Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Med Genet ; 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38531626

ABSTRACT

BACKGROUND: Mismatch repair deficiency (dMMR) is a characteristic feature of cancers linked to Lynch syndrome. However, in most cases, it results from sporadic somatic events rather than hereditary factors. The term 'Lynch-like syndrome' (LLS) has been used to guide colorectal cancer surveillance for relatives of individuals with a dMMR tumour when somatic and germline genomic testing is uninformative. As the assessment of mismatch repair through immunohistochemistry and/or microsatellite instability is increasingly applied across various tumour types for treatment planning, dMMR is increasingly detected in tumours where suspicion of hereditary aetiology is low. Our objective was to establish current practices and develop national guidance for investigating, and managing relatives of, patients with cancers demonstrating unexplained dMMR. METHODS: This was achieved through a virtual consensus meeting involving key stakeholders from the UK, through premeeting surveys, structured discussions and in-meeting polling to formulate best practice guidance. RESULTS: We identified variability in the availability of diagnostic technologies across specialist centres. It was agreed that equitable access to baseline testing is required, acknowledging the need for a pragmatic approach to investigating dMMR cancers not traditionally associated with Lynch syndrome. Factors such as family history, age, tumour type, protein loss pattern and extent of the investigation were deemed crucial in guiding family management. The term 'unexplained dMMR' was recommended over LLS. CONCLUSION: Decisions regarding investigations and future cancer risk management in patients and relatives should be nuanced, considering factors like clinical suspicion of hereditary predisposition to allocate limited resources efficiently and avoid unnecessary investigations in low-suspicion families.

2.
Breast Cancer Res ; 25(1): 72, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37340476

ABSTRACT

INTRODUCTION: Height, body mass index (BMI), and weight gain are associated with breast cancer risk in the general population. It is unclear whether these associations also exist for carriers of pathogenic variants in the BRCA1 or BRCA2 genes. PATIENTS AND METHODS: An international pooled cohort of 8091 BRCA1/2 variant carriers was used for retrospective and prospective analyses separately for premenopausal and postmenopausal women. Cox regression was used to estimate breast cancer risk associations with height, BMI, and weight change. RESULTS: In the retrospective analysis, taller height was associated with risk of premenopausal breast cancer for BRCA2 variant carriers (HR 1.20 per 10 cm increase, 95% CI 1.04-1.38). Higher young-adult BMI was associated with lower premenopausal breast cancer risk for both BRCA1 (HR 0.75 per 5 kg/m2, 95% CI 0.66-0.84) and BRCA2 (HR 0.76, 95% CI 0.65-0.89) variant carriers in the retrospective analysis, with consistent, though not statistically significant, findings from the prospective analysis. In the prospective analysis, higher BMI and adult weight gain were associated with higher postmenopausal breast cancer risk for BRCA1 carriers (HR 1.20 per 5 kg/m2, 95% CI 1.02-1.42; and HR 1.10 per 5 kg weight gain, 95% CI 1.01-1.19, respectively). CONCLUSION: Anthropometric measures are associated with breast cancer risk for BRCA1 and BRCA2 variant carriers, with relative risk estimates that are generally consistent with those for women from the general population.


Subject(s)
Breast Neoplasms , Genes, BRCA2 , Adult , Female , Humans , Body Mass Index , BRCA1 Protein/genetics , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , BRCA2 Protein/genetics , Risk , Retrospective Studies , Weight Gain/genetics , Heterozygote , Genetic Predisposition to Disease
3.
J Med Genet ; 60(5): 417-429, 2023 05.
Article in English | MEDLINE | ID: mdl-36411032

ABSTRACT

Germline pathogenic variants (GPVs) in the cancer predisposition genes BRCA1, BRCA2, MLH1, MSH2, MSH6, BRIP1, PALB2, RAD51D and RAD51C are identified in approximately 15% of patients with ovarian cancer (OC). While there are clear guidelines around clinical management of cancer risk in patients with GPV in BRCA1, BRCA2, MLH1, MSH2 and MSH6, there are few guidelines on how to manage the more moderate OC risk in patients with GPV in BRIP1, PALB2, RAD51D and RAD51C, with clinical questions about appropriateness and timing of risk-reducing gynaecological surgery. Furthermore, while recognition of RAD51C and RAD51D as OC predisposition genes has been established for several years, an association with breast cancer (BC) has only more recently been described and clinical management of this risk has been unclear. With expansion of genetic testing of these genes to all patients with non-mucinous OC, new data on BC risk and improved estimates of OC risk, the UK Cancer Genetics Group and CanGene-CanVar project convened a 2-day meeting to reach a national consensus on clinical management of BRIP1, PALB2, RAD51D and RAD51C carriers in clinical practice. In this paper, we present a summary of the processes used to reach and agree on a consensus, as well as the key recommendations from the meeting.


Subject(s)
Breast Neoplasms , DNA-Binding Proteins , Fanconi Anemia Complementation Group N Protein , Genetic Predisposition to Disease , Ovarian Neoplasms , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Consensus , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Genetic Predisposition to Disease/genetics , Germ Cells/pathology , Germ-Line Mutation/genetics , MutS Homolog 2 Protein/genetics , Ovarian Neoplasms/genetics , United Kingdom
4.
Eur J Hum Genet ; 29(7): 1110-1120, 2021 07.
Article in English | MEDLINE | ID: mdl-33654309

ABSTRACT

The MCM2-7 helicase is a heterohexameric complex with essential roles as part of both the pre-replication and pre-initiation complexes in the early stages of DNA replication. Meier-Gorlin syndrome, a rare primordial dwarfism, is strongly associated with disruption to the pre-replication complex, including a single case described with variants in MCM5. Conversely, a biallelic pathogenic variant in MCM4 underlies immune deficiency with growth retardation, features also seen in individuals with pathogenic variants in other pre-initiation complex encoding genes such as GINS1, MCM10, and POLE. Through exome and chromium genome sequencing, supported by functional studies, we identify biallelic pathogenic variants in MCM7 and a strong candidate biallelic pathogenic variant in MCM3. We confirm variants in MCM7 are deleterious and through interfering with MCM complex formation, impact efficiency of S phase progression. The associated phenotypes are striking; one patient has typical Meier-Gorlin syndrome, whereas the second case has a multi-system disorder with neonatal progeroid appearance, lipodystrophy and adrenal insufficiency. We provide further insight into the developmental complexity of disrupted MCM function, highlighted by two patients with a similar variant profile in MCM7 but disparate clinical features. Our results build on other genetic findings linked to disruption of the pre-replication and pre-initiation complexes, and the replisome, and expand the complex clinical genetics landscape emerging due to disruption of DNA replication.


Subject(s)
Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/genetics , Congenital Microtia/diagnosis , Congenital Microtia/genetics , Growth Disorders/diagnosis , Growth Disorders/genetics , Lipodystrophy/diagnosis , Lipodystrophy/genetics , Micrognathism/diagnosis , Micrognathism/genetics , Minichromosome Maintenance Complex Component 3/genetics , Minichromosome Maintenance Complex Component 7/genetics , Patella/abnormalities , Adolescent , Alleles , Amino Acid Sequence , Cell Cycle/genetics , Child , Child, Preschool , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Infant , Male , Minichromosome Maintenance Complex Component 3/chemistry , Minichromosome Maintenance Complex Component 7/chemistry , Models, Molecular , New Zealand , Phenotype , Protein Conformation
5.
Am J Med Genet A ; 185(3): 871-876, 2021 03.
Article in English | MEDLINE | ID: mdl-33338304

ABSTRACT

Meier-Gorlin syndrome is an autosomal recessively inherited disorder of growth retardation, accompanied by microtia and patellae a/hypoplasia and characteristic facies. Pathogenic variants in genes associated with the initiation of DNA replication underlie the condition, with biallelic variants in CDT1 the most common cause. Using 10× Chromium genome sequencing, we report CDT1 variants in an adult female, with an inframe amino acid deletion inherited in trans with a deep intronic variant which likely serves as the branchpoint site in Intron 8. Splicing defects arising from this variant were confirmed through in vitro analysis. At 49 years, she represents the oldest patient with a molecular diagnosis described in the literature and is the first reported patient with Meier-Gorlin syndrome to have carried a successful pregnancy to term. Both of her pregnancies were complicated by postpartum hemorrhage and upon subsequent necessary hysterectomy, revealed uterine abnormalities. There is scant knowledge on reproductive ability and success in patients with Meier-Gorlin syndrome. Successful pregnancies among other clinically recognizable forms of primordial dwarfism have also not been described previously. This case is therefore of clinical interest for many forms of inherited growth retardation, and will assist in providing more information and clinical guidance for females of reproductive age.


Subject(s)
Cell Cycle Proteins/genetics , Congenital Microtia/genetics , Frameshift Mutation , Growth Disorders/genetics , Micrognathism/genetics , Patella/abnormalities , Point Mutation , Pregnancy Complications/genetics , Alleles , Alternative Splicing , Base Sequence , Cell Cycle Proteins/deficiency , Codon, Nonsense/genetics , Female , Haplotypes/genetics , Humans , Introns/genetics , Middle Aged , Parity , Phenotype , Postpartum Hemorrhage/genetics , Pregnancy , Sequence Deletion , Uterus/abnormalities , Uterus/pathology , Whole Genome Sequencing
6.
J Med Genet ; 57(3): 195-202, 2020 03.
Article in English | MEDLINE | ID: mdl-31784481

ABSTRACT

MATERIAL: Linked-read whole genome sequencing (WGS) presents a new opportunity for cost-efficient singleton sequencing in place of traditional trio-based designs while generating informative-phased variants, effective for recessive disorders when parental DNA is unavailable. METHODS: We have applied linked-read WGS to identify novel causes of Meier-Gorlin syndrome (MGORS), a condition recognised by short stature, microtia and patella hypo/aplasia. There are eight genes associated with MGORS to date, all encoding essential components involved in establishing and initiating DNA replication. RESULTS: Our successful phasing of linked-read data led to the identification of biallelic rare variants in four individuals (24% of our cohort) in DONSON, a recently established DNA replication fork surveillance factor. The variants include five novel missense and one deep intronic variant. All were demonstrated to be deleterious to function; the missense variants all disrupted the nuclear localisation of DONSON, while the intronic variant created a novel splice site that generated an out-of-frame transcript with no residual canonical transcript produced. CONCLUSION: Variants in DONSON have previously been associated with extreme microcephaly, short stature and limb anomalies and perinatal lethal microcephaly-micromelia syndrome. Our novel genetic findings extend the complicated spectrum of phenotypes associated with DONSON variants and promote novel hypotheses for the role of DONSON in DNA replication. While our findings reiterate that MGORS is a disorder of DNA replication, the pathophysiology is obviously complex. This successful identification of a novel disease gene for MGORS highlights the utility of linked-read WGS as a successful technology to be considered in the genetic studies of recessive conditions.


Subject(s)
Cell Cycle Proteins/genetics , Congenital Microtia/genetics , Genetic Predisposition to Disease , Growth Disorders/genetics , Micrognathism/genetics , Nuclear Proteins/genetics , Patella/abnormalities , Adult , Alleles , Base Sequence/genetics , Child , Congenital Microtia/physiopathology , DNA Replication/genetics , Female , Genome, Human/genetics , Growth Disorders/physiopathology , Humans , Male , Micrognathism/physiopathology , Patella/metabolism , Patella/physiopathology , Pregnancy
7.
Hum Mutat ; 40(8): 1063-1070, 2019 08.
Article in English | MEDLINE | ID: mdl-31045292

ABSTRACT

Microcephalic primordial dwarfism (MPD) is a group of rare single-gene disorders characterized by the extreme reduction in brain and body size from early development onwards. Proteins encoded by MPD-associated genes play important roles in fundamental cellular processes, notably genome replication and repair. Here we report the identification of four MPD individuals with biallelic variants in DNA2, which encodes an adenosine triphosphate (ATP)-dependent helicase/nuclease involved in DNA replication and repair. We demonstrate that the two intronic variants (c.1764-38_1764-37ins(53) and c.74+4A>C) found in these individuals substantially impair DNA2 transcript splicing. Additionally, we identify a missense variant (c.1963A>G), affecting a residue of the ATP-dependent helicase domain that is highly conserved between humans and yeast, with the resulting substitution (p.Thr655Ala) predicted to directly impact ATP/ADP (adenosine diphosphate) binding by DNA2. Our findings support the pathogenicity of these variants as biallelic hypomorphic mutations, establishing DNA2 as an MPD disease gene.


Subject(s)
DNA Helicases/genetics , Dwarfism/genetics , Genetic Variation , Microcephaly/genetics , Adolescent , Alleles , DNA Helicases/chemistry , Female , Genetic Predisposition to Disease , Humans , Introns , Male , Middle Aged , Models, Molecular , Mutagenesis, Insertional , Mutation, Missense , Polymorphism, Single Nucleotide
8.
Am J Hum Genet ; 103(6): 1038-1044, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30503519

ABSTRACT

During genome replication, polymerase epsilon (Pol ε) acts as the major leading-strand DNA polymerase. Here we report the identification of biallelic mutations in POLE, encoding the Pol ε catalytic subunit POLE1, in 15 individuals from 12 families. Phenotypically, these individuals had clinical features closely resembling IMAGe syndrome (intrauterine growth restriction [IUGR], metaphyseal dysplasia, adrenal hypoplasia congenita, and genitourinary anomalies in males), a disorder previously associated with gain-of-function mutations in CDKN1C. POLE1-deficient individuals also exhibited distinctive facial features and variable immune dysfunction with evidence of lymphocyte deficiency. All subjects shared the same intronic variant (c.1686+32C>G) as part of a common haplotype, in combination with different loss-of-function variants in trans. The intronic variant alters splicing, and together the biallelic mutations lead to cellular deficiency of Pol ε and delayed S-phase progression. In summary, we establish POLE as a second gene in which mutations cause IMAGe syndrome. These findings add to a growing list of disorders due to mutations in DNA replication genes that manifest growth restriction alongside adrenal dysfunction and/or immunodeficiency, consolidating these as replisome phenotypes and highlighting a need for future studies to understand the tissue-specific development roles of the encoded proteins.


Subject(s)
Adrenal Insufficiency/genetics , DNA Polymerase II/genetics , Fetal Growth Retardation/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Urogenital Abnormalities/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA Replication/genetics , Female , Humans , Infant , Male , Middle Aged , Phenotype , Young Adult
10.
Nat Genet ; 49(4): 537-549, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28191891

ABSTRACT

To ensure efficient genome duplication, cells have evolved numerous factors that promote unperturbed DNA replication and protect, repair and restart damaged forks. Here we identify downstream neighbor of SON (DONSON) as a novel fork protection factor and report biallelic DONSON mutations in 29 individuals with microcephalic dwarfism. We demonstrate that DONSON is a replisome component that stabilizes forks during genome replication. Loss of DONSON leads to severe replication-associated DNA damage arising from nucleolytic cleavage of stalled replication forks. Furthermore, ATM- and Rad3-related (ATR)-dependent signaling in response to replication stress is impaired in DONSON-deficient cells, resulting in decreased checkpoint activity and the potentiation of chromosomal instability. Hypomorphic mutations in DONSON substantially reduce DONSON protein levels and impair fork stability in cells from patients, consistent with defective DNA replication underlying the disease phenotype. In summary, we have identified mutations in DONSON as a common cause of microcephalic dwarfism and established DONSON as a critical replication fork protein required for mammalian DNA replication and genome stability.


Subject(s)
DNA Replication/genetics , DNA-Binding Proteins/genetics , Dwarfism/genetics , Genomic Instability/genetics , Microcephaly/genetics , Mutation/genetics , Cell Line , DNA Damage/genetics , Female , Humans , Male
11.
Genes Dev ; 30(19): 2158-2172, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27737959

ABSTRACT

Compaction of chromosomes is essential for accurate segregation of the genome during mitosis. In vertebrates, two condensin complexes ensure timely chromosome condensation, sister chromatid disentanglement, and maintenance of mitotic chromosome structure. Here, we report that biallelic mutations in NCAPD2, NCAPH, or NCAPD3, encoding subunits of these complexes, cause microcephaly. In addition, hypomorphic Ncaph2 mice have significantly reduced brain size, with frequent anaphase chromatin bridge formation observed in apical neural progenitors during neurogenesis. Such DNA bridges also arise in condensin-deficient patient cells, where they are the consequence of failed sister chromatid disentanglement during chromosome compaction. This results in chromosome segregation errors, leading to micronucleus formation and increased aneuploidy in daughter cells. These findings establish "condensinopathies" as microcephalic disorders, with decatenation failure as an additional disease mechanism for microcephaly, implicating mitotic chromosome condensation as a key process ensuring mammalian cerebral cortex size.


Subject(s)
Adenosine Triphosphatases/genetics , DNA-Binding Proteins/genetics , Microcephaly/genetics , Mitosis/genetics , Multiprotein Complexes/genetics , Mutation/genetics , Aneuploidy , Animals , Catenanes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromosomal Instability/genetics , Chromosome Segregation/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Micronuclei, Chromosome-Defective , Neurons/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Stem Cells
12.
Am J Hum Genet ; 99(1): 125-38, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27374770

ABSTRACT

DNA replication precisely duplicates the genome to ensure stable inheritance of genetic information. Impaired licensing of origins of replication during the G1 phase of the cell cycle has been implicated in Meier-Gorlin syndrome (MGS), a disorder defined by the triad of short stature, microtia, and a/hypoplastic patellae. Biallelic partial loss-of-function mutations in multiple components of the pre-replication complex (preRC; ORC1, ORC4, ORC6, CDT1, or CDC6) as well as de novo stabilizing mutations in the licensing inhibitor, GMNN, cause MGS. Here we report the identification of mutations in CDC45 in 15 affected individuals from 12 families with MGS and/or craniosynostosis. CDC45 encodes a component of both the pre-initiation (preIC) and CMG helicase complexes, required for initiation of DNA replication origin firing and ongoing DNA synthesis during S-phase itself, respectively, and hence is functionally distinct from previously identified MGS-associated genes. The phenotypes of affected individuals range from syndromic coronal craniosynostosis to severe growth restriction, fulfilling diagnostic criteria for Meier-Gorlin syndrome. All mutations identified were biallelic and included synonymous mutations altering splicing of physiological CDC45 transcripts, as well as amino acid substitutions expected to result in partial loss of function. Functionally, mutations reduce levels of full-length transcripts and protein in subject cells, consistent with partial loss of CDC45 function and a predicted limited rate of DNA replication and cell proliferation. Our findings therefore implicate the preIC as an additional protein complex involved in the etiology of MGS and connect the core cellular machinery of genome replication with growth, chondrogenesis, and cranial suture homeostasis.


Subject(s)
Cell Cycle Proteins/genetics , Congenital Microtia/genetics , Craniosynostoses/genetics , Growth Disorders/genetics , Micrognathism/genetics , Mutation , Patella/abnormalities , Adolescent , Adult , Alleles , Alternative Splicing/genetics , Amino Acid Sequence , Amnion/cytology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Cell Line , Cells, Cultured , Child , Child, Preschool , DNA Mutational Analysis , DNA Replication , Exome/genetics , Exons/genetics , Female , Genetic Association Studies , Humans , Male , Models, Molecular , Protein Conformation , Syndrome , Young Adult
13.
Am J Hum Genet ; 96(3): 412-24, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25728776

ABSTRACT

Non-homologous end joining (NHEJ) is a key cellular process ensuring genome integrity. Mutations in several components of the NHEJ pathway have been identified, often associated with severe combined immunodeficiency (SCID), consistent with the requirement for NHEJ during V(D)J recombination to ensure diversity of the adaptive immune system. In contrast, we have recently found that biallelic mutations in LIG4 are a common cause of microcephalic primordial dwarfism (MPD), a phenotype characterized by prenatal-onset extreme global growth failure. Here we provide definitive molecular genetic evidence supported by biochemical, cellular, and immunological data for mutations in XRCC4, encoding the obligate binding partner of LIG4, causing MPD. We report the identification of biallelic mutations in XRCC4 in five families. Biochemical and cellular studies demonstrate that these alterations substantially decrease XRCC4 protein levels leading to reduced cellular ligase IV activity. Consequently, NHEJ-dependent repair of ionizing-radiation-induced DNA double-strand breaks is compromised in XRCC4 cells. Similarly, immunoglobulin junctional diversification is impaired in cells. However, immunoglobulin levels are normal, and individuals lack overt signs of immunodeficiency. Additionally, in contrast to individuals with LIG4 mutations, pancytopenia leading to bone marrow failure has not been observed. Hence, alterations that alter different NHEJ proteins give rise to a phenotypic spectrum, from SCID to extreme growth failure, with deficiencies in certain key components of this repair pathway predominantly exhibiting growth deficits, reflecting differential developmental requirements for NHEJ proteins to support growth and immune maturation.


Subject(s)
DNA-Binding Proteins/genetics , Dwarfism, Pituitary/genetics , Dwarfism/genetics , Microcephaly/genetics , Mutation , Alleles , Amino Acid Sequence , Child , Child, Preschool , DNA Breaks, Double-Stranded , DNA Ligase ATP , DNA Ligases/genetics , DNA Ligases/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Pulsed-Field , Exome , Facies , Female , Humans , Infant , Male , Molecular Sequence Data , Phenotype , Protein Conformation , Severe Combined Immunodeficiency/genetics
14.
Nat Genet ; 46(12): 1283-1292, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25344692

ABSTRACT

Centrioles are essential for ciliogenesis. However, mutations in centriole biogenesis genes have been reported in primary microcephaly and Seckel syndrome, disorders without the hallmark clinical features of ciliopathies. Here we identify mutations in the genes encoding PLK4 kinase, a master regulator of centriole duplication, and its substrate TUBGCP6 in individuals with microcephalic primordial dwarfism and additional congenital anomalies, including retinopathy, thereby extending the human phenotypic spectrum associated with centriole dysfunction. Furthermore, we establish that different levels of impaired PLK4 activity result in growth and cilia phenotypes, providing a mechanism by which microcephaly disorders can occur with or without ciliopathic features.


Subject(s)
Growth Disorders/genetics , Microcephaly/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Retinal Degeneration/genetics , Adolescent , Adult , Animals , Centrioles/ultrastructure , Child , Child, Preschool , Family Health , Female , Fibroblasts/metabolism , Genotype , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Microsatellite Repeats , Microtubule-Associated Proteins/genetics , Mitosis , Pakistan , Pedigree , Phenotype , Young Adult , Zebrafish
15.
Eur J Med Genet ; 57(10): 587-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25195018

ABSTRACT

Pierre Robin sequence (PRS) is an aetiologically distinct subgroup of cleft palate. We aimed to define the critical genomic interval from five different 5q22-5q31 deletions associated with PRS or PRS-associated features and assess each gene within the region as a candidate for the PRS component of the phenotype. Clinical array-based comparative genome hybridisation (aCGH) data were used to define a 2.08 Mb minimum region of overlap among four de novo deletions and one mother-son inherited deletion associated with at least one component of PRS. Commonly associated anomalies were talipes equinovarus (TEV), finger contractures and crumpled ear helices. Expression analysis of the orthologous genes within the PRS critical region in embryonic mice showed that the strongest candidate genes were FBN2 and PHAX. Targeted aCGH of the critical region and sequencing of these genes in a cohort of 25 PRS patients revealed no plausible disease-causing mutations. In conclusion, deletion of ∼2 Mb on 5q23 region causes a clinically recognisable subtype of PRS. Haploinsufficiency for FBN2 accounts for the digital and auricular features. A possible critical region for TEV is distinct and telomeric to the PRS region. The molecular basis of PRS in these cases remains undetermined but haploinsufficiency for PHAX is a plausible mechanism.


Subject(s)
Chromosomes, Human, Pair 5 , Gene Deletion , Microfilament Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Phosphoproteins/genetics , Pierre Robin Syndrome/genetics , Sequence Deletion/genetics , Adolescent , Child , Cleft Palate/genetics , Clubfoot/complications , Contracture/congenital , Ear, External/abnormalities , Female , Fibrillin-2 , Fibrillins , Fingers , Haploinsufficiency/genetics , Humans , Male , Mutation, Missense , Phenotype , Syndrome , Young Adult
16.
Hum Mutat ; 35(1): 76-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24123394

ABSTRACT

Ligase IV syndrome is a rare differential diagnosis for Nijmegen breakage syndrome owing to a shared predisposition to lympho-reticular malignancies, significant microcephaly, and radiation hypersensitivity. Only 16 cases with mutations in LIG4 have been described to date with phenotypes varying from malignancy in developmentally normal individuals, to severe combined immunodeficiency and early mortality. Here, we report the identification of biallelic truncating LIG4 mutations in 11 patients with microcephalic primordial dwarfism presenting with restricted prenatal growth and extreme postnatal global growth failure (average OFC -10.1 s.d., height -5.1 s.d.). Subsequently, most patients developed thrombocytopenia and leucopenia later in childhood and many were found to have previously unrecognized immunodeficiency following molecular diagnosis. None have yet developed malignancy, though all patients tested had cellular radiosensitivity. A genotype-phenotype correlation was also noted with position of truncating mutations corresponding to disease severity. This work extends the phenotypic spectrum associated with LIG4 mutations, establishing that extreme growth retardation with microcephaly is a common presentation of bilallelic truncating mutations. Such growth failure is therefore sufficient to consider a diagnosis of LIG4 deficiency and early recognition of such cases is important as bone marrow failure, immunodeficiency, and sometimes malignancy are long term sequelae of this disorder.


Subject(s)
DNA Ligases/deficiency , DNA Ligases/genetics , Dwarfism/genetics , Fetal Growth Retardation/genetics , Leukopenia/genetics , Thrombocytopenia/genetics , Abnormalities, Multiple/genetics , Adaptive Immunity , Adolescent , Cell Line , Child , Child, Preschool , DNA Ligase ATP , Exome , Female , Fetal Growth Retardation/etiology , Genetic Variation , Genotype , Heterozygote , Humans , Infant , Male , Microcephaly/genetics , Neoplasms/genetics , Nijmegen Breakage Syndrome/genetics , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Syndrome
18.
Am J Med Genet A ; 158A(10): 2577-82, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22887808

ABSTRACT

Primary microcephaly is a genetically heterogeneous condition characterized by reduced head circumference (-3 SDS or more) and mild-to-moderate learning disability. Here, we describe clinical and molecular investigations of a microcephalic child with sensorineural hearing loss. Although consanguinity was unreported initially, detection of 13.7 Mb of copy neutral loss of heterozygosity (cnLOH) on chromosome 9 implicated the CDK5RAP2 gene. Targeted sequencing identified a homozygous E234X mutation, only the third mutation to be described in CDK5RAP2, the first in an individual of non-Pakistani descent. Sensorineural hearing loss is not generally considered to be consistent with autosomal recessive microcephaly and therefore it seems likely that the deafness in this individual is caused by the co-occurrence of a further gene mutation, independent of CDK5RAP2. Nevertheless, further detailed clinical descriptions of rare CDK5RAP2 patients, including hearing assessments will be needed to resolve fully the phenotypic range associated with mutations in this gene. This study also highlights the utility of SNP-array testing to guide disease gene identification where an autosomal recessive condition is plausible.


Subject(s)
Codon, Nonsense , Hearing Loss, Sensorineural/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Cell Cycle Proteins , Child , Female , Humans , Pedigree , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Somalia
19.
Am J Med Genet A ; 158A(11): 2719-25, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22821869

ABSTRACT

Microcephalic primordial dwarfism (MPD) is a class of disorders characterized by intrauterine growth restriction (IUGR), impaired postnatal growth and microcephaly. Majewski osteodysplastic primordial dwarfism type II (MOPD II) is one of the more common conditions within this group. MOPD II is caused by truncating mutations in pericentrin (PCNT) and is inherited in an autosomal recessive manner. Detailed growth curves for length, weight, and OFC are presented here and derived from retrospective data from 26 individuals with MOPD II confirmed by molecular or functional studies. Severe pre- and postnatal growth failure is evident in MOPD II patients. The length, weight, and OFC at term (when corrected for gestational age) were -7.0, -3.9, and -4.6 standard deviation (SD) below the population mean and equivalent to the 50th centile of a 28-29-, 31-32-, and 30-31-week neonate, respectively. While at skeletal maturity, the height, weight, and OFC were -10.3, -14.3, and -8.5 SD below the population mean and equivalent to the size of 3-year 10- to 11-month-old, a 5-year 2- to 3-month-old, and 5- to 6-month-old, respectively. During childhood, MOPD II patients grow with slowed, but fairly constant growth velocities and show no evidence of any pubertal growth spurt. Treatment with human growth hormone (n = 11) did not lead to any significant improvement in final stature. The growth charts presented here will be of assistance with diagnosis and management of MOPD II, and should have particular utility in nutritional management of MOPD II during infancy.


Subject(s)
Antigens , Dwarfism/diagnosis , Dwarfism/genetics , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/genetics , Growth Charts , Microcephaly/diagnosis , Microcephaly/genetics , Mutation , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Adolescent , Adult , Body Height , Body Weight , Child , Child, Preschool , Dwarfism/drug therapy , Female , Fetal Growth Retardation/drug therapy , Human Growth Hormone/therapeutic use , Humans , Male , Microcephaly/drug therapy , Osteochondrodysplasias/drug therapy , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...