Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Cancer Res ; 29(7): 1220-1231, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36815791

ABSTRACT

PURPOSE: Patients with resected localized clear-cell renal cell carcinoma (ccRCC) remain at variable risk of recurrence. Incorporation of biomarkers may refine risk prediction and inform adjuvant treatment decisions. We explored the role of tumor genomics in this setting, leveraging the largest cohort to date of localized ccRCC tissues subjected to targeted gene sequencing. EXPERIMENTAL DESIGN: The somatic mutation status of 12 genes was determined in 943 ccRCC cases from a multinational cohort of patients, and associations to outcomes were examined in a Discovery (n = 469) and Validation (n = 474) framework. RESULTS: Tumors containing a von-Hippel Lindau (VHL) mutation alone were associated with significantly improved outcomes in comparison with tumors containing a VHL plus additional mutations. Within the Discovery cohort, those with VHL+0, VHL+1, VHL+2, and VHL+≥3 tumors had disease-free survival (DFS) rates of 90.8%, 80.1%, 68.2%, and 50.7% respectively, at 5 years. This trend was replicated in the Validation cohort. Notably, these genomically defined groups were independent of tumor mutational burden. Amongst patients eligible for adjuvant therapy, those with a VHL+0 tumor (29%) had a 5-year DFS rate of 79.3% and could, therefore, potentially be spared further treatment. Conversely, patients with VHL+2 and VHL+≥3 tumors (32%) had equivalent DFS rates of 45.6% and 35.3%, respectively, and should be prioritized for adjuvant therapy. CONCLUSIONS: Genomic characterization of ccRCC identified biologically distinct groups of patients with divergent relapse rates. These groups account for the ∼80% of cases with VHL mutations and could be used to personalize adjuvant treatment discussions with patients as well as inform future adjuvant trial design.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Kidney Neoplasms/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Neoplasm Recurrence, Local/genetics , Mutation
2.
Nat Genet ; 53(11): 1553-1563, 2021 11.
Article in English | MEDLINE | ID: mdl-34663923

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) shows remarkable variation in incidence that is not fully explained by known lifestyle and environmental risk factors. It has been speculated that an unknown exogenous exposure(s) could be responsible. Here we combine the fields of mutational signature analysis with cancer epidemiology to study 552 ESCC genomes from eight countries with varying incidence rates. Mutational profiles were similar across all countries studied. Associations between specific mutational signatures and ESCC risk factors were identified for tobacco, alcohol, opium and germline variants, with modest impacts on mutation burden. We find no evidence of a mutational signature indicative of an exogenous exposure capable of explaining differences in ESCC incidence. Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like (APOBEC)-associated mutational signatures single-base substitution (SBS)2 and SBS13 were present in 88% and 91% of cases, respectively, and accounted for 25% of the mutation burden on average, indicating that APOBEC activation is a crucial step in ESCC tumor development.


Subject(s)
Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/epidemiology , Esophageal Squamous Cell Carcinoma/genetics , