Skip to main content
Erschienen in: Journal of Translational Medicine 1/2019

Open Access 01.12.2019 | Review

The evolving role of genetic tests in reproductive medicine

verfasst von: Federica Cariati, Valeria D’Argenio, Rossella Tomaiuolo

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2019

Abstract

Infertility is considered a major public health issue, and approximately 1 out of 6 people worldwide suffer from infertility during their reproductive lifespans. Thanks to technological advances, genetic tests are becoming increasingly relevant in reproductive medicine. More genetic tests are required to identify the cause of male and/or female infertility, identify carriers of inherited diseases and plan antenatal testing. Furthermore, genetic tests provide direction toward the most appropriate assisted reproductive techniques. Nevertheless, the use of molecular analysis in this field is still fragmented and cumbersome. The aim of this review is to highlight the conditions in which a genetic evaluation (counselling and testing) plays a role in improving the reproductive outcomes of infertile couples. We conducted a review of the literature, and starting from the observation of specific signs and symptoms, we describe the available molecular tests. To conceive a child, both partners' reproductive systems need to function in a precisely choreographed manner. Hence to treat infertility, it is key to assess both partners. Our results highlight the increasing importance of molecular testing in reproductive medicine.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
aCGH
array-based Comparative Genomic Hybridization
AOA
assisted oocyte activation
ART
assisted reproductive technology
AZF
azoospermia factor
cffDNA
cell-free fetal DNA
CFTR
Cystic Fibrosis Transmembrane Conductance Regulator
CVS
chronic villus sampling
DAZ
Deleted in Azoospermia
DNA
deoxyribonucleic acid
ESHG
European Society of Human Genetics
ESHRE
European Society of Human Reproduction and Embryology
FISH
fluorescence in situ hybridization
FMR
Fragile X Mental Retardation
HLA
Human Leukocyte Antigen
ICSI
Intracytoplasmic Sperm Injection
IVF
in vitro fertilization
mCGH
metaphase Comparative Genomic Hybridization
NGS
next generation sequencing
NIPT
non-invasive prenatal testing
OMIM
Online Mendelian Inheritance in Man
PCR
polymerase chain reaction
PGD
preimplantation genetic diagnosis
PGDIS
Preimplantation Genetic Diagnosis International Society
PGT-A
Preimplantation Genetic Test for Aneuploidy
PGT
Preimplantation Genetic Testing
PND
prenatal diagnosis
POI
primary ovarian insufficiency
qPCR
quantitative polymerase chain reaction
SNP
single nucleotide polymorphism
STR
short tandem repeat
STS
sequence tagged site
WGA
whole genome amplification

Introduction

During the last few decades, there have been a series of striking advancements in reproductive and laboratory medicine that have essentially caused these two fields to become inextricably connected. Laboratory medicine now plays a critical role in all stages of the reproductive process, from diagnostic approaches to the choice of the most complex therapy.
In particular, genetic tests are carried out for three main purposes in reproductive medicine: the identification of the infertility causes, identification of genetic diseases transmissible to offspring, and optimization of the assisted reproductive technology (ART) (Fig. 1).
The overall fertility rate is decreasing; for example, in the US, 12% of women receive fertility treatment over the course of their lifetimes, so it is important to emphasize the fertility journey of couples [1]. The reproductive systems of both partners function in a combined and precisely coordinated way to conceive a child; for this reason, evaluation of both members of the couple is mandatory.
A medical evaluation is indicated when the couple fails to achieve pregnancy after 12 months of regular, unprotected sexual intercourse [2]. Currently, the diagnostic timeline of infertile couples includes biochemical and instrumental analyses that allow for a diagnosis in 65% of cases; in the remaining 35% of cases, which are undiagnosed, genetic tests are performed. Considering that approximately 15% of genetic disorders are associated with infertility and that similar clinical signs can have genetic and nongenetic causes, it is important that an infertility diagnosis be determined by the combination of an accurate medical history and instrument- and laboratory-based evaluations, including targeted genetic tests [3]. Confirmation of the clinical diagnosis through genetic evaluation (counselling and testing) can lead to more specific and targeted medical management.
In addition, genetic tests are also indicated for the identification of genetic diseases that are transmissible to the offspring: preconception screening allows couples who are planning to become pregnant to know their reproductive risk a priori. Normally, gametes with genetic or chromosomal alterations have reduced reproductive potential. Thanks to ART, many of these difficulties can be overcome, and therefore, genetic tests (carrier screening, preimplantation and prenatal diagnosis) have the crucial impact of monitoring the possible transmission of these genetic alterations to the offspring [4, 5].
Another application field of molecular diagnostics is related to the antenatal diagnosis. To date, the diagnostic options for couples at risk of transmitting a specific inherited disorder to their offspring are preimplantation genetic testing (PGT) and prenatal diagnosis (PND). These two diagnostic procedures share the same purpose but differ in diagnostic time, type of sampling, and laboratory procedures. In addition to the more traditional laboratory investigations, it is now undisputed that molecular biology methods for PGT support the efficacy of ART techniques, contributing significantly to their success (reductions in time, effort and cost) [5, 6].
To optimize the application of genetic tests in clinical practice, in this review, we discuss (1) the genetic conditions related to infertility, including the common and rare ones that are case appropriate; (2) the diagnostic strategies in families at risk of known monogenic disease transmission; and (3) the impact of PGT in the optimization of ART techniques.

Materials and methods

The literature review was carried out according to PRISMA guidelines. No temporal restrictions were applied. The research was performed using the following keywords: genetic cause of infertility, genetic cause of male infertility, genetic cause of female infertility, mutations and infertility, molecular diagnostics in infertile couples, molecular diagnostics and reproductive medicine, PGT techniques, and PGT and ART. All the papers found were carefully read and evaluated before their inclusion. No unpublished studies were taken into consideration. In addition, the following databases were also consulted to verify gene/phenotype associations: NIH (https://​www.​nih.​gov), OMIM (https://​www.​omim.​org) and OrphaNet (https://​www.​orpha.​net/​consor/​cgi-bin/​index.​php).

Results

Genetic tests in the identification of the causes of infertility

It has been estimated that every healthy subject is a carrier of 5/8 genetic alterations associated with recessive genetic disorders; therefore, even in the absence of specific symptoms, family planning and reproduction can be risky. Moreover, it has been reported that almost 50% of infertility cases are related to genetic disorders [7, 8]. In the presence or high suspicion of a genetically based reproductive risk, the genetic test provides a more accurate diagnosis of infertility and provides the opportunity to inform the couple about the possible risk of transmission to the offspring. Unfortunately, genetic tests for examining infertility are based on a standard algorithm directed to investigate the most frequent genetic causes without taking into account the patient’s personal or family history. Consequently, the results are quite discouraging: a genetic diagnosis is reached in approximately 4% of all infertile males, and approximately 20% of infertile couples remain undiagnosed. In contrast, an accurate medical and familial history aimed at identifying genetically based syndromes (characterized by typical dimorphisms, associated disabilities and even infertility) could direct patients to specific genetic tests [9]. Starting from this consideration, we examined the genetic disorders related to male and female infertility and subdivided them according to the signs and symptoms observed by the specialist during the first medical examination. Genes associated with specific and rare clinical conditions were not excluded either; they could be useful, in the context of a specific clinical picture, to request an in-depth analysis using a targeted genetic test. Therefore, we provide the main points on the genetic pathology, current test execution modality and management of the patient (Tables 1, 2, 3, 4, 5, 6, 7).
Table 1
The chromosome aberrations related to testicular male infertility: from the first observation to the report
Indications for genetic test
Genetic condition
Frequency
Test
Chromosome/genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Azoospermia/oligozoospermia; Sertoli cell syndrome type I and type II (presence of some tubules with normal spermatogenesis) and hypospermatogenesis diagnosis by histological evaluation
Microdeletion Y chromosome AZFc
1/2.500; (AZFc 60%, AZFb 15%, AZFb-c 22%, AZFa 3%); 13% of azoospermia cases; 3–7% of oligozoospermia cases
Molecular diagnosis by PCR of STS sequences
Interstitial deletion of AZFc Y region (recombination between palindromes b2 and b4); DAZ, BPY2, PRY2, CDY1
✓: testicular sperm retrieval + ICSI
Y linked
Other causes of azoospermia or oligozoospermia
[10]
Azoospermia; spermatogenesis arrest by histological evaluation
Microdeletion Y chromosome AZFb
Interstitial deletion of AZFb Y region (deletions P5/proximal-P1); RBMY, CDY, HSFY, PRY
Azoospermia
Microdeletion Y chromosome AZFb-c
Combined deletion AZFb + AZFc (P5/distal-P1 or P4/distal-P1)
✓: donor
NA
NA
Azoospermia; Sertoli cell syndrome type I diagnosis by histological evaluation (i.e., complete absence of germ cells in seminiferous tubules)
Microdeletion Y chromosome AZFa
Deletion of AZFa Y (recombination between HERV15yq1 and HERV15yq2)
Database sources: NIH, OMIM and OrphaNet
AZF, azoospermia factor; ✓, yes; ✗, no; NA, not applicable; donor, heterologous fertilization with sperm donor; STS, sequence tagged sites
Table 2
The chromosome aberrations related to pretesticular male infertility: from the first observation to the report
Indications for genetic test
Genetic condition
Frequency
Test
Chromosome/genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Hypergonadotropic hypogonadism, ↑FSH ↑LH ↓T, azoospermia, oligozoospermia; small testes, infertility, gynecomastia; neurocognitive deficits; metabolic syndrome, type 2 diabetes. Approximately 10% of these subjects have spermatozoa in the ejaculate, and in 30–50% of cases there is intratesticular spermatogenesis
Klinefelter’s syndrome
1/660 newborns; > 5% in severe oligozoospermia; 10% in azoospermia
Karyotype
47,XXY (85–90%)
46,XY/47,XXY mosaicism (6–7%)
46,XX/47,XXY or multiple X aneuploidy (3–8%)
✓ Testicular sperm retrieval + ICSI
De novo mutation
NA
46,XX testicular DSD
[11, 12]
Short stature; gynecomastia, male external genitalia, small testes, cryptorchidism, hypospadias, infertility, ↑FSH ↑LH↓T; azoospermia/oligozoospermia
Nonsyndromic 46,XX Testicular Disorders of Sex Development (De la Chapelle syndrome)
1/20.000; 0,9% in azoospermia; 1–3% normospermia
FISH or CMA
SRY+ XX (80–90%)
✗ Testicular sperm retrieval; ✓ heterologous fertilization
AD
Syndromic forms of 46,XX testicular DSD; 45X/46,XY; 47,XXY; 46,XX; sex chromosome mosaicisms; Prenatal exposure of 46,XX fetuses to androgens
[13]
Penoscrotal hypospadias, cryptorchidism, infertility; ↑FSH ↑LH↓T; azoospermia/oligozoospermia
SRY XX (< 10%)
Unknown
Short stature; small testes, infertility; ↑FSH ↑LH↓T; azoospermia/oligozoospermia
CMA or molecular diagnostic by PCR
CNV or rearrangements in SOX9, SOX3, RSPO1 and WNT4 (rare)
✓AD for SOX9; AR for RSPO1 or WNT4
46,XX; 46,XY disorders of sex development
[1418]
Tall stature, delayed development of speech, language or motor skills, autism spectrum disorder, hypotonia, motor tics, clinodactyly, scoliosis, attention deficit hyperactivity disorder; ↑FSH normal or ↓T; from normal to azoospermia; from 0.57 to 77.8% sperm mosaicism, a- or hyper diploidy
Double Y syndrome (Jacobs syndrome)
1/1.000; 0.4% in oligozoospermia
Cytogenetics tests
47,XYY; 46,XY/47,XYY mosaics
✓IVF or ICSI in case of oligospermic patients
Does not have a clear pattern of inheritance
46,XY
[14, 19]
Subfertility or uneventful andrological history; oligozoospermia
Balanced structural chromosome aberrations
5% of infertile men
FISH
t(SRY; X); der(13, 14); der(14, 21); der(14, 15)
NA
✓ PGT
Other causes of oligozoospermia
[20]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; ICSI, intracytoplasmic sperm injection; IVF, in vitro fertilization; FISH, fluorescence in situ hybridization; PGT: preimplantation genetic testing; CMA, chromosomal microarray analysis

