ArticlePDF AvailableLiterature Review

Mitochondrial Dysfunction in Primary Ovarian Insufficiency

Authors:

Abstract and Figures

Primary ovarian insufficiency (POI) is defined by the loss or dysfunction of ovarian follicles associated with amenorrhea before the age of 40. Symptoms include hot flashes, sleep disturbances, and depression, as well as reduced fertility and increased long-term risk of cardiovascular disease. POI occurs in ∼1-2% of women, although the etiology of the majority of cases remains unexplained. Approximately 10-20% of POI cases are due to mutations in a single gene or a chromosomal abnormality, which has provided considerable molecular insight into the biological underpinnings of POI. Many of the genes for which mutations have been associated with POI, either isolated or syndromic cases, function within mitochondria, including MRPS22, POLG, TWNK, LARS2, HARS2, AARS2, CLPP, and LRPPRC. Collectively, these genes play roles in mitochondrial DNA replication, gene expression, and protein synthesis and degradation. Although mutations in these genes clearly implicate mitochondrial dysfunction in rare cases of POI, data is scant as to whether these genes in particular, and mitochondrial dysfunction in general, contribute to the majority of POI cases that lack a known etiology. Further studies will be needed to better elucidate the contribution of mitochondria to POI and whether there is a common molecular defect in mitochondrial function that distinguishes mitochondrial-related genes that when mutated cause POI versus those that do not. Nonetheless, the clear implication of mitochondrial dysfunction in POI suggests that manipulation of mitochondrial function represents an important therapeutic target for the treatment or prevention of POI.
Content may be subject to copyright.
MINI-REVIEW
Mitochondrial Dysfunction in Primary
Ovarian Insufficiency
Dov Tiosano,
1,2
Jason A. Mears,
3,4
and David A. Buchner
5,6,7
1
Division of Pediatric Endocrinology, Ruth Rappaport Childrens Hospital, Rambam Medical Center, Haifa
3109601, Israel;
2
Rappaport Family Faculty of Medicine, TechnionIsrael Institute of Technology, Haifa
3200003, Israel;
3
Center for Mitochondrial Diseases, Case Western Reserve University, Cleveland, Ohio
44106;
4
Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106;
5
Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio 44106;
6
Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; and
7
Research
Institute for Childrens Health, Case Western Reserve University, Cleveland, Ohio 44106
ORCiD numbers: 0000-0003-3920-4871 (D. A. Buchner).
Primary ovarian insufficiency (POI) is defined by the loss or dysfunction of ovarian follicles as-
sociated with amenorrhea before the age of 40. Symptoms include hot flashes, sleep distur-
bances, and depression, as well as reduced fertility and increased long-term risk of cardiovascular
disease. POI occurs in ;1% to 2% of women, although the etiology of most cases remains un-
explained. Approximately 10% to 20% of POI cases are due to mutations in a single gene or a
chromosomal abnormality, which has provided considerable molecular insight into the biological
underpinnings of POI. Many of the genes for which mutations have been associated with POI,
either isolated or syndromic cases, function within mitochondria, including MRPS22,POLG,
TWNK,LARS2,HARS2,AARS2,CLPP,andLRPPRC. Collectively, these genes play roles in mito-
chondrial DNA replication, gene expression, and protein synthesis and degradation. Although
mutations in these genes clearly implicate mitochondrial dysfunction in rare cases of POI, data are
scant as to whether these genes in particular, and mitochondrial dysfunction in general, con-
tribute to most POI cases that lack a known etiology. Further studies are needed to better
elucidate the contribution of mitochondria to POI and determine whether there is a common
molecular defect in mitochondrial function that distinguishes mitochondria-related genes that
when mutated cause POI vs those that do not. Nonetheless, the clear implication of mitochondrial
dysfunction in POI suggests that manipulation of mitochondrial function represents an impor-
tant therapeutic target for the treatment or prevention of POI. (Endocrinology 160: 23532366,
2019)
Primary ovarian insufficiency (POI), which is also
commonly referred to as premature ovarian failure, is
defined by the loss or dysfunction of ovarian follicles
associated with amenorrhea before the age of 40 (1). POI
is a major cause of female infertility, with a prevalence
between 1% and 2%. It can occur spontaneously or be
the result of medical interventions such as removal of
the ovaries, chemotherapy, or radiation treatment.
Symptoms of POI are similar to menopause and com-
monly include hot flashes, sleep disturbances, vaginal
dryness, and night sweats (2). Other less commonly re-
ported symptoms include decreased energy, dry eyes, hair
loss, urinary incontinence, cold intolerance, cognitive
changes, depression, and joint pain, among others (3).
ISSN Online 1945-7170
Copyright © 2019 Endocrine Society
Received 14 June 2019. Accepted 1 August 2019.
First Published Online 8 August 2019
Abbreviations: aaRS, aminoacyl-tRNA synthetase; AARS2, alanyl-tRNA synthetase 2,
mitochondrial; CLPP, caseinolytic mitochondrial matrix peptidase proteolytic subunit;
CLPX, caseinolytic mitochondrial matrix peptidase chaperone subunit; HARS2, histidyl-
tRNA synthetase 2, mitochondrial; LRPPRC, leucine-rich pentatricopeptide repeat containing;
MII, meiosis II; MRPS22, mitochondrial ribosomal pr otein S22; mtDNA, mitochondrial DNA;
OXPHOS, oxidative phosphorylation; POI, primary ov arian insufficiency; POLG, DNA polymerase
g, catalytic subunit; PRORP, protein-only RNase P catalytic subunit; ROS, reactive oxygen specie s;
TCA, tricarboxylic acid; TWNK, twinkle mtDNA helicase; WES, whole-exome sequencing.
doi: 10.1210/en.2019-00441 Endocrinology, October 2019, 160(10):23532366 https://academic.oup.com/endo 2353
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
Longer-term consequences associated with the prema-
ture decrease in estrogen levels include increased risk of
cardiovascular disease and decreased bone mineral density
(4, 5). Symptom severity can be correlated with ovarian
function, as POI encompasses a range of decreased ovarian
functions from individuals with partial function who retain
fertility to those who completely lack ovarian function and
are infertile. Treatment will vary depending on the symp-
toms, but it can include hormone replacement therapy,
fertility management, and psychosocial support, as well as
annual screenings of thyroid and adrenal function (2).
In cases of POI that are not induced by chemotherapy
or radiation, the etiology is determined for only ;20% of
all cases (6). Approximately 5% of all POI cases are of
autoimmune origin, and 15% of cases have a clear ge-
netic origin, either in the form of a chromosomal ab-
normality or a mutation in an individual gene (7, 8).
Among the chromosomal abnormalities that are frequent
causes of POI are a number of X chromosome defects,
including Turner syndrome, triple X syndrome, and
fragile X syndrome. A number of monogenic disorders
resulting in POI have also been identified, with variants
in .50 genes having been associated with POI (8). The
genes that have been implicated in POI fall within a
number of pathways that are critical for ovarian devel-
opment and function, including DNA repair, meiosis,
germ cell recruitment, and steroidogenesis. Additionally,
mutations in many genes involved in mitochondrial
function have been identified as causes of POI. Thus, this
review summarizes the role of mitochondria in oocytes as
well as discusses the genetic causes of POI related to
mitochondrial dysfunction.
Mitochondrial Composition and Function
Mitochondria are maternally inherited double-
membrane organelles that are best known for their
role as the powerhouses of a cell, based on their gener-
ation of ATP via the process of oxidative phosphoryla-
tion (OXPHOS). The mitochondrial genome is ;16.7 kb
and encodes 13 proteins that function within the
OXPHOS pathway, as well as 22 tRNAs and 2 rRNAs.
However, estimates of the complete human mitochon-
drial proteome suggest the presence of at least 1500
proteins, and thus the vast majority of proteins localized
to mitochondria are encoded by the nuclear genome (9).
The proteomic complexity of mitochondria enables them
to perform many critical cellular functions beyond en-
ergy production such as macromolecule biogenesis (i.e.,
protein and nucleic acids), lipid synthesis, regulation of
cell death, calcium handling/homeostasis, generation
of reactive oxygen species (ROS), and antioxidant pro-
tection (10).
OXPHOS is the process by which nutrients are oxi-
dized by a series of five multisubunit protein complexes
(complex I to complex V) within the mitochondrial inner
membrane, ultimately resulting in the conversion of ADP
to ATP. During the process of ATP generation, mito-
chondria also release ROS in the form of superox-
ide. Although typically thought of as a byproduct
of OXPHOS that can damage cells, ROS possess anti-
microbial properties, act as signaling molecules, and
regulate autophagy (11). Nonetheless, excessive or
mislocalized ROS can have deleterious effects on the cell
and can induce oxidative damage to mitochondrial DNA
(mtDNA), the latter of which may be particularly sen-
sitive to damage due to the relative absence of DNA
repair enzymes in mitochondria (12). ROS are eliminated
in the mitochondria by three superoxide dismutases that
convert superoxide to hydrogen peroxide, which can
then be further converted to water by peroxiredoxins and
glutathione peroxidases.
Mitochondria are not static organelles, rather their
numbers are tightly regulated by balancing the processes
of mitochondrial biogenesis and clearance (13). Mito-
chondrial clearance can be mediated by the process of
mitophagy, which describes the degradation and recy-
cling of mitochondria via autophagy. Triggers for
mitophagy include mild increases in ROS (14) and the
inability to maintain an electrochemical gradient that can
result from the accumulation of mtDNA damage (15).
Mitochondrial biogenesis is then used to repopulate the
cellular mitochondrial population with more functional
mitochondria. The process of mitochondrial biogenesis
involves both fusion and fission events, and it is regulated
by a cascade of transcription and translation factors,
both nuclear and mitochondrial encoded, which serve to
generate the materials necessary for organelle duplication
(16). Mitochondrial fusion describes the merging of the
outer and then inner membranes of two previously
separate mitochondria, whereas mitochondrial fission is
the process by which a single mitochondrion divides into
two or more independent mitochondria. Mitochondrial
fusion, fission, and mitophagy collectively serve not only
an important quality control function, but can link
changes in mitochondrial abundance to cellular energy
demands (17). For example, nutrient deprivation pre-
vents mitochondrial fission, which together with other
molecular and structural changes to the mitochondria
serves to increase ATP synthesis capacity, whereas
thermogenesis or AMP-activated protein kinase activa-
tion serves to increase fission (1820). Nutrient excess
inhibits fusion, resulting in an increase in fragmentation,
which can cause mitochondrial dysfunction and in-
crease ROS production (21). OXPHOS activity is posi-
tively correlated with fusion, although the causal nature
2354 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
of this relationship remains unclear (22). Mitophagy
can be triggered by energy stress via activation of
AMP-activated protein kinase, hypoxia, and glucose
deprivation (22). Collectively, the regulation of mito-
chondrial dynamics ensures that these organelles can help
the cell respond and adapt to a variety of cellular states to
match the bioenergetic and other mitochondrial func-
tions with the cellular energy needs.
Mitochondrial Function in Ovarian
Somatic Cells
Beyond the role of mitochondria in OXPHOS, ROS
homeostasis, apoptosis, and thermogenesis, mitochon-
dria have an important role in steroidogenesis. Ste-
roidogenesis occurs in the cumulus granulosa and theca
cells, which surround the oocyte, according to the two-
cell, two-gonadotropin model. The first step in ste-
roidogenesis, which is also the rate-limiting step, involves
the transport of cholesterol from the outer mitochondrial
membrane to the inner mitochondrial membrane by the
steroid acute regulatory protein (STAR) and other ac-
cessory proteins (23). Once inside the mitochondria,
cholesterol is metabolized to pregnenolone by cyto-
chrome P450 family 11 subfamily A member 1 (CYP11A1,
also known as P450SCC), which is then further me-
tabolized in the endoplasmic reticulum to form the final
steroid products, including estradiol and progesterone
(24, 25).
Mitochondrial Function in Oocytes
Oogenesis, the process by which a haploid oocyte is
formed within the ovary, originates from primordial
germ cells that arise in the extraembryonic mesoderm,
which then migrate to the genital ridge where they
proliferate by mitosis and transform to oogonia (26).
Following the arrest of mitosis, human germ cells initiate
meiosis in the fetal ovary at 11 to 12 weeks of gestation
and become primary oocytes (27). Oocytes enter meiosis
in the prophase stage, during which time homologous
chromosomes undergo synapsis and recombination.
Subsequent to the crossing over of genetic material
during recombination, oocytes progress to the diplotene
stage, which is defined by a protracted arrest. During
this resting state, oocytes become surrounded by pre-
granulosa cells to form primordial follicles, although
most oocytes undergo apoptosis during this process and
do not survive to form follicles (28). Oocyte survival
appears in part to depend on the transfer of organelles,
including Golgi and mitochondria, from nurse cells
connected by intercellular bridges that are associated
with the formation of a Balbiani body in the oocyte, a
structure that is densely packed with Golgi, endoplasmic
reticulum, and mitochondria (29, 30). The Balbiani body
is a transient formation and disappears in late-stage
oocytes. A subset of primordial follicles are further in-
duced via intra-oocyte and extra-oocyte factors involving
the phosphoinositide 3-kinase and mammalian target of
rapamycin pathways to transition to primary follicles
(31). The primary follicles then further develop to form
secondary follicles, which are surrounded by at least two
layers of granulosa cells and an outermost layer of theca
cells. The theca cells contain many mitochondria with
vesicular cristae (32). Further development preceding
ovulation includes the formation of a cavity called the
antrum and the selection of dominant follicles that will
eventually ovulate. Reinitiation of meiosis in the domi-
nant follicles can follow a surge of endogenous LH
during puberty, but these follicles can also remain in the
dormant state from the time of their formation during
fetal development until menopause, a period of ;50
years (33).
There is a tremendous increase in the number of mi-
tochondria per cell that occurs during oogenesis, from the
10 to 100 mitochondria found in primordial germ cells to
the .100,000 mitochondria found in a mature pre-
ovulatory oocyte (34). The small number of mitochon-
dria in primordial germ cells contributes to the
establishment of an inheritance bottleneck, in which
rapid shifts in the inheritance of mitochondrial variants
can occur between generations based on the small
number of mitochondria that form the basis of the rapid
expansion (35). Coupled with a dramatic reduction in
mtDNA in primordial germ cells, there exists a strong
selective pressure, including influence from the nuclear
genome, to eliminate deleterious mitochondrial variants
that are frequently present at low levels (3638).
Although the numbers of mitochondria rapidly in-
crease during oogenesis, they maintain a state of re-
latively low activity until the blastocyst stage.
Mitochondria found within oocytes have a morphology
that is more rounded than the typical rod-shaped mor-
phology found in somatic cells, and there are fewer
cristae found in the inner membrane (39). Oocyte mi-
tochondria also have a relatively lower rate of oxygen
consumption, together suggesting that mitochondria
within the oocyte remain in a state of relatively low
activity. It has been hypothesized that the large numbers
of mitochondria functioning at minimal levels are suf-
ficient to provide just the right amount of energy to the
oocyte while minimizing the frequency of mtDNA mu-
tations and the production of ROS (40). This activity is
nonetheless critical for oocyte maturation because gly-
colysis is limited during oogenesis, creating a heavier
reliance on ATP generated by OXPHOS. In addition to
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2355
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
the energy generated by oocyte mitochondria, energy
transfer from the surrounding granulosa and cumulus
cells, in the form of extracellular pyruvate and glutamine,
also contributes toward the energetic requirements of
oocyte maturation and early embryogenesis (41). The
relatively large numbers of quietmitochondria during
oogenesis further suggests that other functions of mito-
chondria beyond ATP production may be important at
this stage, including the production of the reduced form
of nicotinamide adenine dinucleotide phosphate and
tricarboxylic acid (TCA) cycle intermediates, which may
be used to decrease oxidative stress and provide sub-
strates for other biosynthetic pathways (42).
In addition to dramatic changes in the numbers of
mitochondria during oogenesis, the subcellular locali-
zation of these organelle is remodeled to suit the changing
energy demands of the cell during maturation. The most
significant shift in subcellular localization occurs
when .40% of mitochondria accumulate to surround
the forming meiosis I spindle, remaining with the spindle
during its movement from the oocyte center to the pe-
riphery (43). This association of mitochondria with the
developing spindle is dependent on their trafficking along
microtubules in a dynein-dependent process. Following
the first meiotic division, there is an asymmetric distri-
bution of mitochondria among the daughter cells fa-
voring the oocyte over the polar body, which is of
particular importance given the paucity of mitochondrial
biogenesis from this stage of oogenesis through im-
plantation (44). Mitochondria again associate with the
meiosis II (MII) spindle during its formation; however,
following the completion of the MII spindle formation
and arrest at the MII stage, the mitochondria no longer
completely encase the spindle, but do remain somewhat
enriched in the immediate vicinity (44).
Mutations in Mitochondrial Proteins in POI
Mitochondrial disease represents a group of clinically
heterogeneous disorders that can typically be grouped
together based on their common feature of having a
primary defect in OXPHOS (45). The prevalence of
mitochondrial disease in children is ;6 of 100,000 and in
adults is ;23 of 100,000 (46, 47). Mitochondrial dis-
eases can be caused by mutations in either mtDNA or
nuclear-encoded genes, with mutations in .350 genes
having been reported as a cause (48). The clinical pre-
sentations are varied in their onset, inheritance pattern,
and features, but they are often found in organs with high
energy demands, including the brain, skeletal muscle,
and heart. Frequent features include hypertrophic car-
diomyopathy, heart conduction defects, myopathy,
sensorineural deafness, cerebellar ataxia, epilepsy,
and peripheral neuropathy, among others (49).
Nonsyndromic presentations of mitochondrial disease
include mutations in the 12S rRNA that cause non-
syndromic deafness and certain individuals with Leber
optic atrophy, which is caused by mutations in multiple
genes, that present with visual loss as the only clinical
feature (50, 51). This brings up one of the more per-
plexing features of mitochondrial disorders, which is that
despite the fact that mitochondria are critical for the
function of every cell in the body, their clinical pre-
sentation can often be organ or cell specific. Addition-
ally, not all patients with mitochondrial disease have
OXPHOS defects, as evidenced by patients with TCA
cycle defects due to mutations in aconitase 2 (ACO2)
who present with infantile cerebellar-retinal degen-
eration. ACO2, which catalyzes the conversion of
citrate to isocitrate within the TCA cycle, is not consis-
tently associated with defects in OXPHOS in patient-
derived tissue samples, but rather is characterized by
abnormal TCA metabolite levels and mtDNA depletion
(5254).
In addition to the frequently observed defects in the
heart, brain, and muscle, the presentation of mitochon-
drial dysfunction often presents with a range of endocrine
features, including diabetes mellitus, GH deficiency,
hypogonadism, thyroid disease, and ovarian dysfunc-
tion, among others (55, 56). Ovarian dysfunction can be
seen as part of a complex syndromic presentation, but it
also includes disorders where it is the only clinical
finding. Mitochondrial mutations that lead to ovarian
dysfunction have been linked to mtDNA, mitochondrial
RNA, and mitochondrial protein synthesis, with func-
tions in multiple distinct biological pathways (57). Much
of what is known about the pathophysiology underlying
ovarian dysfunction has been learned from the identifi-
cation of disease genes identified in rare monogenic
disorders (58). Below is a discussion of eight genes that
when mutated give rise to either isolated POI or POI as
part of a syndrome (Table 1), as well as three additional
genes that have recently been described in a single patient
or family, suggesting a potential link between those genes
and POI as well.
Rare Monogenic Mitochondrial Disorders
Associated With POI
DNA polymerase g, catalytic subunit
DNA polymerase g, catalytic subunit (POLG) is the
catalytic subunit of the mitochondrial DNA polymerase
that is responsible for replication of the mitochondrial
genome (59). Mutations in POLG cause depletion of the
mtDNA and/or the accumulation of deletions in the
mtDNA and are the most commonly inherited form
2356 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
of monogenic mitochondrial disease (60). Mutations
in POLG cause a spectrum of disorders, including
myocerebrohepatopathy spectrum, AlpersHuttenlocher
syndrome, and progressive external ophthalmoplegia,
among others, but they lack a clear cut genotype
phenotype correlation between the .300 described
pathogenic mutations and the clinical presentation (61).
Among the disparate clinical features associated with
POLG mutations are epilepsy, ataxia, parkinsonism,
hearing loss, cataracts, as well as POI (60, 62). Although
the clinical presentation of POLG mutations can vary,
POI has typically been described as part of a syndrome
including the previously mentioned neurologic features,
and not in isolation (55). However, a recent case report
described identifying the first case of nonsyndromic
ovarian dysfunction associated with a mutation in
POLG (p.R964C), although caution must be taken in
interpreting this study until additional cases are identi-
fied (63). Finally, although candidate genebased stud-
ies have previously failed to find evidence of POLG
mutations as a common cause of POI (64, 65), a recent
metagenome-wide association study analysis identified
an association between a single-nucleotide poly-
morphism located near POLG (rs1054875) and the age
at natural menopause, suggesting that variation in
POLG may contribute to the more common polygenic
forms of POI (66).
Twinkle mtDNA helicase
Twinkle mtDNA helicase (TWNK; also known as
C10ORF2) encodes a mitochondrial helicase that to-
gether with the above-mentioned POLG, the DNA
polymerase gaccessory subunit POLG2, and the mi-
tochondrial single-stranded DNA-binding protein
SSBP1 form the core factors required for mtDNA rep-
lication (67, 68). Similar to POLG, mutations in TWNK
can cause a spectrum of genetic disorders, including
progressive external ophthalmoplegia, mtDNA depletion
syndrome 7 (hepatocerebral type), and Perrault syn-
drome, with clinical presentations that can contain
POI in association with other neurologic findings. For
example, 2 unrelated families each had 2 individuals
present with ataxia, neuropathy, hyporeflexia, pro-
gressive hearing loss, and ovarian dysgenesis, with af-
fected individuals in both families found to be compound
heterozygous for missense mutations in TWNK (69).
Given the functional overlap between POLG and TWNK
in mtDNA replication, it is perhaps not surprising that
mutations in both genes can cause a similar constella-
tion of clinical features, including POI, and together
clearly implicates the importance of mtDNA replication in
ovarian development. However, thus far mutations in the
other core components of mtDNA replication, POLG2
and SSBP1, have not been associated with ovarian dys-
function, although mutations have been observed in
POLG2 that, similar to POLG, cause progressive external
ophthalmoplegia (70).
Leucyl-tRNA synthetase 2, mitochondrial and
histidyl-tRNA synthetase 2, mitochondrial
Histidyl-tRNA synthetase 2, mitochondrial (HARS2)
and leucyl-tRNA synthetase 2, mitochondrial (LARS2)
both encode aminoacyl-tRNA synthetases (aaRSs) that
are used for the translation of mitochondrial-encoded
genes. Mitochondrial aaRSs can be categorized into two
major classification groups, class I and class II, based on
their structural motifs, with LARS2 encoding a class I
aaRS based on the presence of a Rossmann fold that
serves as an ATP biding region and HARS2 encoding a
class II aaRS based on three structural motifs referred to
as 1, 2, and 3 (71). Mutations in both HARS2 and
LARS2 are associated with Perrault syndrome, which
Table 1. Mitochondria-Related Genes in Which Mutations Can Cause POI
Gene Inheritance Clinical Presentation Gene Function Molecular Function in Mitochondria
POLG AR, AD Progressive external ophthalmoplegia DNA polymerase mtDNA replication and maintenance
Mitochondrial DNA depletion syndrome
Mitochondrial recessive ataxia syndrome
TWNK AR Perrault syndrome mtDNA helicase mtDNA replication and proofreading
Mitochondrial DNA depletion syndrome
Progressive external ophthalmoplegia
LARS2 AR Perrault syndrome Leucine tRNA mRNA translation
HARS2 AR Perrault syndrome Histidine tRNA mRNA translation
AARS2 AR Ovarioleukodystrophy Alanine tRNA mRNA translation
Combined OXPHOS deficiency
CLPP AR Perrault syndrome Protease Protein degradation
LRPPRC AR Leigh syndrome RNA binding protein Gene expression
MRPS22 AR Isolated POI Ribosomal subunit mRNA translation
Combined OXPHOS deficiency
Abbreviations: AD, autosomal dominant; AR, autosomal recessive.
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2357
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
describes a rare recessively inherited condition composed
of sensorineural hearing loss in both males and females
and ovarian dysfunction, including ovarian dysgenesis
and primary amenorrhea, and can include neurologic
features including ataxia, learning disability, and neu-
ropathy (7274). Testicular dysfunction has not been
observed in males with the same mutations that cause
ovarian dysfunction in females (72). However, the
number of deaf males with Perrault syndrome mutations
whose sperm function has been clinically evaluated is
small, and a mouse model of Perrault syndrome due to
complete Clpp deficiency (discussed below) is associated
with infertility in both males and females (75, 76).
Among the 19 mitochondrial aaRSs, it remains unclear
why mutations in these two particular genes result in
hearing loss and ovarian dysfunction, whereas mutations
in other mitochondrial aaRSs result in multiorgan sys-
temic disorders (77). However, although most disease-
causing mutations in aaRSs occur at residues that are not
evolutionarily conserved, mutations in both HARS2 and
LARS2 have been described at conserved positions,
suggesting that the mutations in these genes may similarly
affect the canonical functions of these proteins (78).
Alanyl-tRNA synthetase 2, mitochondrial
Alanyl-tRNA synthetase 2, mitochondrial (AARS2),
similar to HARS2 and LARS, is a mitochondrial aaRS;
however, mutations in AARS2 do not cause Perrault
syndrome, but instead are associated with recessively
inherited ovarioleukodystrophy syndrome in females
(7983). Mutations in AARS2 have also been linked to
infantile mitochondrial cardiomyopathy and primary
pulmonary hypoplasia resulting in early childhood le-
thality (84, 85). The ovarioleukodystrophy syndrome
consists of neurologic features stemming from neurologic
deterioration, including ataxia, spasticity, tremor, and
cognitive decline, with ages of onset ranging from
childhood to adulthood, in the absence of cardiomyop-
athy. Ovarian failure occurs in the 20s or 30s and has
been reported with both primary amenorrhea or sec-
ondary amenorrhea. AARS2 mutations that lead to in-
fantile cardiomyopathy reduce protein stability with
minimal effects on aminoacylation, whereas the molec-
ular basis underlying the ovarioleukodystrophy syn-
drome remains unknown (86). However, it has been
hypothesized that the ovarioleukodystrophy is associated
with a reduction in aminoacylation efficiency, although
this remains to be experimentally validated (87).
Caseinolytic mitochondrial matrix peptidase
proteolytic subunit
Caseinolytic mitochondrial matrix peptidase pro-
teolytic subunit (CLPP) is an ATP-dependent protease
that is localized to the mitochondrial matrix where, to-
gether with caseinolytic mitochondrial matrix peptidase
chaperone subunit (CLPX), it is part of the quality
control system that degrades oxidized and denatured
proteins (88). A number of CLPP substrates for degra-
dation have been identified and include proteins involved
in electron transport, metabolic processes, the TCA
cycle, and mitochondrial mRNA, among others (8991).
Consistent with the identified substrates, CLPP de-
ficiency in experimental model systems leads to reduced
mitochondrial respiration, increased ROS, and impaired
mitochondrial protein synthesis associated with defects
in assembly of the mitochondrial ribosome (89, 92).
Mutations in CLPP cause recessively inherited Perrault
syndrome type 3 associated with ovarian dysfunction,
hearing loss, and neurologic defects such as lower limb
spasticity, epilepsy, microcephaly, and learning difficul-
ties (9396). Functional characterization of the identified
mutations in CLPP resulted in a variety of functional
defects, including decreased peptidase activity, inhibition
of CLPX binding, prevention of oligomerization, and
enhanced peptidase activity, suggesting that despite the
clustering of most CLPP mutations near the CLPX-
docking site or the active site of the peptidase, there
may not be a common defect in the molecular function of
CLPP (97). A mouse knockout of Clpp has also been
generated, which similarly demonstrated hearing loss
and infertility due to the failure of ovarian follicular
differentiation in females, as well as growth retardation,
decreased activity, impaired survival, and disruption of
spermatogenesis in males, providing a model to better
study the cellular and molecular defects associated with
CLPP deficiency (76).
Leucine-rich pentatricopeptide repeat containing
Leucine-rich pentatricopeptide repeat containing
(LRPPRC) is an RNA-binding protein that plays a critical
role in regulating mitochondrial gene expression.
LRPPRC together with the interacting protein SRA stem-
loop interacting RNA binding protein (SLIRP) directly
bind to and coat nearly all mitochondrial mRNAs,
thereby regulating their structure to improve their
translational fidelity and increase their stability (98, 99).
Although SLIRP and LRPPRC both depend on the other
for stability, they also have unique functions within their
complex, with only LRPPRC being required for poly-
adenylation of mitochondrial mRNAs, whereas SLIRP
functions to target mRNAs to the mitochondrial ribosome
(100). Nuclear functions have also been ascribed to
LRPPRC, including as a cofactor for eukaryotic translation
initiation factor 4E (eIF4e) that regulates nuclear export
and translation, and a component of the PGC1atran-
scription regulation complex that controls the expression
2358 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
of key metabolic genes such as phosphoenolpyruvate
carboxykinase (PEPCK) and glucose-6-phosphatase
catalytic subunit (G6PC) in addition to genes involved
in mitochondrial biogenesis (101, 102).
Mutations in LRPPRC cause a rare type of Leigh
syndrome referred to as French-Canadian, because most
patients originate from the Saguenay-Lac-Saint-Jean
region of Qu´ebec. A p.A354V founder mutation is
present in this region at a relatively high allele fre-
quency, resulting in an incidence of ;1 of 2000 births
(103106). Leigh syndrome, French-Canadian variety,
has an autosomal recessive inheritance pattern with
clinical features that include developmental delay,
failure to thrive, characteristic facial features, and hy-
potonia. Patients are at high risk of death due to neu-
rologic or acidotic crises that can be instigated by
triggers, including infection, stress, diet, illness, or ex-
ercise. Although life expectancy is typically ,5years,
patients can survive to adulthood (107). Female patients
who survive until at least adolescence, in addition to the
neurologic features, present with POI, including ele-
vated FSH, small ovaries that lack follicles, and primary
amenorrhea (106). Although LRPPRC clearly has a
significant effect on expression of mtDNA-encoded
genes, caution must be taken when interpreting this
as conclusive support for the role of mitochondrial
dysfunction in POI, given that LRPPRC also regulates
nuclear gene expression.
Mitochondrial ribosomal protein S22
Mitochondrial ribosomal protein S22 (MRPS22) is a
component of the 28S small subunit of the mitochon-
drial ribosome that enables the translation of mtDNA-
encoded polypeptides (108). MRPS22 is also required
for maintaining the stability of the mitochondrial ri-
bosomal complex (109). Mutations in MRPS22 have
been identified to cause autosomal recessive inheritance
of POI (110, 111). Unlike the syndromes mentioned
above, mutations in MRPS22 caused isolated POI in the
absence of other neurologic or other syndromic fea-
tures. Surprisingly, MRPS22 deficiency did not cause
defects in OXPHOS, suggesting that the etiology of POI
was related to the nonbioenergetic functions of mito-
chondria. In Drosophila,germcellspecific deficiency
of the MRPS22 ortholog resulted in infertility,
suggesting a cell-autonomous role for MRPS22 in germ
cell development (110). Mutations in MRPS22, which
are presumably more deleterious to protein function,
have been identified that impair OXPHOS activity and
lead to systemic disease, including cardiomyopathy,
hypotonia, brain abnormalities, and in certain instances
lethality (112115). Thus, variants in MRPS22 are as-
sociated with a phenotypic spectrum of disorders ranging
from isolated POI to infantile mitochondrial lethal-
ity (110).
Mitochondria-Related Candidate Genes
for POI Identified in Single Families
or Probands
Mitochondrial ribosomal protein S7, protein only
RNase P catalytic subunit, and required for meiotic
nuclear division 1 homolog
Caution must be exercised when interpreting muta-
tions that have thus far been identified in only a single
family, as the sheer number of potentially deleterious
variants present in all individuals makes it difficult to
identify definitively the true causal variants. Nonetheless,
the initial identification and reporting of these mutations
may facilitate other investigators who encounter muta-
tions in those genes and is thus worthwhile. As such,
mutations have been reported in protein-only RNase P
catalytic subunit (PRORP) as a novel cause of Perrault
syndrome (116). A single consanguineous family was
identified with three individuals presenting with senso-
rineural hearing loss and POI that were each homozy-
gous for a missense variant in PRORP (p.A485V).
Functional studies in patient-derived fibroblasts sup-
ported the presence of mitochondrial dysfunction as
evidenced by impaired processing of mitochondrial RNA
transcripts and an accompanying decrease in protein
levels of multiple OXPHOS components (116). Similarly,
another consanguineous family was identified with two
female siblings who presented with congenital sensori-
neural deafness and lactic acidemia. One sibling had a
significantly more severe presentation, including failure
to thrive, hepatomegaly, and hypoglycemia, and ulti-
mately died at 14 years of age. The sibling has hypo-
glycemia in early childhood that resolved, mild learning
difficulties, and was found at age 16 to have POI (117).
Both siblings were homozygous for a mutation in the
mitochondrial ribosomal protein S7 (MRPS7) at a highly
conserved amino acid residue (p.M184V), and functional
studies in patient-derived fibroblasts identified OXPHOS
defects consistent with a mitochondrial disorder (117).
Another study identified a homozygous missense muta-
tion in required for meiotic nuclear division 1 homolog
(RMND1; p.N238S) that had previously been reported
as a disease causing mutation associated with neurologic,
muscle, and kidney defects (118, 119). However, the
proband in this study carrying the RMND1 variant had
the characteristic features of Perrault syndrome, in-
cluding POI that was observed at 10 years of age, as well
as kidney and growth defects (118). Mutations in
RMND1 have previously been associated with defects in
mitochondrial mRNA translation (120, 121). Although
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2359
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
these studies each describe a strong candidate gene for
POI, with the studies for MRPS7 and PRORP in par-
ticular elegantly combining both genetic and functional
data, caution must be taken until additional patients are
identified to more concretely establish the genotype
phenotype correlations.
Mitochondrial Dysfunction Associated
With Common POI
Rare monogenic forms of POI have been instrumental
into opening a window into the key genes and pathways
that are important for ovarian function (Fig. 1) (122).
However, with ;1% of women affected by POI, it is
not a rare disorder, and even cumulatively, the rare
monogenic causes of POI fail to account for most cases.
Nonetheless, that so many genes involved in mitochon-
drial function, and mitochondrial mRNA translation in
particular, have been implicated as causes of rare syn-
dromic or isolated POI suggests that this organelle may
be involved in the more common and genetically and
environmentally complex presentations of POI that ac-
count for most cases. Toward this end, pilot studies of
POI patients with no known etiology have identified an
increased frequency of mutations in the mitochondrially
encoded ATP synthase 6 gene (MT-ATP6) and the
mitochondrially encoded cytochrome coxidase I gene
(MT-CO1); however, in both cases the number of patients
screened was small (n 524 cases and n 563 cases, re-
spectively) (123, 124). The need for cautionin interpreting
small studies for a common disease is evident, as the
conclusions linking MT-ATP6 variants to POI have been
called into question, based on potential ancestry differ-
ences between the case and control populations analyzed
in that study (125). In addition to genetic mutations in
mitochondria, elevated levels of ROS and decreased levels
of OXPHOS activity and ATP production in oocytes have
also been correlated with POI, although again these
studies are limited in size (123, 126128). Interestingly,
treatment of mice with coenzyme Q10, a product of the
TCA cycle that controls multiple aspects of mitochondrial
biology, including transcription and succinate de-
hydrogenase activity, prevented the onset of POI in
both genetic- and aging-based mouse models of POI
(129). Although these preliminary studies are in-
triguing, future studies with increased power and
further studies in model organisms will be needed to
determine whether mitochondrial dysfunction is a
common contributing factor to the onset of POI.
Summary and Future Directions
It is clear from studies of rare monogenic disorders that
functional mitochondria are necessary to prevent ovarian
dysfunction and infertility. However,
larger and better powered studies are
necessary to access the impact and
causal role of dysfunctional mito-
chondria in most cases of POI.
Nonetheless, we have learned a tre-
mendous amount about the genetics
and pathophysiology of POI by
studying the rare disorders in cases
where a disease-causing mutation has
been identified (Fig. 1). These studies,
which have identified mutations in
genes including MRPS22,HARS2,
LARS2,andAARS2, suggest that one
putative therapeutic target for treating
or preventing POI will be to improve
mitochondrial mRNA translation.
Proof-of-principle experiments have
shown that supplementation with L-
cysteine is able to improve OXPHOS
activity in cell lines from a patient
with a genetic defect that impairs mi-
tochondrial mRNA translation (130).
In other cellular studies, overexpression
of mitochondrial tRNAs were able
to correct function defects in mRNA
Figure 1. Molecular functions of mitochondria-related proteins implicated in POI. A cartoon
depicts the various molecular functions in mitochondria of the proteins in which mutations
have been identified that can cause POI.
2360 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
translation and cellular respiration (131134). Treat-
ment with acetyl-L-carnitine has also long been known
to have beneficial effects on mitochondrial function in
vivo, including improved efficiency of mitochondrial
mRNA translation, and has been proposed as a treat-
ment of a variety of mitochondrial disorders (135).
Treatment with acetyl-L-carnitine improves in vitro
maturation of oocytes and reduces the number of ab-
normal mitochondria, although it is unclear what mo-
lecular mechanism underlies these improvements and
whether these finding will translate to in vivo treatment
or prevention of POI (136138). These studies have
shown that it is possible to manipulate the mitochon-
drial mRNA translational machinery, although it re-
mains to be seen whether this can be accomplished on a
therapeutic basis and what, if any, clinical benefits
would be seen.
The discoveries that mutations in the mitochondria-
related genes described above and in Table 1 can cause
POI were largely made possible by advances in DNA
sequencing technologies that greatly reduced the cost of
whole-exome sequencing (WES) (139). The initial studies
linking mutations in 8 of the 11 genes to POI used WES in
the process of identifying the causal variant. However,
given the rarity of mutations in these genes as a cause of
POI, the current guidelines for diagnosing and treating
nonsyndromic POI do not recommend testing for mu-
tations in these genes or other monogenic causes of POI
(140). Nonetheless, this may change as WES analyses
continue to become more integrated into routine clinical
practice. There is mounting evidence from numerous
clinical settings demonstrating benefits in terms of both
cost reductions and improved clinical care by in-
corporating WES early into the diagnostic process
(141146). However, to make this a clinical reality for
POI, more accurate predictions of whether variants are
likely to be pathogenic will be required. Fortunately,
progress can be made toward this end with limited
sample sizes. The studies utilizing WES discussed above
were each based on analyses of individuals with POI from
just one to three families, illustrating the progress that
can result from even small-scale studies.
Despite all that we have learned, many pressing
questions remain unanswered. (i) Why do mutations in
the genes described above cause POI, whereas mutations
in many more related mitochondrial genes do not? (ii)
Why do mutations in genes such as MRPS22 cause a
spectrum of disorders, ranging from isolated POI to
multiorgan systemic disease, and can we predict the
genotypephenotype correlations? (iii) Why does mito-
chondrial dysfunction preferentially affect the ovary? (iv)
Is mitochondria-associated POI due to cell-autonomous
defects in the oocyte, or rather defects in ovarian somatic
cells or other systemic changes? (v) Is ovarian dysfunction
in rare monogenic cases of POI strictly due to impaired
cellular respiration or are there nonbioenergetic defects
that underlie the disorder? Answers to these questions
will provide insight into the underlying pathophysiology
of POI and may identify new therapeutic targets to im-
prove the diagnosis and treatment of POI.
Acknowledgments
Financial Support: This work was supported by National
Institute of Diabetes and Digestive and Kidney Diseases Grant
DK112846 (to D.A.B.).
Additional Information
Correspondence: David A. Buchner, PhD, Case Western
Reserve University School of Medicine, 10900 Euclid Avenue,
Cleveland, Ohio 44106. E-mail: david.buchner@case.edu;or
Dov Tiosano, MD, Division of Pediatric Endocrinology, Ruth
Rappaport Childrens Hospital, Rambam Medical Center,
HaAliya HaShniya Street 8, Haifa 3109601, Israel. E-mail:
d_tiosano@rambam.health.gov.il.
Disclosure Summary: The authors have nothing to
disclose.
Data Availability: Data sharing is not applicable to this
article as no datasets were generated or analyzed during the
current study.
References and Notes
1. Santoro NF, Cooper AR, eds. Primary Ovarian Insufficiency.
Cham, Switzerland: Springer International Publishing; 2016.
https://doi.org/10.1007/978-3-319-22491-6
2. Cox L, Liu JH. Primary ovarian insufficiency: an update. Int J
Womens Health. 2014;6:235243.
3. Allshouse AA, Semple AL, Santoro NF. Evidence for prolonged
and unique amenorrhea-related symptoms in women with pre-
mature ovarian failure/primary ovarian insufficiency. Menopause.
2015;22(2):166174.
4. Christ JP, Gunning MN, Palla G, Eijkemans MJC, Lambalk CB,
Laven JS, Fauser BC. Estrogen deprivation and cardiovascular
disease risk in primary ovarian insufficiency. Fertil Steril. 2018;
109(4):594600.e1.
5. Podfigurna-Stopa A, Czyzyk A, Grymowicz M, Smolarczyk R,
Katulski K, Czajkowski K, Meczekalski B. Premature ovarian
insufficiency: the context of long-term effects. J Endocrinol Invest.
2016;39(9):983990.
6. Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary
ovarian insufficiency. Clin Genet. 2017;91(2):183198.
7. Silva CA, Yamakami LY, Aikawa NE, Araujo DB, Carvalho JF,
Bonf´a E. Autoimmune primary ovarian insufficiency. Autoimmun
Rev. 2014;13(45):427430.
8. Tucker EJ, Jaillard S, Sinclair AH. Genetics and genomics of pri-
mary ovarian insufficiency. In: Leung PC, Qiao J, eds. Human
Reproductive andPrenatal Genetics. San Diego, CA: Elsevier; 2019:
427445. https://doi.org/10.1016/B978-0-12-813570-9.00019-X
9. Pfanner N, Warscheid B, Wiedemann N. Mitochondrial proteins:
from biogenesis to functional networks. Nat Rev Mol Cell Biol.
2019;20(5):267284.
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2361
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
10. Picard M, Wallace DC, Burelle Y. The rise of mitochondria in
medicine. Mitochondrion. 2016;30:105116.
11. Dan Dunn J, Alvarez LA, Zhang X, Soldati T. Reactive oxygen
species and mitochondria: a nexus of cellular homeostasis. Redox
Biol. 2015;6:472485.
12. Larsson NG. Somatic mitochondrial DNA mutations in mam-
malian aging. Annu Rev Biochem. 2010;79(1):683706.
13. Ploumi C, Daskalaki I, Tavernarakis N. Mitochondrial biogenesis
and clearance: a balancing act. FEBS J. 2017;284(2):183195.
14. Frank M, Duvezin-Caubet S, Koob S, Occhipinti A, Jagasia R,
Petcherski A, Ruonala MO, Priault M, Salin B, Reichert AS.
Mitophagy is triggered by mild oxidative stress in a mitochondrial
fission dependent manner. Biochim Biophys Acta. 2012;
1823(12):22972310.
15. Kulikov AV, Luchkina EA, Gogvadze V, Zhivotovsky B.
Mitophagy: link to cancer development and therapy. Biochem
Biophys Res Commun. 2017;482(3):432439.
16. Tandler B, Hoppel CL, Mears JA. Morphological pathways of
mitochondrial division. Antioxidants. 2018;7(2):E30.
17. Liesa M, Shirihai OS. Mitochondrial dynamics in the regulation of
nutrient utilization and energy expenditure. Cell Metab. 2013;
17(4):491506.
18. Rambold AS, Kostelecky B, Elia N, Lippincott-Schwartz J. Tu-
bular network formation protects mitochondria from autopha-
gosomal degradation during nutrient starvation. Proc Natl Acad
Sci USA. 2011;108(25):1019010195.
19. Wikstrom JD, Mahdaviani K, Liesa M, Sereda SB, Si Y, Las G,
Twig G, Petrovic N, Zingaretti C, Graham A, Cinti S, Corkey BE,
Cannon B, Nedergaard J, Shirihai OS. Hormone-induced mito-
chondrial fission is utilized by brown adipocytes as an amplifi-
cation pathway for energy expenditure. EMBO J. 2014;33(5):
418436.
20. Toyama EQ, Herzig S, Courchet J, Lewis TL Jr, Los´on OC,
Hellberg K, Young NP, Chen H, Polleux F, Chan DC, Shaw RJ.
AMP-activated protein kinase mediates mitochondrial fission in
response to energy stress. Science. 2016;351(6270):275281.
21. Molina AJ, Wikstrom JD, Stiles L, Las G, Mohamed H, Elorza A,
Walzer G, Twig G, Katz S, Corkey BE, Shirihai OS. Mitochondrial
networking protects b-cells from nutrient-induced apoptosis.
Diabetes. 2009;58(10):23032315.
22. Mishra P, Chan DC. Metabolic regulation of mitochondrial dy-
namics. J Cell Biol. 2016;212(4):379387.
23. Stocco DM. StAR protein and the regulation of steroid hormone
biosynthesis. Annu Rev Physiol. 2001;63(1):193213.
24. Chien Y, Rosal K, Chung BC. Function of CYP11A1 in the mi-
tochondria. Mol Cell Endocrinol. 2017;441:5561.
25. Takae S, Suzuki N. Ovarian endocrinology. In: Chian RC,
Nargund G, Huang JYJ, eds. Development of In Vitro Maturation
for Human Oocytes. Cham, Switzerland: Springer International
Publishing; 2017:335.
26. S´anchez F, Smitz J. Molecular control of oogenesis. Biochimica
et Biophysica Acta (BBA) - Molecular Basis of Disease. 2012;
1822(12):18961912.
27. Gondos B, Westergaard L, Byskov AG. Initiation of oogenesis in
the human fetal ovary: ultrastructural and squash preparation
study. Am J Obstet Gynecol. 1986;155(1):189195.
28. Pepling ME, Spradling AC. Mouse ovarian germ cell cysts un-
dergo programmed breakdown to form primordial follicles. Dev
Biol. 2001;234(2):339351.
29. Pepling ME, Wilhelm JE, OHara AL, Gephardt GW, Spradling
AC. Mouse oocytes within germ cell cysts and primordial follicles
contain a Balbiani body. Proc Natl Acad Sci USA. 2007;104(1):
187192.
30. Lei L, Spradling AC. Mouse oocytes differentiate through or-
ganelle enrichment from sister cyst germ cells. Science. 2016;
352(6281):9599.
31. Rimon-Dahari N, Yerushalmi-Heinemann L, Alyagor L, Dekel N.
Ovarian folliculogenesis. In: Piprek RP, ed. Molecular Mechanisms
of Cell Differentiation in Gonad Development. Vol. 58. Cham,
Switzerland: Springer International Publishing; 2016:167190.
32. Magoffin DA. Ovarian theca cell. Int J Biochem Cell Biol. 2005;
37(7):13441349.
33. Reddy P, Zheng W, Liu K. Mechanisms maintaining the dor-
mancy and survival of mammalian primordial follicles. Trends
Endocrinol Metab. 2010;21(2):96103.
34. Poulton J, Marchington DR. Segregation of mitochondrial DNA
(mtDNA) in human oocytes and in animal models of mtDNA
disease: clinical implications. Reproduction. 2002;123(6):
751755.
35. Cao L, Shitara H, Horii T, Nagao Y, Imai H, Abe K, Hara T,
Hayashi J, Yonekawa H. The mitochondrial bottleneck occurs
without reduction of mtDNA content in female mouse germ cells.
Nat Genet. 2007;39(3):386390.
36. Chen D, Clark AT. Mitochondrial DNA selection in human germ
cells. Nat Cell Biol. 2018;20(2):118120.
37. Floros VI, Pyle A, Dietmann S, Wei W, Tang WCW, Irie N, Payne
B, Capalbo A, Noli L, Coxhead J, Hudson G, Crosier M, Strahl H,
Khalaf Y, Saitou M, Ilic D, Surani MA, Chinnery PF. Segregation
of mitochondrial DNA heteroplasmy through a developmental
genetic bottleneck in human embryos [published correction ap-
pears in Nat Cell Biol. 2018;20(8):991]. Nat Cell Biol. 2018;
20(2):144151.
38. Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL,
Bennett DL, Gale DP, Bitner-Glindzicz MA, Black GC, Brennan P,
Elliott P, Flinter FA, Floto RA, Houlden H, Irving M, Koziell A,
Maher ER, Markus HS, Morrell NW, Newman WG, Roberts I,
Sayer JA, Smith KG, Taylor JC, Watkins H, Webster AR, Wilkie
AO, Williamson C, Ashford S, Penkett CJ, Stirrups KE, Rendon A,
Ouwehand WH, Bradley JR, Raymond FL, Caulfield M, Turro E,
Chinnery PF; NIHR BioResourceRare Diseases; 100,000 Ge-
nomes ProjectRare Diseases Pilot. Germline selection shapes
human mitochondrial DNA diversity. Science. 2019;364(6442):
eaau6520.
39. de Paula WB, Lucas CH, Agip AN, Vizcay-Barrena G, Allen JF.
Energy, ageing, fidelity and sex: oocyte mitochondrial DNA as a
protected genetic template. Philos Trans R Soc Lond B Biol Sci.
2013;368(1622):20120263.
40. Leese HJ, Guerif F, Allgar V, Brison DR, Lundin K, Sturmey RG.
Biological optimization, the Goldilocks principle, and how much
is lagom in the preimplantation embryo. Mol Reprod Dev. 2016;
83(9):748754.
41. Collado-Fernandez E, Picton HM, Dumollard R. Metabolism
throughout follicle and oocyte development in mammals. Int J
Dev Biol. 2012;56(1012):799808.
42. Dumollard R, Carroll J, Duchen MR, Campbell K, Swann K.
Mitochondrial function and redox state in mammalian embryos.
Semin Cell Dev Biol. 2009;20(3):346353.
43. Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC,
Brambillasca F, Turchi D, Novara PV, Fadini R. Oocyte matu-
ration: gamete-somatic cells interactions, meiotic resumption,
cytoskeletal dynamics and cytoplasmic reorganization. Hum
Reprod Update. 2015;21(4):427454.
44. Dalton CM, Carroll J. Biased inheritance of mitochondria during
asymmetric cell division in the mouse oocyte. J Cell Sci. 2013;
126(Pt 13):29552964.
45. Lightowlers RN, Taylor RW, Turnbull DM. Mutations causing
mitochondrial disease: What is new and what challenges remain?
Science. 2015;349(6255):14941499.
46. Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, Alston
CL, Feeney C, Horvath R, Yu-Wai-Man P, Chinnery PF, Taylor
RW, Turnbull DM, McFarland R. Prevalence of nuclear and
mitochondrial DNA mutations related to adult mitochondrial
disease. Ann Neurol. 2015;77(5):753759.
47. Skladal D, Halliday J, Thorburn DR. Minimum birth prevalence
of mitochondrial respiratory chain disorders in children. Brain.
2003;126(Pt 8):19051912.
2362 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
48. Rahman J, Rahman S. Mitochondrial medicine in the omics era.
Lancet. 2018;391(10139):25602574.
49. Alston CL, Rocha MC, Lax NZ, Turnbull DM, Taylor RW. The
genetics and pathology of mitochondrial disease. J Pathol. 2017;
241(2):236250.
50. Prezant TR, Agapian JV, Bohlman MC, Bu X, ¨
Oztas S, Qiu WQ,
Arnos KS, Cortopassi GA, Jaber L, Rotter JI, Shohat M, Fischel-
Ghodsian N. Mitochondrial ribosomal RNA mutation associated
with both antibiotic-induced and non-syndromic deafness. Nat
Genet. 1993;4(3):289294.
51. Wallace DC, Lott MT. Leber hereditary optic neuropathy: ex-
emplar of an mtDNA disease. In: Singh H, Sheu SS, eds. Phar-
macology of Mitochondria. Vol. 240. Cham, Switzerland:
Springer International Publishing; 2017:339376.
52. Abela L, Spiegel R, Crowther LM, Klein A, Steindl K, Papuc SM,
Joset P, Zehavi Y, Rauch A, Plecko B, Simmons TL. Plasma
metabolomics reveals a diagnostic metabolic fingerprint for mi-
tochondrial aconitase (ACO2) deficiency. PLoS One. 2017;12(5):
e0176363.
53. Sadat R, Barca E, Masand R, Donti TR, Naini A, De Vivo DC,
DiMauro S, Hanchard NA, Graham BH. Functional cellular
analyses reveal energy metabolism defect and mitochondrial DNA
depletion in a case of mitochondrial aconitase deficiency. Mol
Genet Metab. 2016;118(1):2834.
54. Spiegel R, Pines O, Ta-Shma A, Burak E, Shaag A, Halvardson J,
Edvardson S, Mahajna M, Zenvirt S, Saada A, Shalev S, Feuk L,
Elpeleg O. Infantile cerebellar-retinal degeneration associated
with a mutation in mitochondrial aconitase, ACO2.Am J Hum
Genet. 2012;90(3):518523.
55. Chow J, Rahman J, Achermann JC, Dattani MT, Rahman S.
Mitochondrial disease and endocrine dysfunction. Nat Rev
Endocrinol. 2017;13(2):92104.
56. Al-Gadi IS, Haas RH, Falk MJ, Goldstein A, McCormack SE.
Endocrine disorders in primary mitochondrial disease. J Endocr
Soc. 2018;2(4):361373.
57. Demain LA, ConwayGS, NewmanWG. Genetics of mitochondrial
dysfunction and infertility. Clin Genet. 2017;91(2):199207.
58. Koopman WJ, Willems PH, Smeitink JA. Monogenic mito-
chondrial disorders. N Engl J Med. 2012;366(12):11321141.
59. Ropp PA, Copeland WC. Cloning and characterization of the
human mitochondrial DNA polymerase, DNA polymerase
gamma. Genomics. 1996;36(3):449458.
60. Rahman S, Copeland WC. POLG-related disorders and their
neurological manifestations. Nat Rev Neurol. 2019;15(1):4052.
61. Longley MJ, Graziewicz MA, Bienstock RJ, Copeland WC.
Consequences of mutations in human DNA polymerase gamma.
Gene. 2005;354:125131.
62. Luoma P, Melberg A, Rinne JO, Kaukonen JA, Nupponen NN,
Chalmers RM, Oldfors A, Rautakorpi I, Peltonen L, Majamaa K,
Somer H, Suomalainen A. Parkinsonism, premature menopause,
and mitochondrial DNA polymerase gmutations: clinical and
molecular genetic study. Lancet. 2004;364(9437):875882.
63. Chen B, Li L, Wang J, Zhou Y, Zhu J, Li T, Pan H, Liu B, Cao Y,
Wang B. Identification of the first homozygous POLG mutation
causing non-syndromic ovarian dysfunction. Climacteric. 2018;
21(5):467471.
64. Tong ZB, Sullivan SD, Lawless LM, Vanderhoof V, Zachman K,
Nelson LM. Five mutations of mitochondrial DNA polymerase-
gamma (POLG) are not a prevalent etiology for spontaneous
46,XX primary ovarian insufficiency. Fertil Steril. 2010;94(7):
29322934.
65. Duncan AJ, Knight JA, Costello H, Conway GS, Rahman S.
POLG mutations and age at menopause. Hum Reprod. 2012;
27(7):22432244.
66. Day FR, Ruth KS, Thompson DJ, Lunetta KL, Pervjakova N,
Chasman DI, Stolk L, Finucane HK, Sulem P, Bulik-Sullivan B,
Esko T, Johnson AD, Elks CE, Franceschini N, He C, Altmaier E,
Brody JA, Franke LL, Huffman JE, Keller MF, McArdle PF, Nutile
T, Porcu E, Robino A, Rose LM, Schick UM, Smith JA, Teumer A,
Traglia M, Vuckovic D, Yao J, Zhao W, Albrecht E, Amin N,
Corre T, Hottenga JJ, Mangino M, Smith AV, Tanaka T, Abecasis
GR, Andrulis IL, Anton-Culver H, Antoniou AC, Arndt V, Arnold
AM, Barbieri C, Beckmann MW, Beeghly-Fadiel A, Benitez J,
Bernstein L, Bielinski SJ, Blomqvist C, Boerwinkle E, Bogdanova
NV, Bojesen SE, Bolla MK, Borresen-Dale AL, Boutin TS, Brauch
H, Brenner H, Br ¨uning T, Burwinkel B, Campbell A, Campbell H,
Chanock SJ, Chapman JR, Chen YD, Chenevix-Trench G, Couch
FJ, Coviello AD, Cox A, Czene K, Darabi H, De Vivo I, Demerath
EW, Dennis J, Devilee P, D ¨ork T, dos-Santos-Silva I, Dunning
AM, Eicher JD, Fasching PA, Faul JD, Figueroa J, Flesch-Janys D,
Gandin I, Garcia ME, Garc´
ıa-Closas M, Giles GG, Girotto GG,
Goldberg MS, Gonz´alez-Neira A, Goodarzi MO, Grove ML,
Gudbjartsson DF, Gu´enel P, Guo X, Haiman CA, Hall P, Hamann
U, Henderson BE, Hocking LJ, Hofman A, Homuth G, Hooning
MJ, Hopper JL, Hu FB, Huang J, Humphreys K, Hunter DJ,
Jakubowska A, Jones SE, Kabisch M, Karasik D, Knight JA,
Kolcic I, Kooperberg C, Kosma VM, Kriebel J, Kristensen V,
Lambrechts D, Langenberg C, Li J, Li X, Lindstr ¨om S, Liu Y, Luan
J, Lubinski J, M¨agi R, Mannermaa A, Manz J, Margolin S,
Marten J, Martin NG, Masciullo C, Meindl A, Michailidou K,
Mihailov E, Milani L, Milne RL, M ¨uller-Nurasyid M, Nalls M,
Neale BM, Nevanlinna H, Neven P, Newman AB, Nordestgaard
BG, Olson JE, Padmanabhan S, Peterlongo P, Peters U,
Petersmann A, Peto J, Pharoah PD, Pirastu NN, Pirie A, Pistis G,
Polasek O, Porteous D, Psaty BM, Pylk ¨as K, Radice P, Raffel LJ,
Rivadeneira F, Rudan I, Rudolph A, Ruggiero D, Sala CF, Sanna
S, Sawyer EJ, Schlessinger D, Schmidt MK, Schmidt F, Schmutzler
RK, Schoemaker MJ, Scott RA, Seynaeve CM, Simard J, Sorice R,
Southey MC, St ¨ockl D, Strauch K, Swerdlow A, Taylor KD,
Thorsteinsdottir U, Toland AE, Tomlinson I, Truong T,
Tryggvadottir L, Turner ST, Vozzi D, Wang Q, Wellons M,
Willemsen G, Wilson JF, Winqvist R, Wolffenbuttel BB, Wright
AF, Yannoukakos D, Zemunik T, Zheng W, Zygmunt M,
Bergmann S, Boomsma DI, Buring JE, Ferrucci L, Montgomery
GW, Gudnason V, Spector TD, van Duijn CM, Alizadeh BZ,
Ciullo M, Crisponi L, Easton DF, Gasparini PP, Gieger C, Harris
TB, Hayward C, Kardia SL, Kraft P, McKnight B, Metspalu A,
Morrison AC, Reiner AP, Ridker PM, Rotter JI, Toniolo D,
Uitterlinden AG, Ulivi S, V ¨olzke H, Wareham NJ, Weir DR,
Yerges-Armstrong LM, Price AL, Stefansson K, Visser JA, Ong
KK, Chang-Claude J, Murabito JM, Perry JR, Murray A;
PRACTICAL Consortium; kConFab Investigators; AOCS In-
vestigators; Generation Scotland; EPIC-InterAct Consortium;
LifeLines Cohort Study. Large-scale genomic analyses link re-
productive aging to hypothalamic signaling, breast cancer sus-
ceptibility and BRCA1-mediated DNA repair. Nat Genet. 2015;
47(11):12941303.
67. Suomalainen A, Battersby BJ. Mitochondrial diseases: the con-
tribution of organelle stress responses to pathology. Nat Rev Mol
Cell Biol. 2018;19(2):7792.
68. Korhonen JA, Pham XH, Pellegrini M, Falkenberg M. Re-
constitution of a minimal mtDNA replisome in vitro. EMBO J.
2004;23(12):24232429.
69. Morino H, Pierce SB, Matsuda Y, Walsh T, Ohsawa R, Newby M,
Hiraki-Kamon K, Kuramochi M, Lee MK, Klevit RE, Martin A,
Maruyama H, King MC, Kawakami H. Mutations in Twinkle
primase-helicase cause Perrault syndrome with neurologic fea-
tures. Neurology. 2014;83(22):20542061.
70. Longley MJ, Clark S, Yu Wai Man C, Hudson G, Durham SE,
Taylor RW, Nightingale S, Turnbull DM, Copeland WC,
Chinnery PF. Mutant POLG2 disrupts DNA polymerase g
subunits and causes progressive external ophthalmoplegia. Am J
Hum Genet. 2006;78(6):10261034.
71. Suzuki T, Nagao A, Suzuki T. Human mitochondrial tRNAs:
biogenesis, function, structural aspects, and diseases. Annu Rev
Genet. 2011;45(1):299329.
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2363
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
72. Pierce SB, Gersak K, Michaelson-Cohen R, Walsh T, Lee MK,
Malach D, Klevit RE, King MC, Levy-Lahad E. Mutations in
LARS2, encoding mitochondrial leucyl-tRNA synthetase, lead to
premature ovarian failure and hearing loss in Perrault syndrome.
Am J Hum Genet. 2013;92(4):614620.
73. Pierce SB, Chisholm KM, Lynch ED, Lee MK, Walsh T, Opitz JM,
Li W, Klevit RE, King MC. Mutations in mitochondrial histidyl
tRNA synthetase HARS2 cause ovarian dysgenesis and sensori-
neural hearing loss of Perrault syndrome. Proc Natl Acad Sci USA.
2011;108(16):65436548.
74. Kosaki R, Horikawa R, Fujii E, Kosaki K. Biallelic mutations in
LARS2 can cause Perrault syndrome type 2 with neurologic
symptoms. Am J Med Genet A. 2018;176(2):404408.
75. Rehman AU, Friedman TB, Griffith AJ. Unresolved questions
regarding human hereditary deafness. Oral Dis. 2017;23(5):
551558.
76. Gispert S, Parganlija D, Klinkenberg M, Dr¨ose S, Wittig I,
Mittelbronn M, Grzmil P, Koob S, Hamann A, Walter M, B ¨uchel
F, Adler T, Hrab´e de Angelis M, Busch DH, Zell A, Reichert AS,
Brandt U, Osiewacz HD, Jendrach M, Auburger G. Loss of mi-
tochondrial peptidase Clpp leads to infertility, hearing loss plus
growth retardation via accumulation of CLPX, mtDNA and in-
flammatory factors. Hum Mol Genet. 2013;22(24):48714887.
77. Gonz´alez-Serrano LE, Chihade JW, Sissler M. When a common
biological role does not imply common disease outcomes: dis-
parate pathology linked to human mitochondrial aminoacyl-
tRNA synthetases. J Biol Chem. 2019;294(14):53095320.
78. Sissler M, Gonz´alez-Serrano LE, Westhof E. Recent advances in
mitochondrial aminoacyl-tRNA synthetases and disease. Trends
Mol Med. 2017;23(8):693708.
79. Dallabona C, Diodato D, Kevelam SH, Haack TB, Wong LJ,
Salomons GS, Baruffini E, Melchionda L, Mariotti C, Strom TM,
Meitinger T, Prokisch H, Chapman K, Colley A, Rocha H, Ounap
K, Schiffmann R, Salsano E, Savoiardo M, Hamilton EM, Abbink
TE, Wolf NI, Ferrero I, Lamperti C, Zeviani M, Vanderver A,
Ghezzi D, van der Knaap MS. Novel (ovario) leukodystrophy
related to AARS2 mutations. Neurology. 2014;82(23):
20632071.
80. Lynch DS, Zhang WJ, Lakshmanan R, Kinsella JA, Uzun GA,
Karbay M, T ¨ufekçioglu Z, Hanagasi H, Burke G, Foulds N,
Hammans SR, Bhattacharjee A, Wilson H, Adams M, Walker M,
Nicoll JA, Chataway J, Fox N, Davagnanam I, Phadke R,
Houlden H. Analysis of mutations in AARS2 in a Series of CSF1R-
negative patients with adult-onset leukoencephalopathy with
axonal spheroids and pigmented glia. JAMA Neurol. 2016;
73(12):14331439.
81. Taglia I, Di Donato I, Bianchi S, Cerase A, Monti L, Marconi R,
Orrico A, Rufa A, Federico A, Dotti MT. AARS2-related ovar-
ioleukodystrophy: clinical and neuroimaging features of three new
cases. Acta Neurol Scand. 2018;138(4):278283.
82. Hamatani M, Jingami N, Tsurusaki Y, Shimada S, Shimojima K,
Asada-Utsugi M, Yoshinaga K, Uemura N, Yamashita H, Uemura
K, Takahashi R, Matsumoto N, Yamamoto T. The first Japanese
case of leukodystrophy with ovarian failure arising from novel
compound heterozygous AARS2 mutations. J Hum Genet. 2016;
61(10):899902.
83. Lee JM, Yang HJ, Kwon JH, Kim WJ, Kim SY, Lee EM, Park JY,
Weon YC, Park SH, Gwon BJ, Ryu JC, Lee ST, Kim HJ, Jeon B.
Two Koreansiblings with recently described ovarioleukodystrophy
related to AARS2 mutations. Eur J Neurol.2017;24(4):e21e22.
84. Kiraly-Borri C, Jevon G, Ji W, Jeffries L, Ricciardi J-L,
Konstantino M, Ackerman KG, Lakhani SA. Siblings with lethal
primary pulmonary hypoplasia and compound heterozygous
variants in the AARS2 gene: further delineation of the phenotypic
spectrum. Cold Spring Harb Mol Case Stud. 2019;5(3):a003699.
85. G¨otz A, Tyynismaa H, Euro L, Ellonen P, Hy ¨otyl¨ainen T, Ojala T,
am ¨al¨ainen RH, Tommiska J, Raivio T, Oresic M, Karikoski R,
Tammela O, Simola KOJ, Paetau A, Tyni T, Suomalainen A. Exome
sequencing identifies mitochondrial alanyl-tRNA synthetase mutations
in infantile mitochondrial cardiomyopathy. Am J Hum Genet. 2011;
88(5):635642.
86. Sommerville EW, Zhou X-L, Ol ´ahov´a M, Jenkins J, Euro L,
Konovalova S, Hilander T, Pyle A, He L, Habeebu S, Saunders C,
Kelsey A, Morris AAM, McFarland R, Suomalainen A, Gorman
GS, Wang ED, Thiffault I, Tyynismaa H, Taylor RW. Instability
of the mitochondrial alanyl-tRNA synthetase underlies fatal
infantile-onset cardiomyopathy. Hum Mol Genet. 2019;28(2):
258268.
87. Euro L, Konovalova S, Asin-Cayuela J, Tulinius M, Griffin H,
Horvath R, Taylor RW, Chinnery PF, Schara U, Thorburn DR,
Suomalainen A, Chihade J, Tyynismaa H. Structural modeling of
tissue-specific mitochondrial alanyl-tRNA synthetase (AARS2)
defects predicts differential effects on aminoacylation. Front
Genet. 2015;6:21.
88. Fischer F, Hamann A, Osiewacz HD. Mitochondrial quality
control: an integrated network of pathways. Trends Biochem Sci.
2012;37(7):284292.
89. Szczepanowska K, Maiti P, Kukat A, Hofsetz E, Nolte H, Senft K,
Becker C, Ruzzenente B, Hornig-Do HT, Wibom R, Wiesner RJ,
Kr ¨uger M, Trifunovic A. CLPP coordinates mitoribosomal as-
sembly through the regulation of ERAL1 levels. EMBO J. 2016;
35(23):25662583.
90. Cole A, Wang Z, Coyaud E, Voisin V, Gronda M, Jitkova Y,
Mattson R, Hurren R, Babovic S, Maclean N, Restall I, Wang X,
Jeyaraju DV, Sukhai MA, Prabha S, Bashir S, Ramakrishnan A,
Leung E, Qia YH, Zhang N, Combes KR, Ketela T, Lin F, Houry
WA, Aman A, Al-Awar R, Zheng W, Wienholds E, Xu CJ, Dick J,
Wang JC, Moffat J, Minden MD, Eaves CJ, Bader GD, Hao Z,
Kornblau SM, Raught B, Schimmer AD. Inhibition of the mito-
chondrial protease ClpP as a therapeutic strategy for human acute
myeloid leukemia. Cancer Cell. 2015;27(6):864876.
91. Fischer F, Langer JD, Osiewacz HD. Identification of potential
mitochondrial CLPXP protease interactors and substrates sug-
gests its central role in energy metabolism. Sci Rep. 2015;5(1):
18375.
92. Deepa SS, Bhaskaran S, Ranjit R, Qaisar R, Nair BC, Liu Y, Walsh
ME, Fok WC, Van Remmen H. Down-regulation of the mito-
chondrial matrix peptidase ClpP in muscle cells causes mito-
chondrial dysfunction and decreases cell proliferation. Free Radic
Biol Med. 2016;91:281292.
93. Lerat J, Jonard L, Loundon N, Christin-Maitre S, Lacombe D,
Goizet C, Rouzier C, Van Maldergem L, Gherbi S, Garabedian
EN, Bonnefont JP, Touraine P, Mosnier I, Munnich A, Denoyelle
F, Marlin S. An application of NGS for molecular investigations in
Perrault syndrome: study of 14 families and review of the liter-
ature. Hum Mutat. 2016;37(12):13541362.
94. Dursun F, Mohamoud HSA, Karim N, Naeem M, Jelani M,
Kırmızıbekmez H. A novel missense mutation in the CLPP gene
causing Perrault Syndrome type 3 in a Turkish family. J Clin Res
Pediatr Endocrinol. 2016;8(4):472477.
95. Ahmed S, Jelani M, Alrayes N, Mohamoud HS, Almramhi MM,
Anshasi W, Ahmed NA, Wang J, Nasir J, Al-Aama JY. Exome
analysis identified a novel missense mutation in the CLPP gene
in a consanguineous Saudi family expanding the clinical spectrum
of Perrault syndrome type-3. J Neurol Sci. 2015;353(12):
149154.
96. Jenkinson EM, Rehman AU, Walsh T, Clayton-Smith J, Lee K,
Morell RJ, Drummond MC, Khan SN, Naeem MA, Rauf B,
Billington N, Schultz JM, Urquhart JE, Lee MK, Berry A, Hanley
NA, Mehta S, Cilliers D, Clayton PE, Kingston H, Smith MJ,
Warner TT, Black GC, Trump D, Davis JR, Ahmad W, Leal SM,
Riazuddin S, King MC, Friedman TB, Newman WG; University of
Washington Center for Mendelian Genomics. Perrault syndrome
is caused by recessive mutations in CLPP, encoding a mito-
chondrial ATP-dependent chambered protease. Am J Hum Genet.
2013;92(4):605613.
2364 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
97. Brodie EJ, Zhan H, Saiyed T, Truscott KN, Dougan DA. Perrault
syndrome type 3 caused by diverse molecular defects in CLPP. Sci
Rep. 2018;8(1):12862.
98. Siira SJ, Sp˚ahr H, Shearwood AJ, Ruzzenente B, Larsson NG,
Rackham O, Filipovska A. LRPPRC-mediated folding of the
mitochondrial transcriptome. Nat Commun. 2017;8(1):1532.
99. Ruzzenente B, Metodiev MD, Wredenberg A, Bratic A, Park CB,
amara Y, Milenkovic D, Zickermann V, Wibom R, Hultenby K,
Erdjument-Bromage H, Tempst P, Brandt U, Stewart JB,
Gustafsson CM, Larsson NG. LRPPRC is necessary for poly-
adenylation and coordination of translation of mitochondrial
mRNAs. EMBO J. 2012;31(2):443456.
100. Lagouge M, Mourier A, Lee HJ, Sp ˚ahr H, Wai T, Kukat C, Silva
Ramos E, Motori E, Busch JD, Siira S, Kremmer E, Filipovska A,
Larsson NG; German Mouse Clinic Consortium. SLIRP regulates
the rate of mitochondrial protein synthesis and protects LRPPRC
from degradation. PLoS Genet. 2015;11(8):e1005423.
101. Volpon L, Culjkovic-Kraljacic B, Sohn HS, Blanchet-Cohen A,
Osborne MJ, Borden KL. A biochemical framework for eIF4E-
dependent mRNA export and nuclear recycling of the export
machinery. RNA. 2017;23(6):927937.
102. Cooper MP, Qu L, Rohas LM, Lin J, Yang W, Erdjument-
Bromage H, Tempst P, Spiegelman BM. Defects in energy ho-
meostasis in Leigh syndrome French Canadian variant through
PGC-1a/LRP130 complex. Genes Dev. 2006;20(21):29963009.
103. Han VX, TanTS, Wang FS, Tay SK. NovelLRPPRC mutation in a
boy with mild Leigh syndrome, FrenchCanadian type outside of
Qu´ebec. Child Neurol Open. 2017;4: 2329048X17737638. doi:
10.1177/2329048X17737638.
104. Ol´ahov ´a M, Hardy SA, Hall J, Yarham JW, Haack TB, Wilson
WC, Alston CL, He L, Aznauryan E, Brown RM, Brown GK,
Morris AAM, Mundy H, Broomfield A, Barbosa IA, Simpson
MA, Deshpande C, Moeslinger D, Koch J, Stettner GM, Bonnen
PE, Prokisch H, Lightowlers RN, McFarland R, Chrzanowska-
Lightowlers ZM, Taylor RW. LRPPRC mutations cause early-onset
multisystem mitochondrial disease outside of the French-Canadian
population. Brain. 2015;138(Pt 12):35033519.
105. Mootha VK, Lepage P, Miller K, Bunkenborg J, Reich M, Hjerrild
M, Delmonte T, Villeneuve A, Sladek R, Xu F, Mitchell GA,
Morin C, Mann M, Hudson TJ, Robinson B, Rioux JD, Lander
ES. Identification of a gene causing human cytochrome coxidase
deficiency by integrative genomics. Proc Natl Acad Sci USA.
2003;100(2):605610.
106. Ghaddhab C, Morin C, Brunel-Guitton C, Mitchell GA, Van Vliet
G, Huot C. Premature ovarian failure in French Canadian Leigh
syndrome. J Pediatr. 2017;184:227229.e1.
107. Debray FG, Morin C, Janvier A, Villeneuve J, Maranda B,
Laframboise R, Lacroix J, Decarie JC, Robitaille Y, Lambert M,
Robinson BH, Mitchell GA. LRPPRC mutations cause a phe-
notypically distinct form of Leigh syndrome with cytochrome c
oxidase deficiency. J Med Genet. 2011;48(3):183189.
108. Amunts A, Brown A, Toots J, Scheres SH, Ramakrishnan V. The
structure of the human mitochondrial ribosome. Science. 2015;
348(6230):9598.
109. Emdadul Haque M, Grasso D, Miller C, Spremulli LL, Saada A.
The effect of mutated mitochondrial ribosomal proteins S16 and
S22 on the assembly of the small and large ribosomal subunits in
human mitochondria. Mitochondrion. 2008;8(3):254261.
110. Chen A, Tiosano D, Guran T, Baris HN, Bayram Y, Mory A,
Shapiro-Kulnane L, Hodges CA, Akdemir ZC, Turan S, Jhangiani
SN, van den Akker F, Hoppel CL, Salz HK, Lupski JR, Buchner
DA. Mutations in the mitochondrial ribosomal protein MRPS22
lead to primary ovarian insufficiency. Hum Mol Genet. 2018;
27(11):19131926.
111. Jolly A, Bayram Y, Turan S, Aycan Z, Tos T, Abali ZY,
Hacihamdioglu B, Coban Akdemir ZH, Hijazi H, Bas S, Atay Z,
Guran T, Abali S, Bas F, Darendeliler F, Colombo R, Barakat TS,
Rinne T, White JJ, Yesil G, Gezdirici A, Gulec EY, Karaca E,
Pehlivan D, Jhangiani SN, Muzny DM, Poyrazoglu S, Bereket A,
Gibbs RA, Posey JE, Lupski JR. Exome sequencing of a primary
ovarian insufficiency cohort reveals common molecular etiologies
for a spectrum of disease. J Clin Endocrinol Metab. 2019;104(8):
30493067.
112. Saada A, Shaag A, Arnon S, Dolfin T, Miller C, Fuchs-Telem D,
Lombes A, Elpeleg O. Antenatal mitochondrial disease caused by
mitochondrial ribosomal protein (MRPS22) mutation. JMed
Genet. 2007;44(12):784786.
113. Smits P, Saada A, Wortmann SB, Heister AJ, Brink M, Pfundt R,
Miller C, Haas D, Hantschmann R, Rodenburg RJ, Smeitink
JAM, van den Heuvel LP. Mutation in mitochondrial ribosomal
protein MRPS22 leads to Cornelia de Lange-like phenotype, brain
abnormalities and hypertrophic cardiomyopathy. Eur J Hum
Genet. 2011;19(4):394399.
114. Baertling F, Haack TB, Rodenburg RJ, Schaper J, Seibt A, Strom
TM, Meitinger T, Mayatepek E, Hadzik B, Selcan G, Prokisch H,
Distelmaier F. MRPS22 mutation causes fatal neonatal lactic
acidosis with brain and heart abnormalities. Neurogenetics. 2015;
16(3):237240.
115. KılıçM,O
˘guz K-K, KılıçE,Y¨uksel D, Demirci H, Sa ˘gıro ˘glu MS
¸,
ucel-Yılmaz D, ¨
Ozg ¨ul RK. A patient with mitochondrial dis-
order due to a novel mutation in MRPS22. Metab Brain Dis.
2017;32(5):13891393.
116. Hochberg I, Demain LA, Urquhart JE, Amberger A, Deutschmann
AJ, Demetz S, Thompson K, OSullivan J, Belyantseva IA, Barzik
M, Williams SG, Bhaskar SS, Jenkinson EM, AlSheqaih N,
Blumenfeld Z, Yalonetsky S, Oerum S, Rossmanith W, Yue WW,
Zschocke J, Taylor RW, Friedman TB, Munro KJ, OKeefe RT,
Newman WG. A homozygous variant in mitochondrial RNase P
subunit PRORP is associated with Perrault syndrome charac-
terized by hearing loss and primary ovarian insufficiency. bio-
Rxiv. 2017:168252.
117. Menezes MJ, Guo Y, Zhang J, Riley LG, Cooper ST, Thorburn
DR, Li J, Dong D, Li Z, Glessner J, Davis RL, Sue CM, Alexander
SI, Arbuckle S, Kirwan P, Keating BJ, Xu X, Hakonarson H,
Christodoulou J. Mutation in mitochondrial ribosomal protein S7
(MRPS7) causes congenital sensorineural deafness, progressive
hepatic and renal failure and lactic acidemia. Hum Mol Genet.
2015;24(8):22972307.
118. Demain LA, Antunes D, OSullivan J, Bhaskhar SS, OKeefe RT,
Newman WG. A known pathogenic variant in the essential mi-
tochondrial translation gene RMND1 causes a Perrault-like
syndrome with renal defects. Clin Genet. 2018;94(2):276277.
119. Ng YS, Alston CL, Diodato D, Morris AA, Ulrick N, Kmoch S,
Houˇstˇek J, Martinelli D, Haghighi A, Atiq M, Gamero MA,
Garcia-Martinez E, Kratochv´
ılov´a H, Santra S, Brown RM,
Brown GK, Ragge N, Monavari A, Pysden K, Ravn K, Casey JP,
Khan A, Chakrapani A, Vassallo G, Simons C, McKeever K,
OSullivan S, Childs AM, Østergaard E, Vanderver A, Goldstein
A, Vogt J, Taylor RW, McFarland R. The clinical, biochemical
and genetic features associated with RMND1-related mito-
chondrial disease. J Med Genet. 2016;53(11):768775.
120. Janer A, Antonicka H, Lalonde E, Nishimura T, Sasarman F,
Brown GK, Brown RM, Majewski J, Shoubridge EA. An RMND1
mutation causes encephalopathy associated with multiple oxi-
dative phosphorylation complex deficiencies and a mitochondrial
translation defect. Am J Hum Genet. 2012;91(4):737743.
121. Garcia-Diaz B, Barros MH, Sanna-Cherchi S, Emmanuele V,
Akman HO, Ferreiro-Barros CC, Horvath R, Tadesse S, El
Gharaby N, DiMauro S, De Vivo DC, Shokr A, Hirano M,
Quinzii CM. Infantile encephaloneuromyopathy and defective
mitochondrial translation are due to a homozygous RMND1
mutation. Am J Hum Genet. 2012;91(4):729736.
122. Posey JE, ODonnell-Luria AH, Chong JX, Harel T, Jhangiani
SN, Coban Akdemir ZH, Buyske S, Pehlivan D, Carvalho CMB,
Baxter S, Sobreira N, Liu P, Wu N, Rosenfeld JA, Kumar S,
Avramopoulos D, White JJ, Doheny KF, Witmer PD, Boehm C,
doi: 10.1210/en.2019-00441 https://academic.oup.com/endo 2365
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
Sutton VR, Muzny DM, Boerwinkle E, G ¨unel M, Nickerson DA,
Mane S, MacArthur DG, Gibbs RA, Hamosh A, Lifton RP,
Matise TC, Rehm HL, Gerstein M, Bamshad MJ, Valle D, Lupski
JR; Centers for Mendelian Genomics. Insights into genetics,
human biology and disease gleaned from family based genomic
studies. Genet Med. 2019;21(4):798812.
123. Venkatesh S, Kumar M, Sharma A, Kriplani A, Ammini AC,
Talwar P, Agarwal A, Dada R. Oxidative stress and ATPase6
mutation is associated with primary ovarian insufficiency. Arch
Gynecol Obstet. 2010;282(3):313318.
124. Zhen X, Wu B, Wang J, Lu C, Gao H, Qiao J. Increased incidence
of mitochondrial cytochrome c oxidase 1 gene mutations in pa-
tients with primary ovarian insufficiency. PLoS One. 2015;10(7):
e0132610.
125. Palanichamy MG, Zhang Y-P. Are ATPase6 polymorphisms
associated with primary ovarian insufficiency? Arch Gynecol
Obstet. 2011;283(3):671672.
126. Kumar M, Pathak D, Venkatesh S, Kriplani A, Ammini AC, Dada
R. Chromosomal abnormalities & oxidative stress in women with
premature ovarian failure (POF). Indian J Med Res. 2012;135(1):
9297.
127. Kumar M, Pathak D, Kriplani A, Ammini AC, Talwar P, Dada R.
Nucleotide variations in mitochondrial DNA and supra-
physiological ROS levels in cytogenetically normal cases of pre-
mature ovarian insufficiency. Arch Gynecol Obstet. 2010;282(6):
695705.
128. Ding Y, Xia BH, Zhuo GC, Zhang CJ, Leng JH. Premature
ovarian insufficiency may be associated with the mutations in
mitochondrial tRNA genes. Endocr J. 2019;66(1):8188.
129. Ben-Meir A, Burstein E, Borrego-Alvarez A, Chong J, Wong E,
Yavorska T, Naranian T, Chi M, Wang Y, Bentov Y, Alexis J,
Meriano J, Sung H-K, Gasser DL, Moley KH, Hekimi S, Casper
RF, Jurisicova A. Coenzyme Q10 restores oocyte mitochondrial
function and fertility during reproductive aging. Aging Cell. 2015;
14(5):887895.
130. Boczonadi V, Smith PM, Pyle A, Gomez-Duran A, Schara U,
Tulinius M, Chinnery PF, Horvath R. Altered 2-thiouridylation
impairs mitochondrial translation in reversible infantile re-
spiratory chain deficiency. Hum Mol Genet. 2013;22(22):
46024615.
131. Wang G, Shimada E, Zhang J, Hong JS, Smith GM, Teitell MA,
Koehler CM. Correcting human mitochondrial mutations with
targeted RNA import. Proc Natl Acad Sci USA. 2012;109(13):
48404845.
132. Perli E, Giordano C, Pisano A, Montanari A, Campese AF, Reyes
A, Ghezzi D, Nasca A, Tuppen HA, Orlandi M, Di Micco P, Poser
E, Taylor RW, Colotti G, Francisci S, Morea V, Frontali L,
Zeviani M, dAmati G. The isolated carboxy-terminal domain of
human mitochondrial leucyl-tRNA synthetase rescues the path-
ological phenotype of mitochondrial tRNA mutations in human
cells. EMBO Mol Med. 2014;6(2):169182.
133. Hornig-Do HT, Montanari A, Rozanska A, Tuppen HA, Almalki
AA, Abg-Kamaludin DP, Frontali L, Francisci S, Lightowlers RN,
Chrzanowska-Lightowlers ZM. Human mitochondrial leucyl
tRNA synthetase can suppress non cognate pathogenic mt-tRNA
mutations. EMBO Mol Med. 2014;6(2):183193.
134. Zhao X, Han J, Zhu L, Xiao Y, Wang C, Hong F, Jiang P, Guan
M-X. Overexpression of human mitochondrial alanyl-tRNA
synthetase suppresses biochemical defects of the mt-tRNA
Ala
mutation in cybrids. Int J Biol Sci. 2018;14(11):14371444.
135. Gadaleta MN, Petruzzella V, Daddabbo L, Olivieri C, Fracasso F,
Loguercio Polosa P, Cantatore P. Mitochondrial DNA tran-
scription and translation in aged rat. Effect of acetyl-L-carnitine.
Ann N Y Acad Sci. 1994;717(1):150160.
136. Liang LF, Qi ST, Xian YX, Huang L, Sun XF, Wang WH.
Protective effect of antioxidants on the pre-maturation aging of
mouse oocytes. Sci Rep. 2017;7(1):1434.
137. Zare Z, Masteri Farahani R, Salehi M, Piryaei A, Ghaffari Novin
M, Fadaei Fathabadi F, Mohammadi M, Dehghani-
Mohammadabadi M. Effect of L-carnitine supplementation on
maturation and early embryo development of immature mouse
oocytes selected by brilliant cresyle blue staining. J Assist Reprod
Genet. 2015;32(4):635643.
138. Yamada T, Imai H, Yamada M. Beneficial effects of acetyl-L-
carnitine treatment during IVM on post-fertilization development
of bovine oocytes in vitro. Reprod Fertil Dev. 2006;18(2):280.
139. Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ,
Nickerson DA, Shendure J. Exome sequencing as a tool for
Mendelian disease gene discovery. Nat Rev Genet. 2011;12(11):
745755.
140. Committee on Adolescent Health Care. Committee opinion no.
605: primary ovarian insufficiency in adolescents and young
women. Obstet Gynecol. 2014;124(1):193197.
141. Stark Z, Schofield D, Alam K, Wilson W, Mupfeki N, Macciocca
I, Shrestha R, White SM, Gaff C. Prospective comparison of the
cost-effectiveness of clinical whole-exome sequencing with that of
usual care overwhelmingly supports early use and reimbursement.
Genet Med. 2017;19(8):867874.
142. Monroe GR, Frederix GW, Savelberg SMC, de Vries TI, Duran
KJ, van der Smagt JJ, Terhal PA, van Hasselt PM, Kroes HY,
Verhoeven-Duif NM, Nijman IJ, Carbo EC, van Gassen KL,
Knoers NV, H ¨ovels AM, van Haelst MM, Visser G, van Haaften
G. Effectiveness of whole-exome sequencing and costs of the
traditional diagnostic trajectory in children with intellectual
disability. Genet Med. 2016;18(9):949956.
143. Tan TY, Dillon OJ, Stark Z, Schofield D, Alam K, Shrestha R,
Chong B, Phelan D, Brett GR, Creed E, Jarmolowicz A, Yap P,
Walsh M, Downie L, Amor DJ, Savarirayan R, McGillivray G,
Yeung A, Peters H, Robertson SJ, Robinson AJ, Macciocca I,
Sadedin S, Bell K, Oshlack A, Georgeson P, Thorne N, Gaff C,
White SM. Diagnostic impact and cost-effectiveness of whole-
exome sequencing for ambulant children with suspected mono-
genic conditions. JAMA Pediatr. 2017;171(9):855862.
144. Stark Z, Tan TY, Chong B, Brett GR, Yap P, Walsh M, Yeung A,
Peters H, Mordaunt D, Cowie S, Amor DJ, Savarirayan R,
McGillivray G, Downie L, Ekert PG, Theda C, James PA, Yaplito-
Lee J, Ryan MM, Leventer RJ, Creed E, Macciocca I, Bell KM,
Oshlack A, Sadedin S, Georgeson P, Anderson C, Thorne N, Gaff
C, White SM; Melbourne Genomics Health Alliance. A pro-
spective evaluation of whole-exome sequencing as a first-tier
molecular test in infants with suspected monogenic disorders.
Genet Med. 2016;18(11):10901096.
145. Vissers LE, van Nimwegen KJ, Schieving JH, Kamsteeg EJ,
Kleefstra T, Yntema HG, Pfundt R, van der Wilt GJ, Krabbenborg
L, Brunner HG, van der Burg S, Grutters J, Veltman JA,
Willemsen MA. A clinical utility study of exome sequencing
versus conventional genetic testing in pediatric neurology. Genet
Med. 2017;19(9):10551063.
146. Stavropoulos DJ, Merico D, Jobling R, Bowdin S, Monfared N,
Thiruvahindrapuram B, Nalpathamkalam T, Pellecchia G,
Yuen RK, Szego MJ, Hayeems RZ, Shaul RZ, Brudno M,
GirdeaM,FreyB,AlipanahiB,AhmedS,Babul-HirjiR,Porras
RB, Carter MT, Chad L, Chaudhry A, Chitayat D, Doust SJ,
Cytrynbaum C, Dupuis L, Ejaz R, Fishman L, Guerin A,
Hashemi B, Helal M, Hewson S, Inbar-Feigenberg M, Kannu P,
Karp N, Kim R, Kronick J, Liston E, MacDonald H, Mercimek-
Mahmutoglu S, Mendoza-Londono R, Nasr E, Nimmo G,
Parkinson N, Quercia N, Raiman J, Roifman M, Schulze A,
Shugar A, Shuman C, Sinajon P, Siriwardena K, Weksberg R,
Yoon G, Carew C, Erickson R, Leach RA, Klein R, Ray PN,
Meyn MS, Scherer SW, Cohn RD, Marshall CR. Whole-
genome sequencing expands diagnostic utility and improves
clinical management in paediatric medicine. NPJ Genom Med.
2016;1(1):15012.
2366 Tiosano et al Mitochondrial Dysfunction in POI Endocrinology, October 2019, 160(10):23532366
Downloaded from https://academic.oup.com/endo/article/160/10/2353/5544515 by guest on 12 June 2023
... Premature ovarian insufficiency (POI) is a disease characterized by early menopause before 40 years of age, accompanied by an elevation of follicle-stimulating hormone (FSH ≥ 25 IU/L on two occasions over 4 weeks apart), and ultimately leads to female infertility [1]. POI is one of the most common reproductive endocrine disorders and affects 1-2% of women of childbearing age [2]. Ovarian granulosa cells (GCs) dysfunction triggered by reactive oxygen species (ROS) exposure is an important causal factor for POI [3]. ...
... Previously, we demonstrated that H-Exs were able to alleviate GCs senescence by reducing ROS damage [31]. Considering that mitochondria are the first sensors of senescence and target organs of ROS production [2,5,32], we hypothesize that Ex-mediated antisenescence effects may be associated with improvements in the mitochondrial function of GCs. To illustrate this, we isolated and characterized H-Exs and evaluated the effect of H-Exs in the repair of oxidative damage-induced mitochondrial dysfunction in KGNs (the in vitro cell line of GCs). ...
Article
Full-text available
Background Premature ovarian insufficiency (POI) is an important cause of female infertility and seriously impacts the physical and psychological health of patients. Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSCs-Exs, H-Exs) have exhibited protective effects on ovarian function with unclear mechanisms. Methods A comprehensive analysis of the Gene Expression Omnibus (GEO) database were used to identify POI-associated circRNAs and miRNAs. The relationship between HucMSC-derived exosomal circBRCA1/miR-642a-5p/FOXO1 axis and POI was examined by RT-qPCR, Western blotting, reactive oxygen species (ROS) staining, senescence-associated β-gal (SA-β-gal) staining, JC-1 staining, TEM, oxygen consumption rate (OCR) measurements and ATP assay in vivo and in vitro. RT-qPCR detected the expression of circBRCA1 in GCs and serum of patients with normal ovarian reserve function (n = 50) and patients with POI (n = 50); then, the correlation of circBRCA1 with ovarian reserve function indexes was analyzed. Results Herein, we found that circBRCA1 was decreased in the serum and ovarian granulosa cells (GCs) of patients with POI and was associated with decreased ovarian reserve. H-Exs improved the disorder of the estrous cycles and reproductive hormone levels, reduced the number of atretic follicles, and alleviated the apoptosis and senescence of GCs in rats with POI. Moreover, H-Exs mitigated mitochondrial damage and reversed the reduced circBRCA1 expression induced by oxidative stress in GCs. Mechanistically, FTO served as an eraser to increase the stability and expression of circBRCA1 by mediating the m⁶A demethylation of circBRCA1, and exosomal circBRCA1 sponged miR-642a-5p to block its interaction with FOXO1. CircBRCA1 insufficiency aggravated mitochondrial dysfunction, mimicking FTO or FOXO1 depletion effects, which was counteracted by miR-642a-5p inhibition. Conclusion H-Exs secreted circBRCA1 regulated by m⁶A modification, directly sponged miR-642a-5p to upregulate FOXO1, resisted oxidative stress injuries in GCs and protected ovarian function in rats with POI. Exosomal circBRCA1 supplementation may be a general prospect for the prevention and treatment of POI. Graphical Abstract
... SLC25A39 is known to play a role in mitochondrial transport and metabolism (Ruprecht and Kunji, 2020). It was reported that mitochondrial dysfunction were associated with some rare cases of POI, and manipulation of mitochondrial function represents an important therapeutic target for the treatment or prevention of POI (Tiosano et al., 2019). Therefore, the gene product of SLC25A39 plays a pivotal role in mitochondrial function by facilitating the transport of glutathione into mitochondria, which is essential for antioxidant defense and maintaining cellular redox balance (Slabbaert et al., 2016). ...
Article
Full-text available
Introduction This study aimed to explore the transcriptomic profile of premature ovarian insufficiency (POI) by investigating alterations in gene expression. Methods A total of sixty-one women, comprising 31 individuals with POI in the POI group and 30 healthy women in the control group (HC group), aged between 24 and 40 years, were recruited for this study. The transcriptomic profiles of peripheral blood samples from all study subjects were analyzed using RNA-sequencing. Results The results revealed 39 differentially expressed genes in individuals with POI compared to healthy controls, with 10 upregulated and 29 downregulated genes. Correlation analysis highlighted the relationship between the expression of SLC25A39, CNIH3, and PDZK1IP1 and hormone levels. Additionally, an effective classification model was developed using SLC25A39, CNIH3, PDZK1IP1, SHISA4, and LOC389834. Functional enrichment analysis demonstrated the involvement of these differentially expressed genes in the “haptoglobin-hemoglobin complex,” while KEGG pathway analysis indicated their participation in the “Proteoglycans in cancer” pathway. Conclusion The identified genes could play a crucial role in characterizing the genetic foundation of POI, potentially serving as valuable biomarkers for enhancing disease classification accuracy.
... Mutations in individual genes and chromosomal abnormalities are important causes of POI. Tiosano et al. found that mutated genes associated with POI, such as MRPS22, POLG, TWNK, and LARS2, are all involved in mitochondrial DNA replication, gene expression, and protein synthesis and degradation [115]. Notably, spermidine plays an important role in improving mitochondrial function. ...
Article
Full-text available
Ovarian aging and disease-related decline in fertility are challenging medical and economic issues with an increasing prevalence. Polyamines are a class of polycationic alkylamines widely distributed in mammals. They are small molecules essential for cell growth and development. Polyamines alleviate ovarian aging through various biological processes, including reproductive hormone synthesis, cell metabolism, programmed cell death, etc. However, an abnormal increase in polyamine levels can lead to ovarian damage and promote the development of ovarian disease. Therefore, polyamines have long been considered potential therapeutic targets for aging and disease, but their regulatory roles in the ovary deserve further investigation. This review discusses the mechanisms by which polyamines ameliorate human ovarian aging and disease through different biological processes, such as autophagy and oxidative stress, to develop safe and effective polyamine targeted therapy strategies for ovarian aging and the diseases.
Article
Full-text available
Female hypogonadism (FH) is a relatively common endocrine disorder in women of premenopausal age, but there are significant uncertainties and wide variation in its management. Most current guidelines are monospecialty and only address premature ovarian insufficiency (POI); some allude to management in very brief and general terms, and most rely upon the extrapolation of evidence from the studies relating to physiological estrogen deficiency in postmenopausal women. The Society for Endocrinology commissioned new guidance to provide all care providers with a multidisciplinary perspective on managing patients with all forms of FH. It has been compiled using expertise from Endocrinology, Primary Care, Gynaecology and Reproductive Health practices, with contributions from expert patients and a patient support group, to help clinicians best manage FH resulting from both POI and hypothalamo‐pituitary disorders, whether organic or functional.
Chapter
Reproductive health implies the total well-being of an individual in all aspects of reproduction, including physical, behavioral, emotional, and social well-being. Maintaining a functional reproductive health status is critical for ensuring community and family health. Reproductive disorders present a global health concern that considerably impacts human health and are more challenging to diagnose and treat, increasing the likelihood of comorbidity. Owing to the involvement of a multitude of factors in the development and pathophysiology of reproductive diseases, the identification of critical therapeutic targets becomes challenging. Factors influencing ovarian function and endometrial receptivity represent ideal treatment targets due to their importance in influencing fertility and successful pregnancy outcomes. Components of the immune system and inflammatory pathways are central to various reproductive events, which can be therapeutically manipulated for the management of reproductive pathologies in which inflammatory dysregulation plays a central role. Additionally, targeting the reproductive tract microbiota and pathogenic microbes involved in reproductive tract infections provides a promising approach to restoring reproductive health. Treatment of reproductive disorders mainly includes drugs for treating menstrual disorders, drugs for fertility treatments, and those for treating reproductive infections. Herbal medicines offer a holistic and safe approach to the management of reproductive disorders, and currently, a number of active compounds derived from natural sources have demonstrated the potential to develop into successful drug candidates. The chapter summarizes data on the different therapeutic targets in reproductive disorders with the description of potential drug leads that have proven promising in the management of reproductive pathologies.
Chapter
Premature ovarian failure is defined as the lack of ovarian hormones or ovarian follicle reserve. This results in aberrant ovarian activity, which causes early oocyte depletion and the start of menopause. Amenorrhea, a review of the family history going back at least three generations, a pelvic sonogram, and a serum FSH level > 40 mIU/ml are all used to diagnose POF illness. The 2016 guidelines from the European Society of Human Reproduction and Embryology (ESHRE) recommended that amenorrhea be monitored for at least 4 months, increased FSH levels be tested for two consecutive months, and then karyotyping be used for genetic testing. 20–25% of the cases are believed to be genetic. A few genes have been discovered to affect the risk of POF, but 50–90% of the cases remain idiopathic. Since normal ovarian function is a complex process, many genes have yet to be discovered with a role in causing the disease. In this chapter, we have focused on genetic testing in POF cases. Different diagnostic methods, such as karyotyping, candidate gene sequencing, genome-wide association studies, whole genome/exome sequencing, array comparative genomic hybridization, and microarray, have been discussed in this chapter.
Preprint
Full-text available
Background: Premature ovarian insufficiency (POI) is an important cause of female infertility and seriously impacts the physical and psychological health of patients. Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSCs-Exs, H-Exs) have exhibited protective effects on ovarian function with unclear mechanisms. Methods: A comprehensive analysis of the Gene Expression Omnibus (GEO) database were used to identify POI-associated circRNAs and miRNAs. The relationship between HucMSC-derived exosomal circBRCA1/miR-642a-5p/FOXO1 axis and POI was examined by RT-qPCR, Western blotting, reactive oxygen species (ROS) staining, senescence-associated β-gal (SA-β-gal) staining, JC-1 staining, TEM, oxygen consumption rate (OCR) measurements and ATP assay in vivo and in vitro. RT-qPCR detected the expression of circBRCA1 in GCs and serum of patients with normal ovarian reserve function (n=50) and patients with POI (n=50); then, the correlation of circBRCA1 with ovarian reserve function indexes was analyzed. Results: Herein, we found that circBRCA1 was decreased in the serum and ovarian granulosa cells (GCs) of patients with POI and was associated with decreased ovarian reserve. H-Exs improved the disorder of the estrous cycles and reproductive hormone levels, reduced the number of atretic follicles, and alleviated the apoptosis and senescence of GCs in rats with POI. Moreover, H-Exs mitigated mitochondrial damage and reversed the reduced circBRCA1 expression induced by oxidative stress in GCs. Mechanistically, FTO served as an eraser to increase the stability and expression of circBRCA1 by mediating the m⁶A demethylation of circBRCA1, and exosomal circBRCA1 sponged miR-642a-5p to block its interaction with FOXO1. CircBRCA1 insufficiency aggravated mitochondrial dysfunction, mimicking FTO or FOXO1 depletion effects, which was counteracted by miR-642a-5p inhibition. Conclusion: H-Exs secreted circBRCA1 regulated by m⁶A modification, directly sponged miR-642a-5p to upregulate FOXO1, resisted oxidative stress injuries in GCs and protected ovarian function in rats with POI. Exosomal circBRCA1 supplementation may be a general prospect for the prevention and treatment of POI.
Article
Full-text available
Heteroplasmy incidence in mitochondrial DNA In humans, mitochondrial DNA (mtDNA) is predominantly maternally inherited. mtDNA is under selection to prevent heteroplasmy—the transmission of multiple genetic variants into the next generation. Wei et al. explored human mtDNA sequences to determine mtDNA genome structure, selection, and transmission. Whole-genome sequencing revealed that about 45% of individuals carry heteroplasmic mtDNA sequences at levels greater than 1% of their total mtDNA. Furthermore, studies of more than 1500 mother-offspring pairs indicated that the female line selected which mtDNA variants were passed on to children. This effect was influenced by the mother's nuclear genetic background. Thus, mtDNA is under selection at specific loci in the human germ line. Science , this issue p. eaau6520
Article
Full-text available
Variants in the mitochondrial alanyl-tRNA synthetase 2 gene AARS2 (OMIM 612035) are associated with infantile mitochondrial cardiomyopathy or later-onset leukoencephalopathy with premature ovarian insufficiency. Here, we report two newborn siblings who died soon after birth with primary pulmonary hypoplasia without evidence of cardiomyopathy. Whole-exome sequencing detected the same compound heterozygous AARS2 variants in both siblings (c.1774C>T, p.Arg592Trp and c.647dup, p.Cys218Leufs*6) that have previously been associated with infantile mitochondrial cardiomyopathy. Segregation analysis in the family confirmed carrier status of the parents and an unaffected sibling. To our knowledge, this is the first report of primary pulmonary hypoplasia in the absence of cardiomyopathy associated with recessive AARS2 variants and further defines the phenotypic spectrum associated with this gene.
Article
Full-text available
Mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs) are essential components of the mitochondrial translation machinery. The correlation of mitochondrial disorders with mutations in these enzymes has raised the interest of the scientific community over the past several years. Most surprising has been the wide-ranging presentation of clinical manifestations in patients with mt-aaRS mutations, despite the enzymes’ common biochemical role. Even among cases where a common physiological system is affected, phenotypes, severity, and age of onset varies depending on which mt-aaRS is mutated. Here we review work done thus far and propose a categorization of diseases based on tissue specificity that highlights emerging patterns. We further discuss multiple in vitro and in cellulo efforts to characterize the behavior of wildtype and mutant mt-aaRSs that have shaped hypotheses about the molecular causes of these pathologies. Much remains to do in order to complete our understanding of these proteins. We expect that futher work is likely to result in the discovery of new roles for the mt-aaRSs in addition to their fundamental function in mitochondrial translation, informing the development of treatment strategies and diagnoses.
Article
Context Primary ovarian insufficiency (POI) encompasses a spectrum of premature menopause, including both primary and secondary amenorrhea. For 75-90% of individuals with hypergonadotropic hypogonadism (HH) presenting as POI, the molecular etiology is unknown. Common etiologies include chromosomal abnormalities, environmental factors, and congenital disorders affecting ovarian development and function, as well as syndromic and non-syndromic single gene disorders suggesting POI represents a complex trait. Objective To characterize the contribution of known disease genes to POI and identify novel molecular etiologies and biological underpinnings of POI. Design, Setting, and Participants We applied exome sequencing (ES) and family-based genomics to 42 affected female individuals from 36 unrelated Turkish families, including 31 with reported parental consanguinity. Results This analysis identified likely damaging, potentially contributing variants and molecular diagnoses in 16 families (44%), including 11 families with likely damaging variants in known genes and 5 families with predicted deleterious variants in novel disease genes (IGSF10, MND1, MRPS22, and SOHLH1). Of the 16 families, 2 (13%) had evidence for potentially pathogenic variants at more than one locus. Absence of heterozygosity (AOH) consistent with identity-by-descent mediated recessive disease burden contributes to molecular diagnosis in 15 of 16 (94%) families. GeneMatcher allowed identification of additional families from diverse genetic backgrounds. Conclusions ES analysis of a POI cohort further characterized locus heterogeneity, re-affirmed the association of genes integral to meiotic recombination, demonstrated the likely contribution of genes involved in hypothalamic development, and documented multi-locus pathogenic variation suggesting the potential for oligogenic inheritance contributing to the development of POI.
Article
Identifying genes and variants contributing to rare disease phenotypes and Mendelian conditions informs biology and medicine, yet potential phenotypic consequences for variation of >75% of the ~20,000 annotated genes in the human genome are lacking. Technical advances to assess rare variation genome-wide, particularly exome sequencing (ES), enabled establishment in the United States of the National Institutes of Health (NIH)-supported Centers for Mendelian Genomics (CMGs) and have facilitated collaborative studies resulting in novel “disease gene” discoveries. Pedigree-based genomic studies and rare variant analyses in families with suspected Mendelian conditions have led to the elucidation of hundreds of novel disease genes and highlighted the impact of de novo mutational events, somatic variation underlying nononcologic traits, incompletely penetrant alleles, phenotypes with high locus heterogeneity, and multilocus pathogenic variation. Herein, we highlight CMG collaborative discoveries that have contributed to understanding both rare and common diseases and discuss opportunities for future discovery in single-locus Mendelian disorder genomics. Phenotypic annotation of all human genes; development of bioinformatic tools and analytic methods; exploration of non-Mendelian modes of inheritance including reduced penetrance, multilocus variation, and oligogenic inheritance; construction of allelic series at a locus; enhanced data sharing worldwide; and integration with clinical genomics are explored. Realizing the full contribution of rare disease research to functional annotation of the human genome, and further illuminating human biology and health, will lay the foundation for the Precision Medicine Initiative.
Article
Mitochondria are essential for the viability of eukaryotic cells as they perform crucial functions in bioenergetics, metabolism and signalling and have been associated with numerous diseases. Recent functional and proteomic studies have revealed the remarkable complexity of mitochondrial protein organization. Protein machineries with diverse functions such as protein translocation, respiration, metabolite transport, protein quality control and the control of membrane architecture interact with each other in dynamic networks. In this Review, we discuss the emerging role of the mitochondrial protein import machinery as a key organizer of these mitochondrial protein networks. The preprotein translocases that reside on the mitochondrial membranes not only function during organelle biogenesis to deliver newly synthesized proteins to their final mitochondrial destination but also cooperate with numerous other mitochondrial protein complexes that perform a wide range of functions. Moreover, these protein networks form membrane contact sites, for example, with the endoplasmic reticulum, that are key for integration of mitochondria with cellular function, and defects in protein import can lead to diseases.
Article
The POLG gene encodes the mitochondrial DNA polymerase that is responsible for replication of the mitochondrial genome. Mutations in POLG can cause early childhood mitochondrial DNA (mtDNA) depletion syndromes or later-onset syndromes arising from mtDNA deletions. POLG mutations are the most common cause of inherited mitochondrial disorders, with as many as 2% of the population carrying these mutations. POLG-related disorders comprise a continuum of overlapping phenotypes with onset from infancy to late adulthood. The six leading disorders caused by POLG mutations are Alpers–Huttenlocher syndrome, which is one of the most severe phenotypes; childhood myocerebrohepatopathy spectrum, which presents within the first 3 years of life; myoclonic epilepsy myopathy sensory ataxia; ataxia neuropathy spectrum; autosomal recessive progressive external ophthalmoplegia; and autosomal dominant progressive external ophthalmoplegia. This Review describes the clinical features, pathophysiology, natural history and treatment of POLG-related disorders, focusing particularly on the neurological manifestations of these conditions.
Article
Premature ovarian insufficiency (POI) is a common endocrine disorder featured by the triad constituting of amenorrhea for at least four months, to date, the molecular pathogenesis of POI is largely undetermined. Despite several investigations have reported an increase in reactive oxygen species (ROS) content in idiopathic POI, the role of mitochondrial DNA (mtDNA) mutations/variants in the progression of POI has not been widely investigated. The current study aimed to explore the association between mt-tRNA mutations/variants and POI; we first used the PCR-Sanger sequencing to detect the mutations/variants in mt-tRNA genes from 50 POI patients and 30 healthy subjects. In addition, we evaluated the mitochondrial functions by using trans-mitochondrial cybrid cells containing these potential pathogenic mt-tRNA mutations. Consequently, five mutations: tRNALeu(UUR) C3303T, tRNAMet A4435G, tRNAGln T4363C, tRNACys G5821A and tRNAThr A15951G were identified. Notably, these mutations occurred at the extremely conserved nucleotides of the corresponding mt-tRNAs and may result the failure in mt-tRNA metabolism and subsequently lead to the impairment in mitochondrial protein synthesis. Furthermore, biochemical and molecular analyses of the cybrid cells containing these mutations showed a significantly lower level of ATP production when compared with the controls, whereas the ROS levels were much higher in POI patients carrying these mt-tRNA mutations, strongly indicated that these mt-tRNA mutations may cause the mitochondrial dysfunction, and play active roles in the progression and pathogensis of POI. Together, this study shaded additional light on the molecular mechanism of POI that was manifestated by mt-tRNA mutations.