ArticlePDF Available

Turner Syndrome

Authors:

Abstract and Figures

Turner syndrome (TS) affects approximately 1 out of every 1500–2500 live female births, with clinical features including short stature, premature ovarian failure, dysmorphic features and other endocrine, skeletal, cardiovascular, renal, gastrointestinal and neurodevelopmental organ system involvement. TS, a common genetic syndrome, is caused by sex chromosome aneuploidy, mosaicism or abnormalities with complete or partial loss of function of the second X chromosome. Advances in genetic and genomic testing have further elucidated other possible mechanisms that contribute to pathogenic variability in phenotypic expression that are not necessarily explained by monosomy or haploinsufficiency of the X chromosome alone. The role of epigenetics in variations of gene expression and how this knowledge can contribute to more individualized therapy is currently being explored. TS is established as a multisystemic condition, with several endocrine manifestations of TS affecting growth, puberty and fertility having significant impact on quality of life. Treatment guidelines are in place for the management of these conditions; however, further data on optimal management is needed.
Content may be subject to copyright.
Citation: Clemente, E.G.; Penukonda,
S.K.; Doan, T.; Sullivan, B.; Kanungo,
S. Turner Syndrome. Endocrines 2022,
3, 240–254. https://doi.org/10.3390/
endocrines3020022
Academic Editor:
Yukihiro Hasegawa
Received: 29 January 2022
Accepted: 4 May 2022
Published: 13 May 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Review
Turner Syndrome
Ethel Gonzales Clemente 1, *, Sasi Kiran Penukonda 2, Tam Doan 3, Brittany Sullivan 3and Shibani Kanungo 3
1Department of Pediatric Endocrinology, Valley Children’s Healthcare, 9300 Valley Children’s Place,
Madera, CA 93636, USA
2WK Children’s Health Specialists, 2032 Elizabeth Avenue, Shreveport, LA 71104, USA;
spenukonda@wkhs.com
3
Department of Pediatric & Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of
Medicine, 1000 Oakland Drive, Kalamazoo, MI 49008, USA; tam.doan@med.wmich.edu (T.D.);
brittany.sullivan@med.wmich.edu (B.S.); shibani.kanungo@med.wmich.edu (S.K.)
*Correspondence: eclemente@valleychildrens.org
Abstract:
Turner syndrome (TS) affects approximately 1 out of every 1500–2500 live female births,
with clinical features including short stature, premature ovarian failure, dysmorphic features and
other endocrine, skeletal, cardiovascular, renal, gastrointestinal and neurodevelopmental organ
system involvement. TS, a common genetic syndrome, is caused by sex chromosome aneuploidy,
mosaicism or abnormalities with complete or partial loss of function of the second X chromosome.
Advances in genetic and genomic testing have further elucidated other possible mechanisms that
contribute to pathogenic variability in phenotypic expression that are not necessarily explained by
monosomy or haploinsufficiency of the X chromosome alone. The role of epigenetics in variations of
gene expression and how this knowledge can contribute to more individualized therapy is currently
being explored. TS is established as a multisystemic condition, with several endocrine manifestations
of TS affecting growth, puberty and fertility having significant impact on quality of life. Treatment
guidelines are in place for the management of these conditions; however, further data on optimal
management is needed.
Keywords: Turner syndrome; epigenetics; growth; puberty
1. Introduction
Turner syndrome (TS) is a complex multisystem genetic condition, first described
in 1938 by an endocrinologist noting growth and congenital hypergonadotropic hypog-
onadism, with a reported prevalence of 1:2000 to 1:4000 among females [
1
,
2
]. It is a
heterogenous genetic disorder with 40–50% of patients having classic genotype 45,X and
the rest having a variety of mosaicisms [
2
,
3
]. There is a diverse range of phenotypic char-
acteristics, influenced by an individual’s karyotype. However, despite extensive research
showing the effects of haploinsufficiency of the X chromosome, exact mechanisms are
still not fully understood. More recent data support the role of altered gene expression
as a result of epigenetic mechanisms as contributory to the varied clinical manifestations
of TS [
4
], and is further discussed in this review. Furthermore, TS is associated with an
increased risk for a variety of health complications, and the particular interest of this review
are the endocrine-related manifestations and their subsequent management.
2. Etiology
2.1. Chromosomal Aberrations in TS
Turner Syndrome is considered to be the most common aneuploidy, although 99% of
congenital occurrences result in spontaneous abortion [
4
]. As with other aneuploidies, the
primary mechanism believed to contribute to TS is meiotic non-disjunction, a process which
leads to an unequal distribution of genetic material amongst daughter cells, such that some
Endocrines 2022,3, 240–254. https://doi.org/10.3390/endocrines3020022 https://www.mdpi.com/journal/endocrines
Endocrines 2022,3241
cells may have additional chromosomes, whereas others will have missing chromosomes
(Figure 1) [5].
Endocrines 2022, 3, FOR PEER REVIEW 2
that some cells may have additional chromosomes, whereas others will have missing chro-
mosomes (Figure 1) [5].
(a)
(b)
(c)
Figure 1. Comparison of (a) normal meiosis with (b) non-disjunction occurring in Meiosis I and (c)
non-disjunction occurring in Meiosis II.
Approximately 4050% of individuals with TS have a monosomy (45,X) karyotype
[2], meaning that all cells only have one X chromosome; 1525% have 45,X/46,XX mosai-
cism and another 1012% have 45,X/46,XY mosaicism, meaning that some cells only have
one X chromosome, whereas others have two X chromosomes or X and Y chromosomes
[2]. Interestingly, in mosaic individuals, the distribution of cells with only one X chromo-
some and cells with two sex chromosomes does not need to be split evenly [6].
Other structural abnormalities of the X chromosome (Table 1), such as deletions,
translocations, ring formation and isochromes contribute to mosaic forms [7]. Large, in-
verted repeats within the short arm (p) of X chromosome at the Xp11.2 locus have been
found to create a complex repetitive architecture that predisposes it to rearrangements
that correlate to the isodicentric X chromosome formation that is the second most common
chromosomal abnormality associated with TS [8]. Additionally, studies have shown that
the parental origin for the single complete X chromosome is maternal (Xm) in 90% of cases
[9]. The origin of the abnormal or missing chromosome is inconclusive at this point, with
some studies demonstrating increased likelihood of maternal origin (Xm) for pseudo
Figure 1.
Comparison of (
a
) normal meiosis with (
b
) non-disjunction occurring in Meiosis I and
(c) non-disjunction occurring in Meiosis II.
Approximately 40–50% of individuals with TS have a monosomy (45,X) karyotype [
2
],
meaning that all cells only have one X chromosome; 15–25% have 45,X/46,XX mosaicism
and another 10–12% have 45,X/46,XY mosaicism, meaning that some cells only have one
X chromosome, whereas others have two X chromosomes or X and Y chromosomes [
2
].
Interestingly, in mosaic individuals, the distribution of cells with only one X chromosome
and cells with two sex chromosomes does not need to be split evenly [6].
Other structural abnormalities of the X chromosome (Table 1), such as deletions,
translocations, ring formation and isochromes contribute to mosaic forms [
7
]. Large,
inverted repeats within the short arm (p) of X chromosome at the Xp11.2 locus have been
found to create a complex repetitive architecture that predisposes it to rearrangements that
correlate to the isodicentric X chromosome formation that is the second most common
chromosomal abnormality associated with TS [
8
]. Additionally, studies have shown that
the parental origin for the single complete X chromosome is maternal (X
m
) in 90% of
cases [
9
]. The origin of the abnormal or missing chromosome is inconclusive at this point,
with some studies demonstrating increased likelihood of maternal origin (X
m
) for pseudo
Endocrines 2022,3242
dicentric and deletions of long arm (q) of X chromosome (X
q
) [
10
], paternal in origin
(X
p
) for deletions of short arm (p) of X chromosome, and the extremely rare cases of
abnormal Y chromosomes, but equally likely to originate in either parent in the case of
either isochromosome Xq or Ring X abnormalities. Other studies, however, postulate that
the majority of Turner syndrome karyotypes are caused by paternal meiotic or mitotic
errors, leading to dominance of the maternal X chromosome [11].
Table 1. TS karyotypes and description of associated structural abnormalities.
TS Karyotype Structural Abnormalities Mechanism
45,X Complete loss of second sex chromosome Monosomy X
45,X/46,XY
some cells only have one X chromosome
whereas others have one X and one
Y chromosome
Mosaicism
45,X/46,XX some cells only have one X chromosome
whereas others have two X chromosomes Mosaicism
46,X,del(X)
(p11.4)
Deletion in the short arm at of one of the
X chromosomes Deletion
46,X,r(X) Ring-like structure with ends of a
chromosome fused
Telomeric aberration resulting
in ring chromosome
46,X,i(Xq)
Duplication of the long arm and loss of short
arm of one of the X chromosomes Isochromosome
The X chromosome is important not just for sex determination, but also contains
several genes responsible for growth and development during the embryonic period and
any aberration or abnormal defects can ultimately lead to pathogenesis of TS disease
conditions. The ‘Gene Dosage Effect’ hypothesis also postulates that TS features can be
mapped to particular regions of the X-chromosome that are gene dosage sensitive [
12
].
However, phenotypic variability in TS cannot be explained by genomic imbalance alone.
It is postulated that other processes such as X-chromosome inactivation and altered gene
expression as a result of epigenetic factors are also contributory [4,13].
2.2. Contributions of X-Inactivation to the TS Phenotype
The genes on the short arm of X chromosome which escape X-inactivation are im-
plicated in the TS phenotype [
14
,
15
]. X-inactivation is the process by which one of the X
chromosomes is silenced to equalize the gene dosages between male and female mammals,
resulting in one functional copy of the X chromosome on all somatic cells [
16
]. Under
normal conditions, an X inactivation epigenetic process randomly methylates one of the
two X chromosomes present in each cell, so that the genes from only one X chromosome are
actively expressed. However, certain genes in the pseudoautosomal region 1 (PAR1) and
pseudoautosomal region 2 (PAR2) of the X chromosome may escape X-inactivation and still
be expressed, even if these genes are on the silenced X chromosome [
17
]. The Y chromo-
some in males also contains genes found on PARs; hence, both 46,XX and 46,XY individuals
express two copies of the pseudoautosomal genes that escape X-inactivation in females [
17
].
In contrast, for TS, cells with only one X chromosome lack the second copy of these pseu-
doautosomal genes or other genes that would typically escape X-inactivation due to not
having a second sex chromosome present. This results in decreased expression of those
genes, which is termed haploinsufficiency [
6
]. The degree of haploinsufficiency involved in
TS depends on the karyotype; the 45,X karyotype involves greater haploinsufficiency than
mosaic karyotypes.
An example of the contribution of haploinsufficiency associated with the skewed
inactivation pattern resulting from Turner syndrome karyotype is with respect to the
previously identified short stature homeobox, or SHOX gene, which is located on Xp22.23,
and thus far is the only gene that has been compellingly associated with TS attributes. SHOX
is expressed in bone marrow fibroblasts, which can give rise to osteogenic genes that can
contribute to bone growth [
18
]. This gene’s function is dosage-dependent, with decreased
Endocrines 2022,3243
gene expression or haploinsufficiency leading to short stature and other features such
as Madelung deformity, high arched palate, scoliosis and micrognathia [
19
,
20
]. Another
gene located in PAR1 is the CSF2RA gene [
17
], which could be implicated in the high
rates of in utero deaths associated with TS [
21
]. One study has shown that expression of
various genes that are active in the placenta, including CSF2RA, was higher in wild-type
human embryonic stem cells than in cells that had spontaneously lost an X chromosome
(mimicking the 45,X karyotype) [
21
]. Although haploinsufficiency of the SHOX gene could
interfere with growth in TS patients, haploinsufficiency of CSF2RA could interfere with
proper placental function.
Other genes on the X chromosome besides those found in PARs can also escape X-
inactivation. For instance, TIMP1 usually escapes X-inactivation, so TS would involve
haploinsufficiency of TIMP1, potentially promoting the formation of a bicuspid aortic valve
(rather than a normal tricuspid aortic valve) [
22
]. Of note, certain variants of TIMP3, a gene
located on chromosome 22, are also associated with bicuspid aortic valve, and decreased
expression of TIMP1 can heighten risk for heart defects if the individual has these TIMP3
variants [
22
]. The relationship between TIMP1 and TIMP3 indicates that the inadequate
expression of X chromosome genes combined with the presence of certain autosomal genes
can bring about the clinical manifestations seen in TS.
2.3. Role of Epigenetics in TS
Epigenetic differences that influence gene expression without altering base sequences
exist between 45,X and 46,XX individuals, with extensive hypomethylation throughout 45,X
genome compared to 46,XX individuals, apart from differences in hypermethylation [
23
,
24
]
Epigenetic modification and resulting altered gene expression can contribute to various
pathogenesis seen in TS. For example, differentially methylated genes such as STAT4 and
KDM6A can affect T helper 1 (T
H
1) cell development and function [
13
], and T follicular helper
(T
FH
) cells development [
25
], respectively, and contribute to resultant inadequate immune
responses and autoimmunity in affected TS individuals. KDM6A, implicated in ovarian
dysfunction in TS [22,26], is also differentially expressed in Klinefelter syndrome [23,27].
Levels of microRNA (miRNA), which can bind to mRNA to block translation into
protein, can also differ between TS and 46,XX patients. For example, blood samples from
TS patients had lower amounts of miR-126-3p compared to controls [
28
]. Furthermore,
miR-126-3p levels were higher in TS patients who had abnormal aortic valves compared to
TS patients who did not; miR-126-3p can limit Bcl-2 expression and lead to dysregulated
apoptosis in the heart [
28
]. Fetal gonadal tissues with a 45,X karyotype have decreased
amounts of KITLG protein compared to tissues with a 46,XX karyotype, and the difference
was found to be mediated by levels of miR-320a. In 46,XX individuals, KDM5C blocks
expression of miR-320a, allowing the expression of KITLG, which supports ovarian devel-
opment. However, since KDM5C escapes X-inactivation, haploinsufficiency of KDM5C
allows for increased levels of miR-320a to be present in 45,X individuals, thus leading to
deficient KITLG expression and possibly impaired ovarian development [
29
]. Thus, this
demonstrates that differences in miRNA abundance can influence the clinical presentation
of TS. Another example is how PPARGC1A promoter DNA methylation status affects
Homeostasis Model Assessment for Insulin Resistance (HOMA-IR) independently of BMI
or age in TS subjects [30].
RPS4X and JPX are other escape genes that are also differentially expressed in TS;
however, their precise roles in TS pathogenesis are still to be elucidated [
31
33
]. The gene
IL3RA, which encodes for a subunit of the IL-3 receptor, is noted to be differentially methy-
lated in TS women. This gene has been linked to increased risk for autoimmune disorders
among these patients [
34
,
35
]. One study compared the transcriptomes (the collection of
mRNA expressed) of 45,X and 46,XX fibroblasts and revealed that the BMP2 and BMPER
genes were dysregulated in 45,X cells such that normal bone mineralization could be im-
paired [
28
]. Other interesting findings in 45,X cells from this transcriptome analysis include
downregulation of STC1 that could lead to dysregulated follicle development, downregu-
Endocrines 2022,3244
lation of IGF2 and ENPP1 that could lead to metabolic pathologies, and upregulation of
CLDN11, which is involved in spermatogenesis [
32
]. Notably, these six genes are all located
on autosomes, indicating that the genetic etiologies of TS manifestations extend beyond the
X chromosome.
Another role of epigenetics that has been explored is its involvement in X chromosome
monosomy. It is postulated that differential methylation of sex chromosomes could cause
errors in chromosome alignment and spindle formation during meiosis or mitosis [
4
].
Herrera et al. studied hypomethylated cells as caused by mutations in DNA methyltransferase
3b gene (DNMT3B), and found delayed centromere separation leading to aneuploidy [
36
].
Underlying pathogenesis mechanism in various TS phenotypic presentation is evolving
with improved understanding of notable difference in RNA expression, autosomal DNA
methylation and X chromosome methylation in TS patients.
3. Clinical Presentation
As alluded to earlier, the karyotype a TS individual possesses can influence which
TS features appear and how severely these features present. A study of 656 individuals
with TS in London found that 45,X patients were diagnosed at a younger age than patients
with other TS karyotypes [
6
]. This data suggests that the 45,X karyotype gives rise to more
serious or noticeable complications to warrant an earlier diagnosis. In the same study,
a significantly higher percentage of 45,X patients had primary amenorrhea compared to
45,X/46,XX, ring X, and isochromosome Xq patients, and a significantly lower percentage of
patients with 45,X/46,XX had hearing loss, hypertension, and obesity compared with 45,X
patients [
6
]. Another study involving adults from the UK Biobank revealed that 45,X/46,XX
and 46,XX individuals had a similar age of menarche, rate of pregnancy, and number
of pregnancies [
37
]. Furthermore, 45,X/46,XX individuals had a smaller height deficit
than 45,X [
37
], and a different study found that 45,X patients had more congenital heart
malformations than other TS patients [
38
]. The difference in phenotype between 45,X and
45,X/46,XX individuals could be explained by genes that escape X-inactivation. Monosomy
X is the most extreme form of haploinsufficiency, whereas 45,X/46,XX individuals have
some cell lines with normal levels of gene expression intact. These normal cell lines in
45,X/46,XX individuals can be protective.
Table 2lists some of the more common clinical features of TS. Clinical presentation
can be very varied, ranging from having all or most classical features to minimal or no
apparent symptoms or signs. Because of heterogeneity of clinical manifestations, TS may
be diagnosed at any age. Prenatal diagnosis has increased throughout the years because
of prenatal testing, particularly for at-risk mothers. Findings suggestive of TS include
chromosome abnormalities detected with chorionic villous sampling or amniocentesis;
increased nuchal translucency, presence of cystic hygroma, coarctation or evidence of left
sided cardiac defects, brachycephaly, renal anomalies, polyhydramnios, oligohydramnios
and growth retardation on ultrasound studies; and abnormal maternal triple or quadruple
testing [
2
]. The presence of lymphedema of the extremities, dysmorphic features, failure to
thrive and developmental delay in a female infant or young child should alert the medical
provider to proceed with further evaluation and testing for TS. However, the diagnosis
can be delayed until the patient presents with short stature later in childhood or delayed
pubertal development during the adolescent period.
Endocrines 2022,3245
Table 2. Clinical features of TS.
Associated Features and Conditions in TS
Characteristic facial and
physical features
Hypertelorism, upward slanting of palpebral fissures, epicanthal
folds, flat nasal bridge, low set ears, high arched palate, low posterior
hairline, webbed neck, broad chest with widely spaced nipples
Cardiovascular
Bicuspid aortic arch, coarctation of the aorta, aortic dilatation
or aneurysm
Hypertension
Endocrine and metabolic
Autoimmune thyroiditis, glucose intolerance/diabetes mellitus,
dyslipidemia, decreased bone mineral density
Gynecologic Absent or delayed puberty development, premature ovarian
failure, infertility
Skeletal
Short stature, delayed bone maturation, spine deformities (scoliosis,
kyphosis), angular deformity of limbs (cubitus valgus, genu valgum,
madelung deformity)
Gastrointestinal Celiac disease, transaminitis, inflammatory bowel disease
Renal
Horseshoe kidney, renal ectopia, abnormal position or duplication of
ureters or vessels
Neurocognitive and
behavioral
Impaired visuospatial and perceptive abilities, deficits in non-verbal
memory and executive function
Increased risk for attention-deficit/hyperactivity disorder
4. Endocrine-Related Manifestations of Turner Syndrome
4.1. Growth and Short Stature
The short stature in TS is attributed to haploinsufficiency of SHOX gene which regu-
lates differentiation of chondrocytes. This also explains other features noted in TS including
high arched palate, obstructive sleep apnea, prominent ears, and chronic otitis media [
39
].
Although individuals with TS do not have true growth hormone deficiency, they respond
to growth hormone as seen in those with isolated SHOX gene deficiency [
40
], and can have
a mean height gain of about 7 cm; however, this is still dependent on age when related to
age of initiation and duration of treatment [
41
]. Results of a French observational multicen-
ter study published in 2016 demonstrated association between karyotype subgroup and
phenotypic variation in spontaneous intrauterine and postnatal growth and adult height
after GH therapy [
42
]. The authors concluded that haploinsufficiency of unknown Xp gene
increases the risk of deficit in prenatal and postnatal growth and short adult height after
GH treatment [42].
4.2. Ovarian Insufficiency
Ovarian insufficiency resulting from rapid and progressive loss of oocytes is another
feature of the syndrome in almost all TS individuals, presenting as absent or delayed
pubertal development in adolescent girls, or infertility in women of childbearing age. This
accelerated degeneration of oocytes is attributed to failure of chromosome pairing in the
early stage of meiotic prophase [
12
]. Spontaneous pubertal development is more common
in mosaics, occurring in about a third of this population; however, only a smaller percentage
of this group will continue to progress to the occurrence of menarche [
43
]. Consistent with
the variability of clinical presentation in TS, there is also significant differences in the size
primordial follicle pool. This explains how mosaics with a large enough pool may undergo
spontaneous puberty and menarche [
43
]; however, they will still have a faster rate of follicle
apoptosis than women with normal 46,XX [
44
]. Anti-Mullerian hormone (AMH) is a good
marker of ovarian reserve of growing follicles and Visser et al., found that AMH levels
corelate with karyotype of TS with measurable levels found in 77% of TS patients with
45,X/4XX mosaicism and in only 10% with classic 45,X karyotype [45].
Endocrines 2022,3246
4.3. Autoimmune Thyroiditis
Individuals with TS are at increased risk of developing autoimmune diseases, par-
ticularly the autoimmune thyroid diseases (ATD). As per the recent meta-analysis report
the overall prevalence of ATD in TS is 38.6% [
46
]. About 41–45% of individuals with TS
are found to have thyroid peroxidase antibodies [
47
,
48
], with prevalence of Hashimoto’s
disease (HD) is 30–50% [
48
,
49
]. Graves’ disease (GD), which is rarer than HD, still occurs
more frequently in TS compared with general population with an estimated prevalence of
1.7–3% [
50
,
51
] Although ATD is observed in all TS karyotypes, several studies have shown
that autoimmune disorders including ATD are more likely to occur in those with isolated
Xp deletion and isochromosome Xq [
47
,
52
54
]. Both these chromosomal anomalies lack
the short arm of X chromosome and the haploinsufficiency of immune regulatory genes
located in the Xter-p11.2 region is the most likely explanation for increased predisposition
to autoimmune disorders [
54
]. Other proposed mechanisms for the cause of increased
frequency of autoimmune disorders in TS include: maternal origin of X chromosome,
hypogonadism and cytokine imbalance with more pro-inflammatory cytokines and less
anti-inflammatory cytokines [55].
4.4. Diabetes and Metabolic Syndrome
The patients with TS are more likely to develop either type 1 or type 2 diabetes mellitus
(DM) with a relative risk of 11.56 and 4.38, respectively [
56
]. The increased risk for Type 1
diabetes is explained by the overall higher risk for development of autoimmune conditions
in TS, as previously discussed.
The associated risk for developing metabolic syndrome including Type 2 diabetes
in TS involves a more complicated pathophysiologic process, with several contributory
factors. The features of metabolic syndrome observed in patients with TS include Increased
abdominal adiposity, impaired vascular function and hypertension, dyslipidemia and
insulin resistance [
57
]. Lebenthal et al. looked at the evolution of metabolic comorbidities
in TS, with the longitudinal and cross-sectional retrospective study that showed that
increasing age and weight gain increase metabolic risks in this cohort of 98 TS patients [
58
].
Similar to previous studies, these cardiometabolic risk factors are already apparent during
childhood, and in a good number of patients who have normal BMI at this age [
58
60
].
X chromosome gene dosage is thought to influence the occurrence of metabolic disorders
in TS, with reports of clustering of risk factors in 45,X monosomy [3,61].
Individuals with TS were noted to have higher body mass index (BMI) and higher
waist circumference when compared with BMI matched women without TS [
62
]. As many
as 20–40% of youth and 60% of adult individuals with TS have systemic hypertension,
which may be due to renal anomalies, or idiopathic [
2
]. An atherogenic lipid profile of high
triglycerides, high low-density lipoprotein (LDL) and low high-density lipoprotein (HDL)
is also observed in some studies, with hypercholesterolemia being reported in 37–50% of
patients with TS [
2
,
61
,
63
]. Altered glucose and insulin metabolism in TS is most likely
due to decreased beta-cell responsiveness with diminished first-phase insulin release [64].
Haploid gene deficiency of PAR1 is believed to result into altered expression of molecules
that the gene encodes for, which include certain types of receptors, transcription factors,
phospholipase and protein lipases involved in appropriate insulin response [
65
]. Another
genetic mechanism explored is the association of the long arm of the X chromosome (iXq)
with higher incidence of DM, with additional copies of Xq triggering overexpression of
diabetes-related genes such as islet cell antigen (ICA) and insulin-like growth factor II [
66
].
This overexpression is then linked to immune-mediated injury to beta-cells, leading to
decreased responsiveness.
5. Diagnosis—Karyotyping and Beyond
A standard 20-cell karyotype using the peripheral blood sample is recommended for
all girls and women suspected with TS. Sampling of another tissue such as buccal mucosa
cells or skin fibroblasts may be needed if the standard karyotype is reported normal and
Endocrines 2022,3247
there is strong clinical suspicion of TS. [
2
,
67
]. If the diagnosis was made prenatally with
chromosomal analysis using chronic villous sampling or amniocentesis, it is recommended
to repeat the karyotyping postnatally [2].
Presence of Y chromosome material suggests increased risk of gonadoblastoma and
germ cell tumors [
68
]. In patients with virilizing features, fluorescent in situ hybridization
(FISH) analysis of at least two to three different tissues using X and Y probes is recom-
mended to detect cryptic Y material [
2
,
67
]. Additional testing has also been recommended
for patients with 45,X karyotype, as true sex chromosome monosomy is incompatible with
life and these patients may have cryptic mosaicism potentially with Y chromosome mate-
rial [
69
]. FISH using a probe to DYZ3 locus at Y-centromere is suggested to detect cryptic
mosaicism for Y chromosome as this region is associated with increased susceptibility
to gonadoblastoma [69].
The polymerase chain reaction test using multiple Y-chromosome-specific DNA probes
is more sensitive than FISH [
70
,
71
]; however, it may be susceptible to contamination [
72
].
Non-invasive prenatal screening tests such as cell-free fetal DNA screening in maternal
plasma by microarray or whole genome sequencing helps to detect aneuploidy, but both
methods have limitations such as failure to identify balanced translocations and triploidies,
or missing X-chromosome structural abnormalities and mosaicism, respectively [
73
,
74
].
Single nucleotide polymorphism (SNP) array genotyping has been compared with kary-
otyping in patients with TS and it was found that SNP array can better detect cryptic
Y chromosome; however, this may cause misinterpretation of rare cell lines and cannot
detect fully balanced translocations of the X chromosome [75].
Further advancements in genetic testing during the past decade have allowed for
understanding genomic mechanisms of TS, elucidating further on what affects growth,
puberty, neurocognitive development and occurrence of associated conditions in these
patients. Advances in epigenetic research has seen the use of assays that started with DNA
methylation and histone acetylation studies that were site-specific, to genome-wide assess-
ments [
76
]. More recently, bioinformatics analysis has been used to identify differentially
expressed genes that may further increase knowledge on the pathogenesis of TS [
77
,
78
].
While newer gene editing technological advances such as Crispr/Cas9 can offer potential
therapeutic options, their use in human disease can elicit multitude of ethical issues and
such discussion is beyond the scope of this review.
6. Treatment and Management
6.1. Growth Hormone Therapy
Growth hormone (GH) treatment started at an early age of 4–6 years in patients with
TS helps to achieve normal growth pattern similar to that for peers of the same age [
2
]. The
mean average height of TS patients untreated with GH is cited to be around
142–144 cm
,
which is approximately 20 cm less than the mean height of general population [
79
]. When
treated with GH, the annual height gain of TS patients increased by 1–2 cm for every year
of GH therapy [
2
,
79
]. Besides promoting growth, GH augments bone mass, regulates lipid
and glucose metabolism and increases amino acid transport in the muscle [
79
]. Long-
term therapy is also associated with positive effects on craniofacial development in TS,
mostly affecting mandibular ramus and posterior facial height [
80
]. A recent systematic
review looking at effects of GH on the cardiovascular system showed positive effect on
lipid profile, reducing the risk of cardiovascular disease, particularly if with concomitant
estradiol therapy [81].
Several factors affect success of growth hormone therapy. Dose, duration, as well as
adherence and compliance to therapy all influence final adult height [
79
]. The “Toddler
Turner” study, which looked at effects of early initiation of GH therapy beginning at
9 months to 4 years of age, revealed that the early treated group were taller all through out,
but with no significant difference in near-adult height when compared with early untreated
group. This was attributed to the catch down growth noted in the early treated group
during lapses of GH therapy, demonstrating the significance of uninterrupted treatment to
Endocrines 2022,3248
promote better adult height [
82
]. Later initiation not only limits adult height predictions but
may delay growth associated with puberty as well. The presence of other health conditions
such as congenital heart disease, hypothyroidism or celiac disease may contribute to growth
deficits, independent of growth hormone effects [
79
]. Other non-modifiable factors include
height of parents and height of the patient at the beginning of GH therapy. Specific genetic
markers have also been linked with response to GH therapy in TS, such as estrogen receptor
alpha (ESR1) and tyrosine-protein phosphatase nonreceptor type1 (PTPN1) both noted to
influence height velocity in TS [
83
], whereas homozygosity for SOCS-2, GHR exon3 full
length and IGFBP3-202 C alleles was associated with poor response to GH therapy [84].
The initial dose of GH is 0.35–0.375 mg/kg/week and patients with poor height
prognosis may be started on higher doses after careful consideration [
2
]. Concomitant
treatment with Oxandrolone is an option to improve final height if there is delay in initiation
of GH therapy due to delayed diagnosis. GH therapy along with Oxandrolone at a dose of
0.03–0.05 mg/kg/day in TS patients 10 years and older results in gain of final height by
2–5 cm as compared to those treated with GH alone [
2
]. Although Oxandrolone can have
synergistic effect on growth acceleration, it is associated with undesirable effects including
virilization and delayed pubertal development [
85
]. These side effects are modest when
treated with doses less than 0.06 mg/kg/day [86].
6.2. Estrogen Replacement
The goal of estrogen replacement therapy in TS is to induce and maintain normal
pubertal development with secondary sexual characteristics including normal breasts and
uterine size and shape. The other goals include achievement of physiological effects of
endogenous estrogens such as bone mineralization and maintenance of cardiovascular
health. The negative effects of estrogen deficiency in TS include poor intrauterine growth,
decreased cognitive and motor reaction time, reduced bone mass, poor cardiovascular
outcomes, low self-esteem and poor quality of life [
87
,
88
]. In a study by Viuff et al., hormone
replace therapy is found to reduce endocrine and cardiovascular morbidity in TS adults
with decreased use of antidiabetics, thyroid hormone replacement and antihypertensives
and reduced hospitalizations due to osteoporotic fractures and stroke [
89
]. Other studies
show that earlier induction of puberty and start of estrogen replacement may be beneficial
for adult bone density [90].
Current recommendations call for starting estrogen replacement at around age
11–12 years
,
with transdermal 17-
β
estradiol being the preferred treatment [
2
,
88
]. Monitoring go-
nadotropins, particularly follicle stimulating hormone (FSH), starting at about 11 years of
age, aid in detecting and confirming hypergonadotropic hypogonadism before puberty
induction [
2
]. Low levels of anti-Mullerian hormone (AMH) can also suggest ovarian
failure [
45
,
89
,
91
]. To mimic the normal physiologic milieu during the peripubertal period,
initiation with low doses of estrogen (3–7
µ
g/kg/day) is recommended, gradually increas-
ing every 6 months, eventually reaching adult doses of up to 100 mg/day in 2–3 years [2].
Ultra-low dose estrogen therapy using oral ethinyl estradiol during the prepubertal period
has been suggested, with studies showing normalization of onset and tempo of puberty
development, as well as improvements in cognition and memory [
92
94
]. However, routine
use of this therapy is not recommended due to lack of long-term safety data [2].
As mentioned, transdermal Estradiol (E
2
) is the more widely used preparation that
theoretically provides a more physiologic, systemic route of delivery. Orally administered
estrogen, on the other hand, reaches the systemic circulation after undergoing metabolism
in the liver. A randomized clinical trial by Torres-Santiago et al. demonstrated no significant
differences between the TS patients treated with oral and transdermal E2 regarding their
body composition, bone mineral density and lipid profile when their estradiol levels
were titrated to those of normal menstruating adolescents [
95
]. However, the increased
amounts of conjugated estrogen precursors and metabolites associated with oral estrogen
preparations pose a higher risk for thromboembolic events as seen in the post-menopausal
women [
96
]. To date, there are no data available to suggest the same risk in TS population, as
Endocrines 2022,3249
long-term studies assessing the optimal dose, route and duration of hormone replacement
treatment are still to be published.
TS individuals usually have normal uterus, and it is recommended to add progestin
therapy 2 years after induction of puberty or once spotting or menstrual bleeding has
commenced [
2
]. Progestins minimize risk of endometrial hyperplasia due to unopposed
estrogen therapy and thereby prevent endometrial cancer [
97
]. There are several proposed
progestin and estrogen/progestin replacement options after puberty induction [
88
]. After
establishing adult dosing for hormone replacement therapy, it needs to be continued until
the usual age of menopause. These patients need to be monitored for risks associated with
estrogen therapy on an annual basis.
6.3. Fertility Preservation
Premature ovarian failure results in infertility in majority of patients with TS with
spontaneous pregnancy reported only in 2–10% [
98
]. In those with mosaicism (45X/46XX)
salvage of existing oocytes using assisted reproductive technologies was proposed [
99
].
Oocyte retrieval and cryopreservation after ovarian stimulation in adults and post pubertal
adolescents with Turner mosaicism has been reported [
100
102
]. Ovarian stimulation was
initiated with FSH (recombinant or highly purified) along with LH supplementation in
the form of human menopausal gonadotropins or recombinant LH due to concerns of
hypothalamic immaturity in post pubertal adolescents with TS [101].
Ovarian tissue cryopreservation which is still experimental fertility preservation tech-
nique may be an option in prepubertal patients with TS who are at increased risk of
accelerated ovarian failure based declining AMH levels (<2 ng/mL) and cannot wait until
sufficient maturity to undergo oocyte cryopreservation [
99
]. Though fertility preservation
is no longer consider experimental in adult patients, ovarian stimulation for oocyte cry-
opreservation and ovarian tissue cryopreservation in young patients needs consent from
parents and patients above nine years of age and also approval of institutional review
board (IRB) is strongly recommended [
99
]. More recently, another approach for fertility
preservation is suggested, primarily based on the patient’s genotype (monosomy vs. mo-
saic), then subsequently based on AMH concentrations over time [
103
]. This approach
also takes into strong consideration the expected maternal risks that may vary significantly
from person to person. Prenatal genetic counseling plays an important role in prenatal
diagnostic procedures in all pregnant women and in future reproductive options such as
in vitro fertilization for TS adults.
7. Conclusions
Scientific advancements have led to improved diagnostic and management options
in the care of patients with TS. Still there is more to discover how the impact of more
detailed genetic and genomic testing could affect health outcomes, help decrease health
burdens, and ultimately improve quality of life. Although clinical manifestations could
be highly variable, more often the condition affects multiple organ systems, thus needing
multidisciplinary team approach to ensure optimal outcomes.
Author Contributions:
Conceptualization: E.G.C. and S.K.; Administrative support: E.G.C. and S.K.;
Provision of study materials or patients: E.G.C. and S.K.; Manuscript writing: E.G.C., S.K.P., T.D., B.S.
and S.K. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict of interest.
Endocrines 2022,3250
References
1.
Kansra, A.; Donohue, P. Hypofunction of the Ovaries. In Nelson Textbook of Pediatrics, 21st ed.; Kliegman, R.M., St. Geme, J., Eds.;
Elsevier Inc.: Philadelphia, PA, USA, 2020; pp. 3001–3007.
2.
Gravholt, C.H.; Andersen, N.H.; Conway, G.S.; Dekkers, O.M.; Geffner, M.E.; Klein, K.O.; Lin, A.E.; Mauras, N.; Quigley, C.A.;
Rugin, K.; et al. Clinical practice guidelines for the care of girls and women with Turner syndrome: Proceedings from the 2016
Cincinnati International Turner Syndrome Meeting. Eur. J. Endocrinol. 2017,177, G1–G70. [CrossRef]
3.
El-Mansoury, M.; Barrenäs, M.L.; Bryman, I.; Hanson, C.; Larsson, C.; Wilhelmsen, L.; Landin-Wilhelmsen, K. Chromosomal
mosaicism mitigates stigmata and cardiovascular risk factors in Turner syndrome. Clin. Endocrinol.
2007
,66, 744–751. [CrossRef]
4. Álvarez-Nava, F.; Lanes, R. Epigenetics in Turner syndrome. Clin. Epigenetics 2018,10, 45. [CrossRef]
5.
Scott, S.A.; Cohen, N.; Brandt, T.; Warburton, P.E.; Edelmann, L. Large inverted repeats within Xp11.2 are present at the
breakpoints of isodicentric X chromosomes in Turner syndrome. Hum. Mol. Genet. 2010,19, 3383–3393. [CrossRef]
6.
Cameron-Pimblett, A.; La Rosa, C.; King, T.F.J.; Davies, M.C.; Conway, G.S. The Turner syndrome life course project: Karyotype-
phenotype analyses across the lifespan. Clin. Endocrinol. 2017,87, 532–538. [CrossRef]
7.
Catovi´c, A. Cytogenetics findings at Turner Syndrome and their correlation with clinical findings. Bosn. J. Basic Med. Sci.
2005
,5,
54–58. [CrossRef]
8.
Monroy, N.; López, M.; Cervantes, A.; Garcia-Cruz, D.; Zafra, G.; Canun, S.; Zenteno, J.C.; Kofman-Alfaro, S. Microsatellite
analysis in Turner syndrome: Parental origin of X chromosomes and possible mechanism of formation of abnormal chromosomes.
Am. J. Med. Genet. 2002,107, 181–189. [CrossRef]
9.
Gottlieb, S.F.; Tupper, C.; Kerndt, C.C.; Tegay, D.H. Genetics, Nondisjunction. In StatPearls; StatPearls Publishing: Treasure Island,
FL, USA, 2022.
10.
Jacobs, P.; Dalton, P.; James, R.; Mosse, K.; Power, M.; Robinson, D.; Skuse, D. Turner syndrome: A cytogenetic and molecular
study. Ann. Hum. Genet. 1997,61 Pt 6, 471–483. [CrossRef]
11.
Uematsu, A.; Yorifuji, T.; Muroi, J.; Kawai, M.; Mamada, M.; Kaji, M.; Yamanaka, C.; Momoi, T.; Nakahata, T. Parental origin
of normal X chromosomes in Turner syndrome patients with various karyotypes: Implications for the mechanism leading to
generation of a 45,X karyotype. Am. J. Med. Genet. 2002,111, 134–139. [CrossRef]
12.
Ogata, T.; Matsuo, N. Turner syndrome and female sex chromosome aberrations: Deduction of the principal factors involved in
the development of clinical features. Hum. Genet. 1995,95, 607–629. [CrossRef]
13.
Nielsen, M.M.; Trolle, C.; Vang, S.; Hornshøj, H.; Skakkebæk, A.; Hedegaard, J.; Nordentoft, I.; Pederson, J.S.; Gravholt,
C.H. Epigenetic and transcriptomic consequences of excess X-chromosome material in 47,XXX syndrome-A comparison with
Turner syndrome and 46,XX females. Am. J. Med. Genet. C Semin. Med. Genet.
2020
,184, 279. Available online: https:
//pubmed.ncbi.nlm.nih.gov/32489015/ (accessed on 2 April 2022). [CrossRef]
14.
Bondy, C.A.; Cheng, C. Monosomy for the X chromosome. Chromosome Res.
2009
,17, 649. Available online: https://pubmed.ncbi.
nlm.nih.gov/19802705/ (accessed on 2 April 2022). [CrossRef]
15.
Gravholt, C.H.; Viuff, M.H.; Brun, S.; Stochholm, K.; Andersen, N.H. Turner syndrome: Mechanisms and management. Nat. Rev.
Endocrinol. 2019,15, 601–614. [CrossRef]
16.
Disteche, C.; Berletch, J. X-chromosome inactivation and escape. J. Genet.
2015
,94, 591. Available online: https://pubmed.ncbi.
nlm.nih.gov/26690513/ (accessed on 2 April 2022). [CrossRef]
17. Ross, M.T.; Grafham, D.V.; Coffey, A.J.; Scherer, S.; McLay, K.; Muzny, D.; Platzer, M.; Howell, G.R.; Burrows, C.; Bird, C.P.; et al.
The DNA sequence of the human X chromosome. Nature 2005,434, 325–337. [CrossRef]
18.
Ellison, J.W.; Wardak, Z.; Young, M.F.; Robey, P.G.; Laig-Webster, M.; Chiong, W. PHOG, a Candidate Gene for Involvement in the
Short Stature of Turner Syndrome. Hum. Mol. Genet. 1997,6, 1341–1347. [CrossRef]
19.
Clement-Jones, M.; Schiller, S.; Rao, E.; Blaschke, R.J.; Zuniga, A.; Zeller, R.; Robson, S.C.; Binder, G.; Glass, I.; Strachan, T.; et al.
The short stature homeobox gene SHOX is involved in skeletal abnormalities in Turner syndrome. Hum. Mol. Genet.
2000
,9,
695–702. [CrossRef]
20.
Fukami, M.; Seki, A.; Ogata, T. SHOX Haploinsufficiency as a Cause of Syndromic and Nonsyndromic Short Stature. Mol.
Syndromol. 2016,7, 3–11. [CrossRef]
21.
Urbach, A.; Benvenisty, N. Studying Early Lethality of 45,XO (Turner’s Syndrome) Embryos Using Human Embryonic Stem Cells.
PLoS ONE 2009,4, e4175. [CrossRef]
22.
Corbitt, H.; Gutierrez, J.; Silberbach, M.; Maslen, C.L. The genetic basis of Turner syndrome aortopathy. Am. J. Med. Genet. C
Semin. Med. Genet. 2019,181, 117–125. [CrossRef]
23.
Trolle, C.; Nielsen, M.M.; Skakkebæk, A.; Lamy, P.; Vang, S.; Hedegaard, J.; Nordentoft, I.; Orntoft, T.F.; Pedersen, J.S.; Gravholt,
C.H. Widespread DNA hypomethylation and differential gene expression in Turner syndrome. Sci. Rep.
2016
,6, 34220. Available
online: https://pubmed.ncbi.nlm.nih.gov/27687697/ (accessed on 2 April 2022). [CrossRef]
24.
Sharma, A.; Jamil, M.A.; Nuesgen, N.; Schreiner, F.; Priebe, L.; Hoffmann, P.; Herns, P.; Nothen, M.M.; Frohlich, H.; Oldenburg, J.;
et al. DNA methylation signature in peripheral blood reveals distinct characteristics of human X chromosome numerical aberra-
tions. Clin. Epigenetics
2015
,7, 76. Available online: https://pubmed.ncbi.nlm.nih.gov/26221191/ (accessed on 2 April 2022).
[CrossRef]
Endocrines 2022,3251
25.
Cook, K.D.; Shpargel, K.B.; Starmer, J.; Whitfield-Larry, F.; Conley, B.; Allard, D.E.; Rager, J.E.; Fry, R.C.; Davvenport, M.;
Magnuson, T.; et al. T Follicular Helper Cell-Dependent Clearance of a Persistent Virus Infection Requires T Cell Expression of
the Histone Demethylase UTX. Immunity 2015,43, 703–714. [CrossRef]
26.
Berletch, J.B.; Deng, X.; Nguyen, D.K.; Disteche, C.M. Female bias in Rhox6 and 9 regulation by the histone demethylase KDM6A.
PLoS Genet. 2013,9, e1003489. [CrossRef]
27.
Skakkebæk, A.; Nielsen, M.M.; Trolle, C.; Vang, S.; Hornshoj, H.; Hedegaard, J.; Wallentin, M.; Bojesen, A.; Hertz, J.M.; Fedder,
J.; et al. DNA hypermethylation and differential gene expression associated with Klinefelter syndrome. Sci Rep.
2018
,8, 13740.
[CrossRef]
28.
Abu-Halima, M.; Oberhoffer, F.S.; El Rahman, M.A.; Jung, A.M.; Zemlin, M.; Rohrer, T.R.; Kahraman, M.; Keller, A.; Meese, E.;
Abdul-Khaliq, H. Insights from circulating microRNAs in cardiovascular entities in turner syndrome patients. PLoS ONE
2020
,
15, e0231402. Available online: https://pubmed.ncbi.nlm.nih.gov/32271829/ (accessed on 2 April 2022). [CrossRef]
29.
Sun, Y.X.; Zhang, Y.X.; Zhang, D.; Xu, C.M.; Chen,S.C.; Zhang, J.Y.; Ruan, Y.C.; Chen, F.; Zhang, R.J.; Qian, Y.Q.; et al. XCI-escaping
gene KDM5C contributes to ovarian development via downregulating miR-320a. Hum. Genet. 2017,136, 227–239. [CrossRef]
30.
Álvarez-Nava, F.; Salinas, M.; Bastidas, D.; Vicuña, Y.; Racines-Orbe, M. PPARGC1A promoter DNA-methylation level and
glucose metabolism in Ecuadorian women with Turner syndrome. Horm. Mol. Biol. Clin. Investig. 2020,42, 159–165. [CrossRef]
31.
Rajpathak, S.N.; Vellarikkal, S.K.; Patowary, A.; Scaria, V.; Sivasubbu, S.; Deobagkar, D.D. Human 45,X fibroblast transcriptome
reveals distinct differentially expressed genes including long noncoding RNAs potentially associated with the pathophysiology
of Turner syndrome. PLoS ONE 2014,9, e100076. [CrossRef]
32.
Zhang, R.; Hao, L.; Wang, L.; Chen, M.; Li, W.; Li, R.; Yu, J.; Xiao, J.; Wu, J. Gene expression analysis of induced pluripotent stem
cells from aneuploid chromosomal syndromes. BMC Genom. 2013,14 (Suppl. 5), S8. [CrossRef]
33.
Bellott, D.W.; Hughes, J.F.; Skaletsky, H.; Brown, L.G.; Pyntikova, T.; Cho, T.J.; Koutseva, N.; Zaghul, S.; Graves, T.; Rock, S.; et al.
Mammalian Y chromosomes retain widely expressed dosage-sensitive regulators. Nature 2014,508, 494–499. [CrossRef]
34.
Lleo, A.; Moroni, L.; Caliari, L.; Invernizzi, P. Autoimmunity and Turner’s syndrome. Autoimmun. Rev.
2012
,11, A538–A543.
[CrossRef]
35.
Berglund, A.; Cleemann, L.; Oftedal, B.E.; Holm, K.; Husebye, E.S.; Gravholt, C.H. 21-hydroxylase autoantibodies are more
prevalent in Turner syndrome but without an association to the autoimmune polyendocrine syndrome type I. Clin. Exp. Immunol.
2019,195, 364–368. [CrossRef]
36.
Herrera, L.A.; Prada, D.; Andonegui, M.A.; Dueñas-González, A. The epigenetic origin of aneuploidy. Curr. Genom.
2008
,9, 43–50.
[CrossRef]
37.
Tuke, M.A.; Ruth, K.S.; Wood, A.R.; Beaumont, R.N.; Tyrrell, J.; Jones, S.E.; Yaghootkar, H.; Turner, C.L.S.; Donohoe, M.E.; Brooke,
A.M.; et al. Mosaic Turner syndrome shows reduced penetrance in an adult population study. Genet. Med.
2019
,21, 877–886.
[CrossRef]
38.
Fiot, E.; Zénaty, D.; Boizeau, P.; Haignere, J.; Dos Santos, S.; Leger, J.; French Turner Syndrome Study Group. X chromosome gene
dosage as a determinant of congenital malformations and of age-related comorbidity risk in patients with Turner syndrome, from
childhood to early adulthood. Eur. J. Endocrinol. 2019,180, 397–406. [CrossRef]
39. Davenport, M.L. Approach to the patient with Turner syndrome. J. Clin. Endocrinol. Metab. 2010,95, 1487–1495. [CrossRef]
40.
Blum, W.F.; Ross, J.L.; Zimmermann, A.G.; Quigley, C.A.; Child, C.J.; Kalifa, G.; Deal, C.; Drop, S.L.S.; Rappold, G.; Butler, G.B., Jr.
GH treatment to final height produces similar height gains in patients with SHOX deficiency and turner syndrome: Results of a
multicenter trial. J. Clin. Endocrinol. Metab. 2013,98, 1383–1392. [CrossRef]
41.
Soriano-Guillen, L.; Coste, J.; Ecosse, E.; Leger, J.; Tauber, M.; Cabrol, S.; Nicolino, M.; Chaussain, J.L.; Carel, J.C. Adult height
and pubertal growth in Turner syndrome after treatment with recombinant growth hormone. J. Clin. Endocrinol. Metab.
2005
,90,
5197–5204. [CrossRef]
42.
Fiot, E.; Zenaty, D.; Boizeau, P.; Haignere, J.; Dos Santos, S.; Leger, J.; French Turner Syndrome Study Group. X-chromosome gene
dosage as a determinant of impaired pre and postnatal growth and adult height in Turner syndrome. Eur. J. Endocrinol.
2016
,174,
281–288. [CrossRef]
43.
Pasquino, A.M.; Passeri, F.; Pucarelli, I.; Segni, M.; Municchi, G. Spontaneous pubertal development in Turner’s syndrome. J. Clin.
Endocrinol. Metab. 1997,82, 1810–1813. [CrossRef]
44.
Reindollar, R.H. Turner syndrome: Contemporary thoughts and reproductive issues. Semin. Reprod. Med.
2011
,29, 342–352.
[CrossRef]
45.
Visser, J.A.; Hokken-Koelega, A.C.S.; Zandwijken, G.R.J.; Limacher, A.; Ranke, M.B.; Flück, C.E. Anti-Müllerian hormone levels in
girls and adolescents with Turner syndrome are related to karyotype, pubertal development and growth hormone treatment.
Hum. Reprod. 2013,28, 1899–1907. [CrossRef]
46.
Mohamed, S.O.O.; Elkhidir, I.H.E.; Abuzied, A.I.H.; Noureddin, A.A.M.H.; Ibrahim, G.A.A.; Mohmoud, A.A.A. Prevalence of
autoimmune thyroid diseases among the Turner Syndrome patients: Meta-analysis of cross-sectional studies. BMC Res. Notes
2018,11, 842. [CrossRef]
47.
Elsheikh, M.; Wass, J.A.H.; Conway, G.S. Autoimmune thyroid syndrome in women with Turner’s syndrome—The association
with karyotype. Clin. Endocrinol. 2001,55, 223–226. [CrossRef]
48.
Mortensen, K.H.; Cleemann, L.; Hjerrild, B.E.; Nexo, E.; Locht, H.; Jeppesen, E.M.; Gravholt, C.H. Increased prevalence of
autoimmunity in Turner syndrome—Influence of age. Clin. Exp. Immunol. 2009,156, 205–210. [CrossRef]
Endocrines 2022,3252
49.
El-Mansoury, M.; Bryman, I.; Berntorp, K.; Hanson, C.; Wilhelmsen, L.; Landin-Wilhelmsen, K. Hypothyroidism is common in
turner syndrome: Results of a five-year follow-up. J. Clin. Endocrinol. Metab. 2005,90, 2131–2135. [CrossRef]
50.
Livadas, S.; Xekouki, P.; Fouka, F.; Kanaka-Gantenbein, C.; Kaloumenou, I.; Mavrou, A.; Constantinidou, N.; Dacou-Voutetakis, C.
Prevalence of thyroid dysfunction in Turner’s syndrome: A long-term follow-up study and brief literature review. Thyroid
2005
,
15, 1061–1066. [CrossRef]
51.
Valenzise, M.; Aversa, T.; Corrias, A.; Mazzanti, L.; Cappa, M.; Ubertini, G.; Scarano, E.; Mussa, A.; Messina, M.F.; De Luca, F.;
et al. Epidemiology, presentation and long-term evolution of Graves’ disease in children, adolescents and young adults with
Turner syndrome. Horm. Res. Paediatr. 2014,81, 245–250. [CrossRef]
52.
Libert, C.; Dejager, L.; Pinheiro, I. The X chromosome in immune functions: When a chromosome makes the difference. Nat. Rev.
Immunol. 2010,10, 594–604. [CrossRef]
53.
Mondal, S.; Bhattacharjee, R.; Chowdhury, S.; Mukhopadhyah, S. Karyotype-Phenotype Correlation in Turner Syndrome at a
Single Center in Eastern India. Indian Pediatr. 2021,58, 34–37. [CrossRef]
54.
Stoklasova, J.; Zapletalova, J.; Frysak, Z.; Hana, V.; Cap, J.; Pavlikova, M.; Soucek, O.; Lebl, J. An isolated Xp deletion is linked to
autoimmune diseases in Turner syndrome. J. Pediatr. Endocrinol. Metab. 2019,32, 479–488. [CrossRef]
55. De Sanctis, V.; Khater, D. Autoimmune diseases in Turner syndrome: An overview. Acta Biomed. 2019,90, 341–344. [CrossRef]
56.
Gravholt, C.H.; Juul, S.; Naeraa, R.W.; Hansen, J. Morbidity in Turner syndrome. J. Clin. Epidemiol.
1998
,51, 147–158. [CrossRef]
57.
Davis, S.M.; Geffner, M.E. Cardiometabolic health in Turner syndrome. Am. J. Med. Genet. C Semin. Med. Genet.
2019
,181, 52–58.
[CrossRef]
58.
Lebenthal, Y.; Levy, S.; Sofrin-Drucker, E.; Nagelberg, N.; Weintrob, N.; Shalitin, S.; de Vries, L.; Tenenbaum, A.; Phillip, M.;
Lazar, L. The Natural History of Metabolic Comorbidities in Turner Syndrome from Childhood to Early Adulthood: Comparison
between 45,X Monosomy and Other Karyotypes. Front. Endocrinol. 2018,9, 27. [CrossRef]
59.
Pirgon, Ö.; Atabek, M.E.; Oran, B.; Güçlü, R. Atherogenic lipid profile and systolic blood pressure are associated with carotid
artery intima-media thickness in children with Turner syndrome. J. Clin. Res. Pediatr. Endocrinol. 2008,1, 62–71. [CrossRef]
60.
O’Gorman, C.S.; Syme, C.; Lang, J.; Bradley, T.J.; Wells, G.D.; Hamilton, J.K. An evaluation of early cardiometabolic risk factors in
children and adolescents with Turner syndrome. Clin. Endocrinol. 2013,78, 907–913. [CrossRef]
61.
Van, P.L.; Bakalov, V.K.; Bondy, C.A. Monosomy for the X-chromosome is associated with an atherogenic lipid profile. J. Clin.
Endocrinol. Metab. 2006,91, 2867–2870. [CrossRef]
62.
Calcaterra, V.; Brambilla, P.; Maffè, G.C.; Klersy, C.; Albertini, R.; Introzzi, F.; Bozzola, E.; Bozzola, M.; Larizza, D. Metabolic
syndrome in Turner syndrome and relation between body composition and clinical, genetic, and ultrasonographic characteristics.
Metab. Syndr. Relat. Disord. 2014,12, 159–164. [CrossRef]
63.
Mavinkurve, M.; O’Gorman, C.S. Cardiometabolic and vascular risks in young and adolescent girls with Turner syndrome. BBA
Clin. 2015,3, 304–309. [CrossRef]
64.
Hjerrild, B.E.; Holst, J.J.; Juhl, C.B.; Christiansen, J.S.; Schmitz, O.; Gravholt, C.H. Delayed
β
-cell response and glucose intolerance
in young women with Turner syndrome. BMC Endocr. Disord. 2011,11, 6. [CrossRef]
65.
Helena Mangs, A.; Morris, B.J. The Human Pseudoautosomal Region (PAR): Origin, Function and Future. Curr. Genom.
2007
,8,
129–136. [CrossRef]
66. Bakalov, V.K.; Cheng, C.; Zhou, J.; Bondy, C.A. X-chromosome gene dosage and the risk of diabetes in Turner syndrome. J. Clin.
Endocrinol. Metab. 2009,94, 3289–3296. [CrossRef]
67. Wolff, D.J.; Van Dyke, D.L.; Powell, C.M. Laboratory guideline for turner syndrome. Genet. Med. 2010,12, 52–55. [CrossRef]
68.
de Oliveira, R.M.R.; do Nascimento Verreschi, I.T.; Lipay, M.V.N.; Eça, L.P.; Guedes, A.D.; Bianco, B. Y chromosome in Turner
syndrome: Review of the literature. Sao Paulo Med. J. 2009,127, 373–378. [CrossRef]
69.
Ackermann, A.; Bamba, V. Current controversies in turner syndrome: Genetic testing, assisted reproduction, and cardiovascular
risks. J. Clin. Transl. Endocrinol. 2014,1, 61–65. [CrossRef]
70.
Knauer-Fischer, S.; Besikoglu, B.; Inta, I.; Kneppo, C.; Vogt, P.H.; Bettendorf, M. Analyses of Gonadoblastoma Y (GBY)-locus and
of Y centromere in Turner syndrome patients. Exp. Clin. Endocrinol. Diabetes 2015,123, 61–65. [CrossRef]
71.
Gravholt, C.H.; Fedder, J.; Naeraa, R.W.; Müller, J. Occurrence of gonadoblastoma in females with Turner syndrome and
Y chromosome material: A population study. J. Clin. Endocrinol. Metab. 2000,85, 3199–3202.
72.
Nishi, M.Y.; Domenice, S.; Medeiros, M.A.; Mendonca, B.B.; Billerbeck, A.E.C. Detection of Y-specific sequences in 122 patients
with Turner syndrome: Nested PCR is not a reliable method. Am. J. Med. Genet. 2002,107, 299–305. [CrossRef]
73.
Wapner, R.J.; Martin, C.L.; Levy, B.; Ballif, B.C.; Eng, C.M.; Zachary, J.M.; Savage, M.; Platt, L.D.; Saltzman, D.; Grobman, W.A.;
et al. Chromosomal microarray versus karyotyping for prenatal diagnosis. N. Engl. J. Med. 2012,367, 2175–2184. [CrossRef]
74.
Mazloom, A.R.; Džakula, Ž.; Oeth, P.; Wang, H.; Jnsen, T.; Tynan, J.; McCullough, R.; Saldivar, J.S.; Ehrich, M.; van den Boom, D.;
et al. Noninvasive prenatal detection of sex chromosomal aneuploidies by sequencing circulating cell-free DNA from maternal
plasma. Prenat. Diagn. 2013,33, 591–597. [CrossRef]
75.
Prakash, S.; Guo, D.; Maslen, C.L.; Silberbach, M.; Milewicz, D.; Bondy, C.A.; GenTAC Investigators. Single-nucleotide
polymorphism array genotyping is equivalent to metaphase cytogenetics for diagnosis of Turner syndrome. Genet. Med.
2014
,16,
53–59. [CrossRef]
76.
Viuff, M.; Skakkebaek, A.; Nielsen, M.M.; Chang, S.; Gravholt, C.H. Epigenetics and genomics in Turner syndrome. Am. J. Med.
Genet. Part C Semin. Med. Genet. 2019,181, 125–132. [CrossRef]
Endocrines 2022,3253
77.
Wang, H.; Zhu, H.; Zhu, W.; Xu, Y.; Wang, N.; Han, B.; Song, H.; Qiao, J. Bioinformatic Analysis Identifies Potential Key Genes in
the Pathogenesis of Turner Syndrome. Front. Endocrinol. 2020,11, 104. [CrossRef]
78.
Manotas, M.C.; Calderón, J.C.; López-Kleine, L.; Suárez-Obando, F.; Moreno, O.M.; Rojas, A. Identification of common differen-
tially expressed genes in Turner (45,X) and Klinefelter (47,XXY) syndromes using bioinformatics analysis. Mol. Genet. Genom.
Med. 2020,8, e1503. [CrossRef]
79.
Los, E.; Rosenfeld, R.G. Growth and growth hormone in Turner syndrome: Looking back, looking ahead. Am. J. Med. Genet. Part.
C Semin. Med. Genet. 2019,181, 86–90. [CrossRef]
80.
Juloski, J.; Dumanˇci´c, J.; Š´cepan, I.; Lauc, T.; Milašin, J.; Kaic, Z.; Dumic, M.; Babic, M. Growth hormone positive effects on
craniofacial complex in Turner syndrome. Arch. Oral Biol. 2016,71, 10–15. [CrossRef]
81.
Dyrka, K.; Rozkiewicz, N.; Obara-Moszynska, M.; Niedziela, M. The influence of growth hormone therapy on the cardiovascular
system in Turner syndrome. J. Pediatr. Endocrinol. Metab. 2020,33, 1363–1372. [CrossRef]
82.
Quigley, C.A.; Fechner, P.Y.; Geffner, M.E.; Eugster, E.A.; Ross, J.L.; Habiby, R.L.; Ugrasbul, F.; Rubin, K.; Travers, S.; Antalis, C.J.;
et al. Prevention of Growth Failure in Turner Syndrome: Long-Term Results of Early Growth Hormone Treatment in the “Toddler
Turner” Cohort. Horm. Res. Paediatr. 2021,94, 18–35. [CrossRef]
83.
Stevens, A.; Murray, P.; Wojcik, J.; Raelson, J.; Koledova, E.; Chatelain, P.; Clayton, P.; PREDICT Investigator Group. Validating
genetic markers of response to recombinant human growth hormone in children with growth hormone deficiency and Turner
syndrome: The PREDICT validation study. Eur. J. Endocrinol. 2016,175, 633–643. [CrossRef] [PubMed]
84.
Braz, A.F.; Costalonga, E.F.; Trarbach, E.B.; Scalco, R.C.; Malaquias, A.C.; Guerra-Junior, G.; Antonini, S.R.R.; Mendonca, B.B.;
Arnhold, I.J.P.; Jorge, A.A.L. Genetic predictors of long-term response to growth hormone (GH) therapy in children with GH
deficiency and Turner syndrome: The influence of a SOCS2 polymorphism. J. Clin. Endocrinol. Metab.
2014
,99, E1808–E1813.
[CrossRef] [PubMed]
85.
Li, P.; Cheng, F.; Xiu, L. Height outcome of the recombinant human growth hormone treatment in Turner syndrome: A meta-
analysis. J. Clin. Endocrinol. Metab. 2014,99, E1808–E1813. [CrossRef] [PubMed]
86.
Sas, T.C.J.; Gault, E.J.; Zeger Bardsley, M.; Menke, L.A.; Freriks, K.; Perry, R.J.; Otten, N.J.; Keizer-Schrama, S.M.; Timmers, H.; Wit,
J.M.; et al. Safety and efficacy of oxandrolone in growth hormone-treated girls with Turner syndrome: Evidence from recent
studies and recommendations for use. Horm. Res. Paediatr. 2014,81, 289–297. [CrossRef]
87.
Cintron, D.; Rodriguez-Gutierrez, R.; Serrano, V.; Latortue-Albino, P.; Erwin, P.J.; Murad, M.H. Effect of estrogen replacement
therapy on bone and cardiovascular outcomes in women with turner syndrome: A systematic review and meta-analysis. Endocrine
2016,55, 366–375. [CrossRef]
88.
Klein, K.O.; Rosenfield, R.L.; Santen, R.J.; Gawlik, A.M.; Backeljauw, P.F.; Gravholt, C.H.; Sas, T.C.J.; Mauras, N. Estrogen
Replacement in Turner Syndrome: Literature Review and Practical Considerations. J. Clin. Endocrinol. Metab.
2018
,103, 1790–1803.
[CrossRef]
89.
Viuff, M.H.; Berglund, A.; Juul, S.; Andersen, N.H.; Stochholm, K.; Gravholt, C.H. Sex Hormone Replacement Therapy in Turner
Syndrome: Impact on Morbidity and Mortality. J. Clin. Endocrinol. Metab. 2020,105, 468–478. [CrossRef]
90.
Cameron-Pimblett, A.; Davies, M.C.; Burt, E.T.; Talaulikar, V.S.; La Rosa, C.; King, T.F.J.; Conway, G.S. Effects of Estrogen
Therapies on Outcomes in Turner Syndrome: Assessment of Induction of Puberty and Adult Estrogen Use. J. Clin. Endocrinol.
Metab. 2019,104, 2820–2826. [CrossRef]
91.
Lunding, S.A.; Aksglaede, L.; Anderson, R.A.; Main, K.M.; Juul, A.; Hagen, C.P.; Pedersen, A.T. AMH as Predictor of Premature
Ovarian Insufficiency: A Longitudinal Study of 120 Turner Syndrome Patients. J. Clin. Endocrinol. Metab.
2015
,100, E1030–E1038.
[CrossRef]
92.
Quigley, C.A.; Wan, X.; Garg, S.; Kowal, K.; Cutler, G.B., Jr.; Ross, J.L. Effects of low-dose estrogen replacement during childhood on
pubertal development and gonadotropin concentrations in patients with Turner syndrome: Results of a randomized, double-blind,
placebo-controlled clinical trial. J. Clin. Endocrinol. Metab. 2014,99, E1754–E1764. [CrossRef]
93.
Ross, J.L.; Roeltgen, D.; Feuillan, P.; Kushner, H.; Cutler, G.B., Jr. Effects of estrogen on nonverbal processing speed and motor
function in girls with Turner ’s syndrome. J. Clin. Endocrinol. Metab. 1998,83, 3198–3204. [CrossRef] [PubMed]
94.
Ross, J.L.; Roeltgen, D.; Feuillan, P.; Kushner, H.; Cutler, G.B., Jr. Use of estrogen in young girls with Turner syndrome: Effects on
memory. Neurology 2000,54, 164. [CrossRef] [PubMed]
95.
Torres-Santiago, L.; Mericq, V.; Taboada, M.; Unanue, N.; Klein, K.O.; Singh, R.; Hossain, J.; Santen, R.J.; Ross, J.L.; Mauras, N.
Metabolic effects of oral versus transdermal 17
β
-estradiol (E
2
): A randomized clinical trial in girls with Turner syndrome. J. Clin.
Endocrinol. Metab. 2013,98, 2716–2724. [CrossRef] [PubMed]
96.
Mohammed, K.; Dabrh, A.M.A.; Benkhadra, K.; Al Nofal, A.; Carranza Leon, B.G.; Prokop, L.J.; Montori, V.M.; Faubion, S.S.;
Hurad, M.H. Oral vs Transdermal Estrogen Therapy and Vascular Events: A Systematic Review and Meta-Analysis. J. Clin.
Endocrinol. Metab. 2015,100, 4012–4020. [CrossRef] [PubMed]
97.
Shifren, J.L.; Gass, M.L.S.; NAMS Recommendations for Clinical Care of Midlife Women Working Group. The North American
Menopause Society recommendations for clinical care of midlife women. Menopause 2014,21, 1038–1062. [CrossRef] [PubMed]
98.
Hovatta, O. Ovarian function and
in vitro
fertilization (IVF) in Turner syndrome. Pediatr. Endocrinol. Rev.
2012
,9(Suppl. 2),
713–717. [PubMed]
Endocrines 2022,3254
99.
Oktay, K.; Bedoschi, G.; Berkowitz, K.; Bronson, R.; Kashani, B.; McGovern, P.; Pal, L.; Quinn, G.; Rubin, K. Fertility Preservation
in Women with Turner Syndrome: A Comprehensive Review and Practical Guidelines. J. Pediatr. Adolesc. Gynecol.
2015
,29,
409–416. [CrossRef]
100.
Kavoussi, S.K.; Fisseha, S.; Smith, Y.R.; Smith, G.D.; Christman, G.M.; Gago, L.A. Oocyte cryopreservation in a woman with
mosaic Turner syndrome: A case report. J. Reprod. Med. 2008,53, 223–226.
101.
Oktay, K.; Bedoschi, G. Oocyte cryopreservation for fertility preservation in postpubertal female children at risk for premature
ovarian failure due to accelerated follicle loss in Turner syndrome or cancer treatments. J. Pediatr. Adolesc. Gynecol.
2014
,27,
342–346. [CrossRef]
102.
Oktay, K.; Rodriguez-Wallberg, K.A.; Sahin, G. Fertility preservation by ovarian stimulation and oocyte cryopreservation in
a 14-year-old adolescent with Turner syndrome mosaicism and impending premature ovarian failure. Fertil. Steril.
2010
,94,
753.e15–753.e19. [CrossRef]
103.
Nawroth, F.; Schüring, A.N.; von Wolff, M. The indication for fertility preservation in women with Turner syndrome should not
only be based on the ovarian reserve but also on the genotype and expected future health status. Acta Obstet. Gynecol. Scand.
2020,99, 1579–1583. [CrossRef] [PubMed]
... Patients with a history of dental trauma, dental treatment, and systematic conditions influence the prevalence of apical root resorption [13]. The severe resorption observed in the patient could be a result of impaired metabolism and endocrinological problems, resulting from insufficient levels of growth hormone and estrogen, as reported among patients with Turner syndrome [14]. The main goal of the second hormone is to prevent bone resorption, so lowering its level often leads to consequences such as increased bone fragility, observed in osteoporosis [15]. ...
Article
Full-text available
External apical root resorption in permanent teeth is a multifactorial process influenced by a variety of local and systemic factors. This report describes a case of multiple and severe apical root resorptions in a patient with Turner syndrome. The condition was discovered in a young female with Turner syndrome after 30 months of orthodontic treatment with fixed appliance. The purpose of this report is to present reports by other authors on the potential causes of the increased risk of tooth resorption in patients with Turner syndrome and to share insights derived from its course, highlighting the implications and lessons learned. Patients with Turner syndrome are not ideal candidates for orthodontic treatment. Prior to commencing orthodontic treatment, it is essential to carefully consider the potential benefits of the therapy compared to the risk associated with exacerbating root resorption. In the case of Turner syndrome patients, where there is an elevated risk of such complications, a thorough analysis should be conducted to determine whether the expected benefits of the treatment outweigh the potential hazards to the patient's dental health.
Article
Full-text available
Objective: To describe clinical features in Indian girls with Turner syndrome along with the phenotype-karyotype correlation. Methods: 103 girls with Turner syndrome were divided into karyotype-groups: Classic (45X), 45,X/46,XX mosaics, isochromosomeXq (46,X,iXq and 45,X/46,X,iXq mosaics), 45,X/46,XYmosaics and structural defects, and analyzed for phenotypic differences. Results: Majority (44.1%) had classic karyotype followed by isochromosome-Xq (26.5%). Classic Turner syndrome had higher prevalence of most skeletal and cutaneous stigmata, cubitus valgus (68.3%) and multiple nevi (68.2%) being the commonest. Bicuspid aortic valve was most common in 45,X/46,XX mosaics (5/15, 33.3%), and aortic coarctation in classic TS (3/42, 7.2%). Congenital renal anomalies occurred mostly in classic TS (6/42,14.3%). Overt hypothyroidism, conductive deafness and recurrent otitis media were commonest in isochromosomes (P<0.03). 45,X/46,XY mosaics had highest IQ (P<0.005). Conclusions: We report some novel associations of karyotype with non-endocrine parameters in Turner syndrome. In resource-limited settings, underlying karyotype may help prioritize screening investigations in girls with Turner syndrome.
Article
Full-text available
Objectives Reduced gene expression of PPARGC1A in subjects with insulin resistance (IR) has been reported. Insulin resistance occurs early on the course of Turner syndrome (TS). The main objective of this study was to evaluate the relationship between PPARGC1A promoter DNA methylation status in lymphocytes and insulin sensitivity and secretion in Ecuadorian females with TS. Methods We examined a cohort of 34 Ecuadorian patients with TS along with a sex-, age- and BMI-matched reference group. All subjects received a standard 75 g oral glucose tolerance test. Insulin resistance and secretion indices were calculated. The PPARGC1A methylated DNA/unmethylated DNA ratio and mitochondrial content (mtDNA/nDNA ratio) were further determined. Results Notably, the PPARGC1A DNA methylation level was significantly higher in TS subjects than the reference group and correlated with IR indices. Conversely, mitochondrial content was significantly lower in the study group than healthy controls and negatively correlated with the PPARGC1A methylated DNA/unmethylated DNA ratio in TS individuals. PPARGC1A promoter DNA methylation status contributed to 20% of the total variability in Homeostasis Model Assessment for Insulin Resistance (HOMA-IR) independently of BMI or age in TS subjects. Conclusions Our collective findings suggest that expression of PPARGC1A and lower mitochondrial number affect the metabolic phenotype in TS subjects.
Article
Full-text available
Background: Analysis of patients with chromosomal abnormalities, including Turner syndrome and Klinefelter syndrome, has highlighted the importance of X-linked gene dosage as a contributing factor for disease susceptibility. Escape from X-inactivation and X-linked imprinting can result in transcriptional differences between normal men and women as well as in patients with sex chromosome abnormalities. Objective: To identify differentially expressed genes among patients with Turner (45,X) and Klinefelter (46,XXY) syndrome using bioinformatics analysis. Methodology: Two gene expression data sets of Turner (45,X) and Klinefelter syndrome (47,XXY) were obtained from the Gene Omnibus Expression (GEO) database of the National Center for Biotechnology Information (NCBI). Statistical analysis was performed using R Bioconductor libraries. Differentially expressed genes (DEGs) were determined using significance analysis of microarray (SAM). The functional annotation of the DEGs was performed with DAVID v6.8 (The Database for Annotation, Visualization, and Integrated Discovery). Results: There are no genes over-expressed simultaneously in both diseases. However, when crossing the list of under-expressed genes for 45,X cells and the list of over-expressed genes for 47,XXY cells, there are 16 common genes: SLC25A6, AKAP17A, ASMTL, KDM5C, KDM6A, ATRX, CSF2RA, DHRSX, CD99, ZBED1, EIF1AX, MVB12B, SMC1A, P2RY8, DOCK7, DDX3X, eight of which are involved in the regulation of gene expression by epigenetic mechanisms, regulation of splicing processes and protein synthesis. Conclusion: Of the 16 identified as under-expressed in 45,X cells and over-expressed in 47,XXY cells, 14 are located in X chromosome and 2 in autosomal chromosome; 8 of these genes are involved in the regulation of gene expression: 5 genes are related to epigenetic mechanisms, 2 in regulation of splicing processes, and 1 in the protein synthesis process. Our results are limited by it being the product of a bioinformatic analysis from mRNA isolated from whole blood, this makes necessary further exploration of the relationships between these genes and Turner syndrome and Klinefelter syndrome in the future.
Article
Full-text available
Fertility preservation in women with Turner syndrome is highly controversial. Some strongly recommend freezing of ovarian tissue at young age, others don’t. The controversy is partly due to different perspectives and professions. Biologists prefer to freeze young ovaries with high follicle density, reproductive physicians want to avoid risky operations and iatrogenic infertility by removing one ovary and cardiologists and obstetricians warn against the risks of later pregnancies. Accordingly, fertility preservation in young women with Turner syndrome is more than freezing of ovarian tissue or oocytes. Fertility preservation requires a balanced decision considering the conservation of fertility, the protection of reproductive health and future health consequences. Therefore, fertility preservation strategies should be based not only on the individual ovarian reserve but also on the genotype and the expected cardiac health status to decide what is the best option: to freeze tissue or alternatively to wait and see.
Article
Full-text available
Background Turner syndrome (TS) is a chromosomal disorder, in which a female is partially or entirely missing one of the two X chromosomes, with a prevalence of 1:2500 live female births. The present study aims to identify a circulating microRNA (miRNA) signature for TS patients with and without congenital heart disease (CHD). Methods Microarray platform interrogating 2549 miRNAs were used to detect the miRNA abundance levels in the blood of 33 TS patients and 14 age-matched healthy volunteer controls (HVs). The differentially abundant miRNAs between the two groups were further validated by RT-qPCR. Results We identified 60 differentially abundant miRNA in the blood of TS patients compared to HVs, from which, 41 and 19 miRNAs showed a higher and a lower abundance levels in TS patients compared to HVs, respectively. RT-qPCR confirmed the significantly higher abundance levels of eight miRNAs namely miR-374b-5p, miR-199a-5p, miR-340-3p, miR-125b-5p, miR-30e-3p, miR-126-3p, miR-5695, and miR-26b-5p in TS patients as compared with the HVs. The abundance level of miR-5695 was higher in TS patients displaying CHD as compared to TS patients without CHD (p = 0.0265; log2-fold change 1.99); whereas, the abundance level of miR-126-3p was lower in TS patients with congenital aortic valve disease (AVD) compared to TS patients without BAV (p = 0.0139, log2-fold change 1.52). The clinical feature statistics revealed that miR-126-3p had a significant correlation with sinotubular junction Z-score (r = 0.42; p = 0.0154). Conclusion The identified circulating miRNAs signature for TS patients with manifestations associated with cardiovascular diseases provide new insights into the molecular mechanism of TS that may guide the development of novel diagnostic approaches.
Article
Full-text available
Background: Turner syndrome (TS) is a sex chromosome aneuploidy with a variable spectrum of symptoms including short stature, ovarian failure and skeletal abnormalities. The etiology of TS is complex, and the mechanisms driving its pathogenesis remain unclear.Methods: In our study, we used the online Gene Expression Omnibus (GEO) microarray expression profiling dataset GSE46687 to identify differentially expressed genes (DEGs) between monosomy X TS patients and normal female individuals. The relevant data on 26 subjects with TS (45,XO) and 10 subjects with the normal karyotype (46,XX) was investigated. Then, tissue-specific gene expression, functional enrichment, and protein-protein interaction (PPI) network analyses were performed, and the key modules were identified.Results: In total, 25 upregulated and 60 downregulated genes were identified in the differential expression analysis. The tissue-specific gene expression analysis of the DEGs revealed that the system with the most highly enriched tissue-specific gene expression was the hematologic/immune system, followed by the skin/skeletal muscle and neurologic systems. The PPI network analysis, construction of key modules and manual screening of tissue-specific gene expression resulted in the identification of the following five genes of interest: CD99, CSF2RA, MYL9, MYLPF, and IGFBP2. CD99 and CSF2RA are involved in the hematologic/immune system, MYL9 and MYLPF are related to the circulatory system, and IGFBP2 is related to skeletal abnormalities. In addition, several genes of interest with possible roles in the pathogenesis of TS were identified as being associated with the hematologic/immune system or metabolism.Conclusion: This discovery-driven analysis may be a useful method for elucidating novel mechanisms underlying TS. However, more experiments are needed to further explore the relationships between these genes and TS in the future.
Article
Introduction: In the randomized "Toddler Turner" study, girls who received growth hormone (GH) starting at ages 9 months to 4 years (early-treated [ET] group) had marked catch-up growth and were 1.6 ± 0.6 SD taller than untreated (early-untreated [EUT]) control girls after 2 years. However, whether the early catch-up growth would result in greater near-adult height (NAH) was unknown. Therefore, this extension study examined the long-term effects of toddler-age GH treatment on height, pubertal development, and safety parameters. Methods: Toddler Turner study participants were invited to enroll in a 10-year observational extension study for annual assessments of growth, pubertal status, and safety during long-term GH treatment to NAH for both ET and EUT groups. Results: The ET group was taller than the EUT group at all time points from preschool to maturity and was significantly taller at the onset of puberty (p = 0.016), however, the difference was not significant at NAH. For the full cohort (ET + EUT combined, n = 50) mean (± SD) NAH was 151.2 ± 7.1 cm at age 15.0 ± 1.3 years. NAH standard deviation score (SDS) was within the normal range (>-2.0) for 76% of ET and 60% of EUT subjects (68% overall) and correlated strongly with height SDS at GH start (r = 0.78; p < 0.01), which in turn had a modest inverse correlation with age at GH start (i.e., height SDS declined with increasing age in untreated girls [r = -0.30; p = 0.016]). No new safety concerns arose. Conclusion: Although the ET group was taller throughout, height SDS at NAH was not significantly different between groups due to catch-down growth of ET girls during lapses in GH treatment after the Toddler study and similar long-term GH exposure overall. Early initiation of GH by age 6 years, followed by uninterrupted treatment during childhood, can prevent ongoing growth failure and enable attainment of height within the normal range during childhood, adolescence, and adulthood.
Article
Short stature, ovarian dysgenesis, infertility, and cardiovascular malformations are classic features in Turner syndrome (TS), but the phenotypical spectrum is wide. Through early diagnosis and appropriate treatment, TS patients have a chance to achieve satisfactory adult height and sexual development. The doses of recombinant growth hormone (rGH) used are usually higher than the substitution dose. The safety aspects of this therapy are very important, especially in terms of the cardiovascular system. The presented study aimed to analyze how the rGH therapy may influence the cardiovascular system in TS based on current literature data. We conducted a systematic search for studies related to TS, cardiovascular system, and rGH therapy. The results show that rGH seems to have a positive effect on lipid parameters, reducing the risk of ischemic disease. It is additionally optimized by estradiol therapy. Although rGH may increase insulin resistance, the metabolic derangement is rare, probably due to lower fat content and an increase in lean body mass. Several studies showed that rGH treatment could cause aorta widening or increase the aorta growth rate. IGF-1 can be independently associated with increased aortic diameters. The studies analyzing the impact of GH on blood pressure show conflicting data. The proper cardiovascular imaging before and during rGH treatment and detecting the known risk factors for aorta dissection in every individual is very important. The long-term effects of growth hormone treatment on the heart and arteries are still not available and clearly estimated and have to be monitored in the future.
Article
47,XXX (triple X) and Turner syndrome (45,X) are sex chromosomal abnormalities with detrimental effects on health with increased mortality and morbidity. In karyotypical normal females, X‐chromosome inactivation balances gene expression between sexes and upregulation of the X chromosome in both sexes maintain stoichiometry with the autosomes. In 47,XXX and Turner syndrome a gene dosage imbalance may ensue from increased or decreased expression from the genes that escape X inactivation, as well as from incomplete X chromosome inactivation in 47,XXX. We aim to study genome‐wide DNA‐methylation and RNA‐expression changes can explain phenotypic traits in 47,XXX syndrome. We compare DNA‐methylation and RNA‐expression data derived from white blood cells of seven women with 47,XXX syndrome, with data from seven female controls, as well as with seven women with Turner syndrome (45,X). To address these questions, we explored genome‐wide DNA‐methylation and transcriptome data in blood from seven females with 47,XXX syndrome, seven females with Turner syndrome, and seven karyotypically normal females (46,XX). Based on promoter methylation, we describe a demethylation of six X‐chromosomal genes (AMOT , HTR2C , IL1RAPL2 , STAG2 , TCEANC , ZNF673 ), increased methylation for GEMIN8 , and four differentially methylated autosomal regions related to four genes (SPEG , MUC4 , SP6 , and ZNF492 ). We illustrate how these changes seem compensated at the transcriptome level although several genes show differential exon usage. In conclusion, our results suggest an impact of the supernumerary X chromosome in 47,XXX syndrome on the methylation status of selected genes despite an overall comparable expression profile.