Male genetic infertility

Genetic factors have been found in all the etiological categories of male fertility (pre-testicular, testicular and post-testicular): OMIM (Online Mendelian Inheritance in Man) reports more than 200 genetic conditions related to male infertility, ranging from the most common clinical presentations of infertility to the rarest complex syndromes in which signs and symptoms are beyond the reproductive problems [103]. In most cases, infertility is only one of the clinical signs of a complex syndrome; on the contrary, in some genetic conditions, infertility is the main phenotypic feature. Moreover, it is important to monitor these infertile patients over time because a greater morbidity and a lower life expectancy have been described for these infertile patients than for the general population and are most likely caused by the genetic abnormalities involved in male sterility [104].
Today, the presence of alterations in the spermiogram is the first indication for genetic tests, particularly in cases of severe oligospermia (< 5 million/ml) (further parameters are hormonal levels, malformations, recurrent abortions, and family history) [105]. Interestingly, a recent study by Oud et al. highlighted how the number of genes that are definitively linked to the more common phenotypes of oligozoospermia or azoospermia remains limited (50%); the other half are genes involved in teratozoospermia, although the monomorphic forms of teratozoospermia are extremely rare [106].
Genetic disorders related to male infertility include whole chromosomal aberrations (structural or numerical), partial chromosomal aberrations (i.e., microdeletions of the Y chromosome) (listed in Tables 1, 2) and monogenic diseases (listed in Tables 3, 4, 5). In particular, abnormalities in sex chromosomes have a greater impact on spermatogenesis, while mutations affecting autosomes are more related, for example, to hypogonadism, teratospermia or asthenozoospermia and to familial forms of obstructive azoospermia.
Table 3
The genetic causes related to pretesticular male infertility: from the first observation to the report
Main indications for genetic test
Hypogonadotropic hypogonadism (CHH)
Other indications for genetic test
Genetic disorder
Frequency
Genetic test
Genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Lack of puberty; micropenis, cryptorchidism; prepubertal testicular volume, absence of secondary sexual features, decreased muscle mass, diminished libido, erectile dysfunction, infertility, low testosterone, estradiol
Kallmann syndrome (olfactogenital syndrome with ano- or hyposmia azoospermia)
Prevalence: 1/30,000; incidence: 1/8000
Molecular diagnosis
ANOS1
X-linked
Syndromes associated with hypogonadotropic hypogonadism
[21, 22]
CHD7, FGFR1, FGF8, SOX10
AD
FEZF1, PROK2, PROKR2
AR
Obesity, retinitis pigmentosa, postaxial polydactyly, kidney dysfunction, behavioral dysfunction; infertility
Bardet–Biedl syndrome (Laurence–Moon–Biedl syndrome)
1:100,000 North America; 1:160,000 Switzerland; 1:17,500 Newfoundland; 1:13,500 Bedouin, Kuwait
Multigene panel
From BBS1 to BBS19
AR
McKusick–Kaufman syndrome (MKS) 
[21, 2325]
Adrenal insufficiency; cryptorchidism, delayed puberty, infertility
X-linked adrenal hypoplasia congenita
 1:12,500 
Molecular diagnosis
NR0B1
X-linked recessive pattern
21-hydroxylase deficiency; 11-hydroxylase deficiency
[26]
Diabetes mellitus, hypothyroidism, alopecia totalis, long, triangular face, hypertelorism; dystonias, dysarthria, dysphagia; infertility
Woodhouse–Sakati syndrome (diabetes-hypogonadism-deafness-intellectual disability syndrome)
Unknown
Molecular diagnosis
DCAF17
AR
Perrault syndrome; Deafness and hereditary hearing loss; Gonadotropin-releasing hormone deficiency
[27]
Adult-onset neurodegenerative disorder; hypogonadotropic hypogonadism
Gordon Holmes syndrome (cerebellar ataxia and hypogonadotropic hypogonadism)
Unknown
Molecular diagnosis
RNF216, PNPLA6 
AR
Cerebellar ataxia
[2830]
Cirrhosis, diabetes, cardiomyopathy, arthritis, skin hyperpigmentation; elevated serum transferrin-iron saturation (TS); elevated serum ferritin concentration; infertility
Hemochromatosis (Hemochromatosis Type 1, HFE-Associated Hemochromatosis, HFE-HH)
2–5:1000 northern European ancestry; 1:200–400 non-Hispanic whites, North America
Gene-targeted or molecular diagnosis
HFE (typically p.Cys282Tyr and p.His63Asp can be performed first)
AR
NA
Rarer primary iron overload disorders and secondary iron overload disorders
[3134]
Azoospermia/oligozoospermia; ↑LH, normal T, hyperandrogenism; feminization of the external genitalia at birth, abnormal secondary sexual development in puberty, and infertility
Androgen insensitivity syndrome (AIS)
2–5:100,000
Screening for AR mutations (> 300)
AR
Donor
X-linked recessive
NA
MRKH syndrome; Hypospadias; MAIS; Undermasculinization of external genitalia and pubertal undervirilization
[35, 36]
Glucocorticoid and mineralocorticoid deficiencies; hypospadias; ambiguous genitalia, infertility
3-β-hydroxysteroid dehydrogenase (HSD) deficiency
Unknown
Molecular diagnosis
HSD3B2 
Donor
AR
NA
Ambiguous genitalia
[37]
Deficiencies in GH,TSH, LH, FSH, PrL, and ACTH; hypothyroidism; neonatal hypoglycemia; micropenis without hypospadias, with or without cryptorchidism; short stature and delayed bone maturation; absent/delayed/incomplete secondary sexual development, infertility
PROP1-related combined pituitary hormone deficiency
1:4000 in England and the US
Molecular diagnosis
PROP1 
AR
CPHD; isolated growth hormone deficiency; isolated hypogonadotropic hypogonadism
[38, 39]
Ambiguous genitalia or external genitalia that appear female; micropenis and hypospadias; not much facial or body hair; infertility
5-Alpha reductase deficiency (familial incomplete male pseudohermaphroditism, type 2)
Unknown
Molecular diagnosis
SRD5A2 
AR
Ambiguous genitalia
[4042]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; donor, heterologous fertilization with sperm donor
Table 4
The genetic causes related to testicular male infertility: from the first observation to the report
Indications for genetic test
Genetic disorder
Frequency
Genetic test
Chromosome/genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Maldescended testes
 Absence of one or both testes from the scrotum; nonobstructive azoospermia; hypogonadotropic hypogonadism
Cryptorchidism
2%; 20% of infertile men; 30/80% of azoospermia 
Molecular diagnosis
INSL3; LGR8
AD
Hypogonadotropic hypogonadism; Noonan and Prader–Willi syndrome
[43, 44]
 Hypertension, hypokalemic alkalosis; lack of secondary sexual characteristics; testicular feminization
17 alpha(α)-hydroxylase/17,20-lyase deficiency
1 in 1 million 
Molecular diagnosis
CYP17A1
Donor
AR
NA
Ambiguous genitalia
[45]
 Severe muscular hypotonia, genital hypoplasia, incomplete pubertal development, infertility; cryptorchidism (93%); obesity, mental retardation (late onset)
Prader–Willi syndrome (PWS, Prader–Labhart–Willi syndrome)
1:10,000 to 1:30,000
DNA methylation testing; Cytogenetic/FISH/chromosomal microarray findings: deletion in bands 15q11.2-q13 (70%)
15q11.2 region
Donor
Paternal deletion; maternal uniparental disomy15
Cryptorchidism; Craniopharyngioma
[21, 4648]
 Short stature, facial dysmorphism, congenital heart defects, skeletal defects, webbed neck, mental retardation, bleeding diathesis; early onset
Noonan syndrome-1 (NS1)
1:1000–2500
Gene sequencing starting with PTPN11, followed by SOS1, KRAS and RAF1
PTPN11 (> 50%), SOS1 (10–15%), KRAS (5%), RAF1 (3–17%)
AD
Turner syndrome; cryptorchidism; azoospermia
[49]
 Gonadal dysgenesis, ambiguous genitalia, infertility; increased risk of Wilms tumor
Denys–Drash syndrome
Unknown
Molecular diagnosis
WT1
AD
Frasier syndrome
[50, 51]
 Atrophy of the abdominal muscles, malformations of the urinary tract
Prune–belly syndrome (other names Syndrom of Eagle–Barret; syndrom of Obrinsky)
1/35,000 and 1/50,000 births and 95% of cases occur in males
Molecular diagnosis
CHRM3
NA
NA
Megacystis/megaureter or posterior urethral valves
[52, 53]
 Osteoporosis; hyperglycemia; ambiguous genitalia
Aromatase deficiency
Unknown
Molecular diagnosis
 CYP19A1 
AR
Other condition of estrogen deficiency
[54, 55]
 Proportionate short stature, delayed closure of fontanelles, prominent forehead, drooping shoulders, abnormal dental development; early onset
Cleidocranial dysplasia
1:1,000,000
Molecular diagnosis
RUNX2 (CBFA1)
AD; de novo pathogenic variant
Pycnodysostosis; mandibuloacral dysplasia; CBFB
[21, 56]
Syndromic without maldescended testes
 Short stature, telangiectatic erythematous skin lesions, high risk for malignancies; early onset; azoospermia or severe oligospermia
Bloom’ s syndrome (Bloom–Torre–Machacek syndrome)
Rare disorder
Molecular diagnosis
BLM 
AR
RECQ-mediated genome instability; Ataxia–telangiectasia; Fanconi; anemia; Nijmegen breakage syndrome; Werner syndrome
[21, 57]
 Short stature, macrocephaly, distinctive face (small, triangular face with prominent forehead, narrow chin, small jaw), delayed development, speech and language problems, learning disabilities; digestive system abnormalities; micropenis; early onset
Russel–Silver syndrome
Prevalence: unknown; estimated incidence ranges from 1 in 30,000–1 in 100,000 people
Methylation 
Methylation involving H19 and IGF2
Sporadic; uniparental disomy
Usually not possible
Intrauterine growth retardation and short stature
[21, 58, 59]
 Keratoconus, glaucoma, and myopia as well as from malformations of the brain, skeleton, and kidney; impairment of respiratory functions; infertility (asthenozoospermia and abnormal flagellar morphology)
Primary ciliary dyskinesia (PCD)
Prevalence: 1:16,000; 1:400 in a Volendam population residing in a fishing village of North Holland 
Molecular diagnosis
DNAH5 (30%), DNAI1 (10%) and TXNDC3, DNAH11, DNAI2 (rare); 60% gene loci unknown
ICSI
AR
Chronic sinopulmonary disease and bronchiectasis
[21, 60]
 Multisystem disorder affecting the skeletal and smooth muscles, the heart, the eyes, and the endocrine and central nervous systems. Mental retardation; infertility
Myotonic dystrophy 1 (Morbus Curschmann–Steinert, Dystrophia myotonica 1, DM1)
1 in 8000
Molecular diagnosis of the CTG repeat expansion in the DMPK gene (> 50 CTG repeats result in DM1)
DMPK
AD
Prader–Willi syndrome, nemaline myopathy, X-linked centronuclear myopathy; DM2; Hereditary distal myopathies; Hereditary myotonia
[21, 61]
Bone marrow failure, hypopigmentation, short stature, physical abnormalities, organ defects (gastrointestinal abnormalities; heart defects; and eye abnormalities, malformed ears and hearing loss); increased risk of certain cancers; and malformations of the reproductive system and infertility
Fanconi anemia
1 in 160,000 (more common among people of Ashkenazi Jewish descent, the Roma population of Spain, and black South Africans)
Molecular diagnosis
FANCA, FANCC and FANCG (90%)
NA
AR; AD:RAD51-related FA; X-linked: FANCB-related FA
Bloom syndrome; ataxia–telangiectasia; NBS; Seckel syndrome; neurofibromatosis 1
[62, 63]
Nonsyndromic infertility
 Abnormal sperm cells (round head and no acrosome) and infertility
Globozoospermia (spermatogenic failure 9)
Rare (1:65,000); common in North Africa: 1:100 cases of male infertility
Molecular diagnosis of DPY19L2, followed by SPATA16
DPY19L2 homozygous deletion, point mutations; SPATA16
✓ ICSI + AOA
AR
Spermatogenic failure
[64, 65]
 Abnormal sperm cells (abnormally large and misshapen heads, contains extra chromosomes; multiple flagella, most often four) and infertility
Macrozoospermia (spermatogenic failure 5)
Unknown;1:10,000 males in North Africa
Molecular diagnosis
AURKC mutations (c.144delC, 85%; p.Y248, DR 13%)
Donor
AR
NA
Spermatogenic failure
[6668]
 Primary infertility; multiple morphological abnormalities of sperm flagella (absent, short, coiled, bent, and irregular flagella); asthenozoospermia
Multiple morphological abnormalities of the sperm flagella (spermatogenic failure 18)
Unknown
Molecular diagnosis
DNAH1 mutation (c.8626-1G > A; c.3860 T > G)
✓ ICSI
AR
NA
Ciliary dyskinesia primary
[69]
 Genital abnormalities; hypoplasia of Leydig cells; micropenis, hypospadias, bifid scrotum, ambiguous genitalia
Leydig cell hypoplasia (hypergonadotropic hypogonadism due to LHCGR defect)
Unknown
Molecular diagnosis
LHCGR
Donor
AR
Hypergonadotropic hypogonadism
[70, 71]
 Asthenozoospermia; absence of any other symptoms
CATSPER-related nonsyndromic male infertility
Unknown
Molecular diagnosis
CATSPER1, GALNTL5
Donor
AR
Male infertility
[69, 72, 73]
 Normal general physical examination, absence of clinical findings involving other organ systems; typical female external genitalia, uterus and fallopian tubes normally formed, gonadal dysgenesis; skeletal abnormalities, campomelic dysplasia
Swyer syndrome (46,XY complete gonadal dysgenesis)
1 in 80,000
Molecular diagnosis
SRY (15%); MAP3K1 (18%); DHH and NR5A1 (rare)
ART
De novo; rare AD
Ambiguous genitalia and/or sex chromosome-phenotype discordance
[13]
 Asthenozoospermia; hearing loss
Deafness-infertility syndrome (DIS)
Unknown
CMA/array-CGH
Homozygous deletion at 15q15.3 including CATSPER2, STRC
Donor
AR
 DFNB16
[74, 75]
Nonobstructive azoospermia
 Small testes and infertility, with severe oligozoospermia or nonobstructive azoospermia due to maturation arrest at the primary spermatocyte stage
Meiotic arrest at primary spermatocyte stage (spermatogenic failure 25)
Unknown
Molecular diagnosis
TEX11
Donor
X-linked
NA
Spermatogenic failure
[7678]
 Nonobstructive azoospermia, infertility, testicular biopsy showing absence of spermatogenic cells and a Sertoli cell-only pattern
Spermatogenic failure 32
Unknown
Molecular diagnosis
SOHLH1
Donor
AD
NA
Spermatogenic failure
[79]
 Azoospermia; testicular histology showing arrest of spermatogenesis at the pachytene stage of primary spermatocytes
Spermatogenic failure 4 (SPGF4)
1%
Molecular diagnosis
SYCP3 (COR1
RPRGL4
SCP3
SPGF4)
Donor
AD
NA
Spermatogenic failure
[80]
 Azoospermia or oligozoospermia
Spermatogenic failure, Y-linked 2
Unknown
Molecular diagnosis
RBMY1A1, DAZ1–4
Donor
Y-linked
NA
Spermatogenic failure
[81, 82]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; donor, heterologous fertilization with sperm donor; AOA, assisted ovarian activation, CMA, chromosomal microarray analysis
Table 5
The genetic causes related to posttesticular male infertility: from the first observation to the report
Main indications for genetic test
Obstructive azoospermia or severe oligospermia
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Genetic disorder
Frequency
Genetic test
Genetic alteration
Abnormalities of seminal vesicles or absence of vas deferens; normal testicular development and function; normal spermatogenesis; a low volume of ejaculated semen with a specific profile (volume < 1.5 ml, ph < 7.0, elevated citric acid concentration, elevated acid phosphatase concentration, low fructose concentration, and failure to coagulate)
Congenital bilateral absence of the vas deferens (CBAVD)
25%; 1–2% in infertility
Screening for CFTR mutations
Two CFTR pathogenic variants identified (46%); one CFTR pathogenic variant identified (79%)
✓ ICSI
AR
Young syndrome; Hereditary urogenital dysplasia
[8385]
Multisystem disease affecting epithelia of the respiratory tract, exocrine pancreas, intestine, hepatobiliary system, and exocrine sweat glands; obstructive azoospermia and male infertility
Cystic fibrosis
1:3200; CF occurs with lower frequency in other ethnic and racial populations (1:15,000 African Americans, and 1:31,000 Asian Americans)
Screening for CFTR mutations
Two CFTR pathogenic variants identified
✓ ICSI
AR
Asthma; congenital airway anomalies; primary ciliary dyskinesia; Shwachman–Diamond syndrome; Bronchiectasis with or without elevated sweat chloride; Isolated hyperchlorhidrosis; Congenital bilateral absence of the vas deferens (CBAVD)
[84, 86]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; ICSI, intracytoplasmic sperm injection
Currently, the main genetic tests routinely used for the diagnosis of male infertility are the karyotype, the study of chromosome Y microdeletions, and the analysis of the CFTR gene. Since it has been reported that several mutated are related to male infertility, it is not surprising that in ~ 40% of all cases of male infertility, the underlying genetic pathogenesis remains unknown [107, 108]. It must also be considered that the role of de novo mutations should be further investigated, especially in light of what happens for Klinefelter syndrome and AZF deletions that occur almost exclusively de novo [106]. Therefore, to improve and personalize the entire diagnostic–therapeutic pathway of male infertility, targeted genetic tests should be performed in the presence of specific clinical pictures, always after appropriate genetic counselling: (1) for diagnostic purposes, (2) during clinical decision-making to establish the most appropriate ART strategy (for example, in the presence of deletions of the AZFa and AZFb regions, the possibility of sperm recovery using testicular biopsy is extremely low), and (3) for prognostic purposes (to establish the risk of transmitting the pathology and plan a prenatal or preimplantation diagnostic procedures).
Whole chromosomal aberrations The prevalence of chromosomal alterations varies from 1.05 to 17% (this gap depends on the characteristics of the studied group) but is 0.84% in newborns [109]. Structural chromosomal rearrangements are more common with respect to numerical abnormalities; this does not apply to sex chromosomes whose abnormalities, accounting for approximately 4.2% of all whole chromosomal aberrations, are represented by sex chromosome aneuploidies in 84% of cases and by structural rearrangements of chromosome Y in the remaining 16% of cases. Klinefelter syndrome (karyotype 47, XXY) is the most frequent type of sex chromosome aneuploidy detected in infertile men [11, 12]; the second most frequent gonosomal abnormality is Double Y syndrome or Jacobs syndrome, characterized by the presence of Y chromosome disomy [14, 110]. In addition to reduced reproductive potential, carriers of chromosomal abnormalities have an increased risk of abortion or generate a child with an abnormal karyotype. For this reason, Table 1 shows the main chromosomal aberrations that could interfere with healthy reproduction, the relative information on the phenotypic aspect, the laboratory tests to highlight them and the indications for antenatal genetic testing.
Partial chromosomal aberrations Microdeletions in the long arm of the Y chromosome (Yq), named the AZF (Azoospermia Factor) region, have been found in 8–12% of azoospermic men and 3–7% of oligozoospermic men [106], resulting in the most common molecular genetic cause of male infertility [110]. The AZF region includes three groups of genes (AZFa, AZFb and AZFc) that are most responsible for spermatogenesis, so partial or complete deletions in this area may impair reproductive capacity. Indications for AZF deletion screening are based on sperm count (< 5 × 106 spermatozoa/ml) associated with primary testiculopathy, and ICSI is required to overcome infertility [111].
Male offspring will carry the same father’s Yq microdeletions or even a worse one; therefore, genetic counselling is mandatory [112]. Parents should be aware of the risk of having a child affected by Turner’s syndrome (45, X0) or other phenotypic anomalies associated with sex chromosome mosaicism [113].
The rearrangement of the AZFc zone is responsible for 60% of all Yq microdeletions [114]. The AZFc region (3.5 Mb) contains several copies of five repeats (b1, b2, b3, b4, and gr), whose similarity and large size predispose an individual to a relatively high incidence of de novo deletions via homologous recombination [115]. The most common is the loss of the whole b2/b4 region, which includes the DAZ family (Deleted in Azoospermia) and leads to spermatogenesis deterioration [115, 116]. More details about AZF are reported in Table 1.
Single gene mutations This section will focus on the noteworthy single gene disorders that have clinical relevance for male infertility (Tables 3, 4, 5). Although thousands of genes are involved in male infertility, today, only a handful of genetic diseases are routinely investigated (e.g., cystic fibrosis) [117, 118]. As shown by several studies, the approaches to identify a single causative gene are not useful considering that more than 2300 genes are expressed in the testis alone and that hundreds of them influence reproductive functions and can contribute to male infertility. Even if nearly 50% of infertility cases are due to single or multiple genetic defects, the genetic causes remain unexplained for 20% of patients [3]. Furthermore, the increasingly widespread use of tools, such as NGS (next-generation sequencing), for both diagnostic and research purposes will allow us to rapidly expand our knowledge of this field [4].
Starting from the clinical and laboratory evaluation, as shown in Tables 3, 4, 5, the main genetic conditions that could interfere with healthy reproduction are reported with the aim of improving the targeted genetic test in the presence of specific clinical pictures.

Female genetic infertility

In contrast to male infertility, little is known about the genetic bases of female infertility. Accordingly, fewer specific tests are routinely recommended to infertile females to investigate the presence of chromosomal disorders or single-gene defects related to their clinical phenotypes. Indeed, isolated infertility due to genetic causes is rare; more commonly, syndromic diseases contribute to female infertility. To date, genetic tests are mainly used for patients with POI, limited to chromosomal aberrations and FMR1 premutations. We therefore focused on the description of these two conditions; however, as shown in Tables 6, 7, more details have been reported concerning the main chromosomal and genetic alterations that could interfere with healthy reproduction; for each of them, the main phenotypic presentations and the laboratory tests that are available in the pre- and postnatal periods are reported.
Table 6
The genetic causes related to ovarian female infertility: from the first observation to the report
 
Indications for genetic test
Genetic disorder
Frequency
Genetic test
Chromosome/genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
POI
Short stature, skeletal abnormalities, kidney problems, webbed neck, lymphedema; ovarian hypofunction or premature ovarian failure, infertility
Turner (45,X) (other names monosomy X, TS)
1 in 2500 
Karyotype
Monosomy X: 45,X0
✓-donor
Not inherited
NA
POF
[87]
Asymptomatic (only 10% of individuals with trisomy X are actually diagnosed); tall stature, epicanthal folds, hypotonia and clinodactyly; renal and genitourinary abnormalities; psychological problems
Trisomy X
1/1000
Karyotype
47XXX or mosaic
NA
  
Irregular menstrual cycles, early menopause, premature ovarian failure, infertility
Fragile X-associated primary ovarian insufficiency (premature ovarian failure 1)
1 in 200 (4/6% of all cases of POI)
Molecular diagnosis of premutations in the FMR1 gene on chromosome Xq27.3 (CGG segment is repeated 55 to 200 times)
FMR1 gene
✓-donor
 X-linked
POF
[88]
Hypogonadotropic hypogonadism; hypotonia, poor feeding, vomiting, weight loss, jaundice; impaired growth, cognitive deficit and cataracts
Galactosemia (galactose-1-phosphate uridyltransferase deficiency)
prevalence unknown; incidence 1/40,000–60,000
Molecular diagnosis
GALT, GALK1, and GALE genes (9p13, 17q24, 1p36) 
AR
POF
 
Chronic mucocutaneous candidiasis, hypoparathyroidism and autoimmune adrenal failure; early onset
Autoimmune polyglandular syndrome (types 1)
Prevalence: 1–9 in 1,000,000; 1/25,000 in Finland
Molecular diagnosis
AIRE gene (21q22.3)
AR
IPEX syndrome; autoimmune polyendocrinopathy type 2
 
Hypertension, hypokalemia; abnormal sexual development, amenorrhea, infertility
17α-hydroxylase deficiency
1 in 1 million 
Molecular diagnosis
CYP17A1 gene
Donor
AR
NA
Severe congenital adrenal hyperplasias
[45]
Mineralization of bones and osteoporosis; hyperglycemia; ambiguous genitalia, ovarian cysts early in childhood, anovulation; hirsutism
Aromatase deficiency
unknown
Molecular diagnosis
 CYP19A1 gene
Donor
AR
NA
PCOS 
[62, 63]
Ophthalmic disorder associated with premature ovarian failure; early onset
Blepharophimosis, ptosis, epicanthus inversus syndrome type I (BPES, type I)
Prevalence: 1–9/100 000
Molecular diagnosis
FOXL2 gene
AD or de novo
PCOS 
[89]
Pre- and postnatal growth retardation, facial sun-sensitive telangiectatic erythema, increased susceptibility to infections, and predisposition to cancer
Bloom syndrome
Unknown; 1/48,000 among people of Ashkenazi Jewish descent
Cytogenetic or molecular diagnosis
15q26.1; BLM gene 
AR
Silver–Russell syndrome, Rothmund–Thomson syndrome, ataxia–telangiectasia, Cockayne syndrome, and Nijmegen breakage syndrome
 
Ovulation disorders (not POI)
Hypergonadotropic amenorrhea; lack of puberty; absence of secondary sexual features, decreased muscle mass, diminished libido, infertility
Kallmann
prevalence: 1/30,000; incidence: 1/8,000
Molecular diagnosis
Type 1: ANOS1
Type 2 and 6: CHD7, FGFR1, FGF8 and SOX10
Type 3: FEZF1, PROK2, PROKR2
X-linked
AD
AR
Syndromes associated with hypogonadotropic hypogonadism
 
Diabetes mellitus, hypothyroidism, alopecia totalis, long, triangular face, hypertelorism; dystonias, dysarthria, dysphagia
Woodhouse–Sakati syndrome
Unknown
Molecular diagnosis
DCAF17 gene
Donor
AR
NA
Diabetes; hypogonadism; deafness-intellectual disability
[27]
Hearing loss; intellectual disability, ataxia, peripheral neuropathy; ovarian dysgenesis, primary amenorrhea, primary ovarian insufficiency, normal external genitalia, infertility
Perrault syndrome
Rare
Molecular diagnosis
 TWNK; CLPP; HARS; LARS2; HSD17B4 
Donor
AR
NA
Gonadal dysgenesis; sensorineural deafness
[90]
Gonadal dysgenesis, XX type, with deafness
Ovarian dysgenesis with sensorineural deafness
Primary amenorrhea, infertility, polycystic ovarian syndrome, hirsutism, ambiguous genitalia
Cytochrome P450 oxidoreductase deficiency
Unknown
Molecular diagnosis
POR gene
Donor
AR
NA
PCOS 
[91]
Skeletal abnormalities, craniosynostosis, a flattened mid-face, a prominent forehead, and low-set ears; arachnodactyly, choanal atresia; primary amenorrhea, infertility, polycystic ovarian syndrome, hirsutism, ambiguous genitalia
Antley–Bixler syndrome
Unknown
Molecular diagnosis
FGFR2 gene
Donor
AR
NA
PCOS 
[92]
Obesity, hirsutism, and amenorrhea are clinical correlates of enlarged polycystic ovaries
Polycystic ovary syndrome (PCOS)
6 to 10% of women worldwide
Molecular diagnosis
AOPEP; AR; DENND1A; ERBB4; FSHB; FSHR; FTO; GATA4; HMGA2; INSR; KRR1; LHCGR; RAB5B; RAD50; SUMO1P1; SUOX; THADA; TOX3; YAP1
Does not have a clear pattern of inheritance
NA
Amenorrhea
[93, 94]
Polycystic ovary syndrome 1 (STEIN-LEVENTHAL SYNDROME HYPERANDROGENEMIA)
Molecular diagnosis
PCOS1
AD
NA
Amenorrhea; HYPERANDROGENEMIA
[93, 94]
Hydropic placental villi, trophoblastic hyperplasia, and poor fetal development
Recurrent hydatidiform mole-type 1 (familial recurrent hydatidiform mole, FRHM)
1:250 in eastern Asia
Molecular diagnosis
 NLRP7 gene (55%); KHDC3L gene (5%)
AR
Hydatidiform mole
[95, 96]
Abnormally developed embryo and placenta that result in the formation of hydatidiform moles
Hydatidiform mole
1:1500 in USA
Molecular diagnosis
C11 or F80, MEI1, REC114
AR
FRHM
[95, 96]
Normal general physical examination, absence of clinical findings involving other organ systems; typical female external genitalia, normally formed uterus and fallopian tubes, gonadal dysgenesis; skeletal abnormalities, campomelic dysplasia
Swyer syndrome (46,XY complete gonadal dysgenesis)
1 in 80,000
Molecular diagnosis
SRY (15%); MAP3K1 (18%); DHH and NR5A1 (rare)
ART
De novo; rare AD
Ambiguous genitalia and/or sex chromosome-phenotype discordance
[69]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; POF, premature ovarian failure; PCOS, polycystic ovarian syndrome
Table 7
The genetic causes related to postovarian female infertility: from the first observation to the report
Indications for genetic test
Genetic disorder
Frequency
Genetic test
Genetic alterations
ART
Inheritance
Antenatal test
Differential diagnosis
Refs.
Underdeveloped or absent uterus and abnormalities of other reproductive organs; normal female external genitalia, breasts; hyperandrogenism; facial hirsutism; primary amenorrhea; infertility
Müllerian aplasia and hyperandrogenism (other names: Biason–Lauber syndrome, WNT4 deficiency)
Rare
Molecular diagnosis
WNT4 gene
NA
AD or de novo
Abnormalities of the reproductive system
[9799]
Vagina and uterus to be underdeveloped or absent, although external genitalia are normal, primary amenorrhea
Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome (type 1)
1 in 4500
Molecular diagnosis
ESR1, OXTR, WNT9B
NA
AD
Abnormalities of the reproductive system
[100102]
Underdeveloped or absent vagina and uterus, although external genitalia are normal; primary amenorrhea; unilateral renal agenesis; skeletal abnormalities; hearing loss or heart defects
Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome (type 2)
Bone marrow failure, hypopigmentation, short stature, physical abnormalities, organ defects (gastrointestinal abnormalities; heart defects; eye abnormalities, malformed ears and hearing loss), and an increased risk of certain cancers; abnormal genitalia or malformations of the reproductive system and infertility
Fanconi anemia (Fanconi pancytopenia
Fanconi panmyelopathy)
1 in 160,000 (more common among people of Ashkenazi Jewish descent, the Roma population of Spain, and black South Africans)
Molecular diagnosis
FANCA, FANCC and FANCG (90%)
NA
AR; AD (RAD51-related FA); X-linked (FANCB-related FA).
Bloom syndrome; ataxia–telangiectasia, Nijmegen breakage syndrome (NBS); Seckel syndrome; neurofibromatosis 1; POI
[62, 63]
Database sources: NIH, OMIM and OrphaNet
✓, yes; ✗, no; NA, not applicable; POI, primary ovarian insufficiency
Whole chromosomal aberrations Considering that chromosomal disorders significantly impact fertility and the miscarriage risk, karyotype analysis is always advisable [119]. The most clinically important structural disorders in infertile females are translocations, both reciprocal (exchange of two terminal segments from different chromosomes) or Robertsonian (centric fusion of two acrocentric chromosomes) responsible for blocks of meiosis and structural alterations of the X chromosome. Patients with reciprocal translocations are at a significantly increased risk of infertility, including hypogonadotropic hypogonadism with primary or secondary amenorrhea or oligomenorrhea. The balanced rearrangements do not create health problems for their carriers because they cause neither loss nor duplication of genetic information, but they can give rise to gametes in which the genetic information is unbalanced and can thus become a cause of infertility or multiple miscarriage. Some abnormalities, such as the XXX karyotype, could not be clearly associated with infertility.
Women with a normal karyotype produce a variable percentage of oocytes with chromosomal abnormalities due to errors occurring during crossing-over and/or meiotic nondisjunction [120, 121]. The three main classes of abnormalities are 45X, trisomy and polyploidy. It is well known that these events increase with maternal age [122]. It is possible to analyze gametes or embryos while undergoing ART thanks to PGT. The efficacy of the technique is increased after screening for aneuploid embryos and transferring only euploid embryos [123, 124].
Fragile X syndrome Fragile X syndrome is an autosomal dominant genetic disorder caused by the presence of over 200 repetitions of the CGG triplet sequence in the FMR1 (Fragile X Mental Retardation 1) gene or by a deletion affecting the FMR2 (Fragile X Mental Retardation 2) gene. Carriers of the female FMR1 premutation (when the number of CGG repeats falls between 55 and 200) or FMR2 microdeletion show menstrual dysfunction, diminished ovarian reserve, and premature ovarian failure [125, 126].
In addition to the family history, in the case of women with these clinical manifestations, the possibility of a molecular test should be considered. The most common genetic contributors to POI are X-chromosome-linked defects. In rare cases, the cause is an alteration in an autosomal chromosome [88]. Identifying the mutations in a timely fashion is of paramount importance to managing the reproductive options and, if necessary, choosing a preimplantation genetic diagnosis program: the aim is to identify the specific clinical pictures in which a targeted genetic test could guide a personalized diagnostic–therapeutic treatment approach.

Molecular approaches in the identification of genetic diseases that are transmissible to offspring

It is well known that in 20–25% of cases, perinatal mortality is caused by inherited chromosomal or genetic alterations [127]. Thanks to medical awareness in recent decades, preconception carrier screening has become widely requested. The identification of couples at risk of transmitting a specific inherited disorder to their offspring offers the possibility of making informed reproductive choices to future parents. If the reproductive partner happens to carry a gene alteration for one of the genetic conditions, the pregnancy would be at risk for a child with that disease.
The American College of Obstetricians and Gynecologists has issued standard recommendations for ethnic and general population genetic screening in couples based on reproductive age [128]. Testing is available for more than 2000 genetic disorders, including common diseases, such as sickle-cell anemia, cystic fibrosis, and spinal muscular atrophy, or more complex conditions, such as mental retardation and congenital heart disease.
In this context, genetic counselling is crucial for recognizing the genetic risk, referring patients appropriately and informing patients about genetic issues that are relevant to decision-making [129]. In fact, preconception carrier screening provides genetic information for multiple disorders; thus, all carrier couples can make reproductive decisions based on their results. The tailored genetic test is a crucial tool to improve short-term and long-term outcomes for mothers and their babies [130, 131].
Currently, during the antenatal period, a variety of techniques are available to identify a transmissible disorder to the offspring in the presence of carrier or affected couples. Each of these techniques can be applied only during a specific time period of pregnancy or at different embryo stages in the IVF protocol.
Invasive PND is usually performed on DNA extracted from fetal cells obtained by chorionic villus sampling (CVS) (between the 11th and 13th weeks of gestation) or from amniocytes (from the 15th to the 20th week), and the result is obtained in 7 or 15 days, respectively [132]. The molecular diagnosis for monogenic disease, as we detailed in a previous publication, is carried out by direct mutation analysis when the parental mutations are known or by linkage analysis when the parental mutations are unknown [5, 132]. Paternity verification and contamination analysis are always performed in addition to the specific analytic phases [5].
An increasing amount of interest has been shown regarding the noninvasive prenatal diagnosis (NIPD) of monogenic disease that is able to detect fetal genetic alterations in maternal blood at an early gestational age (approximately 10 weeks). However, although noninvasive prenatal testing (NIPT) of cell-free fetal DNA (cffDNA) for the screening of chromosomes 21, 18, 13, X and Y has been clinically adopted, NIPD remains a challenge [133]. Very recently, NIPD for clinical use has been adopted in cases of sex-linked disorders and RHD [134]. Several studies have tested the application of NIPD in monogenic diseases, such as β-thalassemia, congenital adrenal hyperplasia, and Duchenne and Becker muscular dystrophy [135137]. The disruptive technology of NGS together with the haplotyping strategy is driving the possibility of using NIPD in clinical cases.
PGT has the same diagnostic motivation as the traditional PND, with the advantage of advancing the timing of diagnosis at the embryo stage. Only disease-free embryos are transferred to the mother, avoiding recourse to therapeutic abortion. Even for couples who are able to conceive naturally, PGT requires the application of IVF techniques, including (a) the collection of gametes from both partners; (b) the fertilization of the oocyte by intracytoplasmic sperm injection (ICSI); (c) the embryo biopsy, which allows one or more cells from the blastomere or trophectoderm to be taken 3 or 5 days, respectively, postfertilization; (d) molecular analysis and (e) the embryo transfer.
PGT protocols are set to start from a small amount of biological sample, ranging from 1 to 10 cells from the embryo at the cleavage stage or blastocyst stage. Conflicting opinions are reported on the detrimental effects of embryo biopsy and mosaicism events between cleavage-stage and blastocyst embryos. Linan et al. demonstrated that the concordance of diagnosis in embryos that were double biopsied on D3 and D5 is 67.8% lower than previously reported, supporting the use of blastocyst biopsies instead of cleavage-stage embryo biopsies [138]. Recently, data from the Preimplantation Genetic Diagnosis International Society (PGDIS) in 2018 showed no difference in the detrimental effects between the embryo stages if experienced operators performed the biopsies [139, 140].
PGT includes whole genome amplification (WGA) to obtain a sufficient quantity of genomic DNA for one or more molecular investigations [141, 142]. Several types of WGA can be used depending on the downstream application [142]. The most used technique for PGT is still represented by “multiplex polymerase chain reaction” (PCR) and capillary electrophoresis analysis for the direct identification of the causative mutation of the disease and the analysis of at least two informative polymorphic markers [the most used are the “short tandem repeats” (STR), microsatellites characterized by short tandem nucleotide repeats] or the analysis of at least 3 polymorphic markers in the event that the causative mutation is not known [5, 143]. However, since PGT tailored to a disease is a laborious and expensive procedure, which is time-consuming in the preliminary phase for the study of the family, several laboratories use genome-wide approaches to analyze gene markers throughout the genome. Alan Handyside tested “karyomapping” based on a single nucleotide polymorphism (SNP) array able to determine the genotype of an individual by analyzing thousands of SNPs distributed throughout the genome [144]. The “karyomapping” involves a “linkage” analysis: with a comparison of the SNPs associated with the causative mutation of the disease to be investigated, that are present in the index case and that are therefore in the parents’ chromosomes, to the SNPs present in the embryo cells, it is possible to identify the presence or absence of the mutation carrier [144146]. In addition, the density of the SNPs allows a higher resolution in the case of crossings between chromosomes close to the mutated gene. Finally, it is possible to use “karyomapping” in families with combinations of more monogenic alterations or that require HLA compatibility, truly demanding investigations to be carried out with conventional methods. “Karyomapping”, however, loses its effectiveness when it is not possible to establish which parental allele is linked to the genetic alteration; for quantitative analysis of mitotic abnormalities (mosaicisms), “karyomapping” does not directly analyze the mutation and cannot detect de novo mutations. The aim of most new approaches is to use a targeted SNP analysis to detect single mutations or groups of common mutations combined with quantitative haplotype analysis or chromosome count. Recently, a genome-wide protocol using NGS has been tested for the identification of family mutations together with cytogenetic screening in embryo biopsies [147149]. The protocol is based on an enlarged panel of disease-associated genes (approximately 5000 genes) and enables, in a single workflow, (a) the direct detection of family mutations and the indirect detection through linkage analysis of heterozygous SNPs (PGT-M); (b) a chromosomal translocation (PGT-SR) analysis; and (c) testing for aneuploidies. However, the limitations of a single NGS protocol are related to its inability to detect haploidies, polyploidies, and mosaicisms. In addition, the analysis of consanguineous families is not recommended. Finally, other limitations regarding the limit of detection or the size of the translocation supported by the protocol could be overcome using haplotyping in the presence of the index case.

Molecular approaches for the optimization of art techniques

Human embryos that are developed in vitro show a great deal of acquired chromosomal abnormalities; for this reason, PGT for aneuploidy (PGT-A) has been developed to select euploid embryos that are suitable for transfer [150].
PGT-A is primarily indicated for couples with advanced maternal age, recurrent implantation failure, recurrent abortions, or severe male infertility. Meiotic errors are one of the main causes of the low success rate (~ 30%) of in vitro fertilization techniques. Randomized studies and meta-analyses have shown that the PGT-A technique does not increase the live birth rate but decreases the miscarriage rate and increases the efficiency of IVF techniques [123, 151].
The evolution of PGT-A techniques started with a limited number of chromosomes analyzed by fluorescence in situ hybridization (FISH) in 1995 [152]. It was soon overcome by the analysis of the whole chromosome set by using different genetic platforms, such as metaphase Comparative Genomic Hybridization (mCGH), array-based Comparative Genomic Hybridization (aCGH), single nucleotide polymorphism (SNP) microarray, quantitative polymerase chain reaction (qPCR), and, most recently, NGS.
Currently, the most commonly used technique is NGS. This method involves the amplification of the genome from a single cell by WGA, the preparation of a DNA library, starting directly from the amplified DNA, and the subsequent sequencing of a pool of libraries in parallel, each identified by a specific “barcode” sequence. Finally, an analysis software that compares the sequences obtained in each sample with respect to the “human hap-map reference genome” allows the identification of the possible presence of chromosome aneuploidies [153]. Literature data confirm that NGS can be successfully applied to the diagnosis of a variety of genetic abnormalities, even in single cells isolated from human embryos following WGA, and has numerous advantages over the technologies traditionally used for PGT-A [153156].
However, it was soon clear that the gold standard was to develop a method for the analysis of both monogenic diseases and PGT-A at the same time. Indeed, as previously discussed, the very recent innovation for this purpose is the use of NGS to analyze single gene mutations and chromosomal copy number variations to select euploid disease-free embryos [157]. Currently, only a novel mutation continues to be a challenge [140].

Discussion

Healthy reproduction can be affected by unhealthy environmental and lifestyle factors, increasing paternal and/or maternal age, anatomical or genetic anomalies, systemic or neurological diseases, infections, trauma, and antibody development [158, 159]. As a consequence, infertility can be the result of nongenetic and genetic factors, and it is often multifactorial, polygenic or a combination of both. Presumably, hundreds of genes must interact in a precise manner during sex determination, gametogenesis, complex hormone actions/interactions, embryo implantation, and early development to generate healthy offspring. Indeed, known genetic causes of infertility include chromosomal aberrations, single gene variants and phenotypes with multifactorial inheritance. To date, specific genes and mutations have been confirmed to be associated with infertility phenotypes in males, females or both, and our knowledge regarding the molecular basis of infertility is continually growing. Confirmation of a clinical diagnosis through genetic testing may lead to personalized medical management. Similar clinical symptoms may be the result of different genetic variations. Specifically, in more rare clinical situations, genetic evaluations (counseling and testing) can contribute to the specific identification of the disease or to the confirmation of a suspected diagnosis. The combination of the detailed clinical information provided and the identified genetic cause will allow the development of a personalized diagnostic–therapeutic strategy.
The first stage in assessing a couple with potential fertility problems involves the substantial synergy between the patient’s medical history, physical examination, instrumentation analysis (pelvic ultrasound in women) and laboratory tests (sperm analysis in males) to identify any underlying pathology and possible risk factors. Although this evaluation should be performed simultaneously for both partners, the male partner analysis is required only in 18% of cases [160]. Figure 2 shows how the integration of couples’ information can stratify patients on the basis of different morpho-functional aspects and outline already differential diagnostic–therapeutic approaches. When a complete and simultaneous assessment of the couple is not performed, the fertility prognosis is compromised, and the opportunity to improve the health outcomes of each subject is lost. For this reason, it is essential to accurately collect, for each member of the couple, data regarding the personal history (life habits, smoking, alcohol use, drug use, sports activities, etc.) and the medical history (genital characteristics at birth and in the first years of life, pubertal development, and any past disease affecting the genital system). The medical history evaluation must focus on the presence of possible metabolic, endocrine, and genetic disorders with the aim of highlighting elements that can lead to subsequent, specific diagnostic tests. At this stage, it is important to identify patients who can benefit from predictive genetic testing: the use of targeted multigene panels can become a useful tool that, by allowing the identification of further causes of infertility, may improve genetic and reproductive counselling and patient stratification. In fact, some cases of infertility are due to unknown genetic mutations; for this reason, collecting data is mandatory to increase the detection rate (which is currently 80%) and to reduce the percentage of idiopathic infertility.
Many couples learn that they are at high risk of having a child that is affected by a genetic disease only when the woman is already pregnant or after giving birth. Thus, a detailed family history for a couple planning to undergo ART is a useful tool for identifying genetically at-risk couples and can improve the medical care of these patients. A number of important factors must be considered when collecting a detailed family history in the context of family planning [129]. These include the history of the patient’s pregnancy (e.g., multiple abortions may indicate a chromosomal abnormality), the degree of kinship (particular attention should be given to first- and second-degree relatives who may have a history of mental retardation, learning difficulties, progressive muscle weakness, early cataracts, infertility, motionless birth, recurrent miscarriages, and coagulation disorders), the consanguinity of the couple [129], and the ethnicity of both groups of grandparents. Furthermore, references to other specialists (e.g., fetal maternal medicine, reproductive endocrinology, gynecology) should be considered to exclude maternal/paternal infertility causes. In the case of identification of pathologies, in one or both partners, standard diagnostic algorithms will be used, while in specific situations, integration will need to be made with specific diagnostic procedures that require integration with eligibility criteria to access ART.
NGS-based strategies offer the opportunity not only to optimize genetic testing in reproductive medicine (since in one step, it may be possible to analyze the potential causes of infertility, perform carrier screening, and support ART) but also to tailor the therapeutic decision on the basis of the specific genomic features of the patients. Common observation reveals that patients with the same diagnosis may respond differently to therapies, and it is becoming increasingly evident that this differential responsiveness may be due to specific DNA variations at the genomic level. The challenge for future medicine is to move from a population-based view to an individually based one. Novel technologies are driving this revolution. In the context of reproductive medicine, for example, it has been established that specific single nucleotide variants in the genes encoding for hormonal receptors are able to influence the efficacy of hormonal therapies in inducing ovulation [161]. In addition, other molecular features of the patients should be taken into account before starting therapeutic protocols to avoid potential side effects. An opportunity comes from the example of the BRCA test. Indeed, BRCA1 and BRCA2 are the most common genes related to hereditary breast and ovarian cancers. Molecular screening for these genes has greatly increased in recent years due to technological simplification and the availability of specific drugs that are suitable for patients with mutations in these genes. As a consequence, several affected and nonaffected young females have been found to be carriers of a BRCA mutation. This information should be taken into account when reproductive choices are planned, considering the potential risk of treating these patients with hormonal therapy.

