Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Genet ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849204

ABSTRACT

INTRODUCTION: Tonne-Kalscheuer syndrome (TOKAS) is a recessive X-linked multiple congenital anomaly disorder caused by RLIM variations. Of the 41 patients reported, only 7 antenatal cases were described. METHOD: After the antenatal diagnosis of TOKAS by exome analysis in a family followed for over 35 years because of multiple congenital anomalies in five male fetuses, a call for collaboration was made, resulting in a cohort of 11 previously unpublished cases. RESULTS: We present a TOKAS antenatal cohort, describing 11 new cases in 6 French families. We report a high frequency of diaphragmatic hernia (9 of 11), differences in sex development (10 of 11) and various visceral malformations. We report some recurrent dysmorphic features, but also pontocerebellar hypoplasia, pre-auricular skin tags and olfactory bulb abnormalities previously unreported in the literature. Although no clear genotype-phenotype correlation has yet emerged, we show that a recurrent p.(Arg611Cys) variant accounts for 66% of fetal TOKAS cases. We also report two new likely pathogenic variants in RLIM, outside of the two previously known mutational hotspots. CONCLUSION: Overall, we present the first fetal cohort of TOKAS, describe the clinical features that made it a recognisable syndrome at fetopathological examination, and extend the phenotypical spectrum and the known genotype of this rare disorder.

2.
Clin Genet ; 106(3): 321-335, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38779778

ABSTRACT

Premature ovarian insufficiency is a common form of female infertility affecting up to 4% of women and characterised by amenorrhea with elevated gonadotropin before the age of 40. Oocytes require controlled DNA breakage and repair for homologous recombination and the maintenance of oocyte integrity. Biallelic disruption of the DNA damage repair gene, Fanconi anemia complementation group A (FANCA), is a common cause of Fanconi anaemia, a syndrome characterised by bone marrow failure, cancer predisposition, physical anomalies and POI. There is ongoing dispute about the role of heterozygous FANCA variants in POI pathogenesis, with insufficient evidence supporting causation. Here, we have identified biallelic FANCA variants in French sisters presenting with POI, including a novel missense variant of uncertain significance and a likely pathogenic deletion that initially evaded detection. Functional studies indicated no discernible effect on DNA damage sensitivity in patient lymphoblasts. These novel FANCA variants add evidence that heterozygous loss of one allele is insufficient to cause DNA damage sensitivity and POI. We propose that intragenic deletions, that are relatively common in FANCA, may be missed without careful analysis, and could explain the presumed causation of heterozygous variants. Accurate variant curation is critical to optimise patient care and outcomes.


Subject(s)
Alleles , Fanconi Anemia Complementation Group A Protein , Primary Ovarian Insufficiency , Humans , Primary Ovarian Insufficiency/genetics , Female , Fanconi Anemia Complementation Group A Protein/genetics , Adult , Fanconi Anemia/genetics , Fanconi Anemia/diagnosis , Siblings , Heterozygote , Genetic Predisposition to Disease , Pedigree , Mutation/genetics
3.
Am J Med Genet A ; 194(7): e63531, 2024 07.
Article in English | MEDLINE | ID: mdl-38421086

ABSTRACT

Duplications of the 3q29 cytoband are rare chromosomal copy number variations (CNVs) (overlapping or recurrent ~1.6 Mb 3q29 duplications). They have been associated with highly variable neurodevelopmental disorders (NDDs) with various associated features or reported as a susceptibility factor to the development of learning disabilities and neuropsychiatric disorders. The smallest region of overlap and the phenotype of 3q29 duplications remain uncertain. We here report a French cohort of 31 families with a 3q29 duplication identified by chromosomal microarray analysis (CMA), including 14 recurrent 1.6 Mb duplications, eight overlapping duplications (>1 Mb), and nine small duplications (<1 Mb). Additional genetic findings that may be involved in the phenotype were identified in 11 patients. Focusing on apparently isolated 3q29 duplications, patients present mainly mild NDD as suggested by a high rate of learning disabilities in contrast to a low proportion of patients with intellectual disabilities. Although some are de novo, most of the 3q29 duplications are inherited from a parent with a similar mild phenotype. Besides, the study of small 3q29 duplications does not provide evidence for any critical region. Our data suggest that the overlapping and recurrent 3q29 duplications seem to lead to mild NDD and that a severe or syndromic clinical presentation should warrant further genetic analyses.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 3 , DNA Copy Number Variations , Phenotype , Humans , Female , Male , Chromosomes, Human, Pair 3/genetics , Chromosome Duplication/genetics , Child , DNA Copy Number Variations/genetics , Child, Preschool , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Adolescent , Cohort Studies , Intellectual Disability/genetics , Intellectual Disability/pathology , Adult , Infant
4.
Hum Genet ; 142(7): 879-907, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37148394

