Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Reviews of Reproduction (1998) 3, 172–182

Mitochondrial DNA in mammalian reproduction


Jim Cummins
Division of Veterinary and Biomedical Sciences, Murdoch University, Murdoch, Western Australia 6150,
Australia

Mitochondrial DNA (mtDNA) forms a semi-autonomous asexually reproducing genome in


eukaryotic organisms. It plays an essential role in the life cycle through the control of energy
production, by the inherently dangerous process of oxidative phosphorylation. The asym-
metric nature of its inheritance – almost exclusively through the female – imposes different
evolutionary constraints on males and females, and may lie at the heart of anisogamy. This
review examines the implications of recent findings on the biology of mtDNA for reproduction
and inheritance in mammals.

Although the existence of mitochondria has been known since encodes for tryptophan in mammalian mitochondria). The in-
the last century, mitochondrial DNA (mtDNA) has been studied heritance of mitochondria through the female lineage remains
most extensively in the past two decades. Mitochondria have one of the central enigmas of reproductive biology.
a profound role to play in mammalian tissue bioenergetics, in Mitochondria have certain tissue-specific configurations that
growth, in ageing and in apoptosis, and yet they descend from presumably reflect local energetic requirements (Fawcett, 1981).
an asexually reproducing independent life form. It has been New techniques for visualizing mitochondria in whole cells,
hypothesized that tensions between the evolutionary ‘interests’ such as the incorporation of green fluorescent protein coupled
of the eukaryotic host and its subservient organelles have led to with confocal microscopy, reveal that mitochondrial form can
asymmetrical inheritance, so that mitochondria derive pre- not only reflect pathological states but may also be extremely
dominantly from the female in most organisms (Hurst, 1992; diverse even within the same cell (Kanazawa et al., 1997). The
Hurst et al., 1996). This has consequences that are only just be- basic design is a double membrane surrounding an inner mito-
coming apparent. There is considerable interest in the potential chondrial matrix that contains one or more circular mtDNA
role of mitochondria and cytoplasmic inheritance on growth molecules. The outer mitochondrial membrane is thought to
and performance factors such as muscle development and milk represent the original invaginated host plasma membrane,
production in domestic animals. As might be expected, the while the inner mitochondrial membrane represents the bac-
maternally inherited mitochondrial genome has significant terial wall and contains the site of oxidative phosphorylation
non-Mendelian effects on steroidogenesis and on respiratory- (OXPHOS) on its inner surface. The matrix also contains ribo-
dependent functions such as growth, oxygen consumption and somes for local protein synthesis.
lean:fat ratios, but not on anaerobic metabolism. Smith and The two membranes differ profoundly in composition. The
Alcivar (1993) have reviewed this topic recently and compre- lipid:protein ratio of the outer membrane is about 50:50 and
hensively; therefore, the focus of this article will be the role of it is permeable to molecules with molecular weights of up to
mtDNA in the life cycle. 10 000 (Lodish et al., 1995). The inner membrane is relatively
Mitochondria are semi-autonomous organelles found in all impermeable and is about 80% protein and is thrown up into
eukaryotic cells (except mature red blood cells and some pro- infoldings – crystae – the sites of OXPHOS enzymes. Here the
tozoans). It is generally accepted that mitochondria originated oxidation of metabolites generates ATP through a series of
in ancestral eukaryotic cells through endosymbiosis of free- integral membrane multi-subunit protein complexes which
living bacteria capable of metabolizing oxygen – a suggestion couple electron transport to ATP synthesis. These complexes
first made over a century ago and noted by Ozawa in his review are unique in that they consist of proteins encoded by two
(Ozawa, 1997a). Our ancestral eukaryotes thus exploited the separate yet cooperating genomes, that of the nucleus and that
capacity of mitochondria to metabolise oxygen. This allowed of the mitochondrion (Poyton and McEwen, 1996; Shadel and
them to flourish despite the increasing concentrations of this Clayton, 1997) There are separate translocase systems in the
highly reactive and potentially poisonous element in the en- inner and outer membranes that coordinate the recognition,
vironment. While most of the mitochondrial genes have moved import and assortment of essential proteins from the cytosol
to the nucleus, mitochondria retain their bacterial facility for (Neupert, 1997).
multiplying by simple fission – and even fusing – indepen-
dently of the host cell cycle. Excess mitochondria are removed
Mitochondrial DNA
by autophagic lysosomal activity. These population control
measures act in response to the energetic demands of different Most cells in the body contain between 103 and 104 copies of
tissues. The mitochondrial genome also has idiosyncrasies in mtDNA. There are much higher copy numbers (about 105) in
RNA processing and in its genetic code that differ from those of mature oocytes. This may be in preparation for the energetic
nuclear DNA (for example UGA is normally a stop codon, but demands of embryogenesis (Pikó and Matsumoto, 1976) but
© 1998 Journals of Reproduction and Fertility
1359-6004/98 $12.50
Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM
via free access
Mitochondrial DNA in the life cycle 173

Table 1. Fate of light and heavy chain transcripts from mitochondrial


DNA
Displaced heavy strand (D-loop)
Light chain transcripts Heavy chain transcripts Heavy strand Nascent heavy
origin strand
8 tRNAs 14 tRNAs
1 mRNA 12 mRNAS Heavy strand
RNA primers for heavy chain replication 2 tRNAs promoter
Transcription
Modified from Shadel and Clayton, 1997
factor binding Light strand
sites promoter
an alternative explanation is that replication does not occur
during early embryogenesis and that high copy numbers are
needed to give a sufficient reservoir (see below). The DNA Light strand origin
exists mainly as a circular molecule of approximately 16.6 kb, (stem-loop)
encoding 13 proteins that are transcribed and translated in
the mitochondrion (Table 1). These are essential subunits of the
electron transport complexes on the inner mitochondrial mem-
brane. The mitochondrial genome also encodes the RNA mol-
ecules that are necessary for the translation of these proteins
(Table 1) (Lodish et al., 1995; Shadel and Clayton, 1997).

mtDNA structure Fig. 1. This diagram summarizes the major features of mtDNA re-
ferred to in the text. (See Shadel and Clayton, 1997.)
The separate strands of the mtDNA molecule differ in buoy-
ant density; the heavier ‘H-strand’ has a higher G + T content
than the light ‘L-strand’. Transcription occurs simultaneously
and in opposite directions and many genes overlap. By con- Box 1 Major mitochondrial import proteins
vention mtDNA is depicted as a circle (Fig. 1), but alternatives,
such as dimer loops and catenated (chain-linked) circles, are Mitochondrial DNA and RNA polymerases
Transcription, translation and transcription termination factors
known (Clayton, 1982). In some single-celled organisms (many
RNA processing enzymes
pathogenic to mammals) aberrant linear mtDNA molecules
Mitochondrial ribosomal proteins
with telomere-like endings are also found (Nosek et al., 1998). Aminoacyl-tRNA synthetases
There is a specialised, somewhat unstable and hypervariable
region called the D-(displacement) loop, where there is a triplex
DNA structure at the site of origin of the H strand (Fig. 1). This
structure is formed by a short nascent H-strand that remains
closely associated with the parental molecule. This region is crit- Neupert, 1997). Besides structural components of the mitochon-
ical for the initiation of transcription and translation (see below). dria, these imports involve factors that regulate and specifically
The mammalian mitochondrial genome is extremely com- recognize mtDNA and regulate gene expression (Box 1).
pressed with no introns. Some genes even overlap. This con-
trasts strikingly with, for example, yeast (five times larger at Nuclear–mitochondrial interactions
78 000 bp) or plants, in which there are multiple recombining
molecules with enormous size variation even within a family. Any alterations that arise in the components of the mtDNA
For example, sizes vary from 330 000 bp in watermelons to or RNA that recognize or bind to nuclear-encoded regulatory
2.5 + 106 bp in muskmelon (Lodish et al., 1995). Most of the 100 elements must be balanced by compensatory mutations in the
or more genes controlling the synthesis of mammalian mito- nuclear genes, as the mitochondrial genome mutates much
chondrial proteins have moved to the nucleus over the course more rapidly than the nuclear genome (see below). This mu-
of evolution. tuality is thought to drive species specificity in nuclear–
mitochondrial interaction (Kenyon and Moraes, 1997; Wallace,
1997), but surprisingly little is known about the coordination of
Import–export mechanisms
expression between the nuclear and mitochondrial genomes.
Proteins are assembled on cytoplasmic ribosomes and trans- However, it is clearly a sensitive system. Nagao et al. (1998)
ported into the mitochondrion through protein-lined channels, found decreased physical performance and growth rates in
with the aid of cytoplasmic chaperone proteins (Box 1). These mice in which the nuclear and mitochondrial genes were mis-
import mechanisms are prime targets for strategies aimed at matched. The mitochondrial genome exists as multiple
mitochondrial genome therapy or modification (Murphy, 1997; copies per cell and yet for unknown reasons proteins encoded
Taylor et al., 1997). Import involves hydrolysis of ATP in both the by single copy nuclear genes are present in the same amounts
cytoplasm and the mitochondrion plus an electrochemical poten- as mtDNA-encoded proteins (Poyton and McEwen, 1996). This
tial across the inner mitochondrial membrane (Lodish et al., 1995; is clearly an issue that will bear on attempts to clone animals,

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
174 J. Cummins

Table 2. Summary of some major mtDNA functional locations in the mtDNA. While the ‘common’ deletion is the most prevalent,
human mitochondrial genome this is also practically convenient as the major difference in
size between the deleted and the intact mtDNA gives an un-
Nucleotide position* Description ambiguous signal after PCR amplification. With ageing the
genome undergoes extensive fragmentation. One study still
110–441 H-strand origin awaiting independent verification suggests that in very old
233–260 mtTF1 (=Tfa) binding site human individuals the amount of wild-type intact mtDNA
276–303 mtTF1 (=Tfa) binding site may be as low as only 11% of the total (Hayakawa et al.,
299–315 Conserved sequence box (CSB) II 1996). The mtDNA in these cases can fragment into mini-circles
317–321 Replication primer lacking replication origins; if and how these replicate is
346–363 CSB III unknown.
371–379 mt4 H-strand control element Life expectancy within a species is closely correlated with
384–391 mt3 H-strand control element overall nuclear DNA repair efficiency and inversely correlated
392–445 L-strand promoter with mtDNA damage; for example mtDNA mutation and
418–445 mtTF1 (=Tfa) binding site general body tumour formation rates are approximately 40 fold
523–550 mtTF1 (=Tfa) binding site higher in mice than in humans (Cortopassi and Wang, 1996).
545–567 Major H-strand promoter Although mitochondria obviously play only a part in the ageing
645–645 Minor H-strand promoter story (Holliday, 1995), it seems clear that they hold many of the
5721–5798 L-strand origin keys to life cycle dynamics, through their mode of inheritance
15925–15499 Membrane attachment site and through tissue-specific control of energy production.
16194–16208 Control element
16499–16506 L-strand control element Replication and transcription
16157–16172 Termination sequence
Although the topology and major events of mitochondrial
(From Mitomap URL http://www.gen.emory.edu/cgi-bin/MITOMAP/bin/ replication and transcription are well established (Shadel and
tbl1gen.pl). Clayton, 1997), their differential control is still rather poorly
*Nucleotide positions are based on the published sequence of human mtDNA by understood. Transcription is from each strand, starting from
Anderson et al. (1981). light and heavy chain promoter regions (Table 2). This is bi-
directional, resulting in the production of large polycistronic
where there may be lack of complementarity between nuclear transcripts that are later processed to form mature RNAs
and mitochondrial genomes that may affect body weight (Table 1).
and performance (Gartner et al., 1998). Nuclear–mitochondrial Although there is interaction between the mitochondrial and
interactions will be discussed later with replication and nuclear genomes, mtDNA replication and mitochondrial div-
transcription. ision are clearly uncoupled from the nuclear division cycles.
The two strands of mtDNA have two separate origins for
replication. That for the H-strand (OH) is within the D-loop,
Function in tissue bioenergetics and ageing
whereas that for the L-strand (OL) is about one third of the way
Mitochondria largely control the production of energy by around the genome (Fig. 1). Replication is dependent on the
oxidative phosphorylation (OXPHOS). Defects caused by in- binding of the nuclear encoded transcription factor A to its
herited or cumulative mutations and deletions are now seen as binding site or sites (Table 2). This triggers transcription from
central to many disease states as well as apoptosis and the OH by the core mtRNA polymerase to form an RNA–DNA
ageing process (Shoffner and Wallace, 1990; Ozawa, 1997a; hybrid, involving some subset of the conserved sequence
Wallace, 1997). The mitochondrial genome is vulnerable to blocks (CSBs). The RNA–DNA hybrid acts as a template for as-
oxidative attack as it lies at the site of the electron transfer chain sembly of a new H-strand by DNA polymerase gamma.
(the site of intensive generation of reactive oxygen species) and Replication proceeds in a clockwise manner and when it
lacks protective histones. Consequently it mutates ten or more reaches position 5721–5798, OL is exposed and replication of
times faster than nuclear DNA. Mutations can take the form of the L strand commences in the opposite direction. The lag in
single or multiple base pair replacements or modifications. replication results in the formation of two daughter molecules,
Replication errors can also generate deletions by internal re- conventionally termed a and b: b consists of the original H
combination, generally around tandem repeat ‘hot spots’. strand plus a partially replicated L strand. This proceeds to
Deletions and mutations accumulate preferentially in post- complete replication of the L strand before circle closure.
mitotic tissues such as the heart and the central nervous Completion of replication involves supercoiling and the syn-
system. This is because, in rapidly dividing tissues, cell lines thesis of a new H strand. The whole process is very slow (E. coli
that accumulate lethal threshold concentrations of mtDNA replication is about 200 times faster), taking approximately 2 h
dysfunction can be eliminated: a form of cytological natural to complete. Replication is also prone to error; deletions of
selection. Post-mitotic tissues have no such way of eliminating large segments occur especially at regions of tandem repeat
‘bad’ mitochondria, which accumulate and eventually lead to ‘hot spots’ (Ozawa, 1997a).
irreparable damage. Laboratories attempting to quantify Replication and transcription appear to be differentially
mtDNA damage generally concentrate on major deletions regulated, and there are a number of regulator and promoter
such as the ‘common’ 4977 base pair deletion seen in human sites on the genome (Table 2).

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
Mitochondrial DNA in the life cycle 175