Conclusions

Currently, both the entire diagnostic pathway and the effectiveness of genetic analysis for infertility suffer from an approach that is ineffective: only a few genetic variables are studied, each through a specific molecular diagnostic procedure. This makes the process of genetic investigation fragmented and cumbersome, with a negative impact on the couple, in addition to the psychological distress caused by infertility. Therefore, despite the large potential of genetic tests to provide real insights into infertility causes, this crucial tool is still used without a method tailored to diagnostic needs regarding infertility. However, recent developments in new sequencing technologies have made it possible to compact one or more tests into a single NGS-based analysis, thus reducing diagnostic costs and time. The European Society of Human Genetics (ESHG) and the European Society of Human Reproduction and Embryology (ESHRE) have recently issued a recommendation for the development and introduction of extended carrier screening [162]. Although it is difficult to predict at this time how much the diagnostic yield of genetic tests for the different subtypes of male and female infertility will increase, it is realistic to expect a decrease in the current percentage of idiopathic infertility.
The general state of health in the reproductive environment is gaining increasing attention and clinical relevance. Therefore, reproductive specialists have the task of evaluating infertile couples by considering both their general and reproductive health, since the relative conditions of comorbidity can influence their reproduction. Medicine is undergoing an important transformation from a reactive to a preventive approach: the future will focus on the integrated diagnosis, treatment and prevention of diseases in individual patients.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat World Health Organization. WHO Laboratory Manual for the examination and processing of human semen. Geneva: World Health Organization; 2010. World Health Organization. WHO Laboratory Manual for the examination and processing of human semen. Geneva: World Health Organization; 2010.
3.
Zurück zum Zitat Ferlin A, Arredi B, Foresta C. Genetic causes of male infertility. Reprod Toxicol. 2006;22:133–41.PubMedCrossRef Ferlin A, Arredi B, Foresta C. Genetic causes of male infertility. Reprod Toxicol. 2006;22:133–41.PubMedCrossRef
4.
Zurück zum Zitat Mastantuoni E, Saccone G, Al-Kouatly HB, Paternoster M, D’Alessandro P, Arduino B, et al. Expanded carrier screening: a current perspective. Eur J Obstet Gynecol Reprod Biol. 2018;230:41–54.PubMedCrossRef Mastantuoni E, Saccone G, Al-Kouatly HB, Paternoster M, D’Alessandro P, Arduino B, et al. Expanded carrier screening: a current perspective. Eur J Obstet Gynecol Reprod Biol. 2018;230:41–54.PubMedCrossRef
5.
Zurück zum Zitat Cariati F, Savarese M, D’Argenio V, Salvatore F, Tomaiuolo R. The SEeMORE strategy: single-tube electrophoresis analysis-based genotyping to detect monogenic diseases rapidly and effectively from conception until birth. Clin Chem Lab Med. 2017;56:40–50.PubMedCrossRef Cariati F, Savarese M, D’Argenio V, Salvatore F, Tomaiuolo R. The SEeMORE strategy: single-tube electrophoresis analysis-based genotyping to detect monogenic diseases rapidly and effectively from conception until birth. Clin Chem Lab Med. 2017;56:40–50.PubMedCrossRef
6.
Zurück zum Zitat Griffin DK, Ogur C. Chromosomal analysis in IVF: just how useful is it? Reproduction. 2018;156:F29–50.PubMedCrossRef Griffin DK, Ogur C. Chromosomal analysis in IVF: just how useful is it? Reproduction. 2018;156:F29–50.PubMedCrossRef
7.
Zurück zum Zitat Hussein N, Weng SF, Kai J, Qureshi N. Preconception risk assessment for thalassaemia, sickle cell disease, cystic fibrosis and Tay-Sachs disease. Cochrane Database Syst Rev. 2015;8:1–29. Hussein N, Weng SF, Kai J, Qureshi N. Preconception risk assessment for thalassaemia, sickle cell disease, cystic fibrosis and Tay-Sachs disease. Cochrane Database Syst Rev. 2015;8:1–29.
8.
Zurück zum Zitat Demain LAM, Conway GS, Newman WG. Genetics of mitochondrial dysfunction and infertility. Clin Genet. 2017;91:199–207.PubMedCrossRef Demain LAM, Conway GS, Newman WG. Genetics of mitochondrial dysfunction and infertility. Clin Genet. 2017;91:199–207.PubMedCrossRef
9.
Zurück zum Zitat Committee on Ethics, American College of Obstetricians and Gynecologists, Committee on Genetics, American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 410: ethical issues in genetic testing. Obstet Gynecol. 2008;111:1495–502.CrossRef Committee on Ethics, American College of Obstetricians and Gynecologists, Committee on Genetics, American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 410: ethical issues in genetic testing. Obstet Gynecol. 2008;111:1495–502.CrossRef
10.
Zurück zum Zitat Silber SJ. The Y chromosome in the era of intracytoplasmic sperm injection: a personal review. Fertil Steril. 2011;95:2439–48.PubMedCrossRef Silber SJ. The Y chromosome in the era of intracytoplasmic sperm injection: a personal review. Fertil Steril. 2011;95:2439–48.PubMedCrossRef
11.
Zurück zum Zitat Gravholt CH, Chang S, Wallentin M, Fedder J, Moore P, Skakkebæk A. Klinefelter syndrome: integrating genetics, neuropsychology, and endocrinology. Endocr Rev. 2018;39:389–423.PubMedCrossRef Gravholt CH, Chang S, Wallentin M, Fedder J, Moore P, Skakkebæk A. Klinefelter syndrome: integrating genetics, neuropsychology, and endocrinology. Endocr Rev. 2018;39:389–423.PubMedCrossRef
12.
Zurück zum Zitat Maiburg M, Repping S, Giltay J. The genetic origin of Klinefelter syndrome and its effect on spermatogenesis. Fertil Steril. 2012;98:253–60.PubMedCrossRef Maiburg M, Repping S, Giltay J. The genetic origin of Klinefelter syndrome and its effect on spermatogenesis. Fertil Steril. 2012;98:253–60.PubMedCrossRef
13.
Zurück zum Zitat Barseghyan H, Délot E, Vilain E. New genomic technologies: an aid for diagnosis of disorders of sex development. Horm Metab Res. 2015;47:312–20.PubMedCrossRef Barseghyan H, Délot E, Vilain E. New genomic technologies: an aid for diagnosis of disorders of sex development. Horm Metab Res. 2015;47:312–20.PubMedCrossRef
14.
15.
Zurück zum Zitat Vetro A, Dehghani MR, Kraoua L, Giorda R, Beri S, Cardarelli L, et al. Testis development in the absence of SRY: chromosomal rearrangements at SOX9 and SOX3. Eur J Hum Genet. 2014;23:1025–32.PubMedPubMedCentralCrossRef Vetro A, Dehghani MR, Kraoua L, Giorda R, Beri S, Cardarelli L, et al. Testis development in the absence of SRY: chromosomal rearrangements at SOX9 and SOX3. Eur J Hum Genet. 2014;23:1025–32.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Sutton E, Hughes J, White S, Sekido R, Tan J, Arboleda V, et al. Identification of SOX3 as an XX male sex reversal gene in mice and humans. J Clin Invest. 2011;121:328–41.PubMedCrossRef Sutton E, Hughes J, White S, Sekido R, Tan J, Arboleda V, et al. Identification of SOX3 as an XX male sex reversal gene in mice and humans. J Clin Invest. 2011;121:328–41.PubMedCrossRef
17.
Zurück zum Zitat Moalem S, Babul-Hirji R, Stavropolous DJ, Wherrett D, Bägli DJ, Thomas P, et al. XX male sex reversal with genital abnormalities associated with a de novo SOX3 gene duplication. Am J Med Genet. 2012;158A:1759–64.PubMedCrossRef Moalem S, Babul-Hirji R, Stavropolous DJ, Wherrett D, Bägli DJ, Thomas P, et al. XX male sex reversal with genital abnormalities associated with a de novo SOX3 gene duplication. Am J Med Genet. 2012;158A:1759–64.PubMedCrossRef
18.
Zurück zum Zitat Lim SL, Qu ZP, Kortschak RD, Lawrence DM, Geoghegan J, Hempfling A-L, et al. HENMT1 and piRNA stability are required for adult male germ cell transposon repression and to define the spermatogenic program in the mouse. PLoS Genet. 2015;23(11):e1005620.CrossRef Lim SL, Qu ZP, Kortschak RD, Lawrence DM, Geoghegan J, Hempfling A-L, et al. HENMT1 and piRNA stability are required for adult male germ cell transposon repression and to define the spermatogenic program in the mouse. PLoS Genet. 2015;23(11):e1005620.CrossRef
19.
Zurück zum Zitat Tuerlings JH, de France HF, Hamers A, Hordijk R, Van Hemel JO, Hansson K, et al. Chromosome studies in 1792 males prior to intra-cytoplasmic sperm injection: the Dutch experience. Eur J Hum Genet. 1998;6:194–200.PubMedCrossRef Tuerlings JH, de France HF, Hamers A, Hordijk R, Van Hemel JO, Hansson K, et al. Chromosome studies in 1792 males prior to intra-cytoplasmic sperm injection: the Dutch experience. Eur J Hum Genet. 1998;6:194–200.PubMedCrossRef
20.
Zurück zum Zitat Dul EC, Groen H, van Ravenswaaij Arts CM, Dijkhuizen T, van Echten-Arends J, Land JA. The prevalence of chromosomal abnormalities in subgroups of infertile men. Hum Reprod. 2012;27:36–43.PubMedCrossRef Dul EC, Groen H, van Ravenswaaij Arts CM, Dijkhuizen T, van Echten-Arends J, Land JA. The prevalence of chromosomal abnormalities in subgroups of infertile men. Hum Reprod. 2012;27:36–43.PubMedCrossRef
21.
Zurück zum Zitat Hempel H, Buchholz T. Rare syndromes associated with infertility. J Reproduktionsmed Endokrinol. 2009;6:24–6. Hempel H, Buchholz T. Rare syndromes associated with infertility. J Reproduktionsmed Endokrinol. 2009;6:24–6.
22.
Zurück zum Zitat Quaynor SD, Bosley ME, Duckworth CG, Porter KR, Kim SH, Kim HG, et al. Targeted next generation sequencing approach identifies eighteen new candidate genes in normosmic hypogonadotropic hypogonadism and Kallmann syndrome. Mol Cell Endocrinol. 2016;437:86–96.PubMedCrossRef Quaynor SD, Bosley ME, Duckworth CG, Porter KR, Kim SH, Kim HG, et al. Targeted next generation sequencing approach identifies eighteen new candidate genes in normosmic hypogonadotropic hypogonadism and Kallmann syndrome. Mol Cell Endocrinol. 2016;437:86–96.PubMedCrossRef
23.
Zurück zum Zitat Katsanis N. The oligogenic properties of Bardet–Biedl syndrome. Hum Mol Genet. 2004;13:R65–71.PubMedCrossRef Katsanis N. The oligogenic properties of Bardet–Biedl syndrome. Hum Mol Genet. 2004;13:R65–71.PubMedCrossRef
24.
Zurück zum Zitat Leitch CC, Zaghloul NA, Davis EE, Stoetzel C, Diaz-Font A, Rix S, et al. Hypomorphic mutations in syndromic encephalocele genes are associated with Bardet–Biedl syndrome. Nat Genet. 2008;40:443–8.PubMedCrossRef Leitch CC, Zaghloul NA, Davis EE, Stoetzel C, Diaz-Font A, Rix S, et al. Hypomorphic mutations in syndromic encephalocele genes are associated with Bardet–Biedl syndrome. Nat Genet. 2008;40:443–8.PubMedCrossRef
25.
Zurück zum Zitat Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer BC, Johnson G, et al. The cardinal manifestations of Bardet–Biedl syndrome, a form of Laurence–Moon–Biedl syndrome. N Engl J Med. 1989;321:1002–9.PubMedCrossRef Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer BC, Johnson G, et al. The cardinal manifestations of Bardet–Biedl syndrome, a form of Laurence–Moon–Biedl syndrome. N Engl J Med. 1989;321:1002–9.PubMedCrossRef
26.
Zurück zum Zitat Raffin-Sanson ML, Oudet B, Salenave S, Brailly-Tabard S, Pehuet M, et al. A man with a DAX1/NR0B1 mutation, normal puberty, and an intact hypothalamic–pituitary–gonadal axis but deteriorating oligospermia during long-term follow-up. Europ J Endocr. 2013;168:K45–50.CrossRef Raffin-Sanson ML, Oudet B, Salenave S, Brailly-Tabard S, Pehuet M, et al. A man with a DAX1/NR0B1 mutation, normal puberty, and an intact hypothalamic–pituitary–gonadal axis but deteriorating oligospermia during long-term follow-up. Europ J Endocr. 2013;168:K45–50.CrossRef
27.
Zurück zum Zitat Al-Semari A, Bohlega S. Autosomal-recessive syndrome with alopecia, hypogonadism, progressive extra-pyramidal disorder, white matter disease, sensory neural deafness, diabetes mellitus, and low IGF1. Am J Med Genet. 2007;43A:149–60.CrossRef Al-Semari A, Bohlega S. Autosomal-recessive syndrome with alopecia, hypogonadism, progressive extra-pyramidal disorder, white matter disease, sensory neural deafness, diabetes mellitus, and low IGF1. Am J Med Genet. 2007;43A:149–60.CrossRef
28.
Zurück zum Zitat Husain N, Yuan Q, Yen YC, Pletnikova O, Sally DQ, Worley P, et al. TRIAD3/RNF216 mutations associated with Gordon Holmes syndrome lead to synaptic and cognitive impairments via Arc misregulation. Aging Cell. 2017;16:281–92.PubMedCrossRef Husain N, Yuan Q, Yen YC, Pletnikova O, Sally DQ, Worley P, et al. TRIAD3/RNF216 mutations associated with Gordon Holmes syndrome lead to synaptic and cognitive impairments via Arc misregulation. Aging Cell. 2017;16:281–92.PubMedCrossRef
29.
Zurück zum Zitat Santens P, Van Damme T, Steyaert W, Willaert A, Sablonniere B, De Paepe A, et al. RNF216 mutations as a novel cause of autosomal recessive Huntington-like disorder. Neurology. 2015;84:1760–6.PubMedCrossRef Santens P, Van Damme T, Steyaert W, Willaert A, Sablonniere B, De Paepe A, et al. RNF216 mutations as a novel cause of autosomal recessive Huntington-like disorder. Neurology. 2015;84:1760–6.PubMedCrossRef
30.
Zurück zum Zitat Margolin DH, Kousi M, Chan Y-M, Lim ET, Schmahmann JD, Hadjivassiliou M, et al. Ataxia, dementia, and hypogonadotropism caused by disordered ubiquitination. N Eng J Med. 2013;368:1992–2003.CrossRef Margolin DH, Kousi M, Chan Y-M, Lim ET, Schmahmann JD, Hadjivassiliou M, et al. Ataxia, dementia, and hypogonadotropism caused by disordered ubiquitination. N Eng J Med. 2013;368:1992–2003.CrossRef
31.
Zurück zum Zitat Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A, et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet. 1996;13:399–408.PubMedCrossRef Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A, et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet. 1996;13:399–408.PubMedCrossRef
32.
Zurück zum Zitat Morrison ED, Brandhagen DJ, Phatak PD, Barton JC, Krawitt EL, El-Serag HB, et al. Serum ferritin level predicts advanced hepatic fibrosis among U.S. patients with phenotypic hemochromatosis. Ann Intern Med. 2003;138:627–33.PubMedCrossRef Morrison ED, Brandhagen DJ, Phatak PD, Barton JC, Krawitt EL, El-Serag HB, et al. Serum ferritin level predicts advanced hepatic fibrosis among U.S. patients with phenotypic hemochromatosis. Ann Intern Med. 2003;138:627–33.PubMedCrossRef
33.
Zurück zum Zitat Adams PC, Barton JC, Guo H, Alter D, Speechley M. Serum ferritin is a biomarker for liver mortality in the Hemochromatosis and Iron Overload Screening Study. Ann Hepatol. 2015;14:348–53.PubMedCrossRef Adams PC, Barton JC, Guo H, Alter D, Speechley M. Serum ferritin is a biomarker for liver mortality in the Hemochromatosis and Iron Overload Screening Study. Ann Hepatol. 2015;14:348–53.PubMedCrossRef
34.
Zurück zum Zitat Adams PC, Reboussin DM, Barton JC, McLaren CE, Eckfeldt JH, McLaren GD, et al. Hemochromatosis and iron-overload screening in a racially diverse population. N Engl J Med. 2005;352:1769–78.PubMedCrossRef Adams PC, Reboussin DM, Barton JC, McLaren CE, Eckfeldt JH, McLaren GD, et al. Hemochromatosis and iron-overload screening in a racially diverse population. N Engl J Med. 2005;352:1769–78.PubMedCrossRef
35.
Zurück zum Zitat Boehmer AL, Brinkmann O, Bruggenwirth H, van Assendelft C, Otten BJ, Verleun-Mooijman MC, et al. Genotype versus phenotype in families with androgen insensitivity syndrome. J Clin Endocrinol Metab. 2001;86:4151–60.PubMedCrossRef Boehmer AL, Brinkmann O, Bruggenwirth H, van Assendelft C, Otten BJ, Verleun-Mooijman MC, et al. Genotype versus phenotype in families with androgen insensitivity syndrome. J Clin Endocrinol Metab. 2001;86:4151–60.PubMedCrossRef
36.
Zurück zum Zitat Bianca S, Cataliotti A, Bartoloni G, Torrente I, Barrano B, Boemi G, et al. Prenatal diagnosis of androgen insensitivity syndrome. Fetal Diagn Ther. 2009;26:167–9.PubMedCrossRef Bianca S, Cataliotti A, Bartoloni G, Torrente I, Barrano B, Boemi G, et al. Prenatal diagnosis of androgen insensitivity syndrome. Fetal Diagn Ther. 2009;26:167–9.PubMedCrossRef
37.
Zurück zum Zitat Paula FJ, Dick-de-Paula I, Pontes A, Schmitt FC, Mendonça BB, Foss MC. Hyperandrogenism due to 3 beta-hydroxysteroid dehydrogenase deficiency with accessory adrenocortical tissue: a hormonal and metabolic evaluation. Braz J Med Biol Res. 1994;27:1149–58.PubMed Paula FJ, Dick-de-Paula I, Pontes A, Schmitt FC, Mendonça BB, Foss MC. Hyperandrogenism due to 3 beta-hydroxysteroid dehydrogenase deficiency with accessory adrenocortical tissue: a hormonal and metabolic evaluation. Braz J Med Biol Res. 1994;27:1149–58.PubMed
38.
Zurück zum Zitat Deladoëy J, Flück C, Büyükgebiz A, Kuhlmann BV, Eblé A, Hindmarsh PC, et al. “Hot spot” in the PROP1 gene responsible for combined pituitary hormone deficiency. J Clin Endocrinol Metab. 1999;84:1645–50.PubMed Deladoëy J, Flück C, Büyükgebiz A, Kuhlmann BV, Eblé A, Hindmarsh PC, et al. “Hot spot” in the PROP1 gene responsible for combined pituitary hormone deficiency. J Clin Endocrinol Metab. 1999;84:1645–50.PubMed
39.
Zurück zum Zitat Abrão MG, Leite MV, Carvalho LR, Billerbeck AE, Nishi MY, Barbosa AS, et al. Combined pituitary hormone deficiency (CPHD) due to a complete PROP1 deletion. Clin Endocrinol. 2006;65:294–300.CrossRef Abrão MG, Leite MV, Carvalho LR, Billerbeck AE, Nishi MY, Barbosa AS, et al. Combined pituitary hormone deficiency (CPHD) due to a complete PROP1 deletion. Clin Endocrinol. 2006;65:294–300.CrossRef
40.
Zurück zum Zitat Lee PA, Houk CP, Ahmed SF, Hughes IA, International Consensus Conference on Intersex organized by the Lawson Wilkins Pediatric Endocrine Society and the European Society for Paediatric Endocrinology. Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics. 2006;118:e488–500.PubMedCrossRef Lee PA, Houk CP, Ahmed SF, Hughes IA, International Consensus Conference on Intersex organized by the Lawson Wilkins Pediatric Endocrine Society and the European Society for Paediatric Endocrinology. Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics. 2006;118:e488–500.PubMedCrossRef
41.
Zurück zum Zitat Andersson S, Berman DM, Jenkins EP, Russell DW. Deletion of steroid 5-alpha-reductase 2 gene in male pseudohermaphroditism. Nature. 1991;354:159–61.PubMedPubMedCentralCrossRef Andersson S, Berman DM, Jenkins EP, Russell DW. Deletion of steroid 5-alpha-reductase 2 gene in male pseudohermaphroditism. Nature. 1991;354:159–61.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Hochberg Z, Chayen R, Reiss N, Falik Z, Makler A, Munichor M, et al. Clinical, biochemical, and genetic findings in a large pedigree of male and female patients with 5-alpha-reductase 2 deficiency. J Clin Endocr Metab. 1996;81:2821–7.PubMed Hochberg Z, Chayen R, Reiss N, Falik Z, Makler A, Munichor M, et al. Clinical, biochemical, and genetic findings in a large pedigree of male and female patients with 5-alpha-reductase 2 deficiency. J Clin Endocr Metab. 1996;81:2821–7.PubMed
43.
Zurück zum Zitat Canto P, Escudero I, Soderlund D, Nishimura E, Carranza-Lira S, Gutierrez J, et al. A novel mutation of the insulin-like 3 gene in patients with cryptorchidism. J Hum Genet. 2003;48:86–90.PubMedCrossRef Canto P, Escudero I, Soderlund D, Nishimura E, Carranza-Lira S, Gutierrez J, et al. A novel mutation of the insulin-like 3 gene in patients with cryptorchidism. J Hum Genet. 2003;48:86–90.PubMedCrossRef
44.
Zurück zum Zitat Ferlin A, Simonato M, Bartoloni L, Rizzo G, Bettella A, Dottorini T, et al. The INSL3-LGR8/GREAT ligand-receptor pair in human cryptorchidism. J Clin Endocr Metab. 2003;88:4273–9.PubMedCrossRef Ferlin A, Simonato M, Bartoloni L, Rizzo G, Bettella A, Dottorini T, et al. The INSL3-LGR8/GREAT ligand-receptor pair in human cryptorchidism. J Clin Endocr Metab. 2003;88:4273–9.PubMedCrossRef
45.
Zurück zum Zitat Auchus RJ. Steroid 17-hydroxylase and 17,20-lyase deficiencies, genetic and pharmacologic. J Steroid Biochem Molec Biol. 2017;165:71–8.PubMedCrossRef Auchus RJ. Steroid 17-hydroxylase and 17,20-lyase deficiencies, genetic and pharmacologic. J Steroid Biochem Molec Biol. 2017;165:71–8.PubMedCrossRef
46.
Zurück zum Zitat McCandless SE, Committee on Genetics. Clinical report-health supervision for children with Prader–Willi syndrome. Pediatrics. 2011;127:195–204.PubMedCrossRef McCandless SE, Committee on Genetics. Clinical report-health supervision for children with Prader–Willi syndrome. Pediatrics. 2011;127:195–204.PubMedCrossRef
47.
Zurück zum Zitat Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader–Willi syndrome. Genet Med. 2012;14:10–26.PubMedCrossRef Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader–Willi syndrome. Genet Med. 2012;14:10–26.PubMedCrossRef
51.
Zurück zum Zitat Patek CE, Little MH, Fleming S, Miles C, Charlieu J-P, Clarke AR, et al. A zinc finger truncation of murine WT1 results in the characteristic urogenital abnormalities of Denys–Drash syndrome. Proc Nat Acad Sci. 1999;96:2931–6.PubMedCrossRefPubMedCentral Patek CE, Little MH, Fleming S, Miles C, Charlieu J-P, Clarke AR, et al. A zinc finger truncation of murine WT1 results in the characteristic urogenital abnormalities of Denys–Drash syndrome. Proc Nat Acad Sci. 1999;96:2931–6.PubMedCrossRefPubMedCentral
52.
Zurück zum Zitat Seidel NE, Arlen AM, Smith EA, Kirsch AJ. Clinical manifestations and management of prune–belly syndrome in a large contemporary pediatric population. Urology. 2015;85:211–5.PubMedCrossRef Seidel NE, Arlen AM, Smith EA, Kirsch AJ. Clinical manifestations and management of prune–belly syndrome in a large contemporary pediatric population. Urology. 2015;85:211–5.PubMedCrossRef
53.
Zurück zum Zitat Zugor V, Schott GE, Labanaris AP. The Prune Belly syndrome: urological aspects and long-term outcomes of a rare disease. Pediatr Rep. 2012;4:e20.PubMedPubMedCentralCrossRef Zugor V, Schott GE, Labanaris AP. The Prune Belly syndrome: urological aspects and long-term outcomes of a rare disease. Pediatr Rep. 2012;4:e20.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Conte FA, Grumbach MM, Ito Y, Fisher CR, Simpson ER. A syndrome of female pseudohermaphrodism, hypergonadotropic hypogonadism, and multicystic ovaries associated with missense mutations in the gene encoding aromatase (P450arom). J Clin Endocr Metab. 1994;78:1287–92.PubMed Conte FA, Grumbach MM, Ito Y, Fisher CR, Simpson ER. A syndrome of female pseudohermaphrodism, hypergonadotropic hypogonadism, and multicystic ovaries associated with missense mutations in the gene encoding aromatase (P450arom). J Clin Endocr Metab. 1994;78:1287–92.PubMed
55.
Zurück zum Zitat Jones MEE, Boon WC, McInnes K, Maffei L, Carani C, Simpson ER. Recognizing rare disorders: aromatase deficiency. Nat Clin Pract Endocr Metab. 2007;3:414–21.CrossRef Jones MEE, Boon WC, McInnes K, Maffei L, Carani C, Simpson ER. Recognizing rare disorders: aromatase deficiency. Nat Clin Pract Endocr Metab. 2007;3:414–21.CrossRef
56.
Zurück zum Zitat Guo YW, Chiu CY, Liu CL, Jap TS, Lin LY. Novel mutation of RUNX2 gene in a patient with cleidocranial dysplasia. Int J Clin Exp Pathol. 2015;8:1057–62.PubMedPubMedCentral Guo YW, Chiu CY, Liu CL, Jap TS, Lin LY. Novel mutation of RUNX2 gene in a patient with cleidocranial dysplasia. Int J Clin Exp Pathol. 2015;8:1057–62.PubMedPubMedCentral
57.
Zurück zum Zitat Ling C, Huang J, Yan Z, Li Y, Ohzeki M, Ishiai M. Bloom syndrome complex promotes FANCM recruitment to stalled replication forks and facilitates both repair and traverse of DNA interstrand crosslinks. Cell Discov. 2016;2:16047.PubMedPubMedCentralCrossRef Ling C, Huang J, Yan Z, Li Y, Ohzeki M, Ishiai M. Bloom syndrome complex promotes FANCM recruitment to stalled replication forks and facilitates both repair and traverse of DNA interstrand crosslinks. Cell Discov. 2016;2:16047.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Giabicani E, Boule M, Galliani E, Netchine I. Sleep apneas in Silver Russell syndrome: a constant finding. Horm Res Paediatr. 2015;84(Suppl 1):262. Giabicani E, Boule M, Galliani E, Netchine I. Sleep apneas in Silver Russell syndrome: a constant finding. Horm Res Paediatr. 2015;84(Suppl 1):262.
59.
Zurück zum Zitat Wakeling EL, Brioude F, Lokulo-Sodipe O, O’Connell SM, Salem J, Bliek J, et al. Diagnosis and management of Silver-Russell syndrome: first international consensus statement. Nat Rev Endocrinol. 2017;13:105–24.PubMedCrossRef Wakeling EL, Brioude F, Lokulo-Sodipe O, O’Connell SM, Salem J, Bliek J, et al. Diagnosis and management of Silver-Russell syndrome: first international consensus statement. Nat Rev Endocrinol. 2017;13:105–24.PubMedCrossRef
60.
Zurück zum Zitat Marshall CR, Scherer SW, Zariwala MA, Lau L, Paton TA, Stockley T. Whole-exome sequencing and targeted copy number analysis in primary ciliary dyskinesia. G3 (Bethesda). 2015;5:1775–81.PubMedCentralCrossRef Marshall CR, Scherer SW, Zariwala MA, Lau L, Paton TA, Stockley T. Whole-exome sequencing and targeted copy number analysis in primary ciliary dyskinesia. G3 (Bethesda). 2015;5:1775–81.PubMedCentralCrossRef
61.
Zurück zum Zitat Kamsteeg EJ, Kress W, Catalli C, Hertz JM, Witsch-Baumgartner M, Buckley MF, et al. Best practice guidelines and recommendations on the molecular diagnosis of myotonic dystrophy types 1 and 2. Eur J Hum Genet. 2012;20:1203–8.PubMedPubMedCentralCrossRef Kamsteeg EJ, Kress W, Catalli C, Hertz JM, Witsch-Baumgartner M, Buckley MF, et al. Best practice guidelines and recommendations on the molecular diagnosis of myotonic dystrophy types 1 and 2. Eur J Hum Genet. 2012;20:1203–8.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Kitao H, Takata M. Fanconi anemia: a disorder defective in the DNA damage response. Int J Hematol. 2011;93:417–24.PubMedCrossRef Kitao H, Takata M. Fanconi anemia: a disorder defective in the DNA damage response. Int J Hematol. 2011;93:417–24.PubMedCrossRef
64.
Zurück zum Zitat Ghedir H, Ibala-Romdhane S, Okutman O, Viot G, Saad A, Viville S. Identification of a new DPY19L2 mutation and a better definition of DPY19L2 deletion breakpoints leading to globozoospermia. Mol Hum Reprod. 2016;22:35–45.PubMedCrossRef Ghedir H, Ibala-Romdhane S, Okutman O, Viot G, Saad A, Viville S. Identification of a new DPY19L2 mutation and a better definition of DPY19L2 deletion breakpoints leading to globozoospermia. Mol Hum Reprod. 2016;22:35–45.PubMedCrossRef
65.
Zurück zum Zitat Perrin A, Coat C, Nguyen MH, Talagas M, Morel F, Amice J, et al. Molecular cytogenetic and genetic aspects of globozoospermia: a review. Andrologia. 2013;45:1–9.PubMedCrossRef Perrin A, Coat C, Nguyen MH, Talagas M, Morel F, Amice J, et al. Molecular cytogenetic and genetic aspects of globozoospermia: a review. Andrologia. 2013;45:1–9.PubMedCrossRef
66.
Zurück zum Zitat Ben Khelifa M, Coutton C, Blum MG, Abada F, Harbuz R, Zouari R, et al. Identification of a new recurrent aurora kinase C mutation in both European and African men with macrozoospermia. Hum Reprod. 2012;27:3337–46.PubMedCrossRef Ben Khelifa M, Coutton C, Blum MG, Abada F, Harbuz R, Zouari R, et al. Identification of a new recurrent aurora kinase C mutation in both European and African men with macrozoospermia. Hum Reprod. 2012;27:3337–46.PubMedCrossRef
67.
Zurück zum Zitat Eloualid A, Rouba H, Rhaissi H, Barakat A, Louanjli N, Bashamboo A, et al. Prevalence of the Aurora kinase C c.144delC mutation in infertile Moroccan men. Fertil Steril. 2014;101:1086–90.PubMedCrossRef Eloualid A, Rouba H, Rhaissi H, Barakat A, Louanjli N, Bashamboo A, et al. Prevalence of the Aurora kinase C c.144delC mutation in infertile Moroccan men. Fertil Steril. 2014;101:1086–90.PubMedCrossRef
68.
Zurück zum Zitat Ounis L, Zoghmar A, Coutton C, Rouabah L, Hachemi M, Martinez D, et al. Mutations of the aurora kinase C gene causing macrozoospermia are the most frequent genetic cause of male infertility in Algerian men. Asian J Androl. 2015;17:68–73.PubMedCrossRef Ounis L, Zoghmar A, Coutton C, Rouabah L, Hachemi M, Martinez D, et al. Mutations of the aurora kinase C gene causing macrozoospermia are the most frequent genetic cause of male infertility in Algerian men. Asian J Androl. 2015;17:68–73.PubMedCrossRef
69.
Zurück zum Zitat Amiri-Yekta A, Coutton C, Kherraf Z-E, Karaouzène T, Le Tanno P, Sanatiet MH, et al. Whole-exome sequencing of familial cases of multiple morphological abnormalities of the sperm flagella (MMAF) reveals new DNAH1 mutations. Hum Reprod. 2016;31:2872–80.PubMedCrossRef Amiri-Yekta A, Coutton C, Kherraf Z-E, Karaouzène T, Le Tanno P, Sanatiet MH, et al. Whole-exome sequencing of familial cases of multiple morphological abnormalities of the sperm flagella (MMAF) reveals new DNAH1 mutations. Hum Reprod. 2016;31:2872–80.PubMedCrossRef
70.
Zurück zum Zitat Zenteno JC, Canto P, Kofman-Alfaro S, Mendez JP. Evidence for genetic heterogeneity in male pseudohermaphroditism due to Leydig cell hypoplasia. J Clin Endocr Metab. 1999;84:3803–6.PubMed Zenteno JC, Canto P, Kofman-Alfaro S, Mendez JP. Evidence for genetic heterogeneity in male pseudohermaphroditism due to Leydig cell hypoplasia. J Clin Endocr Metab. 1999;84:3803–6.PubMed
71.
Zurück zum Zitat Wu S-M, Chan W-Y. Male pseudohermaphroditism due to inactivating luteinizing hormone receptor mutations. Arch Med Res. 1999;30:495–500.PubMedCrossRef Wu S-M, Chan W-Y. Male pseudohermaphroditism due to inactivating luteinizing hormone receptor mutations. Arch Med Res. 1999;30:495–500.PubMedCrossRef
72.
Zurück zum Zitat Avenarius MR, Hildebrand MS, Zhang Y, Meyer NC, Smith LL, Kahrizi K, et al. Human male infertility caused by mutations in the CATSPER1 channel protein. Am J Hum Genet. 2009;84:505–10.PubMedPubMedCentralCrossRef Avenarius MR, Hildebrand MS, Zhang Y, Meyer NC, Smith LL, Kahrizi K, et al. Human male infertility caused by mutations in the CATSPER1 channel protein. Am J Hum Genet. 2009;84:505–10.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Takasaki N, Tachibana K, Ogasawara S, Matsuzaki H, Hagiuda J, Ishikawa H, et al. A heterozygous mutation of GALNTL5 affects male infertility with impairment of sperm motility. Proc Natl Acad Sci USA. 2014;111:1120–5.PubMedCrossRefPubMedCentral Takasaki N, Tachibana K, Ogasawara S, Matsuzaki H, Hagiuda J, Ishikawa H, et al. A heterozygous mutation of GALNTL5 affects male infertility with impairment of sperm motility. Proc Natl Acad Sci USA. 2014;111:1120–5.PubMedCrossRefPubMedCentral
74.
Zurück zum Zitat Zhang Y, Malekpour M, Al-Madani N, Kahrizi K, Zanganeh M, Lohr NJ, et al. Sensorineural deafness and male infertility: a contiguous gene deletion syndrome. J Med Genet. 2007;44:233–40.PubMedCrossRef Zhang Y, Malekpour M, Al-Madani N, Kahrizi K, Zanganeh M, Lohr NJ, et al. Sensorineural deafness and male infertility: a contiguous gene deletion syndrome. J Med Genet. 2007;44:233–40.PubMedCrossRef
75.
Zurück zum Zitat Gu X, Guo L, Ji H, Sun S, Chai R, Wang L, et al. Genetic testing for sporadic hearing loss using targeted massively parallel sequencing identifies 10 novel mutations. Clin Genet. 2015;87:588–93.PubMedCrossRef Gu X, Guo L, Ji H, Sun S, Chai R, Wang L, et al. Genetic testing for sporadic hearing loss using targeted massively parallel sequencing identifies 10 novel mutations. Clin Genet. 2015;87:588–93.PubMedCrossRef
76.
Zurück zum Zitat Okutman O, Muller J, Baert Y, Serdarogullari M, Gultomruk M, Piton A, et al. Exome sequencing reveals a nonsense mutation in TEX15 causing spermatogenic failure in a Turkish family. Hum Mol Genet. 2015;24:5581–8.PubMedCrossRef Okutman O, Muller J, Baert Y, Serdarogullari M, Gultomruk M, Piton A, et al. Exome sequencing reveals a nonsense mutation in TEX15 causing spermatogenic failure in a Turkish family. Hum Mol Genet. 2015;24:5581–8.PubMedCrossRef
77.
Zurück zum Zitat Yatsenko AN, Georgiadis AP, Röpke A, Berman AJ, Jaffe T, Olszewska M, et al. X-linked TEX11 mutations, meiotic arrest, and azoospermia in infertile men. N Engl J Med. 2015;372:2097–107.PubMedPubMedCentralCrossRef Yatsenko AN, Georgiadis AP, Röpke A, Berman AJ, Jaffe T, Olszewska M, et al. X-linked TEX11 mutations, meiotic arrest, and azoospermia in infertile men. N Engl J Med. 2015;372:2097–107.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Colombo R, Pontoglio A, Bini M. Two novel TEX15 mutations in a family with nonobstructive azoospermia. Gynecol Obstet Invest. 2017;82:283–6.PubMedCrossRef Colombo R, Pontoglio A, Bini M. Two novel TEX15 mutations in a family with nonobstructive azoospermia. Gynecol Obstet Invest. 2017;82:283–6.PubMedCrossRef