ABSTRACT

Premature ovarian insufficiency (POI) is a common cause of infertility in women, characterised by amenorrhea and elevated FSH under the age of 40 years. In some cases, POI is syndromic in association with other features such as sensorineural hearing loss in Perrault syndrome. POI is a heterogeneous disease with over 80 causative genes known so far; however, these explain only a minority of cases. Using whole-exome sequencing (WES), we identified a MRPL50 homozygous missense variant (c.335T > A; p.Val112Asp) shared by twin sisters presenting with POI, bilateral high-frequency sensorineural hearing loss, kidney and heart dysfunction. MRPL50 encodes a component of the large subunit of the mitochondrial ribosome. Using quantitative proteomics and western blot analysis on patient fibroblasts, we demonstrated a loss of MRPL50 protein and an associated destabilisation of the large subunit of the mitochondrial ribosome whilst the small subunit was preserved. The mitochondrial ribosome is responsible for the translation of subunits of the mitochondrial oxidative phosphorylation machinery, and we found patient fibroblasts have a mild but significant decrease in the abundance of mitochondrial complex I. These data support a biochemical phenotype associated with MRPL50 variants. We validated the association of MRPL50 with the clinical phenotype by knockdown/knockout of mRpL50 in Drosophila, which resulted abnormal ovarian development. In conclusion, we have shown that a MRPL50 missense variant destabilises the mitochondrial ribosome, leading to oxidative phosphorylation deficiency and syndromic POI, highlighting the importance of mitochondrial support in ovarian development and function.


Subject(s)
Gonadal Dysgenesis, 46,XX , Hearing Loss, Sensorineural , Primary Ovarian Insufficiency , Female , Humans , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/genetics , Mitochondria/genetics , Mutation, Missense , Primary Ovarian Insufficiency/genetics , Animals , Drosophila melanogaster
5.
Prenat Diagn ; 43(6): 734-745, 2023 06.
Article in English | MEDLINE | ID: mdl-36914926

ABSTRACT

OBJECTIVE: We aimed to gather fetal cases carrying a 7q11.23 copy number variation (CNV) and collect precise clinical data to broaden knowledge of antenatal features in these syndromes. METHODS: We retrospectively recruited unrelated cases with 7q11.23 deletion, known as Williams-Beuren syndrome (WBS), or 7q11.23 duplication who had prenatal ultrasound findings. We collected laboratory and clinical data, fetal ultrasound, cardiac ultrasound and fetal autopsy reports from 18 prenatal diagnostic centers throughout France. RESULTS: 40 fetuses with WBS were collected and the most common features were intra-uterine growth retardation (IUGR) (70.0%, 28/40), cardiovascular defects (30.0%, 12/40), polyhydramnios (17.5%, 7/40) and protruding tongue (15.0%, 6/40). Fetal autopsy reports were available for 11 cases and were compared with ultrasound prenatal features. Four cases of fetuses with 7q11.23 microduplication were collected and prenatal ultrasound signs were variable and often isolated. CONCLUSION: This work strengthens the fact that 7q11.23 CNVs are associated with a broad spectrum of antenatal presentations. IUGR and cardiovascular defects were the most frequent ultrasound signs. By reporting the biggest series of antenatal WBS, we aim to better delineate distinctive signs in fetuses with 7q11.23 CNVs.


