ArticlePDF AvailableLiterature Review

Oxidative stress and male reproductive health

Authors:

Abstract and Figures

One of the major causes of defective sperm function is oxidative stress, which not only disrupts the integrity of sperm DNA but also limits the fertilizing potential of these cells as a result of collateral damage to proteins and lipids in the sperm plasma membrane. The origins of such oxidative stress appear to involve the sperm mitochondria, which have a tendency to generate high levels of superoxide anion as a prelude to entering the intrinsic apoptotic cascade. Unfortunately, these cells have very little capacity to respond to such an attack because they only possess the first enzyme in the base excision repair (BER) pathway, 8-oxoguanine glycosylase 1 (OGG1). The latter successfully creates an abasic site, but the spermatozoa cannot process the oxidative lesion further because they lack the downstream proteins (APE1, XRCC1) needed to complete the repair process. It is the responsibility of the oocyte to continue the BER pathway prior to initiation of S-phase of the first mitotic division. If a mistake is made by the oocyte at this stage of development, a mutation will be created that will be represented in every cell in the body. Such mechanisms may explain the increase in childhood cancers and other diseases observed in the offspring of males who have suffered oxidative stress in their germ line as a consequence of age, environmental or lifestyle factors. The high prevalence of oxidative DNA damage in the spermatozoa of male infertility patients may have implications for the health of children conceivedin vitro and serves as a driver for current research into the origins of free radical generation in the germ line.
Proposed cycle of cause and effect by which oxidative stress in the male germ line impacts upon the health and well-being of future generations. (1) A variety of primary factors can initiate oxidative stress in the male germ line including infection, age, obesity and exposure to a variety of adverse environmental influences. (2) This initial oxidative stress induces lipid peroxidation culminating in the production of lipid aldehydes such as 4HNE, which bind to proteins in the mitochondrial electron transport chain, stimulating the generation of reactive oxygen species (ROS). The latter stimulate yet more lipid peroxidation in a self-propagating cycle that culminates in apoptosis. (3) One of the most sensitive targets of oxidative stress is the DNA in the sperm nucleus, generating 8-hydroxy, 2’deoxyguanosine (8OHdG) base adducts. (4) The first enzyme in the base excision repair pathway, 8-oxoguanine glycosylase 1 (OGG1), is present in spermatozoa and its activity creates abasic sites. The remainder of the DNA repair pathway is present in the oocyte. The oocyte has to repair the DNA damage brought into the zygote by the fertilizing spermatozoon before the initiation of S-phase for the first mitotic division. (5) If the oocyte makes a mistake at this stage of DNA repair, it has the potential to create a mutation that will be represented in every cell in the body and could account for the range of pathologies seen in the offspring of fathers exhibiting high levels of oxidative DNA damage in their spermatozoa. Abbreviations: IVF, in vitro fertilization; ROS, reactive oxygen species.
… 
Content may be subject to copyright.
Asian Journal of Andrology (2014) 16, (31–38)
© 2014 AJA, SIMM & SJTU. All rights reserved 1008-682X
www.asiaandro.com; www.ajandrology.com
and Curson5 con rmed the presence of such an oxidase in bovine
spermatozoa and demonstrated that it was the dead cells in any given
ejaculate that were particularly active in generating hydrogen peroxide
in response to phenylalanine and that the oxidative stress generated
in this manner could have an impact on the live cells present in the
immediate vicinity.  e cytotoxic e ect of ROS generated on exposure
to the phenylalanine in cryostorage medium could be rescued by the
concomitant presence of catalase, con rming hydrogen peroxide as
the cytotoxic principle.
e notion that oxidative stress might also be a factor in the etiology
of defective sperm function in our species was advanced independently
by Aitken and Clarkson6 and Alvarez etal.7 in 1987. An important but
o en overlooked catalyst for this discovery was the development of
a technique for objectively measuring sperm function, in the form of
the zona-free hamster oocyte penetration assay introduced by another
pioneer of modern andrology, Ryuzo Yanagimachi.8 Up until this point,
the  eld had lacked objective methods for the measurement of human
sperm function aside from motility.  e hamster oocyte penetration
assay provided an objective means of determining the competence
of human spermatozoa to capacitate, undergo the acrosome reaction
and generate a fusogenic equatorial segment capable of initiating
fusion with the vitelline membrane of the oocyte. In the age of
intracytoplasmic sperm injection(ICSI), the hamster oocyte model
can also provide critical information on the ability of spermatozoa
to form a pronucleus.9–11 When combined with objective methods
for assessing sperm motility, this assay has been shown to give a very
accurate assessment of the fertilizing potential of human ejaculates.12,13
One of the interesting results secured with this assay was to demonstrate
INTRODUCTION
Male infertility is a relatively common condition a ecting approximately
1 in 20 of the male population. In a vast majority of infertile subjects
su cient numbers of spermatozoa are generated to initiate a pregnancy;
however, the functionality of the spermatozoa has been compromised.
As a result, defective sperm function is held to be the largest, single
and de ned cause of human infertility.1 e primary causes of defective
sperm function are undoubtedly multifactorial, involving a range of
primary genetic, lifestyle and environmental factors, acting alone or,
more frequently, in combination. However at the level of the gamete,
the integration of these various forces frequently culminates in a state
of oxidative stress that impairs the functional and structural integrity
of these highly di erentiated cells.  e rst suggestion that oxidative
stress might play a role in the etiology of defective sperm function came
from one of the pioneers of modern andrology, DrJohn MacLeod.2
He published an important paper in 1943 which demonstrated that
in oxygenated medium human spermatozoa rapidly lost motility via
mechanisms that could be rescued by the concomitant presence of
catalase, a speci c scavenger of hydrogen peroxide.  e fundamental
notion that spermatozoa could generate reactive oxygen species(ROS),
speci cally hydrogen peroxide, was con rmed by Tosic and Walton in
a paper published in Nature in 1946.3 In this, and a follow-up paper
published in 1950,4 these authors presented impressive biochemical
evidence that bovine spermatozoa could not only generate hydrogen
peroxide but also that this reactive oxygen metabolite was damaging
to sperm function. In this speci c case, the authors demonstrated the
involvement of an L-amino acid oxidase with a particular a nity for
aromatic amino acids such as phenylalanine. Many years later Shannon
INVITED REVIEW
Oxidative stress and male reproductive health
Robert J Aitken, Tegan B Smith, Matthew S Jobling, Mark A Baker , Geo ry N DeIuliis
One of the major causes of defective sperm function is oxidative stress, which not only disrupts the integrity of sperm DNA but also
limits the fertilizing potential of these cells as a result of collateral damage to proteins and lipids in the sperm plasma membrane.
The origins of such oxidative stress appear to involve the sperm mitochondria, which have a tendency to generate high levels of
superoxide anion as a prelude to entering the intrinsic apoptotic cascade. Unfortunately, these cells have very little capacity to
respond to such an attack because they only possess the fi rst enzyme in the base excision repair (BER) pathway, 8-oxoguanine
glycosylase 1 (OGG1). The latter successfully creates an abasic site, but the spermatozoa cannot process the oxidative lesion further
because they lack the downstream proteins (APE1, XRCC1) needed to complete the repair process. It is the responsibility of the
oocyte to continue the BER pathway prior to initiation of S-phase of the fi rst mitotic division. If a mistake is made by the oocyte
at this stage of development, a mutation will be created that will be represented in every cell in the body. Such mechanisms may
explain the increase in childhood cancers and other diseases observed in the offspring of males who have suffered oxidative stress
in their germ line as a consequence of age, environmental or lifestyle factors. The high prevalence of oxidative DNA damage in the
spermatozoa of male infertility patients may have implications for the health of children conceived
in vitro
and serves as a driver
for current research into the origins of free radical generation in the germ line.
Asian Journal of Andrology (2014) 16, (31–38); doi: 10.4103/1008-682X.122203; published online: 16 December 2013
Keywords: DNA damage; oxidative stress; oxoguanine glycosylase 1; oocyte; spermatozoa
Priority Research Centre in Reproductive Science, Discipline of Biological Sciences, Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia.
Correspondence: Prof. RJ Aitken (john.aitken@newcastle.edu.au)
Received: 09-08-2013; Revised: 12-09-2013; Accepted: 22-09-2013
Open Access
Reproductive Health
Oxidative stress
RJ Aitken et al
30
Asian Journal of Andrology
that defective sperm function was evident in infertile men, even
when their spermatozoa had been treated with the divalent cation
ionophore, A23187 in order to induce an acrosome reaction.14 is
result indicated that whatever the lesions are in defective spermatozoa,
they lay downstream of the calcium in ux normally triggered when
the spermatozoa make contact with the cumulus-oocyte complex.
Such results suggested that there must be some defect in the plasma
membrane of functionally compromised human spermatozoa that
prevents them from fusing with the vitelline membrane of the oocyte.
It was this quest for an explanation of failed membrane fusion in the
hamster oocyte assay that led us to the concept that lipid peroxidation
was a key factor in the etiology of defective sperm function.
Spermatozoa are particularly vulnerable to lipid peroxidation because
they contain high concentrations of unsaturated fatty acids, particularly
docosahexaenoic acid with six double bonds per molecule.15 e latter
are vulnerable to free radical attack because the carbon hydrogen
dissociation energies are lowest at the bisallylic methylene position.