79.
Zurück zum Zitat Choi Y, Jeon S, Choi M, Lee MH, Park M, Lee DR, et al. Mutations in SOHLH1 gene associate with nonobstructive azoospermia. Hum Mutat. 2010;31:788–93.PubMedCrossRef Choi Y, Jeon S, Choi M, Lee MH, Park M, Lee DR, et al. Mutations in SOHLH1 gene associate with nonobstructive azoospermia. Hum Mutat. 2010;31:788–93.PubMedCrossRef
80.
Zurück zum Zitat Miyamoto T, Hasuike S, Yogev L, Maduro MR, Ishikawa M, et al. Azoospermia in patients heterozygous for a mutation in SYCP3. Lancet. 2003;362:1714–9.PubMedCrossRef Miyamoto T, Hasuike S, Yogev L, Maduro MR, Ishikawa M, et al. Azoospermia in patients heterozygous for a mutation in SYCP3. Lancet. 2003;362:1714–9.PubMedCrossRef
81.
Zurück zum Zitat Venables JP, Elliott DJ, Makarova OV, Makarov EM, Cooke HJ, Eperon IC. RBMY, a probable human spermatogenesis factor, and other hnRNP G proteins interact with Tra2-beta and affect splicing. Hum Mol Genet. 2000;9:685–94.PubMedCrossRef Venables JP, Elliott DJ, Makarova OV, Makarov EM, Cooke HJ, Eperon IC. RBMY, a probable human spermatogenesis factor, and other hnRNP G proteins interact with Tra2-beta and affect splicing. Hum Mol Genet. 2000;9:685–94.PubMedCrossRef
82.
Zurück zum Zitat Saxena R, de Vries JWA, Repping S, Alagappan RK, Skaletsky H, Brown LG, et al. Four DAZ genes in two clusters found in the AZFc region of the human Y chromosome. Genomics. 2000;67:256–67.PubMedCrossRef Saxena R, de Vries JWA, Repping S, Alagappan RK, Skaletsky H, Brown LG, et al. Four DAZ genes in two clusters found in the AZFc region of the human Y chromosome. Genomics. 2000;67:256–67.PubMedCrossRef
83.
Zurück zum Zitat Yu J, Chen Z, Ni Y, Li Z. CFTR mutations in men with congenital bilateral absence of the vas deferens (CBAVD): a systemic review and meta-analysis. Hum Reprod. 2012;27:25–35.PubMedCrossRef Yu J, Chen Z, Ni Y, Li Z. CFTR mutations in men with congenital bilateral absence of the vas deferens (CBAVD): a systemic review and meta-analysis. Hum Reprod. 2012;27:25–35.PubMedCrossRef
84.
Zurück zum Zitat Tomaiuolo R, Fausto M, Elce A, Strina I, Ranieri A, Amato F, et al. Enhanced frequency of CFTR gene variants in couples who are candidates for assisted reproductive technology treatment. Clin Chem Lab Med. 2011;49:1289–93.PubMedCrossRef Tomaiuolo R, Fausto M, Elce A, Strina I, Ranieri A, Amato F, et al. Enhanced frequency of CFTR gene variants in couples who are candidates for assisted reproductive technology treatment. Clin Chem Lab Med. 2011;49:1289–93.PubMedCrossRef
85.
Zurück zum Zitat Amato F, Bellia C, Cardillo G, Castaldo G, Ciaccio M, Elce A, et al. Extensive molecular analysis of patients bearing CFTR-related disorders. J Mol Diagn. 2012;14:81–9.PubMedCrossRef Amato F, Bellia C, Cardillo G, Castaldo G, Ciaccio M, Elce A, et al. Extensive molecular analysis of patients bearing CFTR-related disorders. J Mol Diagn. 2012;14:81–9.PubMedCrossRef
86.
Zurück zum Zitat Tomaiuolo R, Nardiello P, Martinelli P, Sacchetti L, Salvatore F, Castaldo G. Prenatal diagnosis of cystic fibrosis: an experience of 181 cases. Clin Chem Lab Med. 2013;51:2227–32.PubMedCrossRef Tomaiuolo R, Nardiello P, Martinelli P, Sacchetti L, Salvatore F, Castaldo G. Prenatal diagnosis of cystic fibrosis: an experience of 181 cases. Clin Chem Lab Med. 2013;51:2227–32.PubMedCrossRef
87.
Zurück zum Zitat Ríos Orbañanos I, Vela Desojo A, Martinez-Indart L, Grau Bolado G, Rodriguez Estevez A, Rica Echevarria I. Turner syndrome: from birth to adulthood. Endocrinol Nutr. 2015;62:499–506.PubMedCrossRef Ríos Orbañanos I, Vela Desojo A, Martinez-Indart L, Grau Bolado G, Rodriguez Estevez A, Rica Echevarria I. Turner syndrome: from birth to adulthood. Endocrinol Nutr. 2015;62:499–506.PubMedCrossRef
88.
Zurück zum Zitat Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary ovarian insufficiency. Clin Genet. 2017;91:183–98.PubMedCrossRef Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary ovarian insufficiency. Clin Genet. 2017;91:183–98.PubMedCrossRef
89.
Zurück zum Zitat Qin Y, Jiao X, Simpson JL, Chen Z-J. Genetics of primary ovarian insufficiency: new developments and opportunities. Hum Reprod Update. 2015;21:787–808.PubMedPubMedCentralCrossRef Qin Y, Jiao X, Simpson JL, Chen Z-J. Genetics of primary ovarian insufficiency: new developments and opportunities. Hum Reprod Update. 2015;21:787–808.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Jenkinson EM, Rehman AU, Walsh T, Clayton-Smith J, Lee K, Morell RJ, et al. Perrault syndrome is caused by recessive mutations in CLPP, encoding a mitochondrial ATP-dependent chambered protease. Am J Hum Genet. 2013;92:605–13.PubMedPubMedCentralCrossRef Jenkinson EM, Rehman AU, Walsh T, Clayton-Smith J, Lee K, Morell RJ, et al. Perrault syndrome is caused by recessive mutations in CLPP, encoding a mitochondrial ATP-dependent chambered protease. Am J Hum Genet. 2013;92:605–13.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Huang N, Pandey AV, Agrawal V, Reardon W, Lapunzina PD, Mowat D, et al. Diversity and function of mutations in P450 oxidoreductase in patients with Antley–Bixler syndrome and disordered steroidogenesis. Am J Hum Genet. 2005;76:729–49.PubMedPubMedCentralCrossRef Huang N, Pandey AV, Agrawal V, Reardon W, Lapunzina PD, Mowat D, et al. Diversity and function of mutations in P450 oxidoreductase in patients with Antley–Bixler syndrome and disordered steroidogenesis. Am J Hum Genet. 2005;76:729–49.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Shackleton C, Marcos J, Malunowicz EM, Szarras-Czapnik M, Jira P, Taylor NF, et al. Biochemical diagnosis of Antley–Bixler syndrome by steroid analysis. Am J Med Genet. 2004;128A:223–31.PubMedCrossRef Shackleton C, Marcos J, Malunowicz EM, Szarras-Czapnik M, Jira P, Taylor NF, et al. Biochemical diagnosis of Antley–Bixler syndrome by steroid analysis. Am J Med Genet. 2004;128A:223–31.PubMedCrossRef
93.
Zurück zum Zitat Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocr Metab. 2004;89:2745–9.PubMedCrossRef Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocr Metab. 2004;89:2745–9.PubMedCrossRef
94.
Zurück zum Zitat Chen Z-J, Zhao H, He L, Shi Y, Qin Y, Shi Y, et al. Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat Genet. 2011;43:55–9.PubMedCrossRef Chen Z-J, Zhao H, He L, Shi Y, Qin Y, Shi Y, et al. Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat Genet. 2011;43:55–9.PubMedCrossRef
95.
Zurück zum Zitat Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, Kuick R, et al. Mutations in NALP7 cause recurrent hydatidiform moles and reproductive wastage in humans. Nat Genet. 2006;38:300–2.PubMedCrossRef Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, Kuick R, et al. Mutations in NALP7 cause recurrent hydatidiform moles and reproductive wastage in humans. Nat Genet. 2006;38:300–2.PubMedCrossRef
96.
Zurück zum Zitat Fallahian M, Sebire NJ, Savage PM, Seckl MJ, Fisher RA. Mutations in NLRP7 and KHDC3L confer a complete hydatidiform mole phenotype on digynic triploid conceptions. Hum Mutat. 2013;34:301–8.PubMedCrossRef Fallahian M, Sebire NJ, Savage PM, Seckl MJ, Fisher RA. Mutations in NLRP7 and KHDC3L confer a complete hydatidiform mole phenotype on digynic triploid conceptions. Hum Mutat. 2013;34:301–8.PubMedCrossRef
97.
Zurück zum Zitat Biason-Lauber A, De Filippo G, Konrad D, Scarano G, Nazzaro A, Schoenle EJ. WNT4 deficiency—a clinical phenotype distinct from the classic Mayer–Rokitansky–Kuster–Hauser syndrome: a case report. Hum Reprod. 2007;22:224–9.PubMedCrossRef Biason-Lauber A, De Filippo G, Konrad D, Scarano G, Nazzaro A, Schoenle EJ. WNT4 deficiency—a clinical phenotype distinct from the classic Mayer–Rokitansky–Kuster–Hauser syndrome: a case report. Hum Reprod. 2007;22:224–9.PubMedCrossRef
98.
Zurück zum Zitat Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ. A WNT4 mutation associated with mullerian-duct regression and virilization in a 46,XX woman. N Eng J Med. 2004;351:792–8.CrossRef Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ. A WNT4 mutation associated with mullerian-duct regression and virilization in a 46,XX woman. N Eng J Med. 2004;351:792–8.CrossRef
99.
Zurück zum Zitat Philibert P, Biason-Lauber A, Rouzier R, Pienkowski C, Paris F, Konrad D, et al. Identification and functional analysis of a new WNT4 gene mutation among 28 adolescent girls with primary amenorrhea and mullerian duct abnormalities: a French collaborative study. J Clin Endocr Metab. 2008;93:895–900.PubMedCrossRef Philibert P, Biason-Lauber A, Rouzier R, Pienkowski C, Paris F, Konrad D, et al. Identification and functional analysis of a new WNT4 gene mutation among 28 adolescent girls with primary amenorrhea and mullerian duct abnormalities: a French collaborative study. J Clin Endocr Metab. 2008;93:895–900.PubMedCrossRef
100.
Zurück zum Zitat Brucker SY, Frank L, Eisenbeis S, Henes M, Wallwiener D, Riess O, et al. Sequence variants in ESR1 and OXTR are associated with Mayer–Rokitansky–Küster–Hauser syndrome. Acta Obstet Gynecol Scand. 2017;96:1338–46.PubMedCrossRef Brucker SY, Frank L, Eisenbeis S, Henes M, Wallwiener D, Riess O, et al. Sequence variants in ESR1 and OXTR are associated with Mayer–Rokitansky–Küster–Hauser syndrome. Acta Obstet Gynecol Scand. 2017;96:1338–46.PubMedCrossRef
101.
Zurück zum Zitat Henes M, Jurow L, Peter A, Schoenfisch B, Taran FA, Huebner M, et al. Hyperandrogenemia and ovarian reserve in patients with Mayer–Rokitansky–Küster–Hauser syndrome type 1 and 2: potential influences on ovarian stimulation. Arch Gynecol Obstet. 2018;297:513–20.PubMedCrossRef Henes M, Jurow L, Peter A, Schoenfisch B, Taran FA, Huebner M, et al. Hyperandrogenemia and ovarian reserve in patients with Mayer–Rokitansky–Küster–Hauser syndrome type 1 and 2: potential influences on ovarian stimulation. Arch Gynecol Obstet. 2018;297:513–20.PubMedCrossRef
102.
Zurück zum Zitat Waschk DE, Tewes AC, Römer T, Hucke J, Kapczuk K, Schippert C, et al. Mutations in WNT9B are associated with Mayer–Rokitansky–Küster–Hauser syndrome. Clin Genet. 2016;89:590–6.PubMedCrossRef Waschk DE, Tewes AC, Römer T, Hucke J, Kapczuk K, Schippert C, et al. Mutations in WNT9B are associated with Mayer–Rokitansky–Küster–Hauser syndrome. Clin Genet. 2016;89:590–6.PubMedCrossRef
103.
Zurück zum Zitat Bracke A, Peeters K, Punjabi U, Hoogewijs D, Dewilde S. A search for molecular mechanisms underlying male idiopathic infertility. Reprod Biomed Online. 2018;36:327–39.PubMedCrossRef Bracke A, Peeters K, Punjabi U, Hoogewijs D, Dewilde S. A search for molecular mechanisms underlying male idiopathic infertility. Reprod Biomed Online. 2018;36:327–39.PubMedCrossRef
104.
Zurück zum Zitat Ventimiglia E, Montorsi F, Salonia A. Comorbidities and male infertility: a worrisome picture. Curr Opin Urol. 2016;26:146–51.PubMedCrossRef Ventimiglia E, Montorsi F, Salonia A. Comorbidities and male infertility: a worrisome picture. Curr Opin Urol. 2016;26:146–51.PubMedCrossRef
105.
Zurück zum Zitat Jungwirth A, Diemer T, Kopa Z, Krausz C, Tournaye H. European Association of Urology guidelines on Male Infertility: the 2012 update. Eur Urol. 2012;62:324–32.PubMedCrossRef Jungwirth A, Diemer T, Kopa Z, Krausz C, Tournaye H. European Association of Urology guidelines on Male Infertility: the 2012 update. Eur Urol. 2012;62:324–32.PubMedCrossRef
107.
108.
Zurück zum Zitat Krausz C, Escamilla AR, Chianese CK. Genetics of male infertility: from research to clinic. Reproduction. 2015;150:R159–74.PubMedCrossRef Krausz C, Escamilla AR, Chianese CK. Genetics of male infertility: from research to clinic. Reproduction. 2015;150:R159–74.PubMedCrossRef
109.
Zurück zum Zitat Pylyp LY, Spinenko LO, Verhoglyad NV, Kashevarova OO, Zukin VD. Chromosomal abnormalities in patients with infertility. Cytol Genet. 2015;49:33–9.CrossRef Pylyp LY, Spinenko LO, Verhoglyad NV, Kashevarova OO, Zukin VD. Chromosomal abnormalities in patients with infertility. Cytol Genet. 2015;49:33–9.CrossRef
110.
Zurück zum Zitat Krausz C, Degl’Innocenti S. Y chromosome and male infertility: update, 2006. Front Biosci. 2006;11:3049–61.PubMedCrossRef Krausz C, Degl’Innocenti S. Y chromosome and male infertility: update, 2006. Front Biosci. 2006;11:3049–61.PubMedCrossRef
111.
Zurück zum Zitat Krausz C, Hoefsloot L, Simoni M, Tüttelmann F, European Academy of Andrology, European Molecular Genetics Quality Network. EAA/EMQN best practice guidelines for molecular diagnosis of Y-chromosomal microdeletions: state-of-the-art 2013. Andrology. 2014;2:5–19.PubMedCrossRef Krausz C, Hoefsloot L, Simoni M, Tüttelmann F, European Academy of Andrology, European Molecular Genetics Quality Network. EAA/EMQN best practice guidelines for molecular diagnosis of Y-chromosomal microdeletions: state-of-the-art 2013. Andrology. 2014;2:5–19.PubMedCrossRef
112.
Zurück zum Zitat Stuppia L, Gatta V, Calabrese G, Guanciali Franchi P, Morizio E, Bombieri C, et al. A quarter of men with idiopathic oligo-azoospermia display chromosomal abnormalities and microdeletions of different types in interval 6 of Yq11. Hum Genet. 1998;102:566–70.PubMedCrossRef Stuppia L, Gatta V, Calabrese G, Guanciali Franchi P, Morizio E, Bombieri C, et al. A quarter of men with idiopathic oligo-azoospermia display chromosomal abnormalities and microdeletions of different types in interval 6 of Yq11. Hum Genet. 1998;102:566–70.PubMedCrossRef
113.
Zurück zum Zitat Patsalis PC, Sismani C, Quintana-Murci L, Taleb-Bekkouche F, Krausz C, McElreavey K. Effects of transmission of Y chromosome AZFc deletions. Lancet. 2002;360:1222–4.PubMedCrossRef Patsalis PC, Sismani C, Quintana-Murci L, Taleb-Bekkouche F, Krausz C, McElreavey K. Effects of transmission of Y chromosome AZFc deletions. Lancet. 2002;360:1222–4.PubMedCrossRef
114.
Zurück zum Zitat Asero P, Calogero AE, Condorelli RA, Mongioi L, Vicari E, Lanzafame F, et al. Relevance of genetic investigation in male infertility. J Endocrinol Investig. 2014;37:415–27.CrossRef Asero P, Calogero AE, Condorelli RA, Mongioi L, Vicari E, Lanzafame F, et al. Relevance of genetic investigation in male infertility. J Endocrinol Investig. 2014;37:415–27.CrossRef
115.
Zurück zum Zitat Kuroda-Kawaguchi T, Skaletsky H, Brown LG, Minx PJ, Cordum HS, Waterston RH, et al. The AZFc region of the Y chromosome features massive palindromes and uniform recurrent deletions in infertile men. Nat Genet. 2001;29:279–86.PubMedCrossRef Kuroda-Kawaguchi T, Skaletsky H, Brown LG, Minx PJ, Cordum HS, Waterston RH, et al. The AZFc region of the Y chromosome features massive palindromes and uniform recurrent deletions in infertile men. Nat Genet. 2001;29:279–86.PubMedCrossRef
116.
Zurück zum Zitat Ferlin A, Garolla A, Foresta C. Chromosome abnormalities in sperm of individuals with constitutional sex chromosomal abnormalities. Cytogenet Genome Res. 2005;111:310–6.PubMedCrossRef Ferlin A, Garolla A, Foresta C. Chromosome abnormalities in sperm of individuals with constitutional sex chromosomal abnormalities. Cytogenet Genome Res. 2005;111:310–6.PubMedCrossRef
117.
Zurück zum Zitat Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci USA. 2003;100:12201–6.PubMedCrossRefPubMedCentral Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci USA. 2003;100:12201–6.PubMedCrossRefPubMedCentral
118.
Zurück zum Zitat Aston KI. Genetic susceptibility to male infertility: news from genome-wide association studies. Andrology. 2014;2:315–21.PubMedCrossRef Aston KI. Genetic susceptibility to male infertility: news from genome-wide association studies. Andrology. 2014;2:315–21.PubMedCrossRef
119.
Zurück zum Zitat Morin SJ, Eccles J, Iturriaga A, Zimmerman RS. Translocations, inversions and other chromosome rearrangements. Fertil Steril. 2017;107:19–26.PubMedCrossRef Morin SJ, Eccles J, Iturriaga A, Zimmerman RS. Translocations, inversions and other chromosome rearrangements. Fertil Steril. 2017;107:19–26.PubMedCrossRef
121.
Zurück zum Zitat Oktay K, Bedoschi G, Berkowitz K, Bronson R, Kashani B, McGovern P, et al. Fertility preservation in women with turner syndrome: a comprehensive review and practical guidelines. J Pediatr Adolesc Gynecol. 2016;29:409–16.PubMedCrossRef Oktay K, Bedoschi G, Berkowitz K, Bronson R, Kashani B, McGovern P, et al. Fertility preservation in women with turner syndrome: a comprehensive review and practical guidelines. J Pediatr Adolesc Gynecol. 2016;29:409–16.PubMedCrossRef
122.
Zurück zum Zitat Collins J, Diedrich K, Franks S, Geraedts JPM, Jacobs PA, Karges B, et al. Genetic aspects of female reproduction. Hum Reprod Update. 2008;14:293–307.CrossRef Collins J, Diedrich K, Franks S, Geraedts JPM, Jacobs PA, Karges B, et al. Genetic aspects of female reproduction. Hum Reprod Update. 2008;14:293–307.CrossRef
123.
Zurück zum Zitat Chen M, Wei S, Hu J, Quan S. Can comprehensive chromosome screening technology improve IVF/ICSI outcomes? A meta-analysis. PLoS ONE. 2015;10:e0140779.PubMedPubMedCentralCrossRef Chen M, Wei S, Hu J, Quan S. Can comprehensive chromosome screening technology improve IVF/ICSI outcomes? A meta-analysis. PLoS ONE. 2015;10:e0140779.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat D’Argenio V, Nunziato M, D’Uonno N, Borrillo F, Vallone R, Conforti A, et al. Indications and limitations for preimplantation genetic diagnosis. Biochim Clin. 2017;41:314–21. D’Argenio V, Nunziato M, D’Uonno N, Borrillo F, Vallone R, Conforti A, et al. Indications and limitations for preimplantation genetic diagnosis. Biochim Clin. 2017;41:314–21.
125.
Zurück zum Zitat Man L, Lekovich J, Rosenwaks Z, Gerhardt J. Fragile X-associated diminished ovarian reserve and primary ovarian insufficiency from molecular mechanisms to clinical manifestations. Front Mol Neurosci. 2017;10:290.PubMedPubMedCentralCrossRef Man L, Lekovich J, Rosenwaks Z, Gerhardt J. Fragile X-associated diminished ovarian reserve and primary ovarian insufficiency from molecular mechanisms to clinical manifestations. Front Mol Neurosci. 2017;10:290.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Hoyos LR, Thakur M. Fragile X premutation in women: recognizing the health challenges beyond primary ovarian insufficiency. Assist Reprod Genet. 2017;34:315–23.CrossRef Hoyos LR, Thakur M. Fragile X premutation in women: recognizing the health challenges beyond primary ovarian insufficiency. Assist Reprod Genet. 2017;34:315–23.CrossRef
127.
128.
Zurück zum Zitat Committee on Genetics. Committee opinion No. 691: carrier screening for genetic conditions. Obstet Gynecol. 2017;129:e41–55.CrossRef Committee on Genetics. Committee opinion No. 691: carrier screening for genetic conditions. Obstet Gynecol. 2017;129:e41–55.CrossRef
130.
Zurück zum Zitat Mathijssen IB, Holtkamp KCA, Ottenheim CPE, van Eeten-Nijman JMC, Lakeman P, Meijers-Heijboer H, et al. Preconception carrier screening for multiple disorders: evaluation of a screening offer in a Dutch founder population. Eur J Hum Genet. 2018;26:166–75.PubMedPubMedCentralCrossRef Mathijssen IB, Holtkamp KCA, Ottenheim CPE, van Eeten-Nijman JMC, Lakeman P, Meijers-Heijboer H, et al. Preconception carrier screening for multiple disorders: evaluation of a screening offer in a Dutch founder population. Eur J Hum Genet. 2018;26:166–75.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Elce A, Boccia A, Cardillo G, Giordano S, Tomaiuolo R, Paolella G, et al. Three novel CFTR polymorphic repeats improve segregation analysis for cystic fibrosis. Clin Chem. 2009;55:1372.PubMedCrossRef Elce A, Boccia A, Cardillo G, Giordano S, Tomaiuolo R, Paolella G, et al. Three novel CFTR polymorphic repeats improve segregation analysis for cystic fibrosis. Clin Chem. 2009;55:1372.PubMedCrossRef
133.
Zurück zum Zitat Allyse M, Minear MA, Berson E, Sridhar S, Rote M, Hung A, et al. Non-invasive prenatal testing: a review of international implementation and challenges. Int J Womens Health. 2015;7:113–26.PubMedPubMedCentralCrossRef Allyse M, Minear MA, Berson E, Sridhar S, Rote M, Hung A, et al. Non-invasive prenatal testing: a review of international implementation and challenges. Int J Womens Health. 2015;7:113–26.PubMedPubMedCentralCrossRef
134.
135.
Zurück zum Zitat Saba L, Masala M, Capponi V, Marceddu G, Massidda M, Rosatelli MC. Non-invasive prenatal diagnosis of beta-thalassemia by semiconductor sequencing: a feasibility study in the sardinian population. Eur J Hum Genet. 2017;25:600–7.PubMedPubMedCentralCrossRef Saba L, Masala M, Capponi V, Marceddu G, Massidda M, Rosatelli MC. Non-invasive prenatal diagnosis of beta-thalassemia by semiconductor sequencing: a feasibility study in the sardinian population. Eur J Hum Genet. 2017;25:600–7.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat New M, Tong YK, Yuen T, Jiang P, Pina C, Chan KC, et al. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab. 2014;99:E1022–30.PubMedPubMedCentralCrossRef New M, Tong YK, Yuen T, Jiang P, Pina C, Chan KC, et al. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab. 2014;99:E1022–30.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Xu Y, Li X, Ge HJ, Xiao B, Zhang YY, Ying XM, et al. Haplotype-based approach for noninvasive prenatal tests of Duchenne muscular dystrophy using cell-free fetal DNA. Genet Med. 2015;17:889–96.PubMedCrossRef Xu Y, Li X, Ge HJ, Xiao B, Zhang YY, Ying XM, et al. Haplotype-based approach for noninvasive prenatal tests of Duchenne muscular dystrophy using cell-free fetal DNA. Genet Med. 2015;17:889–96.PubMedCrossRef
138.
Zurück zum Zitat Liñán A, Lawrenz B, El Khatib I, Bayram A, Arnanz A, Rubio C, et al. Clinical reassessment of human embryo ploidy status between cleavage and blastocyst stage by Next Generation Sequencing. PLoS ONE. 2018;13:e0201652.PubMedPubMedCentralCrossRef Liñán A, Lawrenz B, El Khatib I, Bayram A, Arnanz A, Rubio C, et al. Clinical reassessment of human embryo ploidy status between cleavage and blastocyst stage by Next Generation Sequencing. PLoS ONE. 2018;13:e0201652.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Rubio C, Bellver J, Rodrigo L, Castillón G, Guillén A, Vidal C, et al. In vitro fertilization with preimplantation genetic diagnosis for aneuploidies in advanced maternal age: a randomized, controlled study. Fertil Steril. 2017;107:1122–9.PubMedCrossRef Rubio C, Bellver J, Rodrigo L, Castillón G, Guillén A, Vidal C, et al. In vitro fertilization with preimplantation genetic diagnosis for aneuploidies in advanced maternal age: a randomized, controlled study. Fertil Steril. 2017;107:1122–9.PubMedCrossRef
140.
Zurück zum Zitat Munné S. Status of preimplantation genetic testing and embryo selection. Reprod Biomed Online. 2018;37:393–6.PubMedCrossRef Munné S. Status of preimplantation genetic testing and embryo selection. Reprod Biomed Online. 2018;37:393–6.PubMedCrossRef
141.
Zurück zum Zitat Vanneste E, Melotte C, Voet T, Robberecht C, Debrock S, Pexsters A, et al. PGD for a complex chromosomal rearrangement by array comparative genomic hybridization. Hum Reprod. 2011;26:941–9.PubMedCrossRef Vanneste E, Melotte C, Voet T, Robberecht C, Debrock S, Pexsters A, et al. PGD for a complex chromosomal rearrangement by array comparative genomic hybridization. Hum Reprod. 2011;26:941–9.PubMedCrossRef
142.
Zurück zum Zitat D’Argenio V, Tomaiuolo R, Cariati F. La, “whole genome amplification” su singola cellula. Biochim Clin. 2016;40:293–301. D’Argenio V, Tomaiuolo R, Cariati F. La, “whole genome amplification” su singola cellula. Biochim Clin. 2016;40:293–301.
143.
Zurück zum Zitat Harton GL, De Rycke M, Fiorentino F, Moutou C, SenGupta S, Traeger-Synodinos J, et al. ESHRE PGD consortium best practice guidelines for amplification-based PGD. Hum Reprod. 2011;26:33–40.PubMedCrossRef Harton GL, De Rycke M, Fiorentino F, Moutou C, SenGupta S, Traeger-Synodinos J, et al. ESHRE PGD consortium best practice guidelines for amplification-based PGD. Hum Reprod. 2011;26:33–40.PubMedCrossRef
144.
Zurück zum Zitat Handyside AH, Harton GL, Mariani B, Thornhill AR, Affara N, Shaw MA, et al. Karyomapping: a universal method for genome wide analysis of genetic disease based on mapping crossovers between parental haplotypes. Med Genet. 2010;47:651–8.CrossRef Handyside AH, Harton GL, Mariani B, Thornhill AR, Affara N, Shaw MA, et al. Karyomapping: a universal method for genome wide analysis of genetic disease based on mapping crossovers between parental haplotypes. Med Genet. 2010;47:651–8.CrossRef
145.
Zurück zum Zitat Natesan SA, Handyside AH, Thornhill AR, Ottolini CS, Sage K, Summers MC, et al. Live birth after PGD with confirmation by a comprehensive approach (karyomapping) for simultaneous detection of monogenic and chromosomal disorders. Reprod Biomed Online. 2014;29:600–5.PubMedCrossRef Natesan SA, Handyside AH, Thornhill AR, Ottolini CS, Sage K, Summers MC, et al. Live birth after PGD with confirmation by a comprehensive approach (karyomapping) for simultaneous detection of monogenic and chromosomal disorders. Reprod Biomed Online. 2014;29:600–5.PubMedCrossRef
146.
Zurück zum Zitat Thornhill AR, Handyside AH, Ottolini C, Natesan SA, Taylor J, Sage K, et al. Karyomapping-a comprehensive means of simultaneous monogenic and cytogenetic PGD: comparison with standard approaches in real time for Marfan syndrome. J Assist Reprod Genet. 2015;32:347–56.PubMedPubMedCentralCrossRef Thornhill AR, Handyside AH, Ottolini C, Natesan SA, Taylor J, Sage K, et al. Karyomapping-a comprehensive means of simultaneous monogenic and cytogenetic PGD: comparison with standard approaches in real time for Marfan syndrome. J Assist Reprod Genet. 2015;32:347–56.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Treff NR, Fedick A, Tao X, Devkota B, Taylor D, Scott RT. Evaluation of targeted next-generation sequencing–based preimplantation genetic diagnosis of monogenic disease. Fertil Steril. 2013;99:1377–84.PubMedCrossRef Treff NR, Fedick A, Tao X, Devkota B, Taylor D, Scott RT. Evaluation of targeted next-generation sequencing–based preimplantation genetic diagnosis of monogenic disease. Fertil Steril. 2013;99:1377–84.PubMedCrossRef
148.
Zurück zum Zitat Peters BA, Kermani BG, Alferov O, Agarwal MR, McElwain MA, Gulbahce N, et al. Detection and phasing of single base de novo mutations in biopsies from human in vitro fertilized embryos by advanced whole-genome sequencing. Genome Res. 2015;25:426–34.PubMedPubMedCentralCrossRef Peters BA, Kermani BG, Alferov O, Agarwal MR, McElwain MA, Gulbahce N, et al. Detection and phasing of single base de novo mutations in biopsies from human in vitro fertilized embryos by advanced whole-genome sequencing. Genome Res. 2015;25:426–34.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Kung A, Munne S, Bankowski B, Coates A, Wells D. Validation of next-generation sequencing for comprehensive chromosome screening of embryos. Reprod Biomed Online. 2015;31:760–9.PubMedCrossRef Kung A, Munne S, Bankowski B, Coates A, Wells D. Validation of next-generation sequencing for comprehensive chromosome screening of embryos. Reprod Biomed Online. 2015;31:760–9.PubMedCrossRef
150.
Zurück zum Zitat Harper JC, Coonen E, De Rycke M, Harton G, Moutou C, Pehlivan T, et al. ESHRE PGD consortium data collection X: cycles from January to December 2007 with pregnancy follow-up to October 2008. Hum Reprod. 2010;25:2685–707.PubMedCrossRef Harper JC, Coonen E, De Rycke M, Harton G, Moutou C, Pehlivan T, et al. ESHRE PGD consortium data collection X: cycles from January to December 2007 with pregnancy follow-up to October 2008. Hum Reprod. 2010;25:2685–707.PubMedCrossRef
151.
Zurück zum Zitat Brezina PR, Kutteh WH. Clinical applications of preimplantation genetic testing. Br Med J. 2015;350:g7611.CrossRef Brezina PR, Kutteh WH. Clinical applications of preimplantation genetic testing. Br Med J. 2015;350:g7611.CrossRef
152.
153.
Zurück zum Zitat Wells D. Next-generation sequencing: the dawn of a new era for preimplantation genetic diagnostics. Fertil Steril. 2014;101:1250–1.PubMedCrossRef Wells D. Next-generation sequencing: the dawn of a new era for preimplantation genetic diagnostics. Fertil Steril. 2014;101:1250–1.PubMedCrossRef
154.
Zurück zum Zitat Handyside AH. 24-chromosome copy number analysis: a comparison of available technologies. Fertil Steril. 2013;100:595–602.PubMedCrossRef Handyside AH. 24-chromosome copy number analysis: a comparison of available technologies. Fertil Steril. 2013;100:595–602.PubMedCrossRef
155.
Zurück zum Zitat Martin J, Cervero A, Mir P, Martinez JAC, Pellicer A, Simón C, et al. The impact of next-generation sequencing technology on preimplantation genetic diagnosis and screening. Fertil Steril. 2013;99:1054–61.PubMedCrossRef Martin J, Cervero A, Mir P, Martinez JAC, Pellicer A, Simón C, et al. The impact of next-generation sequencing technology on preimplantation genetic diagnosis and screening. Fertil Steril. 2013;99:1054–61.PubMedCrossRef
156.
Zurück zum Zitat Rubio C. Next-generation sequencing: challenges in reproductive genetics. Fertil Steril. 2014;101:1252–3.PubMedCrossRef Rubio C. Next-generation sequencing: challenges in reproductive genetics. Fertil Steril. 2014;101:1252–3.PubMedCrossRef
157.
Zurück zum Zitat Sermon K. Novel technologies emerging for preimplantation genetic diagnosis and preimplantation genetic testing for aneuploidy. Expert Rev Mol Diagn. 2017;17:71–82.PubMedCrossRef Sermon K. Novel technologies emerging for preimplantation genetic diagnosis and preimplantation genetic testing for aneuploidy. Expert Rev Mol Diagn. 2017;17:71–82.PubMedCrossRef
158.
Zurück zum Zitat Cariati F, D’Uonno N, Borrillo F, Iervolino S, Galdiero G, Tomaiuolo R. Bisphenol a: an emerging threat to male fertility. Reprod Biol Endocrinol. 2019;17:6.PubMedPubMedCentralCrossRef Cariati F, D’Uonno N, Borrillo F, Iervolino S, Galdiero G, Tomaiuolo R. Bisphenol a: an emerging threat to male fertility. Reprod Biol Endocrinol. 2019;17:6.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Eisenberg ML, Lathi RB, Baker VL, Westphal LM, Milki AA, Nangia AK. Frequency of the male infertility evaluation: data from the national survey of family growth. J Urol. 2013;189:1030–4.PubMedCrossRef Eisenberg ML, Lathi RB, Baker VL, Westphal LM, Milki AA, Nangia AK. Frequency of the male infertility evaluation: data from the national survey of family growth. J Urol. 2013;189:1030–4.PubMedCrossRef
161.
Zurück zum Zitat Alviggi C, Conforti A, Santi D, Esteves SC, Andersen CY, Humaidan P, et al. Clinical relevance of genetic variants of gonadotrophins and their receptors in controlled ovarian stimulation: a systematic review and meta-analysis. Hum Reprod Update. 2018;24:599–614.PubMedCrossRef Alviggi C, Conforti A, Santi D, Esteves SC, Andersen CY, Humaidan P, et al. Clinical relevance of genetic variants of gonadotrophins and their receptors in controlled ovarian stimulation: a systematic review and meta-analysis. Hum Reprod Update. 2018;24:599–614.PubMedCrossRef
162.
Zurück zum Zitat Harper JC, Aittomäki K, Borry P, Cornel MC, de Wert G, Dondorp W, Geraedts J, et al. Recent developments in genetics and medically assisted reproduction: from research to clinical applications. Eur J Hum Genet. 2018;26:12–33.PubMedCrossRef Harper JC, Aittomäki K, Borry P, Cornel MC, de Wert G, Dondorp W, Geraedts J, et al. Recent developments in genetics and medically assisted reproduction: from research to clinical applications. Eur J Hum Genet. 2018;26:12–33.PubMedCrossRef
Metadaten
Titel
The evolving role of genetic tests in reproductive medicine
verfasst von
Federica Cariati
Valeria D’Argenio
Rossella Tomaiuolo
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-2019-8

Weitere Artikel der Ausgabe 1/2019

Journal of Translational Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.