Subject(s)
Williams Syndrome , Humans , Female , Pregnancy , Williams Syndrome/diagnostic imaging , Williams Syndrome/genetics , Williams Syndrome/complications , DNA Copy Number Variations , Retrospective Studies , Fetal Growth Retardation , Ultrasonography
6.
Hum Mutat ; 43(9): 1125-1148, 2022 09.
Article in English | MEDLINE | ID: mdl-35451537

ABSTRACT

PBX1 is a highly conserved atypical homeodomain transcription factor (TF) belonging to the TALE (three amino acid loop extension) family. Dimerized with other TALE proteins, it can interact with numerous partners and reach dozens of regulating sequences, suggesting its role as a pioneer factor. PBX1 is expressed throughout the embryonic stages (as early as the blastula stage) in vertebrates. In human, PBX1 germline variations are linked to syndromic renal anomalies (CAKUTHED). In this review, we summarized available data on PBX1 functions, PBX1-deficient animal models, and PBX1 germline variations in humans. Two types of genetic alterations were identified in PBX1 gene. PBX1 missense variations generate a severe phenotype including lung hypoplasia, cardiac malformations, and sexual development defects (DSDs). Conversely, truncating variants generate milder phenotypes (mainly cryptorchidism and deafness). We suggest that defects in PBX1 interactions with various partners, including proteins from the HOX (HOXA7, HOXA10, etc.), WNT (WNT9B, WNT3), and Polycomb (BMI1, EED) families are responsible for abnormal proliferation and differentiation of the embryonic mesenchyme. These alterations could explain most of the defects observed in humans. However, some phenotype variability (especially DSDs) remains poorly understood. Further studies are needed to explore the TALE family in greater depth.


Subject(s)
Homeodomain Proteins , Pre-B-Cell Leukemia Transcription Factor 1 , Transcription Factors , Animals , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Phenotype , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Hum Mutat ; 43(10): 1443-1453, 2022 10.
Article in English | MEDLINE | ID: mdl-35801529

ABSTRACT

Premature ovarian insufficiency (POI) is a leading form of female infertility, characterised by menstrual disturbance and elevated follicle-stimulating hormone before age 40. It is highly heterogeneous with variants in over 80 genes potentially causative, but the majority of cases having no known cause. One gene implicated in POI pathology is TP63. TP63 encodes multiple p63 isoforms, one of which has been shown to have a role in the surveillance of genetic quality in oocytes. TP63 C-terminal truncation variants and N-terminal duplication have been described in association with POI, however, functional validation has been lacking. Here we identify three novel TP63 missense variants in women with nonsyndromic POI, including one in the N-terminal activation domain, one in the C-terminal inhibition domain, and one affecting a unique and poorly understood p63 isoform, TA*p63. Via blue-native page and luciferase reporter assays we demonstrate that two of these variants disrupt p63 dimerization, leading to constitutively active p63 tetramer that significantly increases the transcription of downstream targets. This is the first evidence that TP63 missense variants can cause isolated POI and provides mechanistic insight that TP63 variants cause POI due to constitutive p63 activation and accelerated oocyte loss in the absence of DNA damage.


Subject(s)
Primary Ovarian Insufficiency , Transcription Factors , Tumor Suppressor Proteins , Female , Humans , Mutation, Missense , Primary Ovarian Insufficiency/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
8.
Cytogenet Genome Res ; 162(3): 124-131, 2022.
Article in English | MEDLINE | ID: mdl-35609520

ABSTRACT

Genetic factors are responsible for 15% of male infertility conditions. Numerical and structural chromosomal anomalies are validated genetic factors leading to spermatogenic quantitative defects, with a frequency depending on the severity of the phenotype. Among the structural chromosomal rearrangements, dicentric chromosomes are generally observed in robertsonian translocations or in cases of Y chromosome isodicentrics. In X-autosome translocations, male carriers are generally infertile, regardless of the position of the breakpoint, due to interrupted spermatogenesis. We report an infertile man bearing an unusual balanced (X;22) translocation, with a centromeric X breakpoint generating a derivative pseudodicentric chromosome psu dic(22;X). Extensive cytogenetic analyses were necessary to determine the precise nature of the derivative chromosome. The likely cause of the reproductive phenotype of the patient is discussed based on meiotic chromosomal conformation.


Subject(s)
Chromosome Disorders , Infertility, Male , Oligospermia , Chromosome Aberrations , Chromosome Disorders/genetics , Humans , Infertility, Male/genetics , Male , Oligospermia/genetics , Translocation, Genetic/genetics , Y Chromosome
9.
Prenat Diagn ; 42(1): 118-135, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34894355

ABSTRACT

OBJECTIVE: Terminal 6q deletion is a rare genetic condition associated with a neurodevelopmental disorder characterized by intellectual disability and structural brain anomalies. Interestingly, a similar phenotype is observed in patients harboring pathogenic variants in the DLL1 gene. Our study aimed to further characterize the prenatal phenotype of this syndrome as well as to attempt to establish phenotype-genotype correlations. METHOD: We collected ultrasound findings from 22 fetuses diagnosed with a pure 6qter deletion. We reviewed the literature and compared our 22 cases with 14 fetuses previously reported as well as with patients with heterozygous DLL1 pathogenic variants. RESULTS: Brain structural alterations were observed in all fetuses. The most common findings (>70%) were cerebellar hypoplasia, ventriculomegaly, and corpus callosum abnormalities. Gyration abnormalities were observed in 46% of cases. Occasional findings included cerebral heterotopia, aqueductal stenosis, vertebral malformations, dysmorphic features, and kidney abnormalities. CONCLUSION: This is the first series of fetuses diagnosed with pure terminal 6q deletion. Based on our findings, we emphasize the prenatal sonographic anomalies, which may suggest the syndrome. Furthermore, this study highlights the importance of chromosomal microarray analysis to search for submicroscopic deletions of the 6q27 region involving the DLL1 gene in fetuses with these malformations.


Subject(s)
Calcium-Binding Proteins/analysis , Chromosome Disorders/complications , Membrane Proteins/analysis , Adult , Calcium-Binding Proteins/genetics , Chromosome Disorders/genetics , Chromosomes, Human, Pair 6/genetics , Female , Humans , Membrane Proteins/genetics , Phenotype , Pregnancy , Retrospective Studies , Trisomy/genetics , Virulence/genetics , Virulence/physiology
10.
Cytogenet Genome Res ; 161(3-4): 143-152, 2021.
Article in English | MEDLINE | ID: mdl-33827072

ABSTRACT

Fetal mosaicism for chromosomal rearrangements remains a challenge to diagnose, even in the era of whole-genome sequencing. We present here a case of fetal mosaicism for a chromosomal rearrangement explored in amniocytes and fetal muscle, consisting of a major cell population (95%) with partial monosomy 4q and a minor population (5%) with additional material replacing the 4qter deleted segment. Molecular techniques (MLPA, array-CGH) failed to assess the origin of this material. Only multicolor-FISH identified the additional segment on chromosome 4 as derived from chromosome 17. Due to the poor prognosis, the couple chose to terminate the pregnancy. Because of low-level mosaicism, chromosomal microarray analysis (CMA), now considered as first-tier prenatal genetic analysis, did not allow the identification of the minor cell line. In case of large CNVs (>5 Mb) detected by CMA, karyotyping may be considered to elucidate the mechanism of the underlying rearrangement and eliminate mosaicism.


Subject(s)
Chromosome Painting/methods , Cytogenetics/methods , Fetus/metabolism , Mosaicism , Prenatal Diagnosis/methods , Translocation, Genetic/genetics , Adult , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 4/genetics , Comparative Genomic Hybridization , Female , Humans , Karyotyping , Maternal Age , Pregnancy
11.
Prenat Diagn ; 41(1): 111-122, 2021 01.
Article in English | MEDLINE | ID: mdl-32920845

ABSTRACT

INTRODUCTION: We aimed to assess the diagnostic yield of ultrasonography in the identification of the etiology of hydramnios, and the added value of MRI or amniocentesis. METHODS: We conducted a single-center retrospective study including pregnancies with confirmed hydramnios (defined as deepest pocket ≥8 cm) between January 2013 and May 2017. Twin pregnancies, secondary hydramnios discovered after the diagnosis of a causal pathology, and pregnancies of unknown outcome were excluded. All pregnancies underwent a targeted scan, and selected cases underwent MRI or amniocentesis. RESULTS: A total of 158 patients with confirmed hydramnios were included. Hydramnios was associated with a fetal pathology in 37 cases (23.4%), with diabetes in 39 (24.6%), isolated macrosomia in 16 (10.1%), and considered idiopathic in 66 (41.7%). Ultrasonography established a diagnosis of the underlying pathology in 73% of cases. Amniocentesis was done in 31 cases (20%) and it allowed diagnosis of chromosome anomalies, esophageal atresia, myotonic dystrophy congenital type, Prader-Willi syndrome, and Bartter syndrome. MRI was done in 15 cases (10%) and it allowed one additional diagnosis of esophageal atresia. The diagnostic yields of MRI and amniocentesis were 91.7% and 95.2%, respectively. There were five false positive diagnoses at ultrasonography, and one false positive diagnosis at MRI. CONCLUSION: Hydramnios can be associated with a wide variety of underlying pathologies. Diagnostic ultrasound can attain a diagnosis in the majority of cases. Amniocentesis offers a valuable complementary assessment.


Subject(s)
Polyhydramnios/diagnostic imaging , Polyhydramnios/etiology , Ultrasonography, Prenatal/statistics & numerical data , Adult , Amniocentesis/statistics & numerical data , Diabetes, Gestational/diagnostic imaging , Female , Humans , Pregnancy , Pregnancy in Diabetics/diagnostic imaging , Retrospective Studies
12.
Hum Genet ; 139(10): 1325-1343, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32399598

ABSTRACT

Perrault syndrome is a rare heterogeneous condition characterised by sensorineural hearing loss and premature ovarian insufficiency. Additional neuromuscular pathology is observed in some patients. There are six genes in which variants are known to cause Perrault syndrome; however, these explain only a minority of cases. We investigated the genetic cause of Perrault syndrome in seven affected individuals from five different families, successfully identifying the cause in four patients. This included previously reported and novel causative variants in known Perrault syndrome genes, CLPP and LARS2, involved in mitochondrial proteolysis and mitochondrial translation, respectively. For the first time, we show that pathogenic variants in PEX6 can present clinically as Perrault syndrome. PEX6 encodes a peroxisomal biogenesis factor, and we demonstrate evidence of peroxisomal dysfunction in patient serum. This study consolidates the clinical overlap between Perrault syndrome and peroxisomal disorders, and highlights the need to consider ovarian function in individuals with atypical/mild peroxisomal disorders. The remaining patients had variants in candidate genes such as TFAM, involved in mtDNA transcription, replication, and packaging, and GGPS1 involved in mevalonate/coenzyme Q10 biosynthesis and whose enzymatic product is required for mouse folliculogenesis. This genomic study highlights the diverse molecular landscape of this poorly understood syndrome.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Amino Acyl-tRNA Synthetases/genetics , DNA-Binding Proteins/genetics , Dimethylallyltranstransferase/genetics , Endopeptidase Clp/genetics , Farnesyltranstransferase/genetics , Genetic Predisposition to Disease , Geranyltranstransferase/genetics , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/genetics , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Adolescent , Adult , Base Sequence , Child , DNA, Mitochondrial/genetics , Female , Gene Expression , Gonadal Dysgenesis, 46,XX/diagnosis , Gonadal Dysgenesis, 46,XX/pathology , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Ovary/metabolism , Ovary/pathology , Pedigree , Peroxisomes/metabolism , Peroxisomes/pathology
13.
Mol Hum Reprod ; 26(9): 665-677, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32634216

ABSTRACT

Infertility, a global problem affecting up to 15% of couples, can have varied causes ranging from natural ageing to the pathological development or function of the reproductive organs. One form of female infertility is premature ovarian insufficiency (POI), affecting up to 1 in 100 women and characterised by amenorrhoea and elevated FSH before the age of 40. POI can have a genetic basis, with over 50 causative genes identified. Non-obstructive azoospermia (NOA), a form of male infertility characterised by the absence of sperm in semen, has an incidence of 1% and is similarly heterogeneous. The genetic basis of male and female infertility is poorly understood with the majority of cases having no known cause. Here, we study a case of familial infertility including a proband with POI and her brother with NOA. We performed whole-exome sequencing (WES) and identified a homozygous STAG3 missense variant that segregated with infertility. STAG3 encodes a component of the meiosis cohesin complex required for sister chromatid separation. We report the first pathogenic homozygous missense variant in STAG3 and the first STAG3 variant associated with both male and female infertility. We also demonstrate limitations of WES for the analysis of homologous DNA sequences, with this variant being ambiguous or missed by independent WES protocols and its homozygosity only being established via long-range nested PCR.


Subject(s)
Azoospermia/genetics , Cell Cycle Proteins/genetics , Mutation, Missense , Primary Ovarian Insufficiency/genetics , Adult , Consanguinity , Female , Homozygote , Humans , Infertility, Female/genetics , Infertility, Male/genetics , Male , Pedigree , Siblings
14.
Genet Med ; 22(3): 524-537, 2020 03.
Article in English | MEDLINE | ID: mdl-31578471

ABSTRACT

PURPOSE: Lamb-Shaffer syndrome (LAMSHF) is a neurodevelopmental disorder described in just over two dozen patients with heterozygous genetic alterations involving SOX5, a gene encoding a transcription factor regulating cell fate and differentiation in neurogenesis and other discrete developmental processes. The genetic alterations described so far are mainly microdeletions. The present study was aimed at increasing our understanding of LAMSHF, its clinical and genetic spectrum, and the pathophysiological mechanisms involved. METHODS: Clinical and genetic data were collected through GeneMatcher and clinical or genetic networks for 41 novel patients harboring various types ofSOX5 alterations. Functional consequences of selected substitutions were investigated. RESULTS: Microdeletions and truncating variants occurred throughout SOX5. In contrast, most missense variants clustered in the pivotal SOX-specific high-mobility-group domain. The latter variants prevented SOX5 from binding DNA and promoting transactivation in vitro, whereas missense variants located outside the high-mobility-group domain did not. Clinical manifestations and severity varied among patients. No clear genotype-phenotype correlations were found, except that missense variants outside the high-mobility-group domain were generally better tolerated. CONCLUSIONS: This study extends the clinical and genetic spectrum associated with LAMSHF and consolidates evidence that SOX5 haploinsufficiency leads to variable degrees of intellectual disability, language delay, and other clinical features.


Subject(s)
DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , SOXD Transcription Factors/genetics , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Humans , Infant , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Language Development Disorders/diagnosis , Language Development Disorders/genetics , Language Development Disorders/pathology , Male , Mutation, Missense/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/pathology , Pedigree , Phenotype , Young Adult
15.
J Med Genet ; 56(8): 526-535, 2019 08.
Article in English | MEDLINE | ID: mdl-30923172

ABSTRACT

BACKGROUND: Balanced chromosomal rearrangements associated with abnormal phenotype are rare events, but may be challenging for genetic counselling, since molecular characterisation of breakpoints is not performed routinely. We used next-generation sequencing to characterise breakpoints of balanced chromosomal rearrangements at the molecular level in patients with intellectual disability and/or congenital anomalies. METHODS: Breakpoints were characterised by a paired-end low depth whole genome sequencing (WGS) strategy and validated by Sanger sequencing. Expression study of disrupted and neighbouring genes was performed by RT-qPCR from blood or lymphoblastoid cell line RNA. RESULTS: Among the 55 patients included (41 reciprocal translocations, 4 inversions, 2 insertions and 8 complex chromosomal rearrangements), we were able to detect 89% of chromosomal rearrangements (49/55). Molecular signatures at the breakpoints suggested that DNA breaks arose randomly and that there was no major influence of repeated elements. Non-homologous end-joining appeared as the main mechanism of repair (55% of rearrangements). A diagnosis could be established in 22/49 patients (44.8%), 15 by gene disruption (KANSL1, FOXP1, SPRED1, TLK2, MBD5, DMD, AUTS2, MEIS2, MEF2C, NRXN1, NFIX, SYNGAP1, GHR, ZMIZ1) and 7 by position effect (DLX5, MEF2C, BCL11B, SATB2, ZMIZ1). In addition, 16 new candidate genes were identified. Systematic gene expression studies further supported these results. We also showed the contribution of topologically associated domain maps to WGS data interpretation. CONCLUSION: Paired-end WGS is a valid strategy and may be used for structural variation characterisation in a clinical setting.


Subject(s)
Chromosome Aberrations , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Gene Rearrangement , Genetic Association Studies , Phenotype , Whole Genome Sequencing , Adolescent , Adult , Biomarkers , Child , Child, Preschool , Chromosome Breakpoints , DNA Copy Number Variations , Female , Genetic Association Studies/methods , Humans , Infant , Male , Structure-Activity Relationship , Translocation, Genetic , Young Adult
16.
Hum Mutat ; 40(7): 886-892, 2019 07.
Article in English | MEDLINE | ID: mdl-30924587

ABSTRACT

Premature ovarian insufficiency involves amenorrhea and elevated follicle-stimulating hormone before age 40, and its genetic basis is poorly understood. Here, we study 13 premature ovarian insufficiency (POI) patients using whole-exome sequencing. We identify PREPL and TP63 causative variants, and variants in other potentially novel POI genes. PREPL deficiency is a known cause of syndromic POI, matching the patients' phenotype. A role for TP63 in ovarian biology has previously been proposed but variants have been described in multiorgan syndromes, and not isolated POI. One patient with isolated POI harbored a de novo nonsense TP63 variant in the terminal exon and an unrelated patient had a different nonsense variant in the same exon. These variants interfere with the repression domain while leaving the activation domain intact. We expand the phenotypic spectrum of TP63-related disorders, provide a new genotype:phenotype correlation for TP63 and identify a new genetic cause of isolated POI.


Subject(s)
Codon, Nonsense , Primary Ovarian Insufficiency/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Female , Genetic Predisposition to Disease , Humans , Pedigree , Prolyl Oligopeptidases , Protein Domains , Serine Endopeptidases/genetics , Transcription Factors/chemistry , Tumor Suppressor Proteins/chemistry , Exome Sequencing/methods
17.
Cytogenet Genome Res ; 159(4): 201-207, 2019.
Article in English | MEDLINE | ID: mdl-31865337

ABSTRACT

Genetic factors are responsible for 15% of male infertility conditions. Numerical and structural chromosomal anomalies (related to the Y chromosome or to the autosomes) are validated genetic factors leading to spermatogenic quantitative defects with a frequency depending on the severity of the phenotype. The most frequent structural chromosomal rearrangements of autosomes are translocations and inversions, whereas dicentric chromosomes involving autosomes are rare. We report a man bearing a pseudodicentric chromosome (9;21) and presenting with oligozoospermia. Extensive cytogenetic analyses were necessary to determine the precise nature of the derivative chromosome and to discount the presence of interstitial telomeric sequences. Defects in spermatogenesis and abnormal segregation at meiosis for existing spermatozoa are proposed and are the likely cause of the reproductive phenotype of the patient.


Subject(s)
Chromosomes/genetics , Oligospermia/genetics , Translocation, Genetic/genetics , Adult , Chromosome Inversion/genetics , Humans , Male , Spermatogenesis/genetics , Spermatozoa/physiology
18.
Am J Med Genet A ; 179(6): 993-1000, 2019 06.
Article in English | MEDLINE | ID: mdl-30888095