As a consequence, the hydrogen abstraction event that initiates lipid
peroxidation is promoted, generating a carbon-centered lipid radical
that then combines with oxygen to generate peroxyl(ROO•) and
alkoxyl(RO•) radicals that, in order to stabilize, abstract hydrogen
atoms from adjacent carbons. These chemical reactions create
additional lipid radicals that then perpetuate the lipid peroxidation
chain reaction, culminating in the generation of small molecular
mass electrophilic lipid aldehydes such as 4-hydroxynonenal(4HNE),
acrolein and malondialdehyde. Added to this vulnerability, we have
shown that sperm mitochondria respond to the presence of free
unsaturated fatty acids with a dramatic increase in ROS generation; the
greater the level of unsaturation, the greater the level of the stimulatory
e ect. Esteri cation of the fatty acid counters this pro-oxidant e ect
suggesting that it is the amphiphilic properties of these molecules
that are central to their ROS-inducing activity, possibly by de ning
the orientation of the fatty acids in relation to the mitochondrial
electron transport chain. In this context, it is signi cant that defective
human spermatozoa possess abnormally high cellular contents of free
polyunsaturated fatty acids, the levels of which are positively correlated
with mitochondrial superoxide generation.17
us, defective human spermatozoa are particularly vulnerable
to oxidative stress because they contain a superabundance of free
unsaturated fatty acids that trigger ROS generation by the sperm
mitochondria and induce high levels of lipid peroxidation. To make
matters worse, the products of lipid peroxidation in the form of small
molecular mass electrophilic aldehydes such as 4HNE or acrolein, are
also capable of triggering ROS generation by the sperm mitochondria.18
is ability of lipid aldehydes generated as a consequence of lipid
peroxidation to trigger mitochondrial ROS generation appears to be a
function of their capacity to adduct onto proteins in the mitochondrial
electron transport chain, such as succinic acid dehydrogenase.18 As a
consequence of these interactions, it is evident that oxidative stress in
human spermatozoa is a self-propagating cycle that, once initiated,
will inevitably lead to oxidative damage, a loss of functionality and
ultimately, cell death(Figure1).
OXIDATIVE STRESS SPERM FUNCTION, DNA INTEGRITY AND
CELL DEATH
One of the first functions affected by oxidative stress and lipid
peroxidation is sperm motility. Correlations between lipid peroxide
formation and sperm movement have been repeatedly observed in a
variety of di erent species.15,19,20 Experiments involving exposure of
mammalian spermatozoa to a variety of ROS using the xanthine oxidase
ROS-generating system have also clearly demonstrated the susceptibility
of sperm motility to oxidative attack and identi ed hydrogen peroxide
as the most cytotoxic oxygen metabolite in this context; catalase, but
not superoxide dismutase, preventing sperm motility loss under such
circumstances.21–24 e mechanisms by which motility is lost when
spermatozoa are under oxidative stress is not known with certainty, but
both oxidative damage to the axoneme and depletion of intracellular
adenosine triphosphate(ATP) appear to be involved.25–27
Notwithstanding the dramatic e ects that high levels of exposure to
ROS have on sperm motility, it is also evident that oxidative stress can
compromise the fertilizing capacity of spermatozoa under conditions
where motility is normal.28,29 Under these circumstances, it is the
capacity of the spermatozoa to fuse with the vitelline membrane of
the oocyte which is impaired. Acareful dose-dependent analysis of
the impact of oxidative stress on sperm-oocyte fusion demonstrated
a biphasic response which beautifully encapsulates the complex
relationship between ROS and sperm function.30 us, at low levels of
oxidative stress, sperm-oocyte fusion rates were enhanced, presumably
as a consequence of:(i) the positive role that ROS are known to play
in driving the tyrosine phosphorylation events associated with sperm
capacitation,31 and(ii) the importance of sterol oxidation in facilitating
the e ux of cholesterol from the sperm plasma membrane.32 However,
at higher levels of oxidative stress the induction of lipid peroxidation in
the plasma membrane is associated with a decline in the competence for
sperm-oocyte fusion, possibly due to the direct induction of oxidative
damage to proteins involved in the fusion process, rather than any
change in the  uidity of the sperm plasma membrane.33
Figure 1: Proposed cycle of cause and effect by which oxidative stress
in the male germ line impacts upon the health and well-being of future
generations. (1) A variety of primary factors can initiate oxidative stress
in the male germ line including infection, age, obesity and exposure to a
variety of adverse environmental infl uences. (2) This initial oxidative stress
induces lipid peroxidation culminating in the production of lipid aldehydes
such as 4HNE, which bind to proteins in the mitochondrial electron
transport chain, stimulating the generation of reactive oxygen species
(ROS). The latter stimulate yet more lipid peroxidation in a self-propagating
cycle that culminates in apoptosis. (3) One of the most sensitive targets
of oxidative stress is the DNA in the sperm nucleus, generating 8-hydroxy,
2’deoxyguanosine (8OHdG) base adducts. (4) The fi rst enzyme in the base
excision repair pathway, 8-oxoguanine glycosylase 1 (OGG1), is present in
spermatozoa and its activity creates abasic sites. The remainder of the DNA
repair pathway is present in the oocyte. The oocyte has to repair the DNA
damage brought into the zygote by the fertilizing spermatozoon before the
initiation of S-phase for the fi rst mitotic division. (5) If the oocyte makes a
mistake at this stage of DNA repair, it has the potential to create a mutation
that will be represented in every cell in the body and could account for the
range of pathologies seen in the offspring of fathers exhibiting high levels
of oxidative DNA damage in their spermatozoa. Abbreviations: IVF, in vitro
fertilization; ROS, reactive oxygen species.
Oxidative stress
RJ Aitken et al
31
Asian Journal of Andrology
OXIDATIVE STRESS AND DNA DAMAGE
When human spermatozoa were exposed to increasing levels of
hydrogen peroxide it was not just the fertilizing potential of the cells
that followed a biphasic pattern of change, the DNA in the sperm
nucleus behaved similarly. At low levels of oxidative stress DNA
damage was diminished, possibly because of the powerful role played
by glutathione peroxidase in e ecting the cross linking of sperm
chromatin. However, at higher levels of oxidative stress, the sperm
chromatin started to fragment.30 Importantly, the losses of fertilizing
potential and DNA integrity occurred at di erent rates, with the latter
being the more sensitive. As a result, spermatozoa that had been driven
to a high state of readiness for fertilization by low levels of oxidative
stress were also found to exhibit signi cantly elevated levels of DNA
damage.30 is is an extremely signi cant observation, since it suggests
a mechanism by which environmental in uences on the paternal germ
line could have a major impact on the health trajectory of any progeny.
e oxidized base adduct, 8-hydroxy, 2deoxyguanosine(8OHdG),
has been used in studies to demonstrate that oxidative DNA damage is
signi cantly elevated in the spermatozoa of patients attending infertility
clinics.34,35 Furthermore, the levels of 8OHdG expression have been
shown to correlate highly with the measurement of DNA damage
in spermatozoa, as measured by the TUNEL or sperm chromatin
dispersion assays.34,36 Indeed, the correlation between 8OHdG
formation and DNA damage is so high that we have been forced
to conclude that most DNA damage in spermatozoa is oxidatively
induced. In order to understand why this would be the case we need
to appreciate the particular architecture of human spermatozoa and
the major points of di erence with somatic cells in terms of the
mechanisms regulating apoptosis.
Apoptosis is the default condition for spermatozoa. In the
absence of fertilization, most spermatozoa will become senescent and
default to an apoptotic state. In somatic cells, apoptosis is associated
with extensive nuclear fragmentation as a consequence of nucleases
released from the mitochondria(e.g., endonuclease G) or activated
in the cytosol(e.g.,caspase-activated DNase). However, spermatozoa
are distinguished from every other cell type in biology in having a
nucleus that is physically separated from the mitochondria and most
of the cytoplasm. As a consequence, even when apoptosis is activated
in these cells using inhibitors of PI3 kinase such as wortmannin,37
the nucleases associated with this process remain resolutely locked
within the midpiece of the cell and do not penetrate the nuclear
compartment(Figure 2). Thus, even when apoptosis is induced
in suspensions of human spermatozoa, the DNA does not become
cleaved by nucleases, at least in the short-term.37 e only products
of apoptosis that can damage sperm DNA are the ROS generated
by the mitochondria. Mitochondria are potent generators of ROS
in spermatozoa and this activity becomes enhanced as soon as the
spermatozoa default to an apoptotic state. Indeed mitochondrial ROS
generation is one of the  rst signs that these cells have engaged the
intrinsic apoptotic cascade.37,38 It is for thi s re aso n th at m ost of t he D NA
damage observed in spermatozoa is oxidative in nature.
If nucleases are ever involved, it would be at the very beginning or
the very end of sperm existence. During late spermatogenesis, spermatid
DNA becomes enzymatically cleaved in order to relieve the torsional
stress associated with sperm chromatin compaction. Such endogenous
nicks are thought to be resolved by topoisomerase before spermiation,
however in pathological cases, such repair mechanisms may be de cient
leading to the persistence of nicked DNA into the mature gamete.39,40
e possibility that DNA damage in spermatozoa has its origins during
spermiation is supported by the profound correlation, which has been
observed between DNA fragmentation and chromatin compaction in
spermatozoa as detected by chromomycin A3  uorescence.34,41 Viewed
in this light, both DNA fragmentation and poor chromatin compaction
may be regarded as independent signs of errors in spermiogenesis. An
alternative explanation is that these two events are causally related.
According to this ‘two-step’ model, errors in spermiogenesis initially lead
to poor chromatin protamination and create a state of vulnerability in the
spermatozoa. In the second step, spermatozoa are exposed to oxidative
stress from a variety of sources including exposure to exogenous ROS
as a consequence of leukocyte in ltration, or endogenous ROS triggered
by entry into the intrinsic apoptotic cascade, ultimately resulting in
enhanced oxidative DNA damage. Of course, this two-step hypothesis42,43
to explain the origins of oxidative DNA damage is not necessarily
exclusive of the concept that nuclease-mediated DNA nicks might
persist in spermatozoa from late spermatogenesis. Nevertheless, the high
correlation that has been observed between oxidative DNA damage and
DNA fragmentation suggests that most of the DNA damage is occurring
following spermiation as a result of enhanced vulnerability to oxidative
stress.42 e only other time that nucleases may contribute to DNA
damage in the male germ line would be at the end of a spermatozoons
life when intracellular nucleases released during the perimortem as the
internal structure of these cells starts to break down, or extracellular
nucleases released from the male reproductive tract, may aid in the  nal
disposal of these cells by the phagocytic armies of the immune system.44,45
DNA REPAIR IN SPERMATOZOA
The importance of oxidative stress in the mechanisms by which
sperm DNA becomes damaged is also indicated by a consideration
Figure 2: The unique architecture of spermatozoa infl uences the impact of
apoptosis on DNA integrity. (a) Conventional somatic cells feature a centrally
placed nucleus surrounded by mitochondria embedded in the cytoplasm.
Under these circumstances, endonucleases activated in the cytoplasm or
released from the mitochondria during apoptosis are able to move into
the nucleus (arrows) and cleave the DNA. (b) Spermatozoa are completely
different from such somatic cells because their mitochondria (stained
black) and most of their cytoplasm are located in the midpiece of the
cell, physically separated from the nucleus. (c) As a consequence of this
compartmentalization key effectors of apoptosis such as apoptosis inducing
factor (AIF) or Endonuclease G (Endo G) remain resolutely locked in the
sperm midpiece even when apoptosis is induced by the powerful PI3 kinase
inhibitor, wortmannin and cannot move into the sperm nucleus. Because
of this physical limitation, most DNA damage in mature spermatozoa is
induced by membrane permeant reactive oxygen species emanating from
the mitochondria, rather than nucleases.
b
ac
Oxidative stress
RJ Aitken et al
32
Asian Journal of Andrology
of the DNA repair strategies these cells are capable of employing.
Incorporated into the subcellular structure of the sperm nucleus and
mitochondria is an 8-oxoguanine glycosylase, known as 8-oxoguanine
glycosylase1(OGG1).46 When sperm DNA experiences an oxidative
attack OGG1 immediately clips the 8OHdG residues out of the DNA
generating an abasic site, releasing the oxidized base into the extracellular
space.  e next enzyme in the base excision repair(BER) pathway, APE1,
then incises DNA at the phosphate groups3’ and 5’ to the baseless site
leaving 3’-OH and 5’-phosphate termini ready for the insertion of a
new base. Spermatozoa do not possess this enzyme.46 As a result, they
carry their abasic sites into the oocyte for continuation of the repair
process(Figure1). For its part, the oocyte engages in a round of DNA
repair immediately a er fertilization and puts S-phase on hold until this
activity has been completed.47,48 If the oocyte should make a mistake
during the completion of this post-fertilization repair process, it creates
the potential for de novo mutations in the o spring which could have a
profound impact on the health and well-being of the latter(Figure1).
LIFESTYLE, AGE AND OXIDATIVE STRESS
Given this propensity for oxidative damage to sperm DNA and a
heavy reliance on OGG1 to cleave out damaged base adducts prior to
fertilization, it would not be surprising if factors that impeded OGG1
activity had a profound impact on fertility and the health of progeny.
e classic inhibitor of OGG1 activity is cadmium and the latter has a
long history of being associated with the etiology of male infertility.49,50
Importantly, cadmium exposure has been shown to increase levels of
DNA damage in spermatozoa51 and positive correlations have been
observed between 8OHdG levels in spermatozoa and the cadmium
concentration in seminal plasma.52 Since one of the classical sources of
cadmium is cigarette smoke, it is also no surprise to learn that men who
smoke heavily exhibit signi cantly elevated levels of oxidative DNA
damage in their spermatozoa.53 Furthermore, the impact of smoking
on 8OHdG levels in human spermatozoa is signi cantly impacted by
the presence of Ser326Cys polymorphism in the OGG1 gene.54 ose
individuals with variant Cys/Cys homozygosity for OGG1 showing
higher levels of sperm 8OHdG than wildtype homozygote carriers
(Ser/Ser).53 e fact that paternal(not maternal) smoking is associated
with a signi cant increase in the risk of childhood cancer in the
o spring55 is further testimony to the lasting clinical consequences of
cigarette smoking and the power of the relationship between oxidative
DNA damage in the paternal germ line and the long-term health
trajectory of the o spring(Table1).
If the oxidative DNA damage induced in the germ line as a
consequence of smoking can impact on the incidence of cancer in the
progeny, then surely any factor capable of inducing oxidative damage in
spermatozoa is potentially capable of profoundly in uencing the health
of children. Furthermore, because there is no particular proposed
order to the nature of the DNA damage or aberrant DNA repair in the
oocyte, we might anticipate that the range of pathologies generated
as a consequence of oxidative stress in the male germ line might be
considerable. Acase in point is paternal aging. It is well-recognized that
as men get older they do not stop producing spermatozoa; however,
the quality of their gametes exhibits a progressive age-related decline
as indicated by a highly signi cant, age-dependent increase in sperm
DNA damage.56,57 Studies on the brown Norway rat indicate that this
age-dependent increase in DNA damage in spermatozoa is associated
with a concomitant down regulation of genes associated with the BER
pathway and a corresponding increase in the levels of oxidative DNA
damage in the spermatozoa.58
This relationship between paternal age and oxidative DNA
damage in spermatozoa has also been indicated by recent studies
on the senescence-accelerated mouse prone 8(SAMP8).  is mouse
strain contains a suite of naturally occurring mutations resulting in
an accelerated senescence phenotype largely mediated by oxidative
stress, which is further enhanced by a mutation in the Ogg1 gene,
greatly reducing the ability of the enzyme to excise 8OHdG adducts. An
analysis of the reproductive phenotype of the SAMP8males revealed
a high level of DNA damage in caudal epididymal spermatozoa as
detected by the alkaline Comet assay. Furthermore, these lesions were
con rmed to be oxidative in nature, as demonstrated by signi cant
increases in 8OHdG adduct formation in the SAMP8 testicular tissue
and mature spermatozoa, relative to a control strain.
If aging is associated with oxidative DNA damage to spermatozoa
then we might expect to see these lesions re ected in the incidence of
morbidity in the o spring of ageing fathers. In fact, we see three major
kinds of paternal age-mediated pathology in the o spring; miscarriage,
dominant genetic mutations and complex neurological conditions, as
set out in Table1. One of the  rst paternally-mediated pathologies
to be detected was an increase in the incidence of dominant genetic
diseases in children as an exponential function of their fathers’ age.59
ese diseases classically include achondroplasia, Apert syndrome
and multiple endocrine neoplasias.60 The traditional explanation
given for the appearance of these conditions is that they represent the
Table 1: Summary of factors that are capable of causing oxidative DNA damage in the male germ line and their consequences for the offspring
Environmental or lifestyle factor Sperm damage Consequences for the offspring References
Smoking Oxidative damage to sperm DNA Increased incidence of childhood cancer 53-55
Age Oxidative damage to sperm DNA Increase in miscarriage
Increase in dominant genetic disease
Increased miscarriage
Increased neurological disorders such as autism, bipolar disease,
spontaneous schizophrenia and epilepsy
Increased death in offspring associated with congenital
malformations, injury and poisoning
Increased risk of cleft palate, diaphragmatic hernia, right
ventricular outfl ow tract obstruction and pulmonary valve stenosis
19,42,56,58-67
Infertility Oxidative damage to sperm DNA Unknown
Possible increase in birth defects
Possible increase in imprinting disorders
Increased hospitalization
42,70-77
Environmental toxicants,
insecticides, herbicides, heavy
metals and so on
Oxidative damage to sperm DNA Unknown 51,52,68
Oxidative stress
RJ Aitken et al
33
Asian Journal of Andrology
consequences of replication error in the germ line. As men age, their
germ cells experience multiple rounds of pre-meiotic replication,
and with each cellular iteration, the risk of a mutation occurring as a
consequence of replication error correspondingly increases. In certain
cases, such as the FGFR2( broblast growth factor receptor 2) mutation
associated with Apert syndrome, there does indeed appear to be a
correspondence between the incidence of this mutation in spermatozoa
and the appearance of the condition in children.60 However, the
underlying cause is not just replication error.61 e mutations that
cause this condition are thought to become over-represented in
the sperm population as a consequence of age-dependent clonal
expansion; mutant spermatogonial stem cells having a proliferative
advantage over non-mutated cells. Recent studies suggest that such
mutations occur in clusters within the seminiferous tubules possibly
as a consequence of failures of asymmetrical division within the germ
line.60 is germ line selection model may also explain the origins
of achondroplasia,62 although in this case there does appear to be a
discrepancy between the incidence of the mutation in spermatozoa
and the appearance of the disease in the progeny.63
An alternative explanation for paternal age e ects may be aberrant
repair of oxidative DNA damage in the fertilized oocyte, as suggested
above in the context of smoking.64 Such a mechanism could account for
the increase in miscarriage rates observed as a function of paternal age65
and could also contribute to the etiology of a range of other complex
polygenic conditions that correlate with the age of the father at the
moment of conception.  us, paternal age is also associated with an
increase in the incidence of complex polygenic neurological conditions
in the offspring including epilepsy, spontaneous schizophrenia,
bipolar disease and autism, as well as an increased rate of death in the
F1 generation associated with congenital malformations, injury and
poisoning.19 An analysis of birth defects has also revealed signi cant
associations between paternal age with the etiology of cle palate,
diaphragmatic hernia, right ventricular out ow tract obstruction and
pulmonary valve stenosis.66 As a result of recent studies conducted
on the Icelandic population, there is now powerful incontrovertible
evidence that the mutational load carried by children is correlated with
the age of their fathers at the moment of conception and that once this
load exceeds a certain critical level, overt pathologies such as autism
appear in the o spring.67 e link between this age-dependent increase
in mutational load in children and the aberrant repair of oxidative
sperm DNA damage in the zygote has yet to be de nitively established,
however such a relationship appears probable. Furthermore, given the
range of environmental and lifestyle factors that can in uence oxidative
stress in the germ line from pesticides to electromagnetic radiation,68
the potential contribution of such mechanisms to the integrity of the
human genome is signi cant(Table1).
Pertinent to this debate is the global increase in the use of assisted
reproductive technology(ART) to solve human infertility. In advanced
western countries such as Australia, nearly 4% of newborn children
are the product of assisted conception therapy.69 Since many of these
conceptions will have been triggered by male factor infertility and the
latter involves a high incidence of oxidative DNA damage in the germ
line, it is inevitable that conceptions are being achieved invitro with
severely DNA damaged spermatozoa, that could never have occurred
in vivo.70 One of the consequences of this trend is that we might
anticipate an increase in disease incidence in children conceived using
ART.  e emerging data on this point is suggestive but unsubstantiated.
us, the incidence of birth defects following ART is approximately
double the background rate and there is also evidence that imprinting
disorders are more frequent in children conceived invitro.71,72 Infants
produced by ART are also signi cantly more likely to be admitted to a
neonatal intensive care unit, to be hospitalized and to stay in hospital
longer than their naturally conceived counterparts.19 Recent studies
have also shown an increase in the hospitalization of ART o spring in
infancy and early childhood compared with spontaneously conceived
children, as well as abnormal patterns of retinal vascularization and an
increase in the incidence of undescended testicles in boys conceived
by ICSI.73–77
Similarly, there are many environmental toxicants(herbicides,
pesticides and so on) that will induce oxidative DNA damage in the
male germ line and are therefore potential contributors to disease
in the offspring.68 Notwithstanding their possible impact, such
transgenerational relationships still remain largely unexplored(Table1 ).
DNA REPAIR IN THE GERM LINE DURING SPERMATOGENESIS
Most of the above discussion has focused on the impact of oxidative
stress at the level of gamete. However, if the oxidative insult is earlier
in spermatogenesis, what are the likely consequences for fertility and
the health and well-being of the o spring? Under these circumstances,
severe oxidative DNA damage in germ cells entering meiosis will
simply precipitate an i ncrease in apoptosis.78 However, milder levels of
oxidative stress might induce compensatory mechanisms on the part
of the germ line that will favor survival of the o spring. An example
of such an e ect might be the impact of paternal ageing on telomere
length. As discussed above, ageing is associated with oxidative stress
in the germ line. One of the ways in which the germ line responds to
the stresses associated with ageing is to upregulate telomerase activity
and increase the length of telomeres in spermatozoa.79 Importantly
telomere length is a paternally inherited trait and so the o spring of
ageing fathers also have longer telomeres.80 Because telomere length
is associated with longevity,81 one of few positive consequences of
having an older father is that he may confer upon his children the
molecular basis for a long life. By contrast, if the paternal germ line
has experienced an oxidative stress post-meiotically, when telomerase
can no longer increase(as is typically the case in infertile patients)
then telomere length in the spermatozoa will be abnormally short and
the implications for the health of ART o spring, potentially serious.82
QUESTIONS FROM THE PANEL
Q1: Which lifestyle factors may cause oxidative stress?
A1: e factors that we know can cause oxidative stress in the
male germ line are age, subfertility and smoking. However, because
mitochondrial free radical generation is an early feature of apoptosis
in spermatozoa, it is probable that any factor capable of compromising
the vitality of male germ cells will initiate a state of oxidative stress.
Alist of potential factors has been compiled68 and includes exposure
to industrial pollutants such as bisphenol A, insecticides, pesticides,
nonionizing electromagnetic radiation, heavy metals and a variety of
small molecular mass toxicants, all of which are potentially in uenced
by interindividual di erences in occupation and lifestyle.
Q2: What is known about oxidative stress in the mitochondria of
male germ cells including spermatozoa, in response to di erent types
of environmental chemicals(e.g.,phthalates, dioxins and so on)? Is
there any speci city in such responses?
A2: Any factor that causes oxidative stress in the germ line will
automatically trigger mitochondrial ROS generation. It is a central
feature of the intrinsic apoptotic cascade. In addition, exposure to
free unsaturated fatty acids will trigger this activity by impeding
the  ow of electrons along the mitochondrial electron transport
chain.  e physiological signi cance of this association is indicated
Oxidative stress
RJ Aitken et al
34
Asian Journal of Andrology
by the correlation observed between the spontaneous levels of
mitochondrial ROS generation by human spermatozoa and their
cellular content of free arachidonic and decosahexaenoic acids.16,17
A variety of synthetic and natural electrophiles are also capable of
triggering superoxide release from the sperm mitochondria. In this
context, the ability of electrophilic aldehydes(e.g.,4HNE, acrolein and
malondialdehyde) generated as a consequence of lipid peroxidation to
trigger mitochondrial ROS generation is particularly signi cant.18 As
a consequence of this pathway, any environmental factor that triggers
oxidative stress in the germ line will potentiate the generation of further
oxidative stress as a direct result of lipid peroxidation. Environmental
factors such as dioxins are certainly capable of eliciting ROS generation
from sperm mitochondria in an experimental situation.84,85 However,
whether such toxicants contribute signi cantly towards the oxidative
stress observed in association with male infertility and sperm DNA
damage is not currently understood.
Q3: Are earlier stages of spermatogenesis sensitive to ROS, and
if so, does oxidative stress during fetal development play a role in the
decline in sperm quality?
A3: Whether maternal exposure to reproductive toxicants
during pregnancy can cause permanent changes in the germ line that
might subsequently impact the fertility of the F1 generation, and the
health trajectory of their o spring, is another fascinating question to
which we do not yet have a de nitive answer. Much will depend on
the nature and intensity of the oxidative stress. In general, DNA proof
reading and DNA repair in the spermatogonial stem cell population is
excellent as indicated by the low risk of birth defects in the children
of men with a history of cancer treatment.86 However, the stability
of the sperm epigenome may be less certain. Studies involving the
maternal administration of the antiandrogenic endocrine disruptor
vinclozolin, have revealed a transgenerational impact on male fertility
that is mediated by a long-lasting epigenetic change in the male germ
line.87 at epigenetic changes in the germ line might be associated
with impaired semen quality is therefore feasible. Furthermore,
oxidative distress is known to alter the pattern of DNA methylation in
spermatozoa.88 However, whether the creation of oxidative stress in the
male germ line during fetal life can subsequently in uence the fertility
of the male o spring, remains an interesting but unresolved possibility.
CONCLUSIONS
Oxidative stress is a major pathological mechanism responsible for
both male infertility and DNA damage in the germ line. When the
oxidative stress occurs in the mature gamete then 8OHdG adducts are
created that are excised by OGG1; however, the remainder of the BER
pathway is completed in the female germ line. Aberrant or ine cient
repair on the part of the oocyte has the potential to create mutations in
the o spring that will impact upon the latters health trajectory.  ere
is strong circumstantial evidence to support such a mechanism in that
high levels of oxidative stress in spermatozoa, due to age or smoking, are
known to increase the burden-of-disease subsequently carried by the
o spring. Mutations in the OGG1 gene are also important contributors
in this respect. Direct evidence for this causative mechanism whereby
the male and female germ lines collude to increase the mutational load
carried by the o spring(oxidative DNA lesions being acquired in the
spermatozoa being followed by imperfect or incomplete repair in the
oocyte) is currently lacking. Furthermore, we do not yet know whether
the range of environmental and lifestyle factors capable of increasing
oxidative DNA damage in human spermatozoa(e.g.,infertility, obesity,
exposure to electromagnetic radiation or environmental toxicants) have
the same degree of impact on the mutation rates in the progeny.  e role
played by the assisted conception industry in facilitating the transfer
of damaged DNA to the oocyte as a consequence of the widespread
use of ICSI is also worthy of detailed scrutiny.
Finally, we do not know whether oxidative insults during fetal or
prepubertal life can have a lasting impact on the genetic integrity of the
germ line with implications for the health trajectory of any o spring.
Studies addressing the impact of ageing on telomere length in the
germ line suggest that early in spermatogenesis, germ cells are capable
of exhibiting adaptive responses that may have a positive impact on
o spring health. As ever, the impact of oxidative stress on reproduction
is a balance of bene t and risk; quantifying the two sides of this delicate
equation will be an important task for the future.
COMPETING INTERESTS
e authors declare that they have no competing interests.
ACKNOWLEDGMENTS
We are grateful to the Australian Research Council, National Health and Medical
Research Council, the University of Newcastle and the Hunter Medical Research
Council for  nancial support.
REFERENCES
1 Hull MG, Glazener CM, Kelly NJ, Conway DI, Foster PA, et al. Population study of
causes, treatment and outcome of infertility. Br Med J (Clin Res Ed) 1985; 291:
1693–7.
2 MacLeod J. The role of oxygen in the metabolism and motility of human spermatozoa.
Am J Physiol 1943; 138: 512–8.
3 Tosic J, Walton A. Formation of hydrogen peroxide by spermatozoa and its inhibitory
effect on respiration. Nature 1946; 158: 485.
4 Tosic J, Walton A. Metabolism of spermatozoa. The formation and elimination of
hydrogen peroxide by spermatozoa and its effects on motility and survival. Biochem J
1950; 47: 199–212.
5 Shannon P, Curson B. Kinetics of the aromatic L-amino acid oxidase from dead
bovine spermatozoa and the effect of catalase on fertility of diluted bovine semen
stored at 5 degrees C and ambient temperatures. J Reprod Fertil 1982; 64: 463–7.
6 Aitken RJ, Clarkson JS. Cellular basis of defective sperm function and its association
with the genesis of reactive oxygen species by human spermatozoa. J Reprod Fertil
1987; 81: 459–69.
7 Alvarez JG, Touchstone JC, Blasco L, Storey BT. Spontaneous lipid peroxidation
and production of hydrogen peroxide and superoxide in human spermatozoa.
Superoxide dismutase as major enzyme protectant against oxygen toxicity. J Androl
1987; 8: 338–48.
8 Yanagimachi R, Yanagimachi H, Rogers BJ. The use of zona-free animal ova as a
test-system for the assessment of the fertilizing capacity of human spermatozoa.
Biol Reprod 1976; 15: 471–6.
9 Aitken RJ, Elton RA. Signifi cance of poisson distribution theory in analysing the
interaction between human spermatozoa and zona-free hamster oocytes. J Reprod
Fertil 1984; 72: 311–21.
10 Aitken RJ. Diagnostic value of the hamster oocyte penetration assay. Int J Androl
1984; 7: 273–5.
11 Twigg JP, Irvine DS, Aitken RJ. Oxidative damage to DNA in human spermatozoa
does not preclude pronucleus formation at intracytoplasmic sperm injection. Hum
Reprod 1998; 13: 1864–71.
12 Aitken RJ, Irvine DS, Wu FC. Prospective analysis of sperm-oocyte fusion and reactive
oxygen species generation as criteria for the diagnosis of infertility. Am J Obstet
Gynecol 1991; 164: 542–51.
13 Irvine DS, Aitken RJ. Predictive value of in-vitro sperm function tests in the context
of an AID service. Hum Reprod 1986; 1: 539–45.
14 Aitken RJ, Buckingham DW, Fang HG. Analysis of the responses of human
spermatozoa to A23187 employing a novel technique for assessing the acrosome
reaction. J Androl 1993; 14: 132–41.
15 Jones R, Mann T, Sherins RJ. Peroxidative breakdown of phospholipids in human
spermatozoa, spermicidal effects of fatty acid peroxides and protective action of
seminal plasma. Fertil Steril 1979; 31: 531–7.
16 Aitken RJ, Wingate JK, De Iuliis GN, Koppers AJ, McLaughlin EA. Cis-unsaturated
fatty acids stimulate reactive oxygen species generation and lipid peroxidation in
human spermatozoa. J Clin Endocrinol Metab 2006; 91: 4154–63.
17 Koppers AJ, Garg ML, Aitken RJ. Stimulation of mitochondrial reactive oxygen species
production by unesterifi ed, unsaturated fatty acids in defective human spermatozoa.
Free Radic Biol Med 2010; 48: 112–9.
18 Aitken RJ, Whiting S, De Iuliis GN, McClymont S, Mitchell LA, et al. Electrophilic
aldehydes generated by sperm metabolism activate mitochondrial reactive oxygen
Oxidative stress
RJ Aitken et al
35
Asian Journal of Andrology
species generation and apoptosis by targeting succinate dehydrogenase. J Biol
Chem 2012; 287: 33048–60.
19 Aitken RJ, Curry BJ. 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 2011; 14: 367–81.
20 Aitken RJ, Gibb Z, Mitchell LA, Lambourne SR, Connaughton HS, et al. Sperm
motility is lost in vitro as a consequence of mitochondrial free radical production
and the generation of electrophilic aldehydes but can be signifi cantly rescued by
the presence of nucleophilic thiols. Biol Reprod 2012; 87: 110.
21 Baumber J, Ball BA, Gravance CG, Medina V, Davies-Morel MC. The effect of
reactive oxygen species on equine sperm motility, viability, acrosomal integrity,
mitochondrial membrane potential, and membrane lipid peroxidation. J Androl
2000; 21: 895–902.
22 Aitken RJ, Buckingham D, Harkiss D. Use of a xanthine oxidase free radical
generating system to investigate the cytotoxic effects of reactive oxygen species on
human spermatozoa. J Reprod Fertil 1993; 97: 441–50.
23 Awda BJ, Mackenzie-Bell M, Buhr MM. Reactive oxygen species and boar sperm
function. Biol Reprod 2009; 81: 553–61.
24 Martínez-Pastor F, Aisen E, Fernández-Santos MR, Esteso MC, Maroto-Morales A,
et al. Reactive oxygen species generators affect quality parameters and apoptosis
markers differently in red deer spermatozoa. Reproduction 2009; 137: 225–35.
25 de Lamirande E, Gagnon C. Reactive oxygen species and human spermatozoa. II.
Depletion of adenosine triphosphate plays an important role in the inhibition of
sperm motility. J Androl 1992; 13: 379–86.
26 de Lamirande E, Gagnon C. Reactive oxygen species and human spermatozoa. I.
Effects on the motility of intact spermatozoa and on sperm axonemes. J Androl
1992; 13: 368–78.
27 Tsunoda S, Kawano N, Miyado K, Kimura N, Fujii J. Impaired fertilizing ability of
superoxide dismutase 1-defi cient mouse sperm during in vitro fertilization. Biol
Reprod 2012; 87: 121.
28 Wishart GJ. Effects of lipid peroxide formation in fowl semen on sperm motility, ATP
content and fertilizing ability. J Reprod Fertil 1984; 71: 113–8.
29 Gomez E, Irvine DS, Aitken RJ. Evaluation of a spectrophotometric assay for the
measurement of malondialdehyde and 4-hydroxyalkenals in human spermatozoa:
relationships with semen quality and sperm function. Int J Androl 1998; 21: 81–94.
30 Aitken RJ, Gordon E, Harkiss D, Twigg JP, Milne P, et al. Relative impact of oxidative
stress on the functional competence and genomic integrity of human spermatozoa.
Biol Reprod 1998; 59: 1037–46.
31 Aitken RJ, Harkiss D, Knox W, Paterson M, Irvine DS. A novel signal transduction
cascade in capacitating human spermatozoa characterised by a redox-regulated,
cAMP-mediated induction of tyrosine phosphorylation. J Cell Sci 1998; 111:
645–56.
32 Brouwers JF, Boerke A, Silva PF, Garcia-Gil N, van Gestel RA, et al. Mass spectrometric
detection of cholesterol oxidation in bovine sperm. Biol Reprod 2011; 85: 128–36.
33 Christova Y, James PS, Jones R. Lipid diffusion in sperm plasma membranes exposed
to peroxidative injury from oxygen free radicals. Mol Reprod Dev 2004; 68: 365–72.
34 De Iuliis GN, Thomson LK, Mitchell LA, Finnie JM, Koppers AJ, et al. DNA damage in
human spermatozoa is highly correlated with the effi ciency of chromatin remodeling
and the formation of 8-hydroxy-2’-deoxyguanosine, a marker of oxidative stress. Biol
Reprod 2009; 81: 517–24.
35 Kodoma H, Yamaguchi R, Fukada J, Kasai H, Tanaka T. Increased oxidative
deoxyribonucleic acid damage in the spermatozoa of infertile men. Fertil Steril
1997; 68: 519–24.
36 Santiso R, Tamayo M, Gosálvez J, Meseguer M, Garrido N, et al. Simultaneous
determination in situ of DNA fragmentation and 8-oxoguanine in human sperm.
Fertil Steril 2010; 93: 314–8.
37 Koppers AJ, Mitchell LA, Wang P, Lin M, Aitken RJ. Phosphoinositide 3-kinase
signalling pathway involvement in a truncated apoptotic cascade associated with
motility loss and oxidative DNA damage in human spermatozoa. Biochem J 2011;
436: 687–98.
38 Koppers AJ, De Iuliis GN, Finnie JM, McLaughlin EA, Aitken RJ. Signifi cance
of mitochondrial reactive oxygen species in the generation of oxidative stress in
spermatozoa. J Clin Endocrinol Metab 2008; 93: 3199–207.
39 McPherson SM, Longo FJ. Localization of DNase I-hypersensitive regions during
rat spermatogenesis: stage-dependent patterns and unique sensitivity of elongating
spermatids. Mol Reprod Dev 1992; 31: 268–79.
40 Leduc F, Nkoma GB, Boissonneault G. Spermiogenesis and DNA repair: a possible
etiology of human infertility and genetic disorders. Syst Biol Reprod Med 2008;
54: 3–10.
41 Manochantr S, Chiamchanya C, Sobhon P. Relationship between chromatin
condensation, DNA integrity and quality of ejaculated spermatozoa from infertile
men. Andrologia 2012; 44: 187–99.
42 Aitken RJ, De Iuliis GN, McLachlan RI. Biological and clinical signifi cance of DNA
damage in the male germ line. Int J Androl 2009; 32: 46–56.
43 Castillo J, Simon L, de Mateo S, Lewis S, Oliva R. Protamine/DNA ratios and DNA
damage in native and density gradient centrifuged sperm from infertile patients.
J Androl 2011; 32: 324–32.
44 Sotolongo B, Huang TT, Isenberger E, Ward WS. An endogenous nuclease in hamster,
mouse, and human spermatozoa cleaves DNA into loop-sized fragments. J Androl
2005; 26: 272–80.
45 Boaz SM, Dominguez K, Shaman JA, Ward WS. Mouse spermatozoa contain a
nuclease that is activated by pretreatment with EGTA and subsequent calcium
incubation. J Cell Biochem 2008; 103: 1636–45.
46 Smith TB, Dun MD, Smith ND, Curry BJ, Connaughton HS, et al. The presence of
a truncated base excision repair pathway in human spermatozoa that is mediated
by OGG1. J Cell Sci 2013; 126: 1488–97.
47 Gawecka JE, Marh J, Ortega M, Yamauchi Y, Ward MA, et al. Mouse zygotes respond
to severe sperm DNA damage by delaying paternal DNA replication and embryonic
development. PLoS One 2013; 8: e56385.
48 Shimura T, Inoue M, Taga M, Shiraishi K, Uematsu N, et al. p53-dependent S-phase
damage checkpoint and pronuclear cross talk in mouse zygotes with X-irradiated
sperm. Mol Cell Biol 2002; 22: 2220–8.
49 Benoff S, Hauser R, Marmar JL, Hurley IR, Napolitano B, et al. Cadmium
concentrations in blood and seminal plasma: correlations with sperm number and
motility in three male populations (infertility patients, artifi cial insemination donors,
and unselected volunteers). Mol Med 2009; 15: 248–62.
50 Benoff S, Jacob A, Hurley IR. Male infertility and environmental exposure to lead
and cadmium. Hum Reprod Update 2000; 6: 107–21.
51 Oliveira H, Spanò M, Santos C, Pereira Mde L. Adverse effects of cadmium exposure
on mouse sperm. Reprod Toxicol 2009; 28: 550–5.
52 Xu DX, Shen HM, Zhu QX, Chua L, Wang QN, et al. The associations among semen
quality, oxidative DNA damage in human spermatozoa and concentrations of
cadmium, lead and selenium in seminal plasma. Mutat Res 2003; 534: 155–63.
53 Shen HM, Chia SE, Ni ZY, New AL, Lee BL, et al. Detection of oxidative DNA
damage in human sperm and the association with cigarette smoking. Reprod Toxicol
1997; 11: 675–80.
54 Ji G, Yan L, Liu W, Qu J, Gu A. OGG1 Ser326Cys polymorphism interacts with
cigarette smoking to increase oxidative DNA damage in human sperm and the risk
of male infertility. Toxicol Lett 2013; 218: 144–9.
55 Lee KM, Ward MH, Han S, Ahn HS, Kang HJ, et al. Paternal smoking, genetic
polymorphisms in CYP1A1 and childhood leukemia risk. Leuk Res 2009; 33: 250–8.
56 Singh NP, Muller CH, Berger RE. Effects of age on DNA double-strand breaks and
apoptosis in human sperm. Fertil Steril 2003; 80: 1420–30.
57 Schmid TE, Eskenazi B, Baumgartner A, Marchetti F, Young S, et al. The effects of male
age on sperm DNA damage in healthy non-smokers. Hum Reprod 2007; 22: 180–7.
58 Paul C, Nagano M, Robaire B. Aging results in differential regulation of DNA repair
pathways in pachytene spermatocytes in the Brown Norway rat. Biol Reprod 2011;
85: 1269–78.
59 Crow JF. The origins, patterns and implications of human spontaneous mutation.
Nat Rev Genet 2000; 1: 40–7.
60 Crow JF. Upsetting the dogma: germline selection in human males. PLoS Genet
2012; 8: e1002535.
61 Goriely A, McVean GA, Röjmyr M, Ingemarsson B, Wilkie AO. Evidence for selective
advantage of pathogenic FGFR2 mutations in the male germ line. Science 2003;
301: 643–6.
62 Shinde DN, Elmer DP, Calabrese P, Boulanger J, Arnheim N, et al. New evidence for
positive selection helps explain the paternal age effect observed in achondroplasia.
Hum Mol Genet 2013; 22: 4117–26.
63 Hurst LD, Ellegren H. Human genetics: mystery of the mutagenic male. Nature
2002; 420: 365–6.
64 Aitken RJ, Krausz C. Oxidative stress, DNA damage and the Y chromosome.
Reproduction 2001; 122: 497–506.
65 Kleinhaus K, Perrin M, Friedlander Y, Paltiel O, Malaspina D, et al. Paternal age
and spontaneous abortion. Obstet Gynecol 2006; 108: 369–77.
66 Green RF, Devine O, Crider KS, Olney RS, Archer N, et al. National Birth Defects
Prevention Study. Association of paternal age and risk for major congenital anomalies
from the National Birth Defects Prevention Study, 1997 to 2004. Ann Epidemiol
2010; 20: 241–9.
67 Kong A, Frigge ML, Masson G, Besenbacher S, Sulem P, et al. Rate of de novo mutations
and the importance of father’s age to disease risk. Nature 2012; 488: 471–5.
68 Aitken RJ, Roman SD. Antioxidant systems and oxidative stress in the testes. Adv
Exp Med Biol 2008; 636: 154–71.
69 Norman RJ. The power of one and its cost. Med J Aust 2011; 195: 564–5.
70 Aitken RJ, Bronson R, Smith TB, De Iuliis GN. The source and signifi cance of DNA
damage in human spermatozoa; a commentary on diagnostic strategies and straw
man fallacies. Mol Hum Reprod 2013; 19: 475–85.
71 Gosden R, Trasler J, Lucifero D, Faddy M. Rare congenital disorders, imprinted genes,
and assisted reproductive technology. Lancet 2003; 361: 1975–7.
72 Hansen M, Kurinczuk JJ, Bower C, Webb S. The risk of major birth defects after
intracytoplasmic sperm injection and in vitro fertilization. N Engl J Med 2002;
346: 725–30.
73 Ericson A, Nygren KG, Olausson PO, Källén B. Hospital care utilization of infants
born after IVF. Hum Reprod 2002; 17: 929–32.
Oxidative stress
RJ Aitken et al
36
Asian Journal of Andrology
74 Källén B, Finnström O, Nygren KG, Olausson PO. In vitro fertilization in Sweden:
child morbidity including cancer risk. Fertil Steril 2005; 84: 605–10.
75 Klemetti R, Sevón T, Gissler M, Hemminki E. Health of children born as a result of
in vitro fertilization. Pediatrics 2006; 118: 1819–27.
76 Ludwig AK, Katalinic A, Thyen U, Sutcliffe AG, Diedrich K, et al. Physical health
at 5.5 years of age of term-born singletons after intracytoplasmic sperm injection:
results of a prospective, controlled, single-blinded study. Fertil Steril 2009; 91:
115–24.
77 Wikstrand MH, Niklasson A, Strömland K, Hellström A. Abnormal vessel morphology
in boys born after intracytoplasmic sperm injection. Acta Paediatr 2008; 97:
1512–7.
78 Aitken RJ, Findlay JK, Hutt KJ, Kerr JB. Apoptosis in the germ line. Reproduction
2011; 141: 139–50.
79 Aston KI, Hunt SC, Susser E, Kimura M, Factor-Litvak P, et al. Divergence of sperm
and leukocyte age-dependent telomere dynamics: implications for male-driven
evolution of telomere length in humans. Mol Hum Reprod 2012; 18: 517–22.
80 Unryn BM, Cook LS, Riabowol KT. Paternal age is positively linked to telomere length
of children. Aging Cell 2005; 4: 97–101.
81 Shammas MA. Telomeres, lifestyle, cancer, and aging. Curr Opin Clin Nutr Metab
Care 2011; 14: 28–34.
82 Desai N, Sabanegh E Jr, Kim T, Agarwal A. Free radical theory of aging: implications
in male infertility. Urology 2010; 75: 14–9.
83 Thilagavathi J, Kumar M, Mishra SS, Venkatesh S, Kumar R, et al. Analysis of
sperm telomere length in men with idiopathic infertility. Arch Gynecol Obstet
2013; 287: 803–7.
84 Fisher MT, Nagarkatti M, Nagarkatti PS. Aryl hydrocarbon receptor-dependent
induction of loss of mitochondrial membrane potential in epididymal spermatozoa
by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Lett 2005; 157: 99–107.
85 Senft AP, Dalton TP, Nebert DW, Genter MB, Puga A, et al. Mitochondrial reactive
oxygen production is dependent on the aromatic hydrocarbon receptor. Free Radic
Biol Med 2002; 33: 1268–78.
86 Ståhl O, Boyd HA, Giwercman A, Lindholm M, Jensen A, et al. Risk of birth
abnormalities in the offspring of men with a history of cancer: a cohort study using
Danish and Swedish national registries. J Natl Cancer Inst 2011; 103: 398–406.
87 Anway MD, Memon MA, Uzumcu M, Skinner MK. Transgenerational effect of the
endocrine disruptor vinclozolin on male spermatogenesis. J Androl 2006; 27: 868–79.
88 Tunc O, Tremellen K. Oxidative DNA damage impairs global sperm DNA methylation
in infertile men. J Assist Reprod Genet 2009; 26: 537–44.
How to cite this article: Aitken RJ, Smith TB, Jobling MS, Baker
MA, De Iuliis GN. Oxidative stress and male reproductive health.
Asian J Androl 2013 Dec 16. doi: 10.4103/1008-682X.122203. [Epub
ahead of print]
... These factors are implicated in cellular dysfunction, affecting the total number, motility, and concentration of spermatozoa [27]. Specifically, LPO products interact with amino residues, leading to protein oxidation and thus affecting the structural and functional aspects of proteins in sperm, particularly in the sperm mitochondrial electron transport chain [28]. Furthermore, LPO products can result in loss of membrane integrity and alterations in motility, thus negatively impacting interactions between sperm and oocytes [29]. ...
... Furthermore, LPO products can result in loss of membrane integrity and alterations in motility, thus negatively impacting interactions between sperm and oocytes [29]. Due to the deficiency of adequate antioxidant systems and a lack of complete DNA repair pathways, spermatozoa are highly susceptible to DNA oxidation [28]. ...
... The outflow of ATP from the inner cell and reduced viability contribute to sperm morphological defects and motility dysfunction, ultimately diminishing sperm quality and fertility [36,37]. Studies have demonstrated that DNA damage, which cannot be repaired due to the absence of a cytoplasmic enzyme repair system [28,31,37], along with poor chromatin packaging, enhances the induction of apoptotic pathways [37]. It has also been demonstrated that a decrease in antioxidant levels may initiate excessive ROS generation [38]. ...
Article
Full-text available
Infertility is a prevalent global issue affecting approximately 17.5% of adults, with sole male factor contributing to 20–30% of cases. Oxidative stress (OS) is a critical factor in male infertility, disrupting the balance between reactive oxygen species (ROS) and antioxidants. This imbalance detrimentally affects sperm function and viability, ultimately impairing fertility. OS also triggers molecular changes in sperm, including DNA damage, lipid peroxidation, and alterations in protein expression, further compromising sperm functionality and potential fertilization. Diagnostic tools discussed in this review offer insights into OS markers, antioxidant levels, and intracellular ROS concentrations. By accurately assessing these parameters, clinicians can diagnose male infertility more effectively and thus tailor treatment plans to individual patients. Additionally, this review explores various treatment options for males with OS-associated infertility, such as empirical drugs, antioxidants, nanoantioxidants, and lifestyle modifications. By addressing the root causes of male infertility and implementing targeted interventions, clinicians can optimize treatment outcomes and enhance the chances of conception for couples struggling with infertility.
... [24] Sperm and oocytes employ 8-oxoguanine glycosylase-1 for repairing damaged DNA; however, they lack downstream DNA repair genes. [25] The limited detection capacity of sperm may lead to substantial damage, causing the accumulation of oxidized DNA adducts in zygote cells. These mutagenic adducts can generate epimutations, leading to genomic instability and an 18-fold increase in mutation rates, including GC > TA transversions. ...
... [44] Infertile men have higher levels of seminal OS and enhanced apoptosis compared to fertile counterparts. [25,45] Studies have proved that yoga-based lifestyle can help to reduce OS induced by these factors; [23,31] hence, yoga improves dysregulation of apoptosis in the process of spermatogenesis. ...
Article
Full-text available
Infertility, a widespread medical condition affecting numerous couples globally, persists as a challenge despite advances in assisted reproductive technologies (ARTs), often burdened by financial, physical, and emotional strains. Complementary and alternative approaches, notably yoga, have garnered attention for potentially enhancing fertility outcomes. Studies reveal yoga’s influence on factors contributing to infertility, including reduced oxidative stress (OS) and oxidative DNA damage (ODD). OS, linked to mutagenic base formation, higher malondialdehyde levels, abnormal methylation, and altered gene expression, can impair sperm genome integrity. Yoga’s efficacy is evident in lowering OS, positively affecting signal transmission, gene expression, and physiological systems. Furthermore, yoga has a positive impact on addressing the dysregulation of apoptosis, resulting in improved processes such as spermatogenesis, sperm maturation, and motility, while also reducing DNA fragmentation. OS correlates with genome-wide hypomethylation, telomere shortening, and mitochondrial dysfunction, contributing to genome instability. Yoga and meditation significantly reduce OS and ODD, ensuring proper reactive oxygen levels and preserving physiological systems. The review explores potential mechanisms underlying yoga’s positive impact on infertility, including enhanced blood flow, reduced inflammation, relaxation response, and modulation of the hypothalamic–pituitary–adrenal axis. Furthermore, a comprehensive review of the literature reveals substantial evidence supporting the positive effects of yoga on infertility factors. These include oxidative stress (OS), oxidative DNA damage (ODD), epigenetic changes, hormonal balance, ovarian function, menstrual irregularities, and stress reduction. In summary, yoga emerges as a promising adjunctive therapy for infertility, demonstrating the potential to mitigate key factors influencing reproductive success. Although preliminary evidence indicates the positive effects of yoga on infertility, further clinical research is imperative to define specific benefits, molecular mechanisms associated, optimal protocols, and long-term effects in infertility treatment plans.
... The opening of the mPTP is one of the main pathways involved in the ability of sperm mitochondria to generate ROS and induce apoptotic cell death in sperm (reviewed in [64]). Therefore, the DNA fragmentation observed here, although low, may be directly associated with the increase in mROS, in agreement with the reports that most of the DNA damage observed in mature human sperm cells results from oxidative stress [65,111,112]. In spermatozoa, the mitochondrial functions, mainly the Ca 2+ homeostasis and production of ROS and ATP, regulate processes such as motility, hyperactivation, capacitation, the acrosome reaction and fertilization [29,31,[113][114][115]. ...
Article
Full-text available
Oxidative stress (OS) and disrupted antioxidant defense mechanisms play a pivotal role in the etiology of male infertility. The alterations in reactive oxygen species (ROS) production and calcium (Ca2+) homeostasis are the main activators for the mitochondrial permeability transition pore (mPTP) opening. The mPTP opening is one of the main mechanisms involved in mitochondrial dysfunction in spermatozoa. This alteration in mitochondrial function adversely affects energy supply, sperm motility, and fertilizing capacity and contributes to the development of male infertility. In human spermatozoa, the mPTP opening has been associated with ionomycin-induced endogenous oxidative stress and peroxynitrite-induced nitrosative stress; however, the effect of exogenous oxidative stress on mPTP opening in sperm has not been evaluated. The aim of this study was to determine the effect of exogenous oxidative stress induced by hydrogen peroxide (H2O2) on mPTP opening, mitochondrial function, motility, and cell death markers in human spermatozoa. Human spermatozoa were incubated with 3 mmol/L of H2O2 for 60 min, and intracellular Ca2+ concentration, mPTP opening, mitochondrial membrane potential (ΔΨm), ATP levels, mitochondrial reactive oxygen species (mROS) production, phosphatidylserine (PS) externalization, DNA fragmentation, viability, and sperm motility were evaluated. H2O2-induced exogenous oxidative stress caused increased intracellular Ca2+, leading to subsequent mPTP opening and alteration of mitochondrial function, characterized by ΔΨm dissipation, decreased ATP levels, increased mROS production, and the subsequent alteration of sperm motility. Furthermore, H2O2-induced opening of mPTP was associated with the expression of apoptotic cell death markers including PS externalization and DNA fragmentation. These results highlight the role of exogenous oxidative stress in causing mitochondrial dysfunction, deterioration of sperm motility, and an increase in apoptotic cell death markers, including PS externalization and DNA fragmentation, through the mPTP opening. This study yielded new knowledge regarding the effects of this type of stress on mitochondrial function and specifically on mPTP opening, factors that can contribute to the development of male infertility, considering that the role of mPTP in mitochondrial dysfunction in human sperm is not completely elucidated. Therefore, these findings are relevant to understanding male infertility and may provide an in vitro model for further research aimed at improving human sperm quality.
... Recent reports have highlighted the significant role of reactive oxygen species (ROS) in both reproductive physiology and pathology [2]. The dual nature of ROS is contingent upon factors such as source, concentration, production site, and exposure time [15]. Physiologically, an appropriate level of ROS is deemed crucial for the successful execution of various functions associated with gamete fertility, including proliferation, maturation, the release of oocytes [16], capacitation, hyperactivation, the acrosome reaction, and fertilization [17]. ...
Article
Full-text available
Cryopreservation poses significant challenges to the preservation of sperm integrity and function, particularly in small ruminants where cryodamage is pronounced. This review explores the molecular mechanisms underlying sperm cryodamage and strategies for improving cryopreservation outcomes, with a focus on the role of antioxidants. Cryopreservation-induced alterations in proteins and RNA transcripts critical for sperm function, including motility, capacitation, fertilization, and embryo development, are discussed. Proteomic, transcriptomic, and epigenomic advancements have provided valuable insights into these mechanisms, offering potential biomarkers for predicting sperm freezability and enhancing cryopreservation strategies. Combining technologies such as mass spectrometry and flow cytometry allows for a comprehensive understanding of molecular and cellular changes induced by the freezing–thawing process. However, challenges remain in optimizing cryoprotectant formulations and antioxidant supplementation to improve post-thaw sperm fertility. Further research is needed to explore a wider range of novel cryoprotectants, antioxidants, and proteins for cryopreservation media, as well as to validate their efficacy in enhancing sperm viability and function. Additionally, investigations into the effects of cryopreservation on RNA transcripts and epigenetic factors in small ruminant species are warranted to advance our understanding of sperm preservation. Overall, this review highlights the importance of antioxidants in mitigating cryodamage and underscores the need for continued research to refine cryopreservation protocols and improve reproductive outcomes in small ruminants.
... During sperm maturation, changes in DNA methylation occur. Ben Maamar et al. [79] found that the sperm DNA methylation profile of male offspring from female mice exposed to toxic environments showed alterations in the epididymis. Factors such as paternal age, diet, and exposure to toxic environments during sperm passage through the epididymis can lead to sperm oxidative stress, affecting DNA methylation. ...
Article
Full-text available
Testicular sperm maturation is critical for establishing male fertility. Spermatozoa undergo remodeling of sperm proteins and changes in lipid and ribonucleic acid composition during transport in the epididymal ducts, which play an important role in sperm maturation. The anatomy, epithelial cell types, physiological functions, and epigenetic inheritance of the epididymis are explored, and recent findings in epididymal research are analyzed. Suggesting possible directions for future research on the epididymis. Using the keywords “epididymis”, “sperm”, and “sperm maturation”, a search of the epididymis was performed through databases and official websites of journals related to reproduction. The epididymis was searched in databases and on the official websites of journals related to reproduction. This review introduces the characteristics of the epididymis, as well as the biological functions of cell types such as principal cells, clear cells, and basal cells, providing a detailed description of the overall physiological functions of the epididymis. It highlights current research hotspots in the field of epididymis, including single-cell analysis, epigenetics, and extracellular vesicles, aiming to offer a comprehensive understanding for beginners. The review emphasizes the importance of the epididymis, its impact on sperm maturation and subsequent embryo development, and how it advances research on epididymal diseases while providing new directions for the study and treatment strategies of infertility.
... Therefore, identifiable metabolites generated due to oxidative damage of cell components (proteins, lipids and DNA) are used to measure the intensity of the redox state-these molecules are known as specific biomarkers; 8-hidroxy-2-deoxyguanosine (8-OHdG) [25], malondialdehyde (MDA) [26] or total antioxidant capacity (TAC) measurements [27] may be pointed as examples [28]. Oxidative stress (OS) is considered to be involved in the development of many various diseases, e.g., cancer [29], diabetes [30], cardiovascular [21] or liver disorders [31], Alzheimer's disease [32], psychological disturbances [33], pathological states affecting reproductive health [34][35][36] any many others. HT is also known to be connected with OS [37], as well as in children and adolescents [27]. ...
Article
Full-text available
Autoimmune thyroid disease (AITD) is the most common organic specific illness of the thyroid gland. It may manifest as the overproduction or the decline of thyroxine and triiodothyronine. Hyperthyroidism develops due to the overproduction of hormones as an answer to the presence of stimulatory antibodies against the TSH receptor. Hashimoto’s thyroiditis (HT) is generally characterized by the presence of thyroid peroxidase and thyroglobulin antibodies, with a concomitant infiltration of lymphocytes in the thyroid. Due to the progressive destruction of cells, AITD can lead to subclinical or overt hypothyroidism. Pathophysiology of AITD is extremely complicated and still not fully understood, with genetic, environmental and epigenetic factors involved in its development. Due to increasing incidence and social awareness of this pathology, there is an urgent need to expand the background concerning AITD. A growing body of evidence suggests possible ways of treatment apart from traditional approaches. Simultaneously, the role of potential new biomarkers in the diagnosis and monitoring of AITD has been highlighted recently, too. Therefore, we decided to review therapeutic trends in the course of AITD based on its pathophysiological mechanisms, mainly focusing on HT. Another aim was to summarize the state of knowledge regarding the role of new biomarkers in this condition.
Article
Full-text available
The management of neurological disorders heavily relies on neurotherapeutic drugs, but notable concerns exist regarding their possible negative effects on reproductive health. Traditional preclinical models often fail to accurately predict reprotoxicity, highlighting the need for more physiologically relevant systems. Organoid models represent a promising approach for concurrently studying neurotoxicity and reprotoxicity, providing insights into the complex interplay between neurotherapeutic drugs and reproductive systems. Herein, we have examined the molecular mechanisms underlying neurotherapeutic drug-induced reprotoxicity and discussed experimental findings from case studies. Additionally, we explore the utility of organoid models in elucidating the reproductive complications of neurodrug exposure. Have discussed the principles of organoid models, highlighting their ability to recapitulate neurodevelopmental processes and simulate drug-induced toxicity in a controlled environment. Challenges and future perspectives in the field have been addressed with a focus on advancing organoid technologies to improve reprotoxicity assessment and enhance drug safety screening. This review underscores the importance of organoid models in unraveling the complex relationship between neurotherapeutic drugs and reproductive health.