Tissue and species specificity not generally transmitted, there is no long-term selective ad-
vantage for mtDNA to be healthy in males – from the ‘selfish
The exact mechanisms by which different tissues control
gene’ point of view, the fitness of the male mitochondrial
their mitochondria are still being debated (Shadel and Clayton,
genome is irrelevant. One could even argue a scenario in which
1997). Replication and transcription do not appear to be regu-
a maternally inherited defective mtDNA in a man may inhibit
lated by availability of DNA polymerase, as expression of the
his fitness and thus allow his sisters to flourish. At least one of
catalytic subunit of DNA polymerase gamma varies little
the mitochondrial DNA diseases (Leigh’s Hereditary Optic
among different tissues and is not increased in muscle mito-
Neuropathy, LHON) is more prevalent and occurs earlier in
chondrial biogenesis after motor nerve stimulation. One
males than in females, which reinforces this hypothesis (Johns,
possibility for control is through the nuclear-encoded RNA
1995). Moreover the free lifespan of spermatozoa, exposed to
polymerase that recognizes promoters in a species-specific
enormous ranges in oxygen tension (Max, 1992), coupled with
manner (Schultz et al., 1998) This could be the mechanism by
poor antioxidant defence systems leaves the mtDNA of the
which higher primate mtDNA can substitute for human
spermatozoa highly vulnerable to damage. There is evidence
mtDNA in a depleted cell (see below), as there are high degrees
that some forms of male infertility may be associated with
of conservation in the D-loop sequences for the promoter re-
defects in mitochondrial function and testicular OXPHOS
gions and for mtTF1 (Kenyon and Moraes, 1997). It is signifi-
(Cummins et al.,1994a).
cant that mtDNA replication within the cell begins close to
the nucleus as this clearly demonstrates the importance of
nuclear-derived signals for activating this process (Davis and Mitochondria in spermatogenesis
Clayton, 1996). Larsson et al. (1998) used targeted gene
It is necessary to trace the fate of the mitochondria in sper-
knockout in mice for the gene for mitochondrial transcription
matogenesis to form a working hypotheses on why the mito-
factor A (mtTFA now renamed Tfam). Heterozygous embryos
chondria from spermatozoa are not generally transmitted after
show reduced mtDNA copy number and respiratory chain
sperm entry (Clermont et al., 1993; Hecht, 1995).
deficiency in cardiac muscle, but homozygous knockout em-
The mitochondria that populate the primordial germ cells
bryos have severe depletion of mtDNA and no OXPHOS and
and the support elements of the testis (Sertoli, Leydig, peri-
die before day 10.5.
tubular myoid and other connective tissue cells) are typical of
The development of ‘clones’ such as the sheep Dolly
somatic cells. With the onset of meiosis, the germ cell mito-
(Wilmut et al., 1997) produced by nuclear transfer highlight the
chondria start to differentiate and the mitochondrial matrix be-
need for understanding the relationship between the nucleus
comes diffuse and vacuolated. Differentiating spermatids also
and the cytoplasm. There is clearly a degree of species spec-
lack heat shock protein 60 (hsp60), a mitochondrial chaperonin,
ificity in mitochondrial function. Kenyon and Moraes (1997)
which suggests that they may also be deficient in protein im-
used a human cell line depleted in mtDNA (Rho 0) and found
port mechanisms (Meinhardt et al., 1995). These ‘germ cell’
that mtDNA from gorillas, chimpanzees and pygmy chim-
mitochondria finally make up the majority (> 80%) of those
panzees could substitute for human mtDNA in supporting res-
found in the testes of mature animals. The mitochondria cluster
piration and mitochondrial protein synthesis, while mtDNA
near the nucleus during meiosis and spermiogenesis and then
from orangutans, and species representative of Old World
differentiate progressively into the crescentic mature sperm
monkeys, New World monkeys and lemurs could not. Within
type. Some proliferation may occur early in spermiogenesis,
species it also appears that there may be factors that select for
as indicated by mtDNA replication, but copy numbers later
and against individual mtDNA types. Jenuth et al. (1997) found
decline so that each mature sperm mitochondrion contains, on
tissue-specific and age-related variations in mtDNA genotype
average, only one copy of mtDNA. However there is no evi-
survival in heteroplasmic mice, suggesting that individual tissue
dence of any gross alteration or selective methylation of the
energetic demands could bias the survival or segregation of
sperm mtDNA that could explain its later loss in the embryo
different mitochondrial genomes. Meirelles and Smith (1997)
(Hecht and Liem, 1984; Hecht et al., 1984; Hecht, 1997). The
found significant variation in the segregation of different mito-
midpiece is delineated by caudal migration of the annulus and
chondrial genomes in mice produced by karyoplast fusion.
then mitochondria migrate along the axoneme in a controlled
However, some of this may have been due to the placement of
stepwise fashion to form a tight array (Otani et al., 1988).
exogenous mitochondria in relation to the nucleus and to em-
Differentiation of the sperm mitochondria into a crescentic
bryonic axes (see below). Nagao et al. (1998) have shown sig-
shape appears to be pre-programmed, as it also occurs in
nificant depression of OXPHOS-dependent exercise capacity in
the superfluous mitochondria of the excess cytoplasm.
interspecific and inter-subspecific congenic mice produced by
Moreover, it appears to depend on specific interaction with
repeated backcrossing, pointing to poor coordination between
the Sertoli cell (Seitz et al.,1995). Formation of the midpiece is
the mismatched nuclear and mitochondrial genomes. The
accompanied by the secretion of a selenoprotein-based mito-
question of species-specific recognition of mitochondria by the
chondrial sheath capsule and a sub-mitochondrial reticulum
host cell returns when inheritance is considered.
that anchors the sheath to the axoneme proper (Olson and
Winfrey, 1992). The capsule consists of at least four proteins
ranging from 17 to 31 kDa (Cataldo et al., 1996). Presumably
Why is there maternal inheritance of mtDNA ?
this structure conveys mechanical strength, as the sperm
Males, like females, receive their mtDNA from their mother. mitochondria are relatively resistant to swelling under
This unequal mode of inheritance may have consequences for osmotic stress compared with other cytoplasmic components
health and fertility. As the mitochondria of spermatozoa are (Willoughby et al., 1996). Spermatozoa contain the highest

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
176 J. Cummins

relative concentration of selenium of any cell line in the body,


What happens at fertilization?
and animals fed a selenium-deficient diet show midpiece
defects (Cataldo et al., 1996). The protein is probably the anti- A flood of misinformation in undergraduate textbooks and
oxidant, phospholipid hydroperoxide glutathione peroxidase popular texts is promoting the ‘African Eve’ model of human
(Roveri et al., 1994), which is intriguing given the known evolution and minimizes and even denies the possibility of
susceptibility of spermatozoa to oxidative damage and lipid paternal mtDNA transmission (Ankel-Simons and Cummins,
peroxidation (Aitken, 1995). 1996). At least one well-respected author (Richard Dawkins)
Other sperm-specific unique mitochondrial proteins also has claimed (but later retracted) that spermatozoa are too small
appear during spermiogenesis. These include a sperm-specific to contain mitochondria, a statement that comes as a surprise to
lactic dehydrogenase isoenzyme that appears early (Machado most biologists (Dawkins, 1986, 1995). In fact, total sperm entry
de Domenech et al., 1972) and a unique isoform of cytochrome c into the egg including the midpiece mitochondria is the rule
(Goldberg et al., 1977). The spermatid nuclei lose the mito- rather than the exception for most mammals. However, it is
chondrial targeting sequence of a testis-specific isoform of nu- now well established that the mitochondria from spermatozoa
clear mitochondrial transcription factor A (Larsson et al., 1994, are targeted for destruction by endogenous proteolytic activity
1997), suggesting that there is no need for mitochondria to during early embryogenesis (see below). Uniparental (gener-
replicate once on the midpiece. However, the mitochondrial ally but not universally maternal) inheritance of cytoplasmic
transcriptional potential of the spermatozoon is apparently un- organelles like mitochondria and, in plants, plastids, is ac-
changed during this process (Alcivar et al., 1989). Larsson et al. complished by a wide variety of strategies and thus is clearly of
(1996) suggest that the nuclear isoform of mtTFA, which is also profound importance to long-term fitness. It is usually argued
present in spermatocytes, plays a secondary structural role in that uniparental inheritance in sexually reproducing organisms
compacting nuclear DNA during spermiogenesis. This would minimizes the risk of lethal genomic conflict between asexually
be consistent with its known capacity to induce conformational reproducing cytoplasmic genes (Hurst, 1992, 1995; Hurst et al.,
changes in DNA (Fisher et al., 1992). 1996) but how this is accomplished remains one of the major
puzzles for evolutionary reproductive biology. Indeed Birky
(1995) states that “... the variety of molecular and cellular mech-
Role of mitochondria in sperm function
anisms found in different organisms is matched only by the
Although the mitochondrial sheath of spermatozoa clearly variety of hypotheses devised to explain the evolution of
has a functional role, rather surprisingly there is little consen- the phenomenon”. Perhaps the most bizarre example is in the
sus about the relative importance of mitochondrial respiration giant spermatozoa of some species of Drosophila, in which
(as opposed to glycolysis) for motility, or even for fertilization the male mitochondria are sequestered to the midgut during
itself (Bedford and Hoskins, 1990). Spermatozoa in which the organogenesis and defaecated by the larvae soon after hatching
mitochondria have been poisoned with cyanide can induce (Pitnick and Karr, 1998).
normal embryo development when microinjected into oocytes
(Ahmadi and Ng, 1997). Spermatozoa are metabolically flexible
Paternal transmission – an anomaly?
and, in some species, can switch between aerobic and anaerobic
metabolism. This perhaps reflects the great range of oxygen Most of the evidence indicating the possibility of paternal
tensions they experience, from near anoxia in the testis and transmission of mtDNA derives from interspecific crosses,
epididymis to ambient tensions in the vagina and in vitro which by definition are uncommon in nature (Kaneda et al.,
(Ford and Rees, 1990; Max, 1992; Cummins et al., 1994b). New 1995). Backcrossing studies in Mus spretus + Mus musculus
techniques, such as flow cytometry using vital dyes and fluoro- hybrids showed a ‘leakage’ of paternal mtDNA of about 1 in
chromes, that can indicate the degree of mitochondrial mem- 1000 to 1 in 10 000 molecules (Gyllensten et al., 1991). This is not
brane potential promise to elucidate the relationship between far from the theoretical ratio of oocyte to sperm mtDNA seen at
mitochondrial function, overall sperm viability and fertilizing fertilization (Ankel-Simons and Cummins, 1996) but this obser-
potential (Garner et al., 1997). Like somatic mtDNA, that of vation of leakage may be an artefact (see below). Kaneda et al.
spermatozoa is highly vulnerable to mutation, and a significant (1995) investigated this question in an ingenious manner, using
number of mtDNA deletions are found in the semen of at least a nested polymerase chain reaction (PCR) assay that could de-
50% of normospermic men (Cummins et al., 1998). Given the tect sperm mtDNA in a single mouse embryo. In embryos pro-
lengthy process of spermiogenesis and epididymal maturation duced by in vitro fertilization of Mus musculus eggs with the
during which the sperm mitochondria have to survive without same strain of spermatozoa, paternal mtDNA could be detected
any interaction with the nuclear genome, and given the likeli- in zygotes but only at the early pronuclear stage, and its dis-
hood that they will be exposed to mutagenic agents, this is per- appearance coincided with loss of membrane potential in
haps not surprising. Indeed, the need to exclude defective sperm-derived mitochondria, as measured by Rhodamine 123
sperm mtDNA from contributing to the embryo is possibly one fluorescence. By contrast, when Mus spretus spermatozoa were
of the major selection pressures against survival of paternal used to fertilize M. musculus eggs, paternal mtDNA was de-
mtDNA. Indeed Short (1998) has suggested that this asym- tected at the pronucleus stage, at the two-cell stage and in
metric inheritance of mtDNA, through the oocyte but not the neonates. This was confirmed by sequencing the PCR product.
spermatozoon, may be a fundamental driving force behind When spermatozoa from the congenic strain b6.mt(spr), which
amphimixis and anisogamy. This is because of the need to con- carries M. spretus mtDNA on a background of M. musculus
serve a healthy stock of mtDNA for embryo development, nuclear genes, was used, the M. Spretus mtDNA was elimi-
through a long period of quiescence in meiosis. nated early in development, as in intraspecific crosses (Kaneda

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
Mitochondrial DNA in the life cycle 177

et al.,1995). Kaneda et al. (1995) proposed that the zygote cyto- Table 3. Possible mechanisms for uniparental inheritance of mammalian
plasm has a species-specific mechanism that recognizes and mitochondria
eliminates sperm mitochondria, on the basis of nuclear DNA
encoded proteins in the sperm midpiece, and not on the Mechanism Evidence in mammals?
mtDNA itself, nor on the proteins it encodes. In further work,
the same group found that the ‘leaked’ paternal mtDNA fol- Prezygotic
lowing interspecific crosses is limited to the first generation, Anisogamy – disparity in numbers between Yes
does not get transmitted to all tissues and does not survive eggs and spermatozoa
into subsequent back-crosses (Shitara et al., 1998). This calls into Exclusion of mitochondria from spermatozoa No
question the original findings of Gyllensten et al. (1991) and Degradation of mitochondria in spermatozoa No
suggests that the congenic mouse strain they used in detecting Degradation of mtDNA in spermatozoa Probable
paternal leakage may have retained a stable heteroplasmy. This Fertilization
may have arisen from mitochondrial fusion as is thought to Exclusion of sperm mitochondria from zygote Very rare
have occurred in at least one line of mice produced by cytoplast
transfer (Meirelles and Smith, 1997). Zygotic/deterministic
Although this picture appears convincing, there are some Selective degradation of sperm mitochondria Yes
problems with these data and the results have not been repli- Selective degradation of sperm mtDNA No
cated by other laboratories. First, nested PCR detection of very Targeted partitioning of sperm mitochondria Remotely possible
small amounts of DNA is fraught with problems of potential into non-embryonic cells
contamination, and most laboratories insist on dual facilities Zygotic/stochastic
with double-blind controls when carrying out procedures such Random partitioning of sperm mitochondria Remotely possible
as embryo sexing. It is not clear whether such precautions into non-embryonic cells
were taken here. Second, the authors observed that midpiece
Rhodamine fluorescence disappeared at the late pronuclear
stage in intraspecific embryos, indicating loss of mitochondrial
membrane potential. This is very different from our own obser- embryos have a generalized defence as opposed to an intra-
vations using the stable fluorochrome MitoTrackerr (Cummins specific defence against exogenous mitochondria. Hecht (1984)
et al., 1997) in which viable midpiece mitochondria were found was unable to detect microinjected conspecific testicular and
up to day 3 in four-cell embryos and up to day 5 in non- liver mitochondria in offspring but, more recently, transmission
activated oocytes and arrested two- and four-cell embryos. of mutant human mtDNA (from an individual who died of
By contrast, other sperm tail components, such as the coarse mitochondrial disease) has been reported in mouse embryos
tail fibres, can be identified up to the blastocyst stage and poss- (Rinaudo et al., 1996). Pinkert at al. (1997) have also reported that
ibly even later. Clearly there is species-specific recognition and Mus spretus liver mitochondria microinjected into Mus domesticus
destruction of the mitochondrial sheath, but it occurs later in zygotes persist to the blastocyst stage. Such models will be very
embryogenesis than was proposed by Kaneda et al. (1995). valuable tools for the study of mtDNA disease transmission
Moreover the mitochondria in the interspecific-cross embryos and in the search for means to target and perhaps even cure
of Kaneda and co-workers (in which the paternal mtDNA per- such disorders (Taylor et al., 1997).
sisted) should have been visible by light microscopy, but the
authors made no comment on this. Findings in other rodent
How are sperm mitochondria recognized by the zygote?
species and cattle (summarized in Cummins et al., 1997) all in-
dicate independently that sperm mitochondria persist into the At present the mechanism that recognizes and targets the
second cell cycle before lysis. Sutovsky et al. (1996) proposed sperm midpiece for destruction is not known although there
that the mitochondrial sheath in cattle may be tagged with is now evidence that it is mediated by ubiquitin (Sutovsky,
ubiquitin for proteolytic destruction and they now have pre- personal communication). This is one of the central mysteries
liminary evidence confirming this interesting idea (Sutovsky, of reproductive biology. The key may lie in the asynchrony
personal communication). between mtDNA transcription and replication in the embryo.
The simplest explanation (apart from dilution beyond the poss-
ibility of subsequent detection) is that the sperm mitochondria
Are sperm mitochondria viable?
cannot maintain membrane potential up to day 6.5 when repli-
Single somatic cell mitochondria can colonise mtDNA- cation of the mtDNA of the embryo begins (Pikó and Taylor,
depleted cell lines under experimental conditions in vitro, albeit 1987; Ebert et al., 1988). Leakage of key components such as
at low efficiency (King and Attardi, 1989). Manfredi et al. (1997) cytochrome c, a known trigger for apoptosis (Ozawa, 1997b)
found even lower long-term colonization when spermatozoa from the mitochondria, may signal multivesicular bodies to
were fused with mitochondrially depleted somatic cell lines surround the mitochondria and digest them (Hiraoka and
in vitro. Only 10–20% of cells had mitochondria demonstrating Hirao, 1988) Against this must be set the observations on
normal transmembrane potential when examined 4 h after persistence of paternal mtDNA in interspecific mouse crosses
fusion. One possibility is that the highly differentiated sperm (Kaneda et al., 1995; Shitara et al., 1998).
mitochondria lack functional protein import mechanisms and Several other possibilities are summarized on the basis of
therefore cannot respond to nuclear-encoded signals when they Birky (1995) (Table 3). The actual mechanism remains to be
receive them (Neupert, 1997). It is not yet clear whether eggs and elucidated fully.

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
178 J. Cummins

The role of leptin in the embryo is obscure but presumably it is


Embryogenesis, oogenesis and the
involved with resource allocation between the ICM and the
‘restriction–amplification’ model of mtDNA transmission
trophectoderm. Clearly our ideas about the ‘totipotency’ of
With very few exceptions such as mussels, most organisms the individual embryo cells up to the eight-cell stage need to
appear intolerant of heteroplasmy for mtDNA, even though be revised in a major way (Edwards and Beard, 1997).
stable heteroplasmic lineages can be created artificially (Jenuth That there is significant polarity in the oocyte and embryo
et al., 1996, 1997). When it does occur it generally arises by may help to explain the skewed segregation of mitochondrial
de novo mutation during oogenesis and in humans is frequently genotypes in heteroplasmic mice created by cytoplast or
associated with progressive mitochondrial bioenergetic dis- karyoplast fusion. When mitochondria were placed in the
eases in offspring. Many homoplasmic mutations are probably periphery of oocytes (cytoplast transplantation) the resulting
lethal in early embryogenesis owing to oxidative phosphoryl- mice showed significantly more variation than when mito-
ation insufficiency (Ozawa, 1997a; Wallace, 1997). Neutral chondria were placed near the (karyoplast derived) nucleus
mutations that arise can become fixed in the germline very (Meirelles and Smith, 1997, 1998). As it is not possible to place
rapidly – within one to three generations (Ashley et al., 1989). cytoplasts accurately to the primordial oocyte axis before
One possibility is that this may arise through a restriction– electrofusion there can be less certainty that they will end up in
amplification process during the exponential growth phase of the ICM and hence in the somatic (and germ) cells of the
oogenesis, in which mitochondria multiply by division from a embryo. Another factor to consider is that mitochondrial repli-
few precursors (Marchington et al., 1997; Nagley et al., 1993). cation starts in regions of the cytoplasm close to the nucleus
Blok et al. (1997) even suggested that the progenitor pool could (Shadel and Clayton, 1997), so spatial positioning may affect
be a single mitochondrion in humans. During oogenesis, this mitochondrial transmission.
pool expands considerably resulting in about 105 mitochondria In mice the amount of mtDNA – and presumably therefore
in the mature oocyte (Pikó and Taylor, 1987). the number of mitochondria – does not change during develop-
Alternatively, Jenuth et al. (1996, 1997) suggest that the ‘bottle- ment up to the blastocyst and early egg cylinder stage (Ebert
neck’ may be an artefact caused by random genetic drift during et al., 1988). There are approximately 910 cells at day 6.5
early embryogenesis. By contrast, Steinborn et al. (1998a,b) found (Hogan et al., 1986) when the primordial germ cells (PGCs)
that the ratio of different mtDNAs found in cattle, produced by first differentiate from the proximal epiblast, so if mitochondria
fusion of nucleated blastomeres with enucleated metaphase IIm are randomly apportioned between cells during this time,
oocytes, was closely related to the stage of the donor embryo: the founder PGC will receive approximately 100 founder
13–18% when the donor was a 24-cell morula and 0.4–0.6% when mitochondria. If mitochondria are not equally segregated to
the donor was a 92-cell morula. Thus there was no doubt that this cell line, the number could be even smaller (Olivo et al.,
donor embryo mtDNA can persist in such heteroplasmic clones. 1983; Ashley et al., 1989). This is a similar order of magnitude
This hypothesis is currently contentious. An anonymous to the numbers in oogonia proposed by Jenuth et al. (1996)
reviewer of the present paper stated “The clonal expansion of (see above). It is possible that only follicles containing oocytes
a limited pool of mtDNA precursors could be likened to an with a high degree of homoplasmy are permitted to achieve
urban myth: there is no direct evidence for it, and perhaps no dominance. One way this could be tested would be to evaluate
need to propose it”. One trusts that the myth, if it is one, will OXPHOS parameters and mtDNA integrity in dominant com-
soon be resolved. pared with atretic oocytes.

Mitochondria in oogenesis and axis formation in embryogenesis Mitochondrial DNA in the ageing ovary
The mammalian oocyte contains 2 × 105 copies of the mito- Unlike spermatogenesis which is continuous from puberty,
chondrial genome in approximately 100 000 mitochondria. oogenesis starts during fetal life (Baker, 1982; Byskov, 1982).
In immature oocytes these appear to be evenly distributed; Oogonia proliferate by mitosis, reaching a peak in mid-
however, Calarco (1995) found that a distinct polarity appeared gestation. In most mammals, oogonia enter meiosis at this time
after meiosis re-started. A cortical group of mitochondria but arrest at dictyotene. Oocyte growth resumes with recruit-
marks the extrusion point of the first polar body. Other periph- ment of primordial follicles during the ovulatory cycle and
eral foci of mitochondria tend to be concentrated in the hemi- meiosis normally completes only at about the time of ovulation
sphere containing the metaphase II spindle. New observations and fertilization. Thus there is an extended time (50 years or
on the distribution of leptin and STAT3 (Antczak and Van more in women) when the nuclear and cytoplasmic integrity of
Blerkom, 1997) demonstrate that the axes of the oocyte and the oocyte need to be maintained without cell division. Atresia
resulting embryo are determined very early – perhaps by pos- at all stages means that the vast majority of oocytes never make
itioning in relation to blood flow and oxygen availability in the it to ovulation: of the 7 million found at mid-gestation in
follicle. Leptin is the 16 kDa cytokine product of the obese gene humans only a few hundred will ever reach ovulatory potential
(ob) and activates STAT3, one of the Signal Transducer and (Baker, 1982). Lactational amenorrhoea coupled with birth
Activation of Transcription family of proteins. The first cell div- spacing of about four years in hunting–gathering societies
ision is meridional resulting in two cells with equal leptin/ means that probably only 10–15 oocytes were ever released in a
STAT3 gradients. However, the second cell cycle has one woman’s life while humans were evolving. Even the menstrual
meridional and one equational division. This results in one cycle may have developed as a non-adaptive consequence of
blastomere being leptin/STAT3 deficient and this cell goes uterine evolution (Finn, 1998). Not surprisingly, the fertility
on to form the inner cell mass (ICM) of the blastomere (Fig. 2). of individual oocytes declines with age along with reduced

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
Mitochondrial DNA in the life cycle 179

Fertilization

Two-cell
mtDNA transcription
starts

Syngamy
Polarity established

Four-cell
Axis commitment
Spermatogenesis Sperm mitochondria
Oogenesis disappear
Reduction in mtDNA
Restriction/amplification copy number
of mtDNA Mitochondrial
Massive atresia differentiation
Prolonged dictyate Loss of replication
meiosis capacity?

Germ cell line

Tissue differentiation Day 6–7 – egg cylinder


Germ cell commitment
mtDNA replication starts
Somatic cell line
Random segregation of mtDNA to tissues
Tissue-specific selection?
Age-specific selection?
Age-related mutations/deletions
Critical threshold effects
Declining OXPHOS efficiency
Ageing and death

Fig. 2. This schematic life-cycle summarizes some of the major ways in which the life cycle of the eukaryotic host interlocks with that of its
mitochondria. The concepts of gradients and axis formation in the embryo are based on those of Antczak and Van Blerkom (1997). OXPHOS,
oxidative phosphorylation.

capacity of the reproductive tract to support pregnancy (Tarin, 1996). However, the data will be hard to evaluate until
(Adams, 1984). There is considerable interest in the potential accurate methods for quantifying mtDNA deletions and point
role of mtDNA in this process, together with the effects of mutations have been developed. Even normal human oocytes
altered OXPHOS and cellular antioxidant systems on the cyto- can be shown to contain mtDNA deletions and rearrangements
skeleton, fertilization and subsequent embryo development using extensive PCR cycling (Chen et al., 1995; Keefe et al.,