ABSTRACT

This report presents two families with interstitial 11q24.2q24.3 deletion, associated with malformations, hematologic features, and typical facial dysmorphism, observed in Jacobsen syndrome (JS), except for intellectual disability (ID). The smallest 700 Kb deletion contains only two genes: FLI1 and ETS1, and a long noncoding RNA, SENCR, narrowing the minimal critical region for some features of JS. Consistent with recent literature, it adds supplemental data to confirm the crucial role of FLI1 and ETS1 in JS, namely FLI1 in thrombocytopenia and ETS1 in cardiopathy and immune deficiency. It also supports that combined ETS1 and FLI1 haploinsufficiency explains dysmorphic features, notably ears, and nose anomalies. Moreover, it raises the possibility that SENCR, a long noncoding RNA, could be responsible for limb defects, because of its early role in endothelial cell commitment and function. Considering ID and autism spectrum disorder, which are some of the main features of JS, a participation of ETS1, FLI1, or SENCR cannot be excluded. But, considering the normal neurodevelopment of our patients, their role would be either minor or with an important variability in penetrance. Furthermore, according to literature, ARHGAP32 and KIRREL3 seem to be the strongest candidate genes in the 11q24 region for other Jacobsen patients.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Jacobsen Distal 11q Deletion Syndrome/diagnosis , Jacobsen Distal 11q Deletion Syndrome/genetics , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA, Long Noncoding , Comparative Genomic Hybridization , Facies , Female , Humans , In Situ Hybridization, Fluorescence , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Karyotyping , Male , Pedigree , Phenotype
19.
Prenat Diagn ; 39(6): 464-470, 2019 05.
Article in English | MEDLINE | ID: mdl-30896039

ABSTRACT

OBJECTIVES: Congenital heart defects (CHDs) may be isolated or associated with other malformations. The use of chromosome microarray (CMA) can increase the genetic diagnostic yield for CHDs by between 4% and 10%. The objective of this study was to evaluate the value of CMA after the prenatal diagnosis of an isolated CHD. METHODS: In a retrospective, nationwide study performed in France, we collected data on all cases of isolated CHD that had been explored using CMAs in 2015. RESULTS: A total of 239 fetuses were included and 33 copy number variations (CNVs) were reported; 19 were considered to be pathogenic, six were variants of unknown significance, and eight were benign variants. The anomaly detection rate was 10.4% overall but ranged from 0% to 16.7% as a function of the isolated CHD in question. The known CNVs were 22q11.21 deletions (n = 10), 22q11.21 duplications (n = 2), 8p23 deletions (n = 2), an Alagille syndrome (n = 1), and a Kleefstra syndrome (n = 1). CONCLUSION: The additional diagnostic yield was clinically significant (3.1%), even when anomalies in the 22q11.21 region were not taken into account. Hence, patients with a suspected isolated CHD and a normal karyotype must be screened for chromosome anomalies other than 22q11.21 duplications and deletions.


Subject(s)
Genetic Testing/methods , Heart Defects, Congenital/genetics , Microarray Analysis/methods , Prenatal Diagnosis/methods , Adult , Chromosome Aberrations , Chromosomes/chemistry , Chromosomes/genetics , Comparative Genomic Hybridization/methods , DNA Copy Number Variations , Female , Fetus/chemistry , Fetus/metabolism , France , Heart Defects, Congenital/diagnosis , Humans , Karyotyping , Pregnancy , Retrospective Studies , Syndrome
20.
J Hum Genet ; 63(5): 691-698, 2018 May.
Article in English | MEDLINE | ID: mdl-29540854

ABSTRACT

Ovarian reserve represents the number of available follicles/oocytes within ovaries and it can be assessed by follicle stimulating hormone levels, anti-Müllerian hormone levels, and/or antral follicle count determined by ultrasounds. A low ovarian reserve is defined by an abnormal ovarian reserve test. This condition can be considered premature if it occurs before the age of 40, leading to premature ovarian insufficiency. Despite the growing knowledge concerning the genetic basis of ovarian deficiency, the majority of cases remain without a genetic diagnosis. Although 22q11.2 deletions and duplications have been associated with genitourinary malformations, ovarian deficiency is not a commonly reported feature. We report here four patients bearing a 22q11.2 rearrangement, identified during the clinical assessment of their low ovarian reserve or premature ovarian insufficiency, and discuss the molecular basis of the ovarian defects.


Subject(s)
Chromosomes, Human, Pair 22 , Ovarian Reserve/genetics , Primary Ovarian Insufficiency/diagnosis , Primary Ovarian Insufficiency/genetics , Translocation, Genetic , Adult , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Humans , In Situ Hybridization, Fluorescence , Karyotype , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL