Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2019

Open Access 01.12.2019 | Research

Outcomes of 4 years of molecular genetic diagnosis on a panel of genes involved in premature aging syndromes, including laminopathies and related disorders

verfasst von: Maude Grelet, Véronique Blanck, Sabine Sigaudy, Nicole Philip, Fabienne Giuliano, Khaoula Khachnaoui, Godelieve Morel, Sarah Grotto, Julia Sophie, Céline Poirsier, James Lespinasse, Laurent Alric, Patrick Calvas, Gihane Chalhoub, Valérie Layet, Arnaud Molin, Cindy Colson, Luisa Marsili, Patrick Edery, Nicolas Lévy, Annachiara De Sandre-Giovannoli

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2019

Abstract

Background

Segmental progeroid syndromes are a heterogeneous group of rare and often severe genetic disorders that have been studied since the twentieth century. These progeroid syndromes are defined as segmental because only some of the features observed during natural aging are accelerated.

Methods

Since 2015, the Molecular Genetics Laboratory in Marseille La Timone Hospital proposes molecular diagnosis of premature aging syndromes including laminopathies and related disorders upon NGS sequencing of a panel of 82 genes involved in these syndromes.
We analyzed the results obtained in 4 years on 66 patients issued from France and abroad.

Results

Globally, pathogenic or likely pathogenic variants (ACMG class 5 or 4) were identified in about 1/4 of the cases; among these, 9 pathogenic variants were novel. On the other hand, the diagnostic yield of our panel was over 60% when the patients were addressed upon a nosologically specific clinical suspicion, excepted for connective tissue disorders, for which clinical diagnosis may be more challenging. Prenatal testing was proposed to 3 families. We additionally detected 16 variants of uncertain significance and reclassified 3 of them as benign upon segregation analysis in first degree relatives.

Conclusions

High throughput sequencing using the Laminopathies/ Premature Aging disorders panel allowed molecular diagnosis of rare disorders associated with premature aging features and genetic counseling for families, representing an interesting first-level analysis before whole genome sequencing may be proposed, as a future second step, by the National high throughput sequencing platforms (“Medicine France Genomics 2025” Plan), in families without molecular diagnosis.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13023-019-1189-z) contains supplementary material, which is available to authorized users.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
aCGH
array comparative genomic hybridization
ACMG
American College of Medical Genetics
APS
Atypical progeroid syndromes
AWS
Atypical Werner Syndrome
BOFS
Branchio-Oculo-Facial Syndrome
BS
Bloom syndrome
cEDS
Classical EDS
CL
Cutis Laxa
clEDS
Classical-Like EDS
CRB-TAC
Biological Resources Center, Tissues-DNA-Cells
DCM
Dilated Cardiomyopathy
DNA
Desoxyribonucleic acid
EDS
Ehlers Danlos Syndrome
HGPS
Hutchinson Gilford Progeria Syndrome
MAD-B
Type B mandibuloacral dysplasia
RD
Restrictive Dermopathy
RTS
Rothmund Thomson Syndrome
SHORT
Short stature, Hyperextensibility of joints, Ocular depression, Rieger anomaly, Teething delay
UPS
Unspecified Progeroid Disorder
VUS
Variant of Unknown Significance
WS
Werner syndrome

Background

Segmental progeroid syndromes are a heterogeneous group of rare genetic disorders, which were clinically described in medical journals since the twentieth century. These syndromes are defined as segmental because only some of the aging features are accelerated [1]. Historically, and based on their main pathophysiological bases, three major groups of progeroid syndromes may be distinguished.
The first consists of syndromes due to alteration of DNA-repair genes. The RecQ family genes, encoding DNA helicases, are mainly involved in this group of syndromes [2, 3]. DNA helicases are enzymes involved in different DNA-repair processes and are referred to as guardians of the genome. They play important roles in genome stability and maintenance [4]. Several syndromes are associated to the RecQ genes’ family including Werner syndrome (WS), Bloom syndrome (BS) and Rothmund Thomson syndrome (RTS) [57], that are respectively associated with pathogenic variants in RECQL2/WRN, RECQL3/BLM, and RECQL4 genes. These syndromes share autosomal recessive inheritance and an increased susceptibility to cancer. WS is an adult-onset progeria syndrome, whereas the first symptoms of BS and RTS appear in childhood. Cockayne syndrome is another autosomal recessive disorder caused by mutations of DNA-repair genes, the CS proteins. Pathogenic variants of CSA/ERCC8 and CSB/ERCC6 genes are responsible for the majority of cases [1, 8, 9].
The second group deals with proteins maintaining the integrity of the nuclear envelope. Lamins A/C and their protein partners are mainly involved in this group of disorders. In 2003, De Sandre-Giovannoli et al. and Eriksson et al. identified a synonymous point mutation of the LMNA gene (NM_170707.4: c.1824C > T; p.Gly608Gly) as causative of the disease in patients affected with Hutchinson Gilford Progeria Syndrome (HGPS) [10, 11]. This synonymous and apparently harmless heterozygous variant activates a cryptic donor splice site in LMNA pre-mRNAs, leading to the production of a prelamin A precursor that lacks the cleavage recognition site for the endoprotease ZMPSTE24. As a result, a truncated and permanently prenylated prelamin A isoform named progerin accumulates and exerts multiple toxic effects in cells’ nuclei [1012]. Other LMNA mutations affecting prelamin A maturation result in more or less severe progeroid syndromes, called HGPS-like, depending essentially on the quantities of progerin/prelamin A isoforms produced [13]. Two other syndromes, restrictive dermopathy (RD), a perinatal lethal genodermatosis, and type B mandibuloacral dysplasia (MAD-B), a relatively milder progeroid syndrome characterized by skeletal, metabolic and cutaneous abnormalities and lipodystrophy, have also been associated to pathological accumulation of prelamin A. Recessive pathogenic variants in ZMPSTE24 have mostly been described in these syndromes, broadening the spectrum of prelamin-A associated disorders [1, 1419]. Furthermore, several atypical progeroid syndromes (APS) or atypical Werner syndrome (AWS) with clinical features overlapping with HGPS and other prelamin A-linked disorders have been associated to missense mutations in the LMNA gene, which are often private and are seen in only a single or few families [2027].
The third group is composed of syndromes characterized by features of premature aging resulting from diverse pathophysiological processes. This group includes all the disorders listed in Additional file 1: Table S1 and not belonging to the 1st and 2nd major groups of progeroid syndromes. It also includes Ehlers Danlos syndromes (EDS), a clinically and genetically heterogeneous group of diseases that affect connective tissues. EDS are classified into 13 subtypes according to the inheritance pattern, phenotype and pathogenetic mechanisms [28] and some of its subtypes present with premature aging signs. Cutis Laxa (CL) syndromes presenting as well with progeroid features are also included in this third group [29].
Since 2015, the molecular genetics laboratory in Marseille La Timone Hospital proposes molecular diagnosis of premature aging syndromes as well as laminopathies and related syndromes upon NGS (next generation sequencing) analysis of a panel of 82 genes involved in those disorders (Additional file 1: Table S1). To the best of our knowledge, this panel is unique in France (Fig. 1) and in Europe (https://​www.​orpha.​net/​consor/​cgi-bin/​ClinicalLabs.​php?​lng=​FR).
We report herein the outcomes of 4 years of NGS molecular diagnosis in 66 index cases coming from France and abroad, affected with syndromes featuring premature aging, and attempt to provide a critical discussion of the results obtained.

Materials and methods

Next generation sequencing and mutation identification procedures

In a diagnostic setting, we performed the targeted analysis of 82 genes (Additional file 1: Table S1). The genes were included into the panel because of their association with laminopathies or with diseases including premature ageing features; they were chosen upon bibliographic search (PubMed) and using genetic/clinical databases (namely OMIM and Orphanet).
Since 2015, 66 index cases were subjected to the NGS analysis and included in this study. The criteria used to include patients in the molecular study were one of the following: (i) patients whose clinical diagnosis fitted with one of the disorders associated with a gene of the panel (Additional file 1: Table S1), whether it was a progeroid syndrome or not, but having a specific nosologic classification, (ii) patients for whom the prescribing physician suspected a disease related to premature aging, due the association of at least two premature aging signs, but without a specific nosologic classification.
DNA was extracted from peripheral blood samples by the Biological Resources Center CRB-TAC (with NF-S 86–900 and ISO-9001 V2015 certifications) of the Department of Medical Genetics.
We created a SureSelectXT 12-24 Mb library, custom 1032 gene panel in collaboration with Agilent Technologies (Santa Clara, California, USA), to be used for enrichment of targeted sequences for several molecular diagnosis indications in the laboratory. The coding regions and flanking intronic regions of the 1032 genes were enriched, in solution, using the SureSelect Target Enrichment System from Agilent (Santa Clara, California, USA), following the manufacturer recommendations. The Ion Proton platform (Thermo Fisher Scientific, USA) was used for high throughput sequencing. Then, raw data were converted to Fastq files and aligned to the reference sequence of the human genome (University of California Santa Cruz, hg19/GRCh37, (https://​genome.​ucsc.​edu/​), using the Torrent Suite software (Thermo Fisher). The same software was used to perform variant calling (germline_low_stringency_targetseq), with the following parameters (min_cov_each_strand: 0, min_variant_score: 10, min_allele_freq: 0.1, snp_min_coverage: 6 snp and indel; strand_bias: 0.98 snp and 0.85 indel). A variant calling format (VCF) file and binary alignment map and index (BAM/BAI) files were then obtained and used for variant annotation using the in-house software VarAFT (Variant Annotation and Filtration Tool, https://​varaft.​eu/​) [30]). The list of genes to be analyzed for each patient was also selected using dedicated bedfiles by VarAFT. Sequence reads and variants’ visualization was performed using IGV (https://​software.​broadinstitute.​org/​software/​igv/​) [31].
The BEDTools-based VarAFT coverage module was used to compute breadth and depth of coverage data using BAM files for the selected gene list, producing a coverage report. Nucleotidic positions were interpreted only over a cutoff of 20X reads depth.
Variants whose allele frequencies were > 1% in GnomAD (https://​gnomad.​broadinstitute.​org) and 1000 genomes (http://​www.​internationalgen​ome.​org/​) were removed, as well as deep intronic variants. The remaining variants were analyzed according to their genomic position and their predicted effects on RNAs and proteins. In order to study the pathogenicity of the variants, we used population databases such as gnomAD (https://​gnomad.​broadinstitute.​org/​), sequence databases such as ClinVar (https://​www.​ncbi.​nlm.​nih.​gov/​clinvar/​), LOVD (http://​www.​lovd.​nl/​3.​0/​home), disease databases such as OMIM (https://​www.​omim.​org/​), integrated databases as ClinGen and in silico predictive algorithms such as Mutation Taster (http://​www.​mutationtaster.​org/​), Human Splicing Finder (http://​www.​umd.​be/​HSF3/​) and UMD Predictor (http://​umd-predictor.​eu/​), together with a review of the literature [3234].
The variants were classified as “pathogenic”, “likely pathogenic”, “of uncertain significance”, “likely benign” or “benign” (respectively classes 5 to 1) according to the American College of Medical Genetics and Genomics (ACMG) classification, using Intervar (http://​wintervar.​wglab.​org/​), ClinVar (https://​www.​ncbi.​nlm.​nih.​gov/​clinvar/​) and ClinGen (https://​www.​clinicalgenome.​org/​); the classification was manually adjusted using further ACMG criteria, whenever necessary, depending on the familial context and literature data [35]. In each case, the additional ACMG criteria allowing the reclassification of variants are given in Table 1.
Table 1
Pathogenic variants and VUS identified from 2015 to 2018 in our laboratory by NGS.
Patient
Age at referral
Indication
Gene
NM
Haplotype
cDNA change
Amino acid change
ACMG (default Intervar)
Details of Intervar
ACMG (Intervar reintepreted or manual-Maryland)
Additional criteria leading to the variant class change
ExAC
GnomAD
Mutation Taster
UMD-predictor
HSF
Literature
P1
11m
CL 2B
PYCR1
NM_006907.3
hoz
c.616G>A
p.(GLy206Arg)
LP
PM1 PM2 PP3 PP5
P
PS1 PP1
1/23214 (4.308e-05)
2/186106 (1.07e-5)
D0.99
P 100
Probably no impact on splicing
Kretz et al. 2011; Reversade et al. 2009
P2
26y
RTS
RECQL4
NM_004260.3
comp.het
c.2263C>T
p.(Arg755Trp)
VUS
PM1 PM2 PP3 BP1
LP
PM3
NA
1/240964 (4.15e-6)
NA
NA
Potential alteration of splicing
THIS REPORT
c.2415_2419dup
p.(Arg807fs113Ter)
NA
 
P
PM2 PVS1 PP3
NA
NA
NA
NA
Potential alteration of splicing
THIS REPORT
P3
9y2m
CL
ALDH18A1
NM_002860.4
het
c.413G>A
p.(Arg138Gln)
LP
PM1 PM2 PP2 PP3 PP5
P
PS1
NA
NA
D0.999
P 84
Potential alteration of splicing
Fischer-Zirnsak et al. 2015
P4
19y
BOFS
TFAP2A
NM_003220.3
het
c.710G>A
p.(Arg237Gln)
LP
PM1 PM2 PP2 PP3 PP5
P
PS1
NA
NA
D0.999
P 87
Potential alteration of splicing
Reiber et al. 2010 and p.(Arg237Gly) in Milunsky et al., 2010
P5
6m
CL
GORAB
NM_152281.2
comp.het
c.546A>T
p.(Glu182Asp)
VUS
PM2
LP
PM3 PP3 PP4
NA
NA
D0.999
PP72
Potential alteration of splicing
THIS REPORT
c.859C>T
p.(Arg287Ter)
VUS
PM2 PP3 PP5
P
PVS1 PM3
2/121244 (1.65e-05)
3/251106 (1.19e-5)
D1
P100
Probably no impact on splicing
THIS REPORT
P6
8m
RTS
RECQL4
NM_004260.3
comp.het
c.1573delT
p.Cys525AlafsTer33
NA
 
P
PVS1 PM2 PM3 PP5
27/104602 (2.581e-04)
69/276484 (2.5e-4)
NA
NA
Potential alteration of splicing
Kitao et al. 1999; Siitonen et al. 2009
c.2269C>T
p.(Gln757Ter)
P
PVS1 PM2 PP3 PP5
P
/
9/113308 (7.943e-05)
32/271122 (1.18e-4)
NA
NA
Potential alteration of splicing
Pujol et al.2000; Wang et al. 2003;Siitonen et al. 2009
P7●
19y
Cockayne Sd
ERCC6
NM_000124.4
het
c.2291T>C
p.(Leu764Ser)
VUS
PM1 PM2 PP3 BP1
LP
PM3 (deletion of the other allele)
NA
NA
D0.999
P90
Probably no impact on splicing
THIS REPORT
P8
41y
Werner Sd
WRN
NM_000553.5
comp.het
c.2313T>A
p.(Cys771Ter)
P
PVS1 PM2 PP3
P
/
NA
1/251244 (3.98e-6)
D1
P100
Probably no impact on splicing
THIS REPORT
c.2665C>T
p.(Arg889Ter)
P
PVS1 PM2 PP3 PP5
P
/
4/120718 (3.314e-05)
6/251074 (2.39e-5)
D1
P100
Potential alteration of splicing
P9
43y
EDS
COL5A1
NM_000093.4
het
c.1884_1891del
p.(Asp629Phefs16Ter)
NA
 
LP
PVS1 PM2
NA
NA
NA
NA
Potential alteration of splicing
THIS REPORT
P10
24y
SHORT Sd
PIK3R1
NM_181523.2
het
c.1945C>T
p.(Arg649Trp)
LP
PM1 PM2 PP3 PP5
P
PS1 PP2
NA
NA
D0.999
P93
Potential alteration of splicing
Dyment et al. 2013
P11
63y
UPS
LMNA
NM_170707.4
het
c.1003C>T
p.(Arg335Trp)
VUS
PM2 PP3 PP5
P
PS1 PM1
NA
NA
D 0.999
P96
Probably no impact on splicing
Zaragova et al. 2017; Lambert et al., 2018
P12
37y
Buschke Ollendorf Sd
LEMD3
NM_014319.4
het
c.1323C>A
p.(Tyr441Ter)
VUS
PM2 PP3
P
PVS1 PS1 PP5
NA
NA
D1
P100
Potential alteration of splicing
Hellemans et al. 2006
P13
2m
CL
ALDH18A1
NM_002860.3
hoz
c.1499G>T
p.(Gly500Val)
LP
PM1 PM2 PP2 PP3
P
PP1 PM3
NA
0/239782 (0)
D0.99
P90
Potential alteration of splicing
THIS REPORT
P14
39y
EDS
COL5A1
NM_000093.4
comp.het
c.4030C>T
p.(Pro1344Ser)
VUS
PM2 PP3 BP1
VUS
/
NA
NA
D 0.999
P96
Potential alteration of splicing
THIS REPORT
c.2374C>T
p.(Arg792Ter)
P
PVS1 PM2 PP3 PP5
P
/
NA
NA
D1
P100
Potential alteration of splicing
Wenstrup et al. 2010
P15
36y
Brittle cornea
PRDM5
NM_018699.2
hoz
c.1036C>T
p.(Arg346Ter)
P
PVS1 PM2 PP3
P
/
3/119938 (2.501e-05)
3/244748 (1.23e-5)
D1
P100
Potential alteration of splicing
THIS REPORT
P16
6y7m
Oculoden-todigital dysplasia
GJA1
NM_000165.5
het
c.287T>A
p.(Val96Glu)
LP
PM1 PM2 PP2 PP3
P
PS1 PP5
NA
NA
D0.99
P100
NA
Wiest et al. 2006
P17
41y
EDS
LMNA
NM_005572.3 (Lamin C isoform)
het
c.1715G>A
p.(Arg572His)
VUS
PM2
VUS
/
NA
0/149540 (0)
Polym 0.992
Prob Polym
Probably no impact on splicing
THIS REPORT
P18
59y
Werner Sd
LMNA
NM_170707.4
het
c.1016C>T
p.(Ala339Val)
VUS
PM2 PP3
VUS
/
NA
NA
D0.999
P84
Potential alteration of splicing
THIS REPORT
P19
4m
UPS
RECQL4
NM_004260.3
hoz
c.2756-8G>T
p.?
NA
 
VUS
/
17/107602 (1.58e-04)
59/276606 (2.13e-4)
NA
NA
Probably no impact on splicing
THIS REPORT
P20
68y
EDS
ALDH18A1
NM_002860.4
het
c.1597G>A
p.(Val533Met)
VUS
PM1 PP2
VUS
/
NA
3/178100 (1.68e-5)
Polym 0,960
PP69
Probably no impact on splicing
THIS REPORT
P21
47y
EDS
COL1A2
NM_000089.3
het
c.798A>C
p.(Glu266Asp)
VUS
PM2
VUS
/
NA
2/282868 (7.07e-6)
D0,999
Polym45
Potential alteration of splicing
THIS REPORT
P22
20y
UPS
SYNE1
NM_182961.4
het
c.21209T>G
p.(Leu7070Trp)
VUS
BP1
VUS
/
NA
NA
Polym 0,890
P78
Potential alteration of splicing
THIS REPORT
P23a
21m
UPS
ATR
NM_001184.4
het
c.4750A>C
p.(Met1584Leu)
VUS
PM2
B
lack of segregation with the disease in the family
NA
3/251454 (1.19e-5)
D0,999
PP72
Potential alteration of splicing
THIS REPORT
P24a
17y
EDS
COL1A1
NM_000088.3
het
c.310G>A
p.(Asp104Asn)
VUS
PM2
B
lack of segregation with the disease in the family
NA
NA
Polym0,999
Polym33
Potential alteration of splicing
THIS REPORT
P25
57y
UPS
COL5A2
NM_000393.5
het
c.463C>T
p.(Arg155Cys)
VUS
PP3
VUS
/
1/49686 (2.013e-5)
7/209120 (3.35e-5)
D0,999
P100
Potential alteration of splicing
THIS REPORT
SYNE2
NM_182914.2
het
c.20632_20634delinsGAA
p.(Ser6878Glu)
NA
 
VUS
/
NA
NA
NA
NA
Potential alteration of splicing
THIS REPORT
P26a
65y
UPS
ATR
NM_001184.4
hoz
c.7701A>G
p.(Lys2567Lys)
VUS
BP4 BP7
B
lack of segregation with the disease in the family
NA
NA
D1
P77
Potential alteration of splicing
THIS REPORT
P27
21y
UPS
ELN
NM_001278913.1
het
c.1063_1086del
p.(355_362del)
NA
 
VUS
/
NA
NA
NA
NA
NA
THIS REPORT
P28
28y
EDS
TNXB
NM_019105.7
het
c.747C>T
p.(Cys249Cys)
LB
PM2 BP4 BP7
VUS
/
1/102780 (9.73e-06)
1/245798 (4.07e-6)
D1
Polym 11
Potential alteration of splicing
THIS REPORT
P29
57y
UPS
FBN1
NM_000138.4
het
c.5347A>T
p.(Ile1783Phe)
VUS
 
VUS
/
NA
NA
D0,999
P78
Potential alteration of splicing
THIS REPORT
P30
19y
EDS
ACAN
NM_013227.3
het
c.7205G>A
p.(Arg2402His)
VUS
PM1 PM2
VUS
/
8/119916 (6.671e-05)
20/248986 (8.03e-5)
D0,999
P87
Probably no impact on splicing
THIS REPORT
Pathogenic variants are shown in bold, VUS are in plain text.athe variant has been searched in family, allowing us to reclassify a VUS into a benign variant, P23: the variant was absent from affected mother, P24 and P26: the variant was present in unaffected first degree relatives ; P7● also carried an heterozygous deletion of ERCC6 . yyears old,mmonths old,Sdsyndrome,RTSRothmund Thomson Syndrome,BOFSBranchiooculofacial syndrome,CLCutis laxa,EDSEhlers Danlos Syndrome,UPSUnspecified Progeroid syndrome,hetheterozygous,comp.hetcompound heterozygous,hozhomozygous,Ppathogenic,LPlikely pathogenic,VUSvariant of unknown significance,LBlikely benign,Bbenign,NAnot availabke,Ddisease causing,Polympolymorphism,Ppathogenic,PPprobably pathogenic. Intervar:http://​wintervar.​wglab.​org/​; Maryland:http://​www.​medschool.​umaryland.​edu/​Genetic_​Variant_​Interpretation_​Tool1.​html/​
Only the variants of classes 5 to 3 were reported in molecular genetics diagnostic reports after Sanger sequencing confirmation. Genes were named following the Hugo gene nomenclature committee guidelines (https://​www.​genenames.​org/​); DNA mutations and predicted protein changes were named following the HGVS nomenclature guidelines (http://​www.​hgvs.​org/​mutnomen/​).

Sanger sequencing for the validation of variants

We designed primers for PCR amplification using the Primer3 software (http://​bioinfo.​ut.​ee/​primer3/​) in order to amplify the gene’s exon. Briefly, Sanger sequencing was performed as follows: purification of PCR products was performed according to the manufacturer’s instructions and both strands were sequenced using the Big Dye Terminator V.1.1 Cycle Sequencing Kit (Applied Biosystems). Sequence reactions were purified on Sephadex G50 (Amersham Pharmacia Biotech, Foster City, California, USA). We performed capillary electrophoresis on a Genetic Analyzer ABI3500XL (Life Technologies, USA). Electropherograms were analyzed on the Sequence Analysis Software V.5.2 (Applied Biosystems) and aligned and interpreted relative to the reference sequence using Sequencher V.5.4.6 (Gene Codes Corporation).

Results

Since 2015, we analyzed 66 patients from all over France and abroad on a panel of genes involved in premature aging syndromes. As depicted in Fig. 1, 50% of the patients were addressed from the southern regions of France and only 3% from abroad (Fig. 2). To the best of our knowledge, this kind of gene panel largely exploring premature aging syndromes and laminopathies is unique in France and Europe (https://​www.​orpha.​net/​consor/​cgi-bin/​index.​php). Some overlap exists with other gene panels, specifically exploring sets of genes involved in particular diseases such as for example connective tissue disorders or Cockayne and related syndromes (Fig. 1).
The average coverage obtained at 20X on the list of genes and corresponding reference transcripts we describe (Additional file 1: Table S1) was 93% (248,228/267,610 nucleotides covered at 20X +/− 10,237 mean nucleotides, i.e. +/− 4% mean SD).
The molecular analysis allowed to identify 20 pathogenic or likely pathogenic variants in 16 patients according to ACMG, using Intervar (http://​wintervar.​wglab.​org/​), literature and databases, as described above (Table 1). Whenever necessary, the clinical interpretation of sequence variants was adjusted manually according to the literature, genetic databases and patients’ clinical data.
Globally, class 1–2 (benign and likely benign) variants were identified in 59% of the patients, class 3 variants (VUS, variant of unknown significance) in 15% and class 5 and 4 variants (pathogenic or likely pathogenic, respectively) in 26% of the patients (about 1/4). In order to analyze the molecular diagnostic yield with respect to the nosologic specificity of the clinical suspicion, we divided the index cases in two, then in three groups according to the clinical diagnosis/indication for NGS: respectively “specific clinical suspicion and unspecified progeroid disorders” then “specific clinical suspicion other than connective tissue disorders, connective tissue disorders (namely including CL and EDS) and unspecified progeroid disorders” (Fig. 3a, b). As expected, when considering the first group of patients with “specific clinical suspicion” we obtained a 39% diagnostic yield for ACMG classes 4 and 5 variants (likely pathogenic or pathogenic), compared to the “unspecified progeroid disorders” for which the same types of variants were retrieved only in 10% of the patients. When we further divided the first group in two, i.e. “specific clinical suspicion other than connective tissue disorders and connective tissue disorders”, we obtained a much higher diagnostic yield (62%) with ACMG classes 4 and 5 variants for patients with a nosologically defined clinical syndrome vs only 26% for connective tissue disorders (Fig. 3b). In the same graph, we can see that the detection rate of class 3 variants was the same for the two subgroups.
Table 1 compiles the 36 variants observed in our cohort from class 3 to 5; 9 of the 20 variants belonging to classes 4 and 5 are described in this report for the first time (Table 1, last column "THIS REPORT").
In the group of nosologically defined, suspected clinical syndromes, we identified compound heterozygous pathogenic variants of RecQ family genes for three patients: RECQL4 in two patients (P2 and P6) presenting with RTS and WRN/RECQL2 in one patient with WS (P8). Both patients with RTS were female and had intrauterine and postnatal growth retardation, poikiloderma (collectively defining the following skin anomalies: reticulated hypo- and hyperpigmentation, punctate atrophy and telangiectasias) (Fig. 4B1-B3) and skeletal anomalies (fractures for patient P2 and decreased bone mineral density for patient P6). Patient P2 was older than patient P6, she developed a metastatic colon cancer and died at age 27. The pathogenic heterozygous variants found in patient P6 had already been described in a patient presenting with RTS [3638], whereas the two pathogenic variants observed in patient P2 (c.2263C > T; p.(Arg755Trp) and c.2415_2419dup; p.(Arg807fs113Ter)) are described for the first time in this report. Patient P8 affected with WS presented with bilateral cataract, premature graying of scalp hair, alterations of the skin (thin skin and hyperpigmentation), soft tissue calcification, Achille’s tendinopathy, muscle atrophy and hypothyroidism at age 38. One of the pathogenic variants identified in the WRN gene (c.2313 T > A; p.(Cys771Ter)) is novel as well. Another patient (P7) was referred to our laboratory with clinical features of late onset Cockayne Syndrome. A deletion of 4,8 Mb encompassing the ERCC6 gene was already identified by array CGH and we observed a missense variant in exon 12 on the other ERCC6 allele (c.2291 T > C; p.(Leu764Ser)).
We identified already described pathogenic variants confirming the clinical diagnoses of SHORT Syndrome in patient P10, of Buschke-Ollendorf syndrome in patient P12, of Branchio-Oculo-Facial Syndrome (BOFS) in patient P4 and of Oculodentodigital Dysplasia in patient P16 (Table 1), respectively in the PIK3R1, LEMD3/MAN1, TFAP2A and GJA1 genes [3942]. As it can be seen in Fig. 4D1, D2, patient P10 affected with SHORT syndrome presented with short stature (below the 3rd SD), triangular face with prominent forehead, deep-set eyes, narrow tip of the nose, low-hanging columella and partial lipodystrophy at age 23. She also presented Axenfeld-Rieger anomaly, diabetes mellitus, hearing loss and ovarian cysts.
In the group of connective tissue disorders, we identified new compound heterozygous or homozygous class 4 or 5 variants in patients presenting with Cutis Laxa: GORAB (c.546A > T; p.(Glu182Asp)) and c.859C > T; p.(Arg287Ter)) for patient P5 and an homozygous novel variant in ALDH18A1 (c.1499G > T; p.(Gly500Val)) for patient P13 (Table 1 and Fig. 4F1-F4). Patient P5 was evaluated in his first year of life: he showed joint laxity, bilateral congenital hip dislocation, fracture of the tibia, skin wrinkling, premature aged appearance of the face, deep set eyes, droopy cheeks and a pinched nose (Fig. 4C1, C2). His nonsense variant was inherited from the mother, who presented with a mild phenotype associating joint laxity and a pinched nose. GORAB compound heterozygous variants and the clinical features of patient P5 were compatible with Geroderma osteodyplastica (GO) or Autosomic Recessive Cutis Laxa type 2 (ARCL2) diagnoses. Patient P13 was the first child of a consanguineous couple from Senegal; IUGR (intrauterine growth retardation) was observed during pregnancy. He was born prematurely with height and weight below the third centile and his head circumference was between the 5th and 10th percentile. Clinical examination at 1 month of life evidenced retarded postnatal growth (below the 3rd centile, as at birth), hypotonia, failure to thrive, large fontanelles, facial progeroid appearance and cutis laxa. He also presented stenosis of the aortic arch. ALDH18A1 pathogenic variants are responsible of De Barsy syndrome, previously known as ARCL3A, defining a molecular diagnosis that was compatible with the clinical features of patient P13 [29]. Other pathogenic variants already described in the literature confirmed the diagnosis of CL for patients P1 and P3 (Table 1 and Fig. 4A1-A4 for patient P1) [4345]. Patient P1 (Fig. 4A1-A4) presented with aged appearance, a prominent forehead, loss of adipose tissue, translucent and wrinkly skin with visible veins. He also showed hypotonia and prenatal and postnatal growth retardation with conserved head circumference. This variant had already been associated with autosomal recessive Cutis Laxa [43, 44]. As mentioned above, EDS is another clinically and genetically heterogeneous group of connective tissue disorders. We identified two heterozygous nonsense variants in COL5A1 in two patients presenting with classical EDS: one of them (c.1884_1891del; p.(Asp629Phefs16Ter) is described in this report for the first time, in patient P9, while the other (c.2374C > T; p.(Arg792Ter) [46]) was observed in a compound heterozygous state with a novel missense VUS in the same gene: (c.4030C > T; p.(Pro1344Ser)) in patient P14 (Table 1). Brittle Cornea syndrome is classified as one of the EDS subtypes [28, 42, 47]. In our cohort, patient P15, carried a homozygous nonsense variant in PRMD5 that was not reported before (c.1036C > T; p.(Arg346Ter), Table 1). Patient P15 was issued from a consanguineous union; at the age of 35 years, he presented with bilateral keratoconus, keratoglobus with bilateral corneal transplants, bilateral cataract surgery, blue sclerae, hearing loss, scoliosis and joint laxity.
We additionally observed 3 LMNA variants, of which one was considered as pathogenic based on previous publications (c.1003C > T; p.(Arg335Trp)) [4854]. This variant was previously described in two patients presenting with acro-osteolysis and dilated cardiomyopathy (DCM), another with a cardiac phenotype, acro-osteolysis and hypertriglyceridemia and other patients presenting with isolated DCM [4852]. As described in Lambert et al., [54] patient P11 presented with a severe progeroid phenotype including acro-osteolysis with painful articulations of hands and feet, she had a pace-maker for heart rhythm disturbances (atrial fibrillation and atrioventricular block) without DCM on echocardiograms and she reported that children laughed at her at school for her “aged face and skin” [54]. Patient P11, shown in Fig. 4E1-E4 at 63 years old, presented with a pinched nose, malar hypoplasia, and an emaciated aspect of the face with aged, thin and dyspigmented skin, brachydactyly due to osteolysis predominating on the first three distal phalanges of both hands, and xerosis of the skin.
We additionally identified 16 variants of unknown significance (VUS, ACMG class 3 variants). Among them, 3 were reclassified as benign upon segregation analysis on DNAs of first-degree relatives. In the remaining cases, we didn’t receive parental DNAs to help reclassify them. Almost all VUS were identified in patients referred to our laboratory for Ehlers Danlos syndrome or unspecified progeroid syndromes. Among the suspected EDS, at least two patients (P20 and P28) had hypermobile EDS for which no gene was identified yet [28].
As mentioned above, one VUS in COL5A1 was associated with a pathogenic variant of the same gene in patient P14. The patient almost fulfilled the diagnostic criteria of Classical-Like EDS (clEDS) syndrome, i.e. as major criteria: skin hyperextensibility, general joint hypermobility (Beighton score: 8/9), spontaneous ecchymoses, autosomal recessive inheritance because she was born form a consanguineous couple and minor criteria: hand acrogeria with clinodactyly, pes planus, hallux valgus. However, she also presented atrophic scarring which is a major criterion for Classical EDS (cEDS) and is normally absent from clEDS. She had one brother with clinical signs of EDS. We did not identify pathogenic variants in TNXB (whose coding sequence NM_019105.6 was entirely covered by the genomic sequencing), but we observed one COL5A1 already known pathogenic variant (c.2374C > T; p.(Arg792Ter)) and one novel VUS (c.4030C > T; p.(Pro1344Ser)) [46]. Compound heterozygosity of COL5A1 has already been described in one family, with one missense COL5A1 variant playing the role of a “modifier gene” [55, 56]. In order to determine the impact of the VUS, the analysis of its segregation in the patient’s family would be useful.
Finally, two unreported heterozygous VUS that we would like to report were identified in the LMNA gene. One of these VUS (c.1016C > T; p.(Ala339Val)) was identified in patient P18, a 59 years old man, who displayed a range of premature-aging symptoms: premature balding since his twenties, short stature (− 2.5 SD), low body weight (− 2.25 DS), atrophic skin, sparse eyebrows and eyelashes, prominent eyes, micrognathia, and sloping shoulders. He suffered from intracranial hemorrhage at age 47 due to a ruptured posterior inferior cerebellar artery aneurysm. He also presented with aortic valve calcification associated with mild aortic insufficiency, incomplete right bundle branch block, hypertension, hypertriglyceridemia, and recurrent bone fractures upon osteoporosis. His cognitive development was normal. He was the only child of unrelated parents and he was childless. None of his relatives presented with premature aging features.
The other VUS in the LMNA gene was detected in patient P17 who was addressed for a clinical phenotype evocative of cEDS. This variant was located in the 3’UTR region of Lamin A-encoding transcripts (NM_170707.4: c.1698 + 17G > A) and in the last codon of Lamin C-encoding transcripts (NM_005572.3) (c.1715G > A; p.(Arg572His)). Unfortunately, no segregation study could be performed in the patient’s first-degree relatives.

Discussion

We report the outcomes of 4 years of molecular genetic diagnosis by high throughput sequencing on a cohort of 66 patients mostly affected with premature aging syndromes, either nosologically classified or not, using a panel of 82 genes. To the best of our knowledge, this panel is the only available in France and in Europe to offer a wide molecular genetics exploration of disorders including features of premature aging. Other panels, mostly involved in the molecular diagnosis of connective tissue disorders or more restrained nosologic entities among premature aging syndromes, partially share genes with the panel we present.
This panel allowed us to provide molecular genetics results for patients from all over France and, in a few cases, from French overseas territories or abroad. As expected, and as seen in Fig. 1, more than half of the patients’ samples received in Marseille La Timone Molecular Genetics Laboratory were issued from the southern part of France. This is due to the strong previous interactions established in other diagnostic contexts with other southern French University Hospitals, and their facilitation by the establishment of financial agreements among regional hospitals in France. Nonetheless, the other cases were addressed from the rest of French territories or abroad, probably due to the unique availability of a large molecular screening in this kind of rare syndromes where, in some cases, large clinical overlaps can be observed. Additionally, this may be explained by the long-lasting involvement of our medical team in translational research and diagnosis of premature aging disorders, namely linked to Lamins A/C [1, 10, 13, 27, 5762].
While for the global cohort of patients (n = 66) the diagnostic yield (i.e. identification of pathogenic or likely pathogenic variants) was of about 1/4 (26%), by further dividing the cohort in two groups, depending on the strength of the clinical suspicion towards a nosologically defined progeroid syndrome vs. an unspecified progeroid disorder (UPS), we observed that the diagnostic yield largely relied on this parameter, with almost 40% resolved cases in the first category of patients. When those groups were further subdivided in three (“specified clinical suspicion other than connective tissue disorders”, “connective tissue disorders” and “unspecified progeroid disorders”), the diagnostic yields were respectively 62, 26 and 10%. Indeed, this allows to observe, as expected, that the lowest yield is obtained in UPS, but a low yield is also retrieved in connective tissue disorders. This group of patients represents about 1/3rd of the cases addressed to our laboratory, with a clinical diagnosis or suspicion of EDS (Fig. 3 and data not shown). The International EDS Consortium recently defined 13 EDS subtypes and listed 19 causal genes [28]. In our panel, we analyze more than 50% of them, including the most frequently mutated ones (11/19). In France, to date, there is no available diagnostic panel exploring the 19 genes. For example, the extended panel of Tübingen in Germany, studies 90% of these 19 genes (17/19). Our panel thus represents the most extensive offer in France for EDS molecular diagnosis to date but it will be further implemented to include 19/19 genes in a future version. On the other hand, the number of patients that we received for this clinical indication points to a probably uncovered diagnostic need in France, together with the fact that it’s often hard to have a precise clinical suspicion given the high phenotypic variability of the EDS subtypes as well as the clinical overlap among EDS subtypes and other connective tissue disorders, making it interesting to use a large panel including most of the genes involved in those disorders with features of premature aging. This was also the reason why, although no specific gene has been identified yet, patients with a clinical diagnosis of hypermobile EDS were accepted and included in the molecular analysis. Indeed, more widely, many syndromes that are associated with the 82 genes included in our panel have overlapping clinical features [28, 63]. We report in this work 20 pathogenic or likely pathogenic variants in different clinical indications, 9 of them being novel. Among these novel variants 4 were non-sense and 2 frameshifting. We detected novel variants and in some cases reclassified them as pathogenic or likely pathogenic according to ACMG criteria, relative to genetic databases, literature and patients’ clinical history (Table 1): for RECQL4 c.2263C > T; p.(Arg755Trp) and c.2415_2419dup; p.(Arg807fs113Ter) variants in patient P2 presenting with clinical RTS, for GORAB c.546A > T; p.(Glu182Asp) and c.859C > T; p.(Arg287Ter) in patient P5 presenting with clinical CL. Indeed, even if most pathogenic variants of GORAB (alias: SCYL1BP1) responsible of GO are nonsense, homozygous missense variants in compound heterozygosity with null alleles have already been associated with GO in previous publications [64, 65].
We also reclassified the ERCC6 (c.2291 T > C; p.(Leu764Ser)) variant in patient P7 carrying a deletion of ERCC6 on the other allele and a clinical diagnosis of late onset Cockayne syndrome; of note, Ghai et al. described a patient affected with Cokayne syndrome carrying almost the same deletion as P7 associated to a splice variant [66]. For ALDH18A1 the homozygous variant: c.1499G > T; p.(Gly500Val) was reclassified in patient P13 presenting with features of CL; for COL5A1, the (c.1884_1891del; p.(Asp629Phefs16Ter)) nonsense variant was reclassified in patient P14 presenting with classical EDS. Other novel variants already classified as pathogenic by Intervar, an ACMG variant classification tool, were observed in PRDM5 (homozygous variant: c.1036C > T; p.(Arg346Ter)) in patient P15 presenting with Brittle Cornea syndrome and in WRN (c.2313 T > A; p.(Cys771Ter)) associated with another already described nonsense variant in patient P8 presenting with classical Werner syndrome.
Other previously described pathogenic variants were identified in patients affected with syndromes as diverse as SHORT, Buschke-Ollendorf syndrome, Branchio-Oculo-Facial Syndrome (BOFS), and Oculodentodigital Dysplasia.
Among the 16 VUS observed, 3 were re-classified as benign by studying their segregation in first degree relatives’ DNAs.
Interestingly, three variants were detected in LMNA: one known pathogenic variant and two novel VUS, whose pathogenicity could not be proven for the moment, due to the lack of segregation studies or functional in vitro analyses. One of the LMNA VUS (NM_170707.4: c.1016C > T; p.(Ala339Val)) was found in a male proband (patient P18) who presented with an adult-onset progeroid phenotype suggestive of Atypical Werner Syndrome (AWS), described in patients presenting with some clinical signs of WS and carrying heterozygous missense LMNA variants [20, 22, 6769]. Furthermore, the WRN gene, whose coding sequence NM_000553.5 was entirely covered by the genomic sequencing, did not show any pathogenic or likely pathogenic variants. We consider the LMNA p.(Ala339Val) variant as likely responsible for the patient’s phenotype, although it doesn’t fulfill ACMG criteria for (likely) pathogenicity [35]. Indeed, Ala339 is a very conserved residue and the LMNA gene has only a few well-known polymorphic variants (SNPs), making every undescribed variant very suspect of being pathogenic, especially if compatible with previously described laminopathies. Moreover, it is located nearby other previously reported AWS mutations [22, 68, 69].
The other LMNA VUS we observed (NM_005572.3: c.1715G > A; p.(Arg572His)) affected only Lamin C-encoding transcripts, being otherwise located in the 3′ UTR region of Lamin A-encoding transcripts, in patient P17, who was addressed for a suspicion of cEDS. It would be very interesting to perform the variant segregation study in this patient’s first relatives since, to the best of our knowledge, no variant affecting uniquely Lamin C isoforms has ever been described in human. Indeed, most laminopathies, including atypical progeria syndromes [24, 26, 7072], have been associated to missense mutations affecting both Lamin A and C isoforms, and the mutation of only Lamin C isoforms may be compatible with a relatively mild clinical phenotype.
The familial segregation of these VUS and eventual functional analyses will be important to determine whether or not they are major pathogenic factors at the origin of the clinical phenotypes of the patients or only possible phenotype modifiers.
Patient P11 carrying the heterozygous c.1003C > T; p.(Arg335Trp) pathogenic variant in LMNA (NM_170707.4), already described in [4854], presented since a young age with a severe progeroid phenotype of acro-osteolysis and cutaneous premature aging associated with heart rhythm disturbances; by giving us the authorization of publishing her pictures, the patient allows the medical community to become more familiar with the spectrum of clinical phenotypes (ranging from isolated dilated cardiomyopathy to premature aging phenotypes) linked to this particular LMNA mutation, as already reported for other LMNA mutations.
Finally, the identification of pathogenic variants allowed us to offer genetic counselling and propose prenatal testing: four prenatal tests were performed in the three families with mutations in GORAB, RECQL4 and TFAP2A.

Conclusion

Globally, the NGS molecular exploration of 66 patients using a panel of 82 genes associated with premature aging syndromes allowed us to confirm or establish a final diagnosis in 26% (about 1/4th) of the cases, while segregation analysis of first-degree relatives would help to reclassify the VUS identified in about 15% of the cohort.
The identification of a molecular diagnosis for these rare but often severe disorders, allowed to provide genetic counseling to the families and to propose prenatal diagnosis, contributing to personalized genomic, medical healthcare.
In order to further improve our panel’s diagnostic yield, clinicians will need to be sensitized to the utility of completing the segregation analyses in the families with VUS and sending at once the trios DNAs (affected patient and parents) whenever possible; also, research projects including functional in vitro analysis on human biological samples may be designed in order to include patients with VUS if the segregation analysis is compatible with a likely pathogenic effect, in order to provide arguments for it and refine the molecular diagnosis. On the other hand, for patients for whom only ACMG classes 1 or 2 were observed (which represent overall, about 60% of the patients of our cohort) further studies in a research context may be proposed.
Alternatively, the French government has financed the first two National high throughput whole genome sequencing platforms for trios; indeed, analysis of both parents and child are well known to improve the diagnostic performance [73, 74]. These WGS platforms have been established in the context of the “Medicine France Genomics 2025” Plan (https://​solidarites-sante.​gouv.​fr/​systeme-de-sante-et-medico-social/​recherche-et-innovation/​france-genomique), aiming to sustain and improve molecular genetic diagnosis of patients affected with rare disorders and therapeutic follow up of patients affected with cancer in the context of genomic, personalized medicine, allowing to further integrate scientific advances into healthcare and to facilitate the access to innovation to all patients. These patients and their relatives may also be candidate for this second level screening in the next years.

Acknowledgements

We thank all the patients and their families for their precious collaboration to this work.
We also thank the AP-HM Data Protection Representative for helping us in guaranteeing patients’ data protection, complying with the RGPD regulation.
This study fully complies with the European RGPD directive (n°2016/679): the use of genetic and clinical data issued from molecular genetics diagnostics procedures were validated from the Data Protection Officer of APHM (Assistance Publique des Hôpitaux de Marseille), in agreement with the European and French regulation.
All the patients or the parents/legal guardians for minors, provided written informed consent for genetic analysis following the French regulation (L1131-1-1 Code de la Santé Publique). Specific consents for publication were additionally obtained from the patients or the parents / legal guardians of the minor patients whose clinical presentation is further developed in the article, namely including their pictures.

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
1.
Zurück zum Zitat Cau P, Navarro C, Harhouri K, Roll P, Sigaudy S, Kaspi E, et al. Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective. Semin Cell Dev Biol. 2014 May;29:125–47.PubMedCrossRef Cau P, Navarro C, Harhouri K, Roll P, Sigaudy S, Kaspi E, et al. Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective. Semin Cell Dev Biol. 2014 May;29:125–47.PubMedCrossRef
2.
Zurück zum Zitat Larsen NB, Hickson ID. RecQ Helicases: Conserved Guardians of Genomic Integrity. Adv Exp Med Biol. 2013;767:161–84. Larsen NB, Hickson ID. RecQ Helicases: Conserved Guardians of Genomic Integrity. Adv Exp Med Biol. 2013;767:161–84.
3.
Zurück zum Zitat Fu W, Ligabue A, Rogers KJ, Akey JM, Monnat RJ. Human RECQ helicase pathogenic variants, population variation and “missing” diseases: HUMAN MUTATION. Hum Mutat. 2017;38(2):193–203.PubMedCrossRef Fu W, Ligabue A, Rogers KJ, Akey JM, Monnat RJ. Human RECQ helicase pathogenic variants, population variation and “missing” diseases: HUMAN MUTATION. Hum Mutat. 2017;38(2):193–203.PubMedCrossRef
4.
Zurück zum Zitat Croteau DL, Popuri V, Opresko PL, Bohr VA. Human RecQ helicases in DNA repair, recombination, and replication. Annu Rev Biochem. 2014;83(1):519–52.PubMedPubMedCentralCrossRef Croteau DL, Popuri V, Opresko PL, Bohr VA. Human RecQ helicases in DNA repair, recombination, and replication. Annu Rev Biochem. 2014;83(1):519–52.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Yu C-E, Oshima J, Fu Y-H, Wijsman EM, Hisama F, Alisch R, et al. Positional cloning of the Werner’s syndrome gene. Science. 1996;272(5259):258–62.PubMedCrossRef Yu C-E, Oshima J, Fu Y-H, Wijsman EM, Hisama F, Alisch R, et al. Positional cloning of the Werner’s syndrome gene. Science. 1996;272(5259):258–62.PubMedCrossRef
6.
Zurück zum Zitat Ellis NA, Groden J, Ye T-Z, Straughen J, Lennon DJ, Ciocci S, et al. The Bloom’s syndrome gene product is homologous to RecQ helicases. Cell. 1995;83(4):655–66.PubMedCrossRef Ellis NA, Groden J, Ye T-Z, Straughen J, Lennon DJ, Ciocci S, et al. The Bloom’s syndrome gene product is homologous to RecQ helicases. Cell. 1995;83(4):655–66.PubMedCrossRef
7.
Zurück zum Zitat Kitao S, Shimamoto A, Goto M, Miller RW, Smithson WA, Lindor NM, et al. Mutations in RECQL4 cause a subset of cases of Rothmund-Thomson syndrome. Nat Genet. 1999;22(1):82–4.PubMedCrossRef Kitao S, Shimamoto A, Goto M, Miller RW, Smithson WA, Lindor NM, et al. Mutations in RECQL4 cause a subset of cases of Rothmund-Thomson syndrome. Nat Genet. 1999;22(1):82–4.PubMedCrossRef
8.
Zurück zum Zitat Wilson BT, Stark Z, Sutton RE, Danda S, Ekbote AV, Elsayed SM, et al. The Cockayne syndrome natural history (CoSyNH) study: clinical findings in 102 individuals and recommendations for care. Genet Med. 2016;18(5):483.PubMedCrossRef Wilson BT, Stark Z, Sutton RE, Danda S, Ekbote AV, Elsayed SM, et al. The Cockayne syndrome natural history (CoSyNH) study: clinical findings in 102 individuals and recommendations for care. Genet Med. 2016;18(5):483.PubMedCrossRef
9.
Zurück zum Zitat Calmels N, Botta E, Jia N, Fawcett H, Nardo T, Nakazawa Y, et al. Functional and clinical relevance of novel mutations in a large cohort of patients with Cockayne syndrome. J Med Genet. 2018;55(5):329–43.PubMedCrossRef Calmels N, Botta E, Jia N, Fawcett H, Nardo T, Nakazawa Y, et al. Functional and clinical relevance of novel mutations in a large cohort of patients with Cockayne syndrome. J Med Genet. 2018;55(5):329–43.PubMedCrossRef
10.
Zurück zum Zitat De Sandre-Giovannoli A, Bernard R, Cau P, Navarro C, Amiel J, Boccaccio I, et al. Lamin a truncation in Hutchinson-Gilford progeria. Science. 2003;300(5628):2055.PubMedCrossRef De Sandre-Giovannoli A, Bernard R, Cau P, Navarro C, Amiel J, Boccaccio I, et al. Lamin a truncation in Hutchinson-Gilford progeria. Science. 2003;300(5628):2055.PubMedCrossRef
11.
Zurück zum Zitat Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, et al. Recurrent de novo point mutations in Lamin a cause Hutchinson-Gilford progeria syndrome. Nature. 2003;423(6937):293–8.PubMedCrossRef Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, et al. Recurrent de novo point mutations in Lamin a cause Hutchinson-Gilford progeria syndrome. Nature. 2003;423(6937):293–8.PubMedCrossRef
12.
Zurück zum Zitat Goldman RD, Shumaker DK, Erdos MR, Eriksson M, Goldman AE, Gordon LB, et al. Accumulation of mutant Lamin a causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. 2004;101(24):8963–8.PubMedPubMedCentralCrossRef Goldman RD, Shumaker DK, Erdos MR, Eriksson M, Goldman AE, Gordon LB, et al. Accumulation of mutant Lamin a causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. 2004;101(24):8963–8.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Barthélémy F, Navarro C, Fayek R, Da Silva N, Roll P, Sigaudy S, et al. Truncated prelamin a expression in HGPS-like patients: a transcriptional study. Eur J Hum Genet. 2015 Aug;23(8):1051–61.PubMedPubMedCentralCrossRef Barthélémy F, Navarro C, Fayek R, Da Silva N, Roll P, Sigaudy S, et al. Truncated prelamin a expression in HGPS-like patients: a transcriptional study. Eur J Hum Genet. 2015 Aug;23(8):1051–61.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Agarwal AK, Fryns J-P, Auchus RJ, Garg A. Zinc metalloproteinase, ZMPSTE24, is mutated in mandibuloacral dysplasia. Hum Mol Genet. 2003;12(16):1995–2001.PubMedCrossRef Agarwal AK, Fryns J-P, Auchus RJ, Garg A. Zinc metalloproteinase, ZMPSTE24, is mutated in mandibuloacral dysplasia. Hum Mol Genet. 2003;12(16):1995–2001.PubMedCrossRef
15.
Zurück zum Zitat Navarro CL, De Sandre-Giovannoli A, Bernard R, Boccaccio I, Boyer A, Genevieve D, et al. Lamin a and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy. Hum Mol Genet. 2004;13(20):2493–503.PubMedCrossRef Navarro CL, De Sandre-Giovannoli A, Bernard R, Boccaccio I, Boyer A, Genevieve D, et al. Lamin a and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy. Hum Mol Genet. 2004;13(20):2493–503.PubMedCrossRef
16.
Zurück zum Zitat Navarro CL, Cadiñanos J, De Sandre-Giovannoli A, Bernard R, Courrier S, Boccaccio I, et al. Loss of ZMPSTE24 (FACE-1) causes autosomal recessive restrictive dermopathy and accumulation of Lamin a precursors. Hum Mol Genet. 2005 Jun 1;14(11):1503–13.PubMedCrossRef Navarro CL, Cadiñanos J, De Sandre-Giovannoli A, Bernard R, Courrier S, Boccaccio I, et al. Loss of ZMPSTE24 (FACE-1) causes autosomal recessive restrictive dermopathy and accumulation of Lamin a precursors. Hum Mol Genet. 2005 Jun 1;14(11):1503–13.PubMedCrossRef
17.
Zurück zum Zitat Navarro CL, Poitelon Y, Lévy N. Lamines A et syndromes progéroïdes-Une farnésylation persistante aux conséquences dramatiques. Méd Sci. 2008;24(10):833–40. Navarro CL, Poitelon Y, Lévy N. Lamines A et syndromes progéroïdes-Une farnésylation persistante aux conséquences dramatiques. Méd Sci. 2008;24(10):833–40.
19.
Zurück zum Zitat Navarro CL, Esteves-Vieira V, Courrier S, Boyer A, Duong Nguyen T, Huong LTT, et al. New ZMPSTE24 (FACE1) mutations in patients affected with restrictive dermopathy or related progeroid syndromes and mutation update. Eur J Hum Genet. 2014;22(8):1002–11.PubMedCrossRef Navarro CL, Esteves-Vieira V, Courrier S, Boyer A, Duong Nguyen T, Huong LTT, et al. New ZMPSTE24 (FACE1) mutations in patients affected with restrictive dermopathy or related progeroid syndromes and mutation update. Eur J Hum Genet. 2014;22(8):1002–11.PubMedCrossRef
20.
Zurück zum Zitat Chen L, Lee L, Kudlow BA, Dos Santos HG, Sletvold O, Shafeghati Y, et al. LMNA mutations in atypical Werner’s syndrome. Lancet. 2003;362(9382):440–5.PubMedCrossRef Chen L, Lee L, Kudlow BA, Dos Santos HG, Sletvold O, Shafeghati Y, et al. LMNA mutations in atypical Werner’s syndrome. Lancet. 2003;362(9382):440–5.PubMedCrossRef
21.
Zurück zum Zitat Madej-Pilarczyk A, Kmieć T, Fidziańska A, Rekawek J, Niebrój-Dobosz I, Turska-Kmieć A, et al. Progeria caused by a rare LMNA mutation p.S143F associated with mild myopathy and atrial fibrillation. Eur J Paediatr Neurol Off J Eur Paediatr Neurol Soc. 2008 Sep;12(5):427–30.CrossRef Madej-Pilarczyk A, Kmieć T, Fidziańska A, Rekawek J, Niebrój-Dobosz I, Turska-Kmieć A, et al. Progeria caused by a rare LMNA mutation p.S143F associated with mild myopathy and atrial fibrillation. Eur J Paediatr Neurol Off J Eur Paediatr Neurol Soc. 2008 Sep;12(5):427–30.CrossRef
23.
Zurück zum Zitat Madej-Pilarczyk A, Rosińska-Borkowska D, Rekawek J, Marchel M, Szaluś E, Jabłońska S, et al. Progeroid syndrome with scleroderma-like skin changes associated with homozygous R435C LMNA mutation. Am J Med Genet A. 2009;149A(11):2387–92.PubMedCrossRef Madej-Pilarczyk A, Rosińska-Borkowska D, Rekawek J, Marchel M, Szaluś E, Jabłońska S, et al. Progeroid syndrome with scleroderma-like skin changes associated with homozygous R435C LMNA mutation. Am J Med Genet A. 2009;149A(11):2387–92.PubMedCrossRef
24.
Zurück zum Zitat Garg A, Subramanyam L, Agarwal AK, Simha V, Levine B, D’Apice MR, et al. Atypical Progeroid syndrome due to heterozygous missense LMNA mutations. J Clin Endocrinol Metab. 2009;94(12):4971–83.PubMedPubMedCentralCrossRef Garg A, Subramanyam L, Agarwal AK, Simha V, Levine B, D’Apice MR, et al. Atypical Progeroid syndrome due to heterozygous missense LMNA mutations. J Clin Endocrinol Metab. 2009;94(12):4971–83.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Liang L, Zhang H, Gu X. Homozygous LMNA mutation R527C in atypical Hutchinson-Gilford progeria syndrome: evidence for autosomal recessive inheritance. Acta Paediatr Oslo Nor 1992. 2009;98(8):1365–8. Liang L, Zhang H, Gu X. Homozygous LMNA mutation R527C in atypical Hutchinson-Gilford progeria syndrome: evidence for autosomal recessive inheritance. Acta Paediatr Oslo Nor 1992. 2009;98(8):1365–8.
26.
Zurück zum Zitat Doubaj Y, De Sandre-Giovannoli A, Vera E-V, Navarro CL, Elalaoui SC, Tajir M, et al. An inherited LMNA gene mutation in atypical progeria syndrome. Am J Med Genet A. 2012;158A(11):2881–7.PubMedCrossRef Doubaj Y, De Sandre-Giovannoli A, Vera E-V, Navarro CL, Elalaoui SC, Tajir M, et al. An inherited LMNA gene mutation in atypical progeria syndrome. Am J Med Genet A. 2012;158A(11):2881–7.PubMedCrossRef
27.
Zurück zum Zitat Soria-Valles C, Carrero D, Gabau E, Velasco G, Quesada V, Bárcena C, et al. Novel LMNA mutations cause an aggressive atypical neonatal progeria without progerin accumulation. J Med Genet. 2016;53(11):776–85.PubMedCrossRef Soria-Valles C, Carrero D, Gabau E, Velasco G, Quesada V, Bárcena C, et al. Novel LMNA mutations cause an aggressive atypical neonatal progeria without progerin accumulation. J Med Genet. 2016;53(11):776–85.PubMedCrossRef
28.
Zurück zum Zitat Malfait F, Francomano C, Byers P, Belmont J, Berglund B, Black J, et al. The 2017 international classification of the Ehlers–Danlos syndromes. Am J Med Genet C Semin Med Genet. 2017;175(1):8–26. Malfait F, Francomano C, Byers P, Belmont J, Berglund B, Black J, et al. The 2017 international classification of the Ehlers–Danlos syndromes. Am J Med Genet C Semin Med Genet. 2017;175(1):8–26.
30.
Zurück zum Zitat Desvignes J-P, Bartoli M, Delague V, Krahn M, Miltgen M, Béroud C, et al. VarAFT: a variant annotation and filtration system for human next generation sequencing data. Nucleic Acids Res. 2018;46:W545–53.PubMedPubMedCentralCrossRef Desvignes J-P, Bartoli M, Delague V, Krahn M, Miltgen M, Béroud C, et al. VarAFT: a variant annotation and filtration system for human next generation sequencing data. Nucleic Acids Res. 2018;46:W545–53.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Desmet F-O, Hamroun D, Lalande M, Collod-Béroud G, Claustres M, Béroud C. Human splicing finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 2009;37(9):e67.PubMedPubMedCentralCrossRef Desmet F-O, Hamroun D, Lalande M, Collod-Béroud G, Claustres M, Béroud C. Human splicing finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 2009;37(9):e67.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Schwarz JM, Rödelsperger C, Schuelke M, Seelow D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods. 2010;7(8):575.PubMedCrossRef Schwarz JM, Rödelsperger C, Schuelke M, Seelow D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods. 2010;7(8):575.PubMedCrossRef
34.
Zurück zum Zitat Salgado D, Desvignes J-P, Rai G, Blanchard A, Miltgen M, Pinard A, et al. UMD-predictor: a high-throughput sequencing compliant system for pathogenicity prediction of any human cDNA substitution. Hum Mutat. 2016;37(5):439–46.PubMedPubMedCentralCrossRef Salgado D, Desvignes J-P, Rai G, Blanchard A, Miltgen M, Pinard A, et al. UMD-predictor: a high-throughput sequencing compliant system for pathogenicity prediction of any human cDNA substitution. Hum Mutat. 2016;37(5):439–46.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med Off J Am Coll Med Genet. 2015;17(5):405–24. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med Off J Am Coll Med Genet. 2015;17(5):405–24.
36.
Zurück zum Zitat Pujol LA, Erickson RP, Heidenreich RA, Cunniff C. Variable presentation of Rothmund-Thomson syndrome. Am J Med Genet. 2000;95(3):204–7.PubMedCrossRef Pujol LA, Erickson RP, Heidenreich RA, Cunniff C. Variable presentation of Rothmund-Thomson syndrome. Am J Med Genet. 2000;95(3):204–7.PubMedCrossRef
37.
Zurück zum Zitat Wang LL, Gannavarapu A, Kozinetz CA, Levy ML, Lewis RA, Chintagumpala MM, et al. Association between osteosarcoma and deleterious mutations in the RECQL4 gene in Rothmund-Thomson syndrome. J Natl Cancer Inst. 2003 May 7;95(9):669–74.PubMedCrossRef Wang LL, Gannavarapu A, Kozinetz CA, Levy ML, Lewis RA, Chintagumpala MM, et al. Association between osteosarcoma and deleterious mutations in the RECQL4 gene in Rothmund-Thomson syndrome. J Natl Cancer Inst. 2003 May 7;95(9):669–74.PubMedCrossRef
38.
Zurück zum Zitat Siitonen HA, Sotkasiira J, Biervliet M, Benmansour A, Capri Y, Cormier-Daire V, et al. The mutation spectrum in RECQL4 diseases. Eur J Hum Genet. 2009;17(2):151–8.PubMedCrossRef Siitonen HA, Sotkasiira J, Biervliet M, Benmansour A, Capri Y, Cormier-Daire V, et al. The mutation spectrum in RECQL4 diseases. Eur J Hum Genet. 2009;17(2):151–8.PubMedCrossRef
39.
Zurück zum Zitat Wiest T, Herrmann O, Stögbauer F, Grasshoff U, Enders H, Koch MJ, et al. Clinical and genetic variability of oculodentodigital dysplasia. Clin Genet. 2006;70(1):71–2.PubMedCrossRef Wiest T, Herrmann O, Stögbauer F, Grasshoff U, Enders H, Koch MJ, et al. Clinical and genetic variability of oculodentodigital dysplasia. Clin Genet. 2006;70(1):71–2.PubMedCrossRef
40.
Zurück zum Zitat Hellemans J, Debeer P, Wright M, Janecke A, Kjaer KW, Verdonk PC, et al. Germline LEMD3 mutations are rare in sporadic patients with isolated melorheostosis. Hum Mutat. 2006;27(3):290.PubMedCrossRef Hellemans J, Debeer P, Wright M, Janecke A, Kjaer KW, Verdonk PC, et al. Germline LEMD3 mutations are rare in sporadic patients with isolated melorheostosis. Hum Mutat. 2006;27(3):290.PubMedCrossRef
41.
Zurück zum Zitat Reiber J, Sznajer Y, Posteguillo EG, Müller D, Lyonnet S, Baumann C, et al. Additional clinical and molecular analyses of TFAP2A in patients with the branchio-oculo-facial syndrome. Am J Med Genet A. 2010;152A(4):994–9.PubMedCrossRef Reiber J, Sznajer Y, Posteguillo EG, Müller D, Lyonnet S, Baumann C, et al. Additional clinical and molecular analyses of TFAP2A in patients with the branchio-oculo-facial syndrome. Am J Med Genet A. 2010;152A(4):994–9.PubMedCrossRef
42.
Zurück zum Zitat Dyment DA, Smith AC, Alcantara D, Schwartzentruber JA, Basel-Vanagaite L, Curry CJ, et al. Mutations in PIK3R1 cause SHORT syndrome. Am J Hum Genet. 2013;93(1):158–66.PubMedPubMedCentralCrossRef Dyment DA, Smith AC, Alcantara D, Schwartzentruber JA, Basel-Vanagaite L, Curry CJ, et al. Mutations in PIK3R1 cause SHORT syndrome. Am J Hum Genet. 2013;93(1):158–66.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Reversade B, Escande-Beillard N, Dimopoulou A, Fischer B, Chng SC, Li Y, et al. Mutations in PYCR1 cause cutis laxa with progeroid features. Nat Genet. 2009;41(9):1016–21.PubMedCrossRef Reversade B, Escande-Beillard N, Dimopoulou A, Fischer B, Chng SC, Li Y, et al. Mutations in PYCR1 cause cutis laxa with progeroid features. Nat Genet. 2009;41(9):1016–21.PubMedCrossRef
44.
Zurück zum Zitat Kretz R, Bozorgmehr B, Kariminejad MH, Rohrbach M, Hausser I, Baumer A, et al. Defect in proline synthesis: pyrroline-5-carboxylate reductase 1 deficiency leads to a complex clinical phenotype with collagen and elastin abnormalities. J Inherit Metab Dis. 2011;34(3):731–9.PubMedCrossRef Kretz R, Bozorgmehr B, Kariminejad MH, Rohrbach M, Hausser I, Baumer A, et al. Defect in proline synthesis: pyrroline-5-carboxylate reductase 1 deficiency leads to a complex clinical phenotype with collagen and elastin abnormalities. J Inherit Metab Dis. 2011;34(3):731–9.PubMedCrossRef
45.
Zurück zum Zitat Fischer-Zirnsak B, Escande-Beillard N, Ganesh J, Tan YX, Al Bughaili M, Lin AE, et al. Recurrent De novo mutations affecting residue Arg138 of Pyrroline-5-carboxylate synthase cause a Progeroid form of autosomal-dominant cutis Laxa. Am J Hum Genet. 2015;97(3):483–92.PubMedPubMedCentralCrossRef Fischer-Zirnsak B, Escande-Beillard N, Ganesh J, Tan YX, Al Bughaili M, Lin AE, et al. Recurrent De novo mutations affecting residue Arg138 of Pyrroline-5-carboxylate synthase cause a Progeroid form of autosomal-dominant cutis Laxa. Am J Hum Genet. 2015;97(3):483–92.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Wenstrup RJ, Florer JB, Willing MC, Giunta C, Steinmann B, Young F, et al. COL5A1 haploinsufficiency is a common molecular mechanism underlying the classical form of EDS. Am J Hum Genet. 2000;66(6):1766–76.PubMedPubMedCentralCrossRef Wenstrup RJ, Florer JB, Willing MC, Giunta C, Steinmann B, Young F, et al. COL5A1 haploinsufficiency is a common molecular mechanism underlying the classical form of EDS. Am J Hum Genet. 2000;66(6):1766–76.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Al-Hussain H, Zeisberger SM, Huber PR, Giunta C, Steinmann B. Brittle cornea syndrome and its delineation from the kyphoscoliotic type of Ehlers–Danlos syndrome (EDS VI): report on 23 patients and review of the literature. Am J Med Genet A. 2004;124(1):28–34.CrossRef Al-Hussain H, Zeisberger SM, Huber PR, Giunta C, Steinmann B. Brittle cornea syndrome and its delineation from the kyphoscoliotic type of Ehlers–Danlos syndrome (EDS VI): report on 23 patients and review of the literature. Am J Med Genet A. 2004;124(1):28–34.CrossRef
48.
Zurück zum Zitat Lakdawala NK, Funke BH, Baxter S, Cirino AL, Roberts AE, Judge DP, et al. Genetic testing for dilated cardiomyopathy in clinical practice. J Card Fail. 2012;18(4):296–303.PubMedPubMedCentralCrossRef Lakdawala NK, Funke BH, Baxter S, Cirino AL, Roberts AE, Judge DP, et al. Genetic testing for dilated cardiomyopathy in clinical practice. J Card Fail. 2012;18(4):296–303.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Bollati M, Barbiroli A, Favalli V, Arbustini E, Charron P, Bolognesi M. Structures of the Lamin a/C R335W and E347K mutants: implications for dilated cardiolaminopathies. Biochem Biophys Res Commun. 2012;418(2):217–21.PubMedCrossRef Bollati M, Barbiroli A, Favalli V, Arbustini E, Charron P, Bolognesi M. Structures of the Lamin a/C R335W and E347K mutants: implications for dilated cardiolaminopathies. Biochem Biophys Res Commun. 2012;418(2):217–21.PubMedCrossRef
50.
Zurück zum Zitat Stallmeyer B, Koopmann M, Schulze-Bahr E. Identification of novel mutations in LMNA associated with familial forms of dilated cardiomyopathy. Genet Test Mol Biomarkers. 2012;16(6):543–9.PubMedCrossRef Stallmeyer B, Koopmann M, Schulze-Bahr E. Identification of novel mutations in LMNA associated with familial forms of dilated cardiomyopathy. Genet Test Mol Biomarkers. 2012;16(6):543–9.PubMedCrossRef
51.
Zurück zum Zitat Carboni N, Politano L, Floris M, Mateddu A, Solla E, Olla S, et al. Overlapping syndromes in laminopathies: a meta-analysis of the reported literature. Acta Myol Myopathies Cardiomyopathies Off J Mediterr Soc Myol. 2013;32(1):7–17. Carboni N, Politano L, Floris M, Mateddu A, Solla E, Olla S, et al. Overlapping syndromes in laminopathies: a meta-analysis of the reported literature. Acta Myol Myopathies Cardiomyopathies Off J Mediterr Soc Myol. 2013;32(1):7–17.
52.
Zurück zum Zitat Pugh TJ, Kelly MA, Gowrisankar S, Hynes E, Seidman MA, Baxter SM, et al. The landscape of genetic variation in dilated cardiomyopathy as surveyed by clinical DNA sequencing. Genet Med. 2014;16(8):601.PubMedCrossRef Pugh TJ, Kelly MA, Gowrisankar S, Hynes E, Seidman MA, Baxter SM, et al. The landscape of genetic variation in dilated cardiomyopathy as surveyed by clinical DNA sequencing. Genet Med. 2014;16(8):601.PubMedCrossRef
53.
Zurück zum Zitat Zaragoza MV, Hakim SA, Hoang V, Elliott AM. Heart-hand syndrome IV: a second family with LMNA -related cardiomyopathy and brachydactyly: letter to the editor. Clin Genet. 2017;91(3):499–500. Zaragoza MV, Hakim SA, Hoang V, Elliott AM. Heart-hand syndrome IV: a second family with LMNA -related cardiomyopathy and brachydactyly: letter to the editor. Clin Genet. 2017;91(3):499–500.
54.
Zurück zum Zitat Lambert J-C, Baudart P, De Sandre-Giovannoli A, Molin A, Marcelli C. Lamin a/C gene (LMNA) mutation associated with laminopathy: a rare cause of idiopathic acro-osteolysis. Joint Bone Spine. 2019;86(4):525–7.PubMedCrossRef Lambert J-C, Baudart P, De Sandre-Giovannoli A, Molin A, Marcelli C. Lamin a/C gene (LMNA) mutation associated with laminopathy: a rare cause of idiopathic acro-osteolysis. Joint Bone Spine. 2019;86(4):525–7.PubMedCrossRef
55.
Zurück zum Zitat Giunta C, Steinmann B. Compound heterozygosity for a disease-causing G1489E [corrected] and disease-modifying G530S substitution in COL5A1 of a patient with the classical type of Ehlers-Danlos syndrome: an explanation of intrafamilial variability? Am J Med Genet. 2000;90(1):72–9.PubMedCrossRef Giunta C, Steinmann B. Compound heterozygosity for a disease-causing G1489E [corrected] and disease-modifying G530S substitution in COL5A1 of a patient with the classical type of Ehlers-Danlos syndrome: an explanation of intrafamilial variability? Am J Med Genet. 2000;90(1):72–9.PubMedCrossRef
56.
Zurück zum Zitat Malfait F, De Paepe A. Molecular genetics in classic Ehlers-Danlos syndrome. Am J Med Genet C: Semin Med Genet. 2005;139C(1):17–23.CrossRef Malfait F, De Paepe A. Molecular genetics in classic Ehlers-Danlos syndrome. Am J Med Genet C: Semin Med Genet. 2005;139C(1):17–23.CrossRef
57.
Zurück zum Zitat Lo Cicero A, Saidani M, Allouche J, Egesipe AL, Hoch L, Bruge C, et al. Pathological modelling of pigmentation disorders associated with Hutchinson-Gilford progeria syndrome (HGPS) revealed an impaired melanogenesis pathway in iPS-derived melanocytes. Sci Rep. 2018;8(1):9112.PubMedPubMedCentralCrossRef Lo Cicero A, Saidani M, Allouche J, Egesipe AL, Hoch L, Bruge C, et al. Pathological modelling of pigmentation disorders associated with Hutchinson-Gilford progeria syndrome (HGPS) revealed an impaired melanogenesis pathway in iPS-derived melanocytes. Sci Rep. 2018;8(1):9112.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Harhouri K, Frankel D, Bartoli C, Roll P, De Sandre-Giovannoli A, Lévy N. An overview of treatment strategies for Hutchinson-Gilford progeria syndrome. Nucl Austin Tex. 2018;9(1):246–57. Harhouri K, Frankel D, Bartoli C, Roll P, De Sandre-Giovannoli A, Lévy N. An overview of treatment strategies for Hutchinson-Gilford progeria syndrome. Nucl Austin Tex. 2018;9(1):246–57.
59.
Zurück zum Zitat Harhouri K, Navarro C, Depetris D, Mattei M-G, Nissan X, Cau P, et al. MG132-induced progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol Med. 2017;9(9):1294–313.PubMedPubMedCentralCrossRef Harhouri K, Navarro C, Depetris D, Mattei M-G, Nissan X, Cau P, et al. MG132-induced progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol Med. 2017;9(9):1294–313.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Elouej S, Beleza-Meireles A, Caswell R, Colclough K, Ellard S, Desvignes JP, et al. Exome sequencing reveals a de novo POLD1 mutation causing phenotypic variability in mandibular hypoplasia, deafness, progeroid features, and lipodystrophy syndrome (MDPL). Metabolism. 2017;71:213–25.PubMedCrossRef Elouej S, Beleza-Meireles A, Caswell R, Colclough K, Ellard S, Desvignes JP, et al. Exome sequencing reveals a de novo POLD1 mutation causing phenotypic variability in mandibular hypoplasia, deafness, progeroid features, and lipodystrophy syndrome (MDPL). Metabolism. 2017;71:213–25.PubMedCrossRef
61.
Zurück zum Zitat Harhouri K, Navarro C, Baquerre C, Da Silva N, Bartoli C, Casey F, et al. Antisense-based Progerin downregulation in HGPS-like patients’ cells. Cells. 2016;5(3):31.PubMedCentralCrossRef Harhouri K, Navarro C, Baquerre C, Da Silva N, Bartoli C, Casey F, et al. Antisense-based Progerin downregulation in HGPS-like patients’ cells. Cells. 2016;5(3):31.PubMedCentralCrossRef
62.
Zurück zum Zitat Ambrosi P, Kreitmann B, Lepidi H, Habib G, Levy N, Philip N, et al. A novel overlapping phenotype characterized by lipodystrophy, mandibular dysplasia, and dilated cardiomyopathy associated with a new mutation in the LMNA gene. Int J Cardiol. 2016;209:317–8.PubMedCrossRef Ambrosi P, Kreitmann B, Lepidi H, Habib G, Levy N, Philip N, et al. A novel overlapping phenotype characterized by lipodystrophy, mandibular dysplasia, and dilated cardiomyopathy associated with a new mutation in the LMNA gene. Int J Cardiol. 2016;209:317–8.PubMedCrossRef
63.
64.
Zurück zum Zitat Hennies HC, Kornak U, Zhang H, Egerer J, Zhang X, Seifert W, et al. Gerodermia osteodysplastica is caused by mutations in SCYL1BP1, a Rab-6 interacting golgin. Nat Genet. 2008;40(12):1410–2.PubMedPubMedCentralCrossRef Hennies HC, Kornak U, Zhang H, Egerer J, Zhang X, Seifert W, et al. Gerodermia osteodysplastica is caused by mutations in SCYL1BP1, a Rab-6 interacting golgin. Nat Genet. 2008;40(12):1410–2.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Al-Dosari M, Alkuraya FS. A novel missense mutation in SCYL1BP1 produces geroderma osteodysplastica phenotype indistinguishable from that caused by nullimorphic mutations. Am J Med Genet A. 2009;149A(10):2093–8.PubMedCrossRef Al-Dosari M, Alkuraya FS. A novel missense mutation in SCYL1BP1 produces geroderma osteodysplastica phenotype indistinguishable from that caused by nullimorphic mutations. Am J Med Genet A. 2009;149A(10):2093–8.PubMedCrossRef
66.
Zurück zum Zitat Ghai SJ, Shago M, Shroff M, Yoon G. Cockayne syndrome caused by paternally inherited 5 Mb deletion of 10q11. 2 and a frameshift mutation of ERCC6. Eur J Med Genet. 2011;54(3):272–6.PubMedCrossRef Ghai SJ, Shago M, Shroff M, Yoon G. Cockayne syndrome caused by paternally inherited 5 Mb deletion of 10q11. 2 and a frameshift mutation of ERCC6. Eur J Med Genet. 2011;54(3):272–6.PubMedCrossRef
67.
Zurück zum Zitat Doh YJ, Kim HK, Jung ED, Choi SH, Kim JG, Kim BW, et al. Novel LMNA gene mutation in a patient with atypical Werner’s syndrome. Korean J Intern Med. 2009;24(1):68–72.PubMedPubMedCentralCrossRef Doh YJ, Kim HK, Jung ED, Choi SH, Kim JG, Kim BW, et al. Novel LMNA gene mutation in a patient with atypical Werner’s syndrome. Korean J Intern Med. 2009;24(1):68–72.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Motegi S, Yokoyama Y, Uchiyama A, Ogino S, Takeuchi Y, Yamada K, et al. First Japanese case of atypical progeroid syndrome/atypical Werner syndrome with heterozygous LMNA mutation. J Dermatol. 2014;41(12):1047–52.PubMedCrossRef Motegi S, Yokoyama Y, Uchiyama A, Ogino S, Takeuchi Y, Yamada K, et al. First Japanese case of atypical progeroid syndrome/atypical Werner syndrome with heterozygous LMNA mutation. J Dermatol. 2014;41(12):1047–52.PubMedCrossRef
69.
Zurück zum Zitat Yanhua X, Suxian Z. Cerebral Haemorrhage in a Young patient with atypical Werner syndrome due to mutations in LMNA. Front Endocrinol. 2018;9:433.CrossRef Yanhua X, Suxian Z. Cerebral Haemorrhage in a Young patient with atypical Werner syndrome due to mutations in LMNA. Front Endocrinol. 2018;9:433.CrossRef
70.
Zurück zum Zitat Csoka AB, Cao H, Sammak PJ, Constantinescu D, Schatten GP, Hegele RA. Novel Lamin a/C gene (LMNA) mutations in atypical progeroid syndromes. J Med Genet. 2004;41(4):304–8.PubMedPubMedCentralCrossRef Csoka AB, Cao H, Sammak PJ, Constantinescu D, Schatten GP, Hegele RA. Novel Lamin a/C gene (LMNA) mutations in atypical progeroid syndromes. J Med Genet. 2004;41(4):304–8.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Brown CA, Lanning RW, McKinney KQ, Salvino AR, Cherniske E, Crowe CA, et al. Novel and recurrent mutations in Lamin a/C in patients with Emery-Dreifuss muscular dystrophy. Am J Med Genet. 2001;102(4):359–67.PubMedCrossRef Brown CA, Lanning RW, McKinney KQ, Salvino AR, Cherniske E, Crowe CA, et al. Novel and recurrent mutations in Lamin a/C in patients with Emery-Dreifuss muscular dystrophy. Am J Med Genet. 2001;102(4):359–67.PubMedCrossRef
72.
Zurück zum Zitat van der Kooi AJ, Bonne G, Eymard B, Duboc D, Talim B, Van der Valk M, et al. Lamin a/C mutations with lipodystrophy, cardiac abnormalities, and muscular dystrophy. Neurology. 2002;59(4):620–3.PubMedCrossRef van der Kooi AJ, Bonne G, Eymard B, Duboc D, Talim B, Van der Valk M, et al. Lamin a/C mutations with lipodystrophy, cardiac abnormalities, and muscular dystrophy. Neurology. 2002;59(4):620–3.PubMedCrossRef
73.
Zurück zum Zitat Wright CF, Fitzgerald TW, Jones WD, Clayton S, McRae JF, van Kogelenberg M, et al. Genetic diagnosis of developmental disorders in the DDD study: a scalable analysis of genome-wide research data. Lancet. 2015;385(9975):1305–14.PubMedPubMedCentralCrossRef Wright CF, Fitzgerald TW, Jones WD, Clayton S, McRae JF, van Kogelenberg M, et al. Genetic diagnosis of developmental disorders in the DDD study: a scalable analysis of genome-wide research data. Lancet. 2015;385(9975):1305–14.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Tammimies K, Marshall CR, Walker S, Kaur G, Thiruvahindrapuram B, Lionel AC, et al. Molecular diagnostic yield of chromosomal microarray analysis and whole-exome sequencing in children with autism spectrum disorder. JAMA. 2015;314(9):895–903.PubMedCrossRef Tammimies K, Marshall CR, Walker S, Kaur G, Thiruvahindrapuram B, Lionel AC, et al. Molecular diagnostic yield of chromosomal microarray analysis and whole-exome sequencing in children with autism spectrum disorder. JAMA. 2015;314(9):895–903.PubMedCrossRef
Metadaten
Titel
Outcomes of 4 years of molecular genetic diagnosis on a panel of genes involved in premature aging syndromes, including laminopathies and related disorders
verfasst von
Maude Grelet
Véronique Blanck
Sabine Sigaudy
Nicole Philip
Fabienne Giuliano
Khaoula Khachnaoui
Godelieve Morel
Sarah Grotto
Julia Sophie
Céline Poirsier
James Lespinasse
Laurent Alric
Patrick Calvas
Gihane Chalhoub
Valérie Layet
Arnaud Molin
Cindy Colson
Luisa Marsili
Patrick Edery
Nicolas Lévy
Annachiara De Sandre-Giovannoli
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2019
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-019-1189-z

Weitere Artikel der Ausgabe 1/2019

Orphanet Journal of Rare Diseases 1/2019 Zur Ausgabe