and Gagnon, C. (2001). Tyrosine nitration in human spermatozoa: a physiological function of peroxynitrite, the reaction product of nitric oxide and superoxide. Mol. Hum. Reprod. 7, 913–921. doi:10.1093/MOLEHR/7.10.913 Hosken, D. J., and Hodgson, D. J. (2014). Why do sperm carry RNA? Relatedness, conflict, and control. Trends Ecol. Evol. 29, 451–455. doi:10.1016/J.TREE.2014.05.006 8 Reproduction, Fertility and Development R. J. Aitken et al.
  • 10. Houston, B., Curry, B., and Aitken, R. J. (2015). Human spermatozoa possess an IL4I1 L-amino acid oxidase with a potential role in sperm function. Reproduction 149, 587–596. doi:10.1530/REP-14-0621 Hull, M. G. R., Glazener, C. M. A., Kelly, N. J., Conway, D. I., Foster, P. A., Hunton, R. A., Coulson, C., Lambert, P. A., Watt, E. M., and Desai, K. M. (1985). Population study of causes, treatment and outcome of infertility. Br. Med. J. (Clin. Res. Ed.) 291, 1693–1697. doi:10.1136/ BMJ.291.6510.1693 Irvine, D. S., Twigg, J. P., Gordon, E. L., Fulton, N., Milne, P. A., and Aitken, R. J. (2000). DNA integrity in human spermatozoa: relationships with semen quality. J. Androl. 21, 33–44. Jones, R., Mann, T., and Sherins, R. J. (1978). Adverse effects of peroxidized lipid on human spermatozoa. Proc. R. Soc. Lond. B Biol. Sci. 201, 413–417. doi:10.1098/RSPB.1978.0053 Jones, R., Mann, T., and Sherins, R. J. (1979). Peroxidative breakdown of phospholipids in human spermatozoa: spermicidal effects of fatty acid peroxides and protective action of seminal plasma. Fertil. Steril. 31, 531–537. Kao, S. H., Chao, H. T., Chen, H. W., Hwang, T. I., Liao, T. L., and Wei, Y. H. (2008). Increase of oxidative stress in human sperm with lower motility. Fertil. Steril. 89, 1183–1190. doi:10.1016/J.FERTNSTERT.2007.05.029 Katen, A. L., and Roman, S. D. (2015). The genetic consequences of paternal acrylamide exposure and potential for amelioration. Mutat. Res. 777, 91–100. doi:10.1016/J.MRFMMM.2015.04.008 Koppers, A. J., De Iuliis, G. N., Finnie, J. M., McLaughlin, E. A., and Aitken, R. J. (2008). Significance of mitochondrial reactive oxygen species in the generation of oxidative stress in spermatozoa. J. Clin. Endocrinol. Metab. 93, 3199–3207. doi:10.1210/JC.2007-2616 Koppers, A. J., Mitchell, L. A., Wang, P., Lin, M., and Aitken, R. J. (2011). Phosphoinositide 3-kinase signalling pathway involvement in a truncated apoptotic cascade associated with motility loss and oxidative DNA damage in human spermatozoa. Biochem. J. 436, 687–698. doi:10.1042/BJ20110114 Lagos-Cabre´, R., and Moreno, R. D. (2012). Contribution of environmental pollutants to male infertility: a working model of germ cell apoptosis induced by plasticizers. Biol. Res. 45, 5–14. doi:10.4067/S0716- 97602012000100001 Lane, M., McPherson, N. O., Fullston, T., Spillane, M., Sandeman, L., Kang, W. X., and Zander-Fox, D. L. (2014). Oxidative stress in mouse sperm impairs embryo development, fetal growth and alters adiposity and glucose regulation in female offspring. PLoS One 9, e100832. doi:10.1371/JOURNAL.PONE.0100832 Liang, R., Senturker, S., Shi, X., Bal, W., Dizdaroglu, M., and Kasprzak, K. S. (1999). Effects of Ni(II) and Cu(II) on DNA interaction with the N-terminal sequence of human protamine P2: enhancement of binding and mediation of oxidative DNA strand scission and base damage. Carcinogenesis 20, 893–898. doi:10.1093/CARCIN/20.5.893 Lundbaek, J. A., and Andersen, O. S. (1994). Lysophospholipids modulate channel function by altering the mechanical properties of lipid bilayers. J. Gen. Physiol. 104, 645–673. doi:10.1085/JGP.104.4.645 MacLeod, J. (1943). The role of oxygen in the metabolism and motility of human spermatozoa. Am. J. Physiol. 138, 512–518. Metzler-Guillemain,C.,Victorero,G.,Lepoivre,C.,Bergon,A.,Yammine,M., Perrin, J., Sari-Minodier, I., Boulanger, N., Rihet, P., and Nguyen, C. (2015). Sperm mRNAs and microRNAs as candidate markers for the impact of toxicants on human spermatogenesis: an application to tobacco smoking. Syst Biol Reprod Med 61, 139–149. doi:10.3109/19396368. 2015.1022835 Moazamian, R., Polhemus, A., Connaughton, H., Fraser, B., Whiting, S., Gharagozloo, P., and Aitken, R. J. (2015). Oxidative stress and human spermatozoa: diagnostic and functional significance of aldehydes gen- erated as a result of lipid peroxidation. Mol. Hum. Reprod. 21, 502–515. doi:10.1093/MOLEHR/GAV014 Morielli, T., and O’Flaherty, C. (2015). Oxidative stress impairs function and increases redox protein modifications in human spermatozoa. Reproduction 149, 113–123. doi:10.1530/REP-14-0240 Morimoto, H., Iwata, K., Ogonuki, N., Inoue, K., Atsuo, O., Kanatsu- Shinohara, M., Morimoto, T., Yabe-Nishimura, C., and Shinohara, T. (2013). ROS are required for mouse spermatogonial stem cell self- renewal. Cell Stem Cell 12, 774–786. doi:10.1016/J.STEM.2013.04.001 Muratori, M., Tamburrino, L., Marchiani, S., Cambi, M., Olivito, B., Azzari, C., Forti, G., and Baldi, E. (2015). Investigation on the origin of sperm DNA fragmentation: role of apoptosis, immaturity and oxida- tive stress. Mol. Med. 21, 109–122. doi:10.2119/MOLMED.2014.00158 Musset,B.,Clark,R.A.,DeCoursey,T.E.,Petheo,G.L.,Geiszt,M., Chen, Y., Cornell, J.E., Eddy, C. A., Brzyski,R. G., and El Jamali, A.(2012).NOX5 in human spermatozoa: expression, function, and regulation. J. Biol. Chem. 287, 9376–9388. doi:10.1074/JBC.M111.314955 Nakamura, H., Kimura, T., Nakajima, A., Shimoya, K., Takemura, M., Hashimoto, K., Isaka, S., Azuma, C., Koyama, M., and Murata, Y. (2002). Detection of oxidative stress in seminal plasma and fractionated sperm from subfertile male patients. Eur. J. Obstet. Gynecol. Reprod. Biol. 105, 155–160. doi:10.1016/S0301-2115(02)00194-X Nishikawa, T., Tomori, Y., Yamashita, S., and Shimizu, S. (1989). Inhibition of Naþ ,Kþ -ATPase activity by phospholipase A2 and several lysopho- spholipids: possible role of phospholipase A2 in noradrenaline release from cerebral cortical synaptosomes. J. Pharm. Pharmacol. 41, 450–458. doi:10.1111/J.2042-7158.1989.TB06499.X Noblanc, A.,Damon-Soubeyrand,C., Karrich, B., Henry-Berger,J., Cadet,R., Saez, F., Guiton, R., Janny, L., Pons-Rejraji, H., Alvarez, J. G., Jr, Drevet, J. R., and Kocer, A. (2013). DNA oxidative damage in mamma- lian spermatozoa: where and why the male nucleus is impacted? Free Radic. Biol. Med. 65, 719–723. doi:10.1016/J.FREERADBIOMED. 2013.07.044 O’Flaherty, C. (2014). Iatrogenic genetic damage of spermatozoa. Adv. Exp. Med. Biol. 791, 117–135. doi:10.1007/978-1-4614-7783-9_8 Ohno, M.,Sakumi,K.,Fukumura, R.,Furuichi,M., Iwasaki,Y.,Hokama, M., Ikemura, T., Tsuzuki, T., Gondo, Y., and Nakabeppu, Y. (2014). 8-Oxoguanine causes spontaneous de novo germline mutations in mice. Sci. Rep. 4, 4689. doi:10.1038/SREP04689 Ostermeier, G. C., Goodrich, R. J., Moldenhauer, J. S., Diamond, M. P., and Krawetz, S. A. (2005). A suite of novel human spermatozoal RNAs. J. Androl. 26, 70–74. Palmer, N. O., Bakos, H. W., Owens, J. A., Setchell, B. P., and Lane, M. (2012). Diet and exercise in an obese mouse fed a high-fat diet improve metabolic health and reverse perturbed sperm function. Am. J. Physiol. Endocrinol. Metab. 302, E768–E780. doi:10.1152/AJPENDO.00401. 2011 Prescott, J., Du, M., Wong, J. Y., Han, J., and De Vivo, I. (2012). Paternal age at birth is associated with offspring leukocyte telomere length in the Nurses’ Health Study. Hum. Reprod. 27, 3622–3631. doi:10.1093/ HUMREP/DES314 Reichart, M., Kahane, I., and Bartoov, B. (2000). In vivo and in vitro impairment of human and ram sperm nuclear chromatin integrity by sexually transmitted Ureaplasma urealyticum infection. Biol. Reprod. 63, 1041–1048. doi:10.1095/BIOLREPROD63.4.1041 Rivlin, J., Mendel, J., Rubinstein, S., Etkovitz, N., and Breitbart, H. (2004). Role of hydrogen peroxide in sperm capacitation and acrosome reaction. Biol. Reprod. 70, 518–522. doi:10.1095/BIOLREPROD.103.020487 Rodriguez, P. C., and Beconi, M. T. (2009). Peroxynitrite participates in mechanisms involved in capacitation of cryopreserved cattle. Anim. Reprod. Sci. 110, 96–107. doi:10.1016/J.ANIREPROSCI.2007.12.017 Sakamoto, Y., Ishikawa, T., Kondo, Y., Yamaguchi, K., and Fujisawa, M. (2008). The assessment of oxidative stress in infertile patients with varicocele. BJU Int. 101, 1547–1552. doi:10.1111/J.1464-410X.2008. 07517.X Oxidative stress in spermatozoa Reproduction, Fertility and Development 9
  • 11. Sanocka, D., Miesel, R., Jedrzejczak, P., and Kurpisz, M. K. (1996). Oxidative stress and male infertility. J. Androl. 17, 449–454. Santiso,R.,Tamayo,M.,Gosa´lvez,J.,Johnston,S.,Marin˜o,A.,Ferna´ndez,C., Losada, C., and Ferna´ndez, J. L. (2012). DNA fragmentation dynamics allows the assessment of cryptic sperm damage in human: evaluation of exposure to ionizing radiation, hyperthermia, acidic pH and nitric oxide. Mutat. Res. 734, 41–49. doi:10.1016/J.MRFMMM. 2012.03.006 Sharma, R. K., and Agarwal, A. (1996). Role of reactive oxygen species in male infertility. Urology 48, 835–850. doi:10.1016/S0090-4295(96) 00313-5 Shen, H., and Ong, C. (2000). Detection of oxidative DNA damage in human sperm and its association with sperm function and male infertility. Free Radic. Biol. Med. 28, 529–536. doi:10.1016/S0891-5849(99) 00234-8 Showell, M. G., Mackenzie-Proctor, R., Brown, J., Yazdani, A., Stankiewicz, M. T., and Hart, R. J. (2014). Antioxidants for male subfertility. Cochrane Database Syst. Rev. 12, CD007411. Shukla, K. K., Mahdi, A. A., and Rajender, S. (2012). Apoptosis, spermato- genesis and male infertility. Front. Biosci. (Elite Ed.) E4, 746–754. doi:10.2741/E415 Sibirtsev, J. T., Shastina, V. V., Menzorova, N. I., Makarieva, T. N., and Rasskazov, V. (2011). A Ca2þ , Mg2þ -dependent DNase involvement in apoptotic effects in spermatozoa of sea urchin Strongylocentrotus intermedius induced by two-headed sphingolipid, rhizochalin. Mar. Biotechnol. (NY) 13, 536–543. doi:10.1007/S10126-010-9324-9 Simo˜es, R., Feitosa, W. B., Siqueira, A. F., Nichi, M., Paula-Lopes, F. F., Marques, M. G., Peres, M. A., Barnabe, V. H., Visintin, J. A., and Assumpc¸a˜o, M. E. (2013). Influence of bovine sperm DNA fragmenta- tion and oxidative stress on early embryo in vitro development outcome. Reproduction 146, 433–441. doi:10.1530/REP-13-0123 Singh, N. P., and Stephens, R. E. (1998). X-Ray induced DNA double- strand breaks in human sperm. Mutagenesis 13, 75–79. doi:10.1093/ MUTAGE/13.1.75 Singh, N. P., Muller, C. H., and Berger, R. E. (2003). Effects of age on DNA double-strand breaks and apoptosis in human sperm. Fertil. Steril. 80, 1420–1430. doi:10.1016/J.FERTNSTERT.2003.04.002 Smith, T. B., De Iuliis, G. N., Lord, T., and Aitken, R. J. (2013a). The senescence-accelerated mouse prone 8 as a model for oxidative stress and impaired DNA repair in the male germ line. Reproduction 146, 253–262. doi:10.1530/REP-13-0186 Smith, T. B., Dun, M. D., Smith, N. D., Curry, B. J., Connaughton, H. S., and Aitken, R. J. (2013b). The presence of a truncated base excision repair pathway in human spermatozoa that is mediated by OGG1. J. Cell Sci. 126, 1488–1497. doi:10.1242/JCS.121657 Sotolongo, B., Huang, T. T., Isenberger, E., and Ward, W. S. (2005). An endogenous nuclease in hamster, mouse, and human spermatozoa cleaves DNA into loop-sized fragments. J. Androl. 26, 272–280. Soubry, A. (2015). Epigenetic inheritance and evolution: a paternal per- spective on dietary influences. Prog. Biophys. Mol. Biol. 118, 79–85. doi:10.1016/J.PBIOMOLBIO.2015.02.008 Tosic, J., and Walton, A. (1946). Formation of hydrogen peroxide by spermatozoa and its inhibitory effect on respiration. Nature 158, 485. doi:10.1038/158485A0 Tosic, J., and Walton, A. (1950). Metabolism of spermatozoa. The formation and elimination of hydrogen peroxide by spermatozoa and effects on motility and survival. Biochem. J. 47, 199–212. van Kuijk, F. J., Handelman, G. J., and Dratz, E. A. (1985). Consecutive action of phospholipase A2 and glutathione peroxidase is required for reduction of phospholipid hydroperoxides and provides a convenient method to determine peroxide values in membranes. J. Free Radic. Biol. Med. 1, 421–427. doi:10.1016/0748-5514(85)90156-4 Vernet, P., Fulton, N., Wallace, C., and Aitken, R. J. (2001). Analysis of reactive oxygen species generating systems in rat epididymal spermato- zoa. Biol. Reprod. 65, 1102–1113. doi:10.1095/BIOLREPROD65.4.1102 Wang, X., Sharma, R. K., Sikka, S. C., Thomas, A. J., Jr, Falcone, T., and Agarwal, A. (2003). Oxidative stress is associated with increased apoptosis leading to spermatozoa DNA damage in patients with male factor infertility. Fertil. Steril. 80, 531–535. doi:10.1016/S0015-0282 (03)00756-8 Weir, C. P., and Robaire, B. (2007). Spermatozoa have decreased antioxi- dant enzymatic capacity and increased reactive oxygen species produc- tion during aging in the Brown Norway rat. J. Androl. 28, 229–240. doi:10.2164/JANDROL.106.001362 Zalata, A., Hafez, T., Mahmoud, A., and Comhaire, F. (1995). Relationship between resazurin reduction test, reactive oxygen species generation, and gamma-glutamyltransferase. Hum. Reprod. 10, 1136–1140. Zhou, D., Wang, H., Zhang, J., Gao, X., Zhao, W., and Zheng, Y. (2010). Di-n-butyl phthalate (DBP) exposure induces oxidative damage in testes of adult rats. Syst. Biol. Reprod. Med. 56, 413–419. doi:10.3109/ 19396368.2010.509902 Zribi, N., Chakroun, N. F., Ben Abdallah, F., Elleuch, H., Sellami, A., Gargouri, J., Rebai, T., Fakhfakh, F., and Keskes, L. A. (2012). Effect of freezing–thawing process and quercetin on human sperm survival and DNA integrity. Cryobiology 65, 326–331. doi:10.1016/J.CRYOBIOL. 2012.09.003 www.publish.csiro.au/journals/rfd 10 Reproduction, Fertility and Development R. J. Aitken et al. View publication statsView publication stats