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
180 J. Cummins

1995). Kitagawa et al. (1993) found large numbers of deletions Conclusions


in women aged 45 and over, and Müller-Hocker et al. (1996)
The role of mitochondria in the reproductive life cycle still
showed age-related declines in OXPHOS enzyme components,
holds many mysteries. Key questions remain to be answered.
and alterations in mitochondrial morphometry that could cor-
What is the mechanism that recognizes and eliminates same-
relate with declining fertility. These alterations could relate to
species but not foreign spermatozoa in the early embryo?
the reduced ATP content and potential for development in the
Do sperm mitochondria have functional mitochondrial
poorer quality oocytes and embryos found in older women
import and export mechanisms? If so can they respond to
(Van Blerkom et al., 1995; Van Blerkom, 1997). This is a rich
transcription factors in vitro, in the embryo and in other cell
potential area for future research, as we may find that oocyte
systems? Can they evade the normal surveillance mechanisms
potential is defined by intra-ovarian factors such as blood
of the embryo?
supply and local oxygen tension.
Is the location of the sperm midpiece after syngamy constant
There are already reports of clinics attempting to improve
in relation to embryonic axes?
IVF results by donor cytoplasmic transfer in women with
What relationship is there between mitochondrial integrity
poor quality oocytes and recurrent implantation failure (Cohen
and oocyte atresia?
et al., 1997, 1998). This was coupled in at least one case with re-
moval of cytoplasmic fragments from embryos, but in this case These ideas could not have been generated without the generosity
mid-pregnancy testing revealed that the mtDNA was of the of a number of colleagues over the past few years, especially Norman
mother not the donor, so that the benefits of manipulation Hecht, Anne Jequier, Roger Martin, Roger Short, Eric Shoubridge,
are unclear. Lawrence Smith, Bayard Storey, Peter Sutovsky and Ryuzo Yanagimachi.
Thanks also to Tim Karr and Scott Pitnick for allowing me to quote
from their paper in print.
Why don’t mitochondria have sex?
Sex, or more specifically homologous recombination (HR) be-
References
tween mtDNA molecules, is assumed not to occur. Even when
there is clear evidence of heteroplasmy within the cytoplasm Key references are indicated by asterisks.
Adams CE (1984) Reproductive senescence. In Reproduction in Mammals. Book 4.
there is no convincing evidence that HR occurs between popu-
Reproductive Fitness (2nd Edn) pp 210–233 Eds CR Austin, RV Short.
lations (Hayashi et al., 1994). However, under some circum- Cambridge University Press, Cambridge
stances mitochondrial DNA may be capable of recombination Ahmadi A and Ng SC (1997) Sperm head decondensation, pronuclear for-
and short-term repair (Thyagarajan et al., 1996; Salazar and mation, cleavage and embryonic development following intracytoplasmic
Vanhouten, 1997; Anson et al., 1998). Howell (1997) reviewed injection of mitochondria-damaged sperm in mammals Zygote 5 247–253
Aitken RJ (1995) Free radicals, lipid peroxidation and sperm function
the evidence and concluded that “_ the physiological signifi- Reproduction, Fertility and Development 7 659–668
cance of this recombinase activity remains inaccessible, because Alcivar AA, Hake LE, Millette CF, Trasler JM and Hecht NB (1989)
there is still neither any evidence for reciprocal recombination Mitochondrial gene expression in male germ cells of the mouse
of mtDNA molecules nor any apparent reason that such an Developmental Biology 135 263–271
Anderson S, Bankier AT, Barrell BG et al. (1981) Sequence and organization
activity would be beneficial.” Howell (1997) suggests that the
of the human mitochondrial genome Nature 290 457–465
HR described by Thyagarajan et al. (1996) may be due to a *Ankel-Simons F and Cummins JM (1996) Misconceptions about mito-
mistake in the topoisomerase–resolvase complex that separates chondria and mammalian fertilization –implications for theories on
and re-seals daughter mtDNA molecules at replication. This is human evolution Proceedings of the National Academy of Sciences USA 93
an imperfect process that frequently results in concatenated 13 859–13 863
Anson RM, Croteau DL, Stierum RH, Filburn C, Parsell R and Bohr VA
oligomers and other configurations (Howell et al., 1984; Shadel (1998) Homogenous repair of singlet oxygen-induced DNA damage in
and Clayton, 1997). Indeed there is not normally much oppor- differentially transcribed regions and strands of human mitochondrial
tunity for mtDNA molecules to meet as they are sequestered DNA Nucleic Acids Research 26 662–668
within the double mitochondrial membrane system. Under Antczak M and Van Blerkom J (1997) Oocyte influences on early develop-
ment: the regulatory proteins leptin and STAT3 are polarized in mouse
certain circumstances mitochondria may have the capacity to
and human oocytes and differentially distributed within the cells of the
fuse and form stable heteroplasmic lineages (Jenuth et al., 1996; preimplantation stage embryo Molecular Human Reproduction 3 1067–1086
Jenuth et al., 1997; Meirelles and Smith, 1997, 1998) Indeed Ashley MV, Laipis PJ and Hauswirth WW (1989) Rapid segregation of
mitochondrial fusion as well as fission is normal in oogenesis heteroplasmic bovine mitochondria Nucleic Acids Research 17 7325–7331
and embryogenesis (Smith and Alcivar, 1993). There is also Baker TG (1982) Oogenesis and ovulation. In Reproduction in Mammals. Book 1.
Germ Cells and Fertilization (2nd Edn) pp 17–45 Eds CR Austin and
a complex tissue-specific and species-specific set of interactions RV Short. Cambridge University Press, Cambridge
between the nuclear and mitochondrial genomes that govern *Bedford JM and Hoskins DD (1990) The mammalian spermatozoon:
both transcription and mtDNA replication (Poyton and McEwen, morphology, biochemistry and physiology. In Marshall’s Physiology of
1996). It is possible that the lack of mitochondrial HR is a Reproduction (4th Edn pp 379–568 Ed. GE Lamming. Churchill Livingston,
London
fundamental aspect of eukaryotic evolution that is possibly
*Birky CW (1995) Uniparental inheritance of mitochondrial and chloroplast
based on the need to provide the next generation with a genes: mechanisms and evolution Proceedings of the National Academy of
highly competent OXPHOS capacity. The generation of di- Sciences, USA 92 11 331–11 338
versity by mitochondrial recombination could lead to lethal Blok RB, Gook DA, Thorburn DR and Dahl HH (1997) Skewed segregation
genomic conflicts and hence reduced fitness (Hurst, 1992). To of the mtDNA nt 8993 (T–>G) mutation in human oocytes American
Journal of Human Genetics 60 1495–1501
coin an analogy: if sex is democracy and mitochondria are Byskov AG (1982) Primordial germ cells and regulation of meiosis. In Germ
the slaves of the organism, then democracy for the slaves is a Cells and Fertilization (2nd Edn) pp 1–16 Eds CR Austin and RV Short.
luxury the organism can well do without. Cambridge University Press, Cambridge

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
Mitochondrial DNA in the life cycle 181

Calarco PG (1995) Polarization of mitochondria in the unfertilized mouse Hecht N (1995) The making of a spermatozoon: a molecular perspective
oocyte Developmental Genetics 16 36–43 Developmental Genetics 16 95–103
Cataldo L, Baig K, Oko R, Mastrangelo MA and Kleene KC (1996) Hecht N (1997) Genetic control of spermatogenesis: where can things go
Developmental expression, intracellular localization, and selenium wrong? In Genetics of Human Male Fertility pp 11–24 Eds C Barratt, C De
content of the cysteine-rich protein associated with the mitochondrial Jonge, D Mortimer and J Parinaud. Editions E.D.K., Paris
capsules of mouse sperm Molecular Reproduction and Development 45 Hecht NB and Liem H (1984) Mitochondrial DNA is synthesized during
320–331 meiosis and spermiogenesis in the mouse Experimental Cell Research 154
Chen X, Prosser R, Simonetti S, Sadlock J, Jagiello G and Schon EA (1995) 293–298
Rearranged mitochondrial genomes are present in human oocytes Hecht NB, Liem H, Kleene KC, Distel RJ and Ho SM (1984) Maternal in-
American Journal of Human Genetics 57 239–247 heritance of the mouse mitochondrial genome is not mediated by a loss or
Clayton DA (1982) Replication of animal mitochondrial DNA Cell 28 693–705 gross alteration of the paternal mitochondrial DNA or by methylation of
Clermont Y, Oko R and Hermo L (1993) Cell biology of mammalian the oocyte mitochondrial DNA Developmental Biology 102 452–461
spermiogenesis. In Cell and Molecular Biology of the Testis pp 332–376 Hiraoka J and Hirao Y (1988) Fate of sperm tail components after incor-
Eds C Desjardins and LL Ewing. Oxford University Press, New York, poration into the hamster egg Gamete Research 19 369–380
Oxford Hogan B, Constantini F and Lacy E (1986) Manipulating the Mouse Embryo.
Cohen J, Scott R, Schimmel T, Levron J and Willadsen S (1997) Birth of A Laboratory Manual (1st Edn) Cold Spring Harbor Laboratory, Cold
infant after transfer of anucleate donor oocyte cytoplasm into recipient Spring Harbour
eggs Lancet 350 186–187 *Holliday R (1995) Understanding Ageing (1st Edn) Cambridge University
Cohen J, Scott R, Alikani M, Schimmel T, Munné S, Levron J, Wu L, Press, Cambridge
Brenner C, Warner C and Willadsen S (1998) Ooplasmic transfer in Howell N (1997) mtDNA recombination: what do in vitro data mean?
mature human oocytes Molecular Human Reproduction 4 269–280 American Journal of Human Genetics 61 19–22
Cortopassi GA and Wang E (1996) There is substantial agreement among Howell N, Huang P and Kolodner RD (1984) Origin, transmission, and
interspecies estimates of DNA repair activity Mechanisms of Ageing and segregation of mitochondrial DNA dimers in mouse hybrid and cybrid
Development 91 211–218 cell lines Somatic and Cellular Molecular Genetics 10 259–274
Cummins JM, Densley E, Jequier AM, Meloni BP and Sutarno (1994a) Hurst LD (1992) Intragenomic conflict as an evolutionary force Philosophical
Human male infertility and mitochondrial DNA. In The Seventh Transactions of the Royal Society of London Series B: Biological Sciences 248
International Symposium on Spermatology Cairns, p 7.5 135–140
Cummins JM, Jequier AM and Kan R (1994b) Molecular biology of human Hurst LD (1995) Selfish genetic elements and their role in evolution – the
male infertility – links with aging, mitochondrial genetics, and oxidative evolution of sex and some of what that entails Philosophical Transactions of
stress? Molecular Reproduction and Development 37 345–362 the Royal Society of London – Series B: Biological Sciences 349 321–332
Cummins JM, Wakayama T and Yanagimachi R (1997) Fate of microinjected *Hurst LD, Atlan A and Bengtsson BO (1996) Genetic conflicts Quarterly
sperm components in the mouse oocyte and embryo Zygote 5 301–308 Review of Biology 71 317–364
Cummins JM, Jequier AM, Martin R, Mehmet D and Goldblatt J (1998) Jenuth JP, Peterson AC, Fu K and Shoubridge EA (1996) Random genetic
Semen levels of mitochondrial DNA deletions in men attending an in- drift in the female germline explains the rapid segregation of mammalian
fertility clinic do not correlate with phenotype Journal of Andrology 21 mitochondrial DNA Nature Genetics 14 146–151
47–52 Jenuth JP, Peterson AC and Shoubridge EA (1997) Tissue-specific selection for
Davis AF and Clayton DA (1996) In situ localization of mitochondrial DNA different mtDNA genotypes in heteroplasmic mice Nature Genetics 16 93–95
replication in intact mammalian cells Journal of Cell Biology 135 883–893 Johns DR (1995) Mitochondrial DNA and disease New England Journal of
Dawkins R (1986) The Blind Watchmaker (1st Edn) p 176 W.W. Norton and Medicine 333 638–644
Company, New York, London Kanazawa M, Yano M, Namchai C, Yamamoto S, Ohtake A, Takayanagi M,
Dawkins R (1995) River out of Eden (1st Edn) Phoenix, London Mori M and Niimi H (1997) Visualization of mitochondria with green
Ebert KM, Liem H and Hecht NB (1988) Mitochondrial DNA in the mouse fluorescent protein in cultured fibroblasts from patients with mitochondrial
preimplantation embryo Journal of Reproduction and Fertility 82 145–149 diseases Biochemical and Biophysical Research Communications 239 580–584
*Edwards RG and Beard HK (1997) Oocyte polarity and cell determination in Kaneda H, Hayashi JI, Takahama S, Taya C, Lindahl KF and Yonekawa H
early mammalian embryos Molecular Human Reproduction 3 863–905 (1995) Elimination of paternal mitochondrial DNA in intraspecific crosses
Fawcett DW (1981) The Cell. An Atlas of Fine Structure (2nd Edn) W. B. Saunders during early mouse embryogenesis Proceedings of the National Academy of
Co, Philadelphia. Sciences USA 92 4542–4546
Finn CA (1998) Menstruation: a nonadaptive consequence of uterine evol- Keefe DL, Niven-Fairchild T, Powell S and Buradagunta S (1995)
ution. Quarterly Review of Biology 73 163–173 Mitochondrial deoxyribonucleic acid deletions in oocytes and repro-
Fisher RP, Lisowsky T, Parisi MA and Clayton DA (1992) DNA wrapping ductive aging in women Fertility and Sterility 64 577–583
and bending by a mitochondrial high mobility group-like transcriptional Kenyon L and Moraes CT (1997) Expanding the functional human mito-
activator protein Journal of Biological Chemistry 267 3358–3367 chondrial DNA database by the establishment of primate xeno-
*Ford WCL and Rees JM (1990) The bioenergetics of mammalian sperm mitochondrial cybrids Proceedings of the National Academy of Sciences USA
motility. In Controls of Sperm Motility: Biological and Clinical Aspects 94 9131–9135
pp 175–202 Ed. C Gagnon. CRC Press, Boca Raton King MP and Attardi G (1989) Human cells lacking mtDNA: repopulation
Garner DL, Thomas CA, Joerg HW, Dejarnette JM and Marshall CE (1997) with exogenous mitochondria by complementation Science 246 500–503
Fluorometric assessments of mitochondrial function and viability in cryo- Kitagawa T, Suganuma N, Nawa A, Kikkawa F, Tanaka M, Ozawa T and
preserved bovine spermatozoa Biology of Reproduction 57 1401–1406 Tomoda Y (1993) Rapid accumulation of deleted mitochondrial deoxy-
Gartner K, Bondioli K, Hill K and Rapp K (1998) High variability of body ribonucleic acid in postmenopausal ovaries Biology of Reproduction 49
sizes within nucleus-transfer-clones of calves – artifacts or a biological 730–736
feature Reproduction in Domestic Animals 33 67–75 Larsson NG, Oldfors A, Holme E and Clayton DA (1994) Low levels of mito-
Goldberg E, Sberna D, Wheat TE, Urbanski GJ and Margoliash E (1977) chondrial transcription factor A in mitochondrial DNA depletion
Cytochrome C: immunofluorescent localization of the testis-specific form Biochemical and Biophysical Research Communications 200 1374–1381
Science 196 1010–1012 Larsson NG, Garman JD, Oldfors A, Barsh GS and Clayton DA (1996)
Gyllensten U, Wharton D, Josefsson A and Wilson AC (1991) Paternal in- A single mouse gene encodes the mitochondrial transcription factor A
heritance of mitochondrial DNA in mice Nature 352 255–257 and a testis-specific nuclear hmg-box protein Nature Genetics 13 296–302
Hayakawa M, Katsumata K, Yoneda M, Tanaka M, Sugiyama S and Larsson NG, Oldfors A, Garman JD, Barsh GS and Clayton DA (1997)
Ozawa T (1996) Age-related extensive fragmentation of mitochondrial Down-regulation of mitochondrial transcription factor A during sper-
DNA into minicircles Biochemical and Biophysical Research Communications matogenesis in humans Human Molecular Genetics 6 185–191
226 369–377 Larsson NG, Wang J, Wilhelmsson H, Oldfors A, Rustin P, Lewandoski M,
Hayashi J, Takemitsu M, Goto Y and Nonaka I (1994) Human mitochondria Barsh GS and Clayton DA (1998) Mitochondrial transcription factor A is
and mitochondrial genome function as a single dynamic cellular unit necessary for mtDNA maintenance and embryogenesis in mice Nature
Journal of Cell Biology 125 43–50 Genetics 18 231–236

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access
182 J. Cummins

*Lodish H, Baltimore D, Berk A, Zipursky SL, Matsudaira P and Darnell J Pitnick S and Karr TL (1998) Paternal products and by-products in Drosophila
(1995) Molecular Cell Biology (3rd Edn) W.H. Freeman and Company, development Proceedings of the Royal Society of London, Series B 265 1–6
New York Poyton RO and McEwen JE (1996) Crosstalk between nuclear and mitochon-
Machado de Domenech E, Domenech CE, Aoki A and Blanco A (1972) drial genomes Annual Review of Biochemistry 65 563–607
Association of the testicular lactate dehydrogenase isozyme with a special Rinaudo P, Niven-Fairchild T, Buradugunta S, Massabrio M and Keefe D
type of mitochondria Biology of Reproduction 6 136–147 (1996) Preimplantation embryo development does not screen out mutant
Manfredi G, Thyagarajan D, Papadopoulou LC, Pallotti F and Schon EA mitochondrial DNA (mtDNA). In American Society for Reproductive
(1997) The fate of human sperm-derived mtDNA in somatic cells American Medicine Annual Conference Boston pp P-015
Journal of Human Genetics 61 953–960 Roveri A, Maiorino M, Nisii C and Ursini F (1994) Purification and charac-
Marchington DR, Hartshorne GM, Barlow D and Poulton J (1997) terization of phospholipid hydroperoxide glutathione peroxidase from rat
Homopolymeric tract heteroplasmy in mtDNA from tissues and single testis mitochondrial membranes Biochimica et Biophysica Acta 1208 211–221
oocytes – support for a genetic bottleneck American Journal of Human Salazar JJ and Vanhouten B (1997) Preferential mitochondrial DNA injury
Genetics 60 408–416 caused by glucose oxidase as a steady generator of hydrogen peroxide in
Max B (1992) This and that: hair pigments, the hypoxic basis of life and the human fibroblasts Mutation Research – DNA Repair 385 139–149
Virgilian journey of the spermatozoon Trends in Pharmacological Science 13 Schultz RA, Swoap SJ, McDaniel LD, Zhang BQ, Koon EC, Garry DJ, Li K
272–276 and Williams RS (1998) Differential expression of mitochondrial DNA
Meinhardt A, Parvinen M, Bacher M, Aumuller G, Hakovirta H, Yagi A and replication factors in mammalian tissues Journal of Biological Chemistry 273
Seitz J (1995) Expression of mitochondrial heat shock protein 60 in dis- 3447–3451
tinct cell types and defined stages of rat seminiferous epithelium Biology of Seitz J, Mobius J, Bergmann M and Meinhardt A (1995) Mitochondrial dif-
Reproduction 52 798–807 ferentiation during meiosis of mole germ cells International Journal of
Meirelles FV and Smith LC (1997) Mitochondrial genotype segregation in a Andrology 18 7–11
mouse heteroplasmic lineage produced by embryonic karyoplast trans- *Shadel GS and Clayton DA (1997) Mitochondrial DNA maintenance in ver-
plantation Genetics 145 445–451 tebrates Annual Review of Biochemistry 66 409–435
Meirelles F and Smith LC (1998) Mitochondrial genotype segregation during Shitara H, Hayashi J, Takahama S, Kaneda H and Yonekawa H (1998)
preimplantation development in mouse heteroplasmic embryos Genetics Maternal inheritance of mouse mtDNA in interspecific hybrids – segre-
148 877–883 gation of the leaked paternal mtDNA followed by the prevention of
Müller-Höcker J, Schäfer S, Weis S, Münscher C and Strowitzki T (1996) subsequent paternal leakage Genetics 148 851–857
Morphological–cytological and molecular genetic analysis of mito- Shoffner JM and Wallace DC (1990) Oxidative phosphorylation diseases.
chondria in isolated human oocytes in the reproductive age Molecular Disorders of two genomes Advances in Human Genetics 19 267–330
Human Reproduction 2 951–958 Short RV (1998) The difference between a testis and an ovary Journal of
Murphy MP (1997) Selective targeting of bioactive compounds to mito- Experimental Zoology 281 (in press)
chondria Trends in Biotechnology 15 326–330 Smith LC and Alcivar AA (1993) Cytoplasmic inheritance and its effects on
Nagao Y, Totsuka Y, Atomi Y, Kaneda H, Lindahl KF, Imai H and development and performance Journal of Reproduction and Fertility
Yonekawa H (1998) Decreased physical performance of congenic mice Supplement 48 31–43
with mismatch between the nuclear and the mitochondrial genome Genes Steinborn R, Zakhartchenko V, Jelyazkov J, Klein D, Wolf E, Muller M and
and Genetic Systems 73 21–27 Brem G (1998a) Composition of parental mitochondrial DNA in cloned
Nagley P, Zhang C, Martinus RD, Vaillant F and Linnane AW (1993) bovine embryos FEBS Letters 426 352–356
Mitochondrial DNA mutation and human aging: molecular biology, bio- Steinborn R, Zakhartchenko V, Wolf E, Muller M and Brem G (1998b)
energetics, and Redox therapy. In Mitochondrial DNA in Human Pathology Non-balanced mix of mitochondrial DNA in cloned cattle produced by
pp 137–157 Eds S DiMauro and D Wallace. Raven Press, New York cytoplast-blastomere fusion FEBS Letters 426 357–361
Neupert W (1997) Protein import into mitochondria Annual Review of *Sutovsky P, Navara CS and Schatten G (1996) Fate of the sperm mito-
Biochemistry 66 863–917 chondria, and the incorporation, conversion, and disassembly of the
Nosek J, Tomaska L, Fukuhara H, Suyama Y and Kovac L (1998) Linear sperm tail structures during bovine fertilization Biology of Reproduction 55
mitochondrial genomes – 30 years down the line Trends in Genetics 14 1195–1205
184–188 Tarin JJ (1996) Potential effects of age-associated oxidative stress on mam-
Olivo PD, Van de Walle MJ, Laipis PJ and Hauswirth WW (1983) malian oocytes/embryos Molecular Human Reproduction 2 717–724
Nucleotide sequence evidence for rapid genotypic shifts in the bovine Taylor RW, Chinnery PF, Clark KM, Lightowlers RN and Turnbull DM
mitochondrial DNA D-loop Nature 306 400–402 (1997) Treatment of mitochondrial disease Journal of Bioenergetics and
Olson GE and Winfrey VP (1992) Structural organization of surface domains Biomembranes 29 195–205
of sperm mitochondria Molecular Reproduction and Development 33 89–98 Thyagarajan B, Padua RA and Campbell C (1996) Mammalian mitochondria
Otani H, Tanaka O, Kasai K and Yoshioka T (1988) Development of mito- possess homologous DNA recombination activity Journal of Biological
chondrial helical sheath in the middle piece of the mouse spermatid tail: Chemistry 271 27 536–27 543
regular dispositions and synchronized changes Anatomical Record 222 Van Blerkom J (1997) Can the developmental competence of early human
26–33 embryos be predicted effectively in the clinical IVF laboratory? Human
*Ozawa T (1997a) Genetic and functional changes in mitochondria associated Reproduction 12 1610–1614
with aging Physiological Reviews 77 425–464 Van Blerkom J, Davis PW and Lee J (1995) ATP content of human oocytes
Ozawa T (1997b) Oxidative damage and fragmentation of mitochondrial and developmental potential and outcome after in vitro fertilization and
DNA in cellular apoptosis Bioscience Reports 17 237–250 embryo transfer Human Reproduction 10 415–424
Pikó L and Matsumoto L (1976) Number of mitochondria and some proper- *Wallace DC (1997) Mitochondrial DNA in aging and disease Scientific
ties of mitochondrial DNA in the mouse egg Developmental Biology 49 1–10 American 277 40–47
Pikó L and Taylor KD (1987) Amounts of mitochondrial DNA and abun- Willoughby CE, Mazur P, Peter AT and Critser JK (1996) Osmotic tolerance
dance of some mitochondrial gene transcripts in early mouse embryos limits and properties of murine spermatozoa Biology of Reproduction 55
Developmental Biology 123 364–374 715–727
Pinkert CA, Irwin MH, Johnson LW and Moffatt RJ (1997) Mitochondria Wilmut I, Schnieke AE, McWhir J, Kind AJ and Campbell KHS (1997) Viable
transfer into mouse ova by microinjection Transgenic Research 6 379–383 offspring derived from fetal and adult mammalian cells Nature 385 810–813

Downloaded from Bioscientifica.com at 06/18/2020 09:26:49PM


via free access

You might also like