Article
Bibliometric analyses serve to identify influential articles that have shaped medical practice and fostered new research ideas. Over the past decade, research in andrology has witnessed exponential growth, with an increasing number of academic publications, collaborations, and research innovations. However, there is a lack of literature that has identified the top-cited andrology articles. We conducted a bibliometric analysis to identify the top 1000 citations in andrology journals, with a focus on the top funding agencies, authors, institutions, countries/regions, and journals. To perform this analysis, we identified the top-cited articles in andrology journals as indexed in the Web of Science Core Collection. From 2013 through 2022, we found a total of 9827 articles published in andrology journals. The top publishers included "Andrology," the "Asian Journal of Andrology," and "Andrologia." The top affiliations contributing to research include the Cleveland Clinic Foundation (269 publications), Egyptian Knowledge Bank (EKB) (265), and Shanghai Jiao Tong University (202). Funding was primarily provided by notable agencies such as the National Natural Science Foundation of China (905 grants), United States Department of Health Human Services (321), and National Institutes of Health (NIH USA) (317). The present bibliometric analysis highlights andrology research from 2013 through 2022, offering key insights into leading contributors, influential authors, prominent funding sources, and major trends in the field.
Article
The pathogenesis of benign prostatic hyperplasia (BPH) in dogs is still not fully understood. Some studies suggest that oxidative stress may have a potential role in the pathogenesis of BPH in dogs. Age‐related hormonal changes may activate a chronic inflammatory response in the prostate. This causes the generation of reactive oxygen species (ROS) and results in oxidative stress. Excessive production of ROS results in DNA damage and hyperplastic transformation of prostatic cells. The use of antioxidants for improvement of treatment outcomes for canine PBH has been discussed. Further research is needed on the importance of oxidative stress in the development of BPH in dogs and the usefulness of antioxidants in the supportive treatment of this condition.
Article
Full-text available
There are certain de novo germline mutations associated with genetic disorders whose mutation rates per generation are orders of magnitude higher than the genome average. Moreover, these mutations occur exclusively in the male germ line and older men have a higher probability of having an affected child than younger ones, known as the paternal age-effect. The classic example of a genetic disorder exhibiting a PAE is achondroplasia, caused predominantly by a single nucleotide substitution (c.1138G>A) in FGFR3. To elucidate what mechanisms might be driving the high frequency of this mutation in the male germline, we examined the spatial distribution of the c.1138G>A substitution in a testis from an 80-year old unaffected man. Using a technology based on bead-emulsion amplification, we were able to measure mutation frequencies in 192 individual pieces of the dissected testis with a false positive rate lower than 2.7x10(-6). We observed that most mutations are clustered in a few pieces with 95% of all mutations occurring in 27% of the total testis. Using computational simulations, we rejected the model proposing an elevated mutation rate per cell division at this nucleotide site. Instead we determined that the observed mutation distribution fits a germline selection model, where mutant spermatogonial stem cells have a proliferative advantage over unmutated cells. Combined with data on several other PAE mutations, our results support the idea that the PAE, associated with a number of Mendelian disorders, may be explained primarily by a selective mechanism.
Article
Full-text available
This article considers the origins of DNA damage in human spermatozoa, the methods that are available to monitor this aspect of semen quality and the clinical significance of such measurements. DNA damage in spermatozoa appears to be largely oxidative in nature, inversely correlated with levels of nuclear protamination and frequently associated with the activation of a truncated apoptotic pathway. DNA base adducts formed as a result of oxidative attack are released from the spermatozoa into the extracellular space through the action of a glycosylase, OGG1. This creates an abasic site, which is not resolved until fertilization because spermatozoa do not possess the molecular machinery needed to continue the base excision repair pathway. The abasic sites so generated in human spermatozoa are readily detected by SCSA or the Comet assay, however no signal is detectable with TUNEL. This is because spermatozoa lack the enzyme (APE1) needed to create the free 3' hydroxyl groups required by this detection system. Nevertheless spermatozoa do eventually become TUNEL positive as they enter the perimortem. The American Society of Reproductive Medicine Practice Committee has suggested that DNA damage in spermatozoa should not be assessed because the correlation with pregnancy is inconsistent across independent studies. However this is a straw man argument. The reason why such assays should be undertaken is not just because they reflect the underlying quality of spermatogenesis but, more importantly, because the DNA damage they reveal may have detrimental effects on the developmental normality of the embryo and health of possible future children.
Article
Full-text available
Mouse zygotes do not activate apoptosis in response to DNA damage. We previously reported a unique form of inducible sperm DNA damage termed sperm chromatin fragmentation (SCF). SCF mirrors some aspects of somatic cell apoptosis in that the DNA degradation is mediated by reversible double strand breaks caused by topoisomerase 2B (TOP2B) followed by irreversible DNA degradation by a nuclease(s). Here, we created zygotes using spermatozoa induced to undergo SCF (SCF zygotes) and tested how they responded to moderate and severe paternal DNA damage during the first cell cycle. We found that the TUNEL assay was not sensitive enough to identify the breaks caused by SCF in zygotes in either case. However, paternal pronuclei in both groups stained positively for γH2AX, a marker for DNA damage, at 5 hrs after fertilization, just before DNA synthesis, while the maternal pronuclei were negative. We also found that both pronuclei in SCF zygotes with moderate DNA damage replicated normally, but paternal pronuclei in the SCF zygotes with severe DNA damage delayed the initiation of DNA replication by up to 12 hrs even though the maternal pronuclei had no discernable delay. Chromosomal analysis of both groups confirmed that the paternal DNA was degraded after S-phase while the maternal pronuclei formed normal chromosomes. The DNA replication delay caused a marked retardation in progression to the 2-cell stage, and a large portion of the embryos arrested at the G2/M border, suggesting that this is an important checkpoint in zygotic development. Those embryos that progressed through the G2/M border died at later stages and none developed to the blastocyst stage. Our data demonstrate that the zygote responds to sperm DNA damage through a non-apoptotic mechanism that acts by slowing paternal DNA replication and ultimately leads to arrest in embryonic development.
Article
Full-text available
DNA repair has long been considered impossible in human spermatozoa due to the high level of DNA compaction observed in these cells. However, detailed examination of the base excision repair pathway in human spermatozoa has revealed the presence of an enzyme critical to this pathway, OGG1. This glycosylase was associated with the sperm nucleus and mitochondria and could actively excise 8-hydrdoxy, 2'-deoxyguanosine, releasing this adduct into the extracellular space. This activity was significantly reduced in the presence of cadmium (II), a recognized inhibitor of OGG1, in a time- and dose- dependent manner (P<0.001). Remarkably, spermatozoa do not possess the downstream components of the base excision repair pathway, APE1 and XRCC1. The absence of these proteins was particularly significant, as APE1 is required to create a 3'-hydroxyl (3'-OH) terminus at the apurinic site created by OGG1, which would be recognized by the TUNEL assay. As a result, TUNEL was unable to detect oxidatively induced DNA damage in spermatozoa following exposure to hydrogen peroxide. In the same cells, intracellular and extracellular 8OHdG could be clearly detected in a manner that was highly correlated with the outcome of SCSA (Sperm Chromatin Structure Assay). However, incubation of these cells for 48 hours revealed a time-dependent increase in TUNEL positivity, suggesting the perimortem activation of a nuclease. These results emphasize the limited capacity of mature spermatozoa to mount a DNA repair response to oxidative stress, and highlight the importance of such mechanisms in the oocyte in order to protect the embryo from paternally mediated genetic damage.
Article
The zona-free hamster ovum was evaluated as a substitute for human o va in the assessment of the fertilizing capacity of human spermatozoa. Zona-intact ova completely resisted sperm penetration. Using nonpreincu bated spermatozoa sperm penetration of zona-free ova began 4-5 hours after insemination. However when spermatozoa were preincubated in a modified Krebs-Ringer solution for 4 hours sperm penetration began within 1 hour. There is some evidence that this is associated with the completion of sperm capacitation and the acrosome reaction. The results suggest that zona-free hamster ova can be substituted for human ova in the preliminary assessment of the fertilizing capacity of human spermatozoa.
Article
Humans are exposed occupationally and environmentally to metal aerosols including lead (Pb2+) and cadmium (Cd2+). These toxicants accumulate in male reproductive organs. Epidemiological studies have been equivocal about effects of Pb2+ and Cd2+ on hormone concentrations, male fertility and sperm parameters. Comparison of Pb2+ and Cd2+ concentrations in fertile and infertile men are problematic. Problem areas include failure to control confounding variables, but genetic polymorphisms as in somatic diseases may modulate Pb2+ and Cd2+ damage. Multiple calcium (Ca2+) and potassium (K⁺) channel isoforms have been identified in human testes and spermatozoa. These Ca2+ and K⁺ channels are involved in early events of acrosome reactions. Ca2+ channel are susceptible to Cd2+ poisoning and K⁺ channels to Pb2+. These channels offer entry paths for metallic toxicants into mature spermatozoa. Ion channel polymorphisms may cause differential sensitivities to Cd2+ and Pb2+, explaining in part prospective blinded studies showing high Cd2+ in varicocele-related human infertility and high Pb2+ in unexplained infertility. In both forms of male infertility the ability to undergo an acrosome reaction decreases. Reverse transcriptase–polymerase chain reaction assays for Ca2+ and K⁺ channel isoforms may identify susceptibility subgroups with lower resistance to environmental exposures.
Article
Apoptosis is a critical process for regulating both the size and the quality of the male and female germ lines. In this review, we examine the importance of this process during embryonic development in establishing the pool of spermatogonial stem cells and primordial follicles that will ultimately define male and female fertility. We also consider the importance of apoptosis in controlling the number and quality of germ cells that eventually determine reproductive success. The biochemical details of the apoptotic process as it affects germ cells in the mature gonad still await resolution, as do the stimuli that persuade these cells to commit to a pathway that leads to cell death. Our ability to understand and ultimately control the reproductive potential of male and female mammals depends upon a deeper understanding of these fundamental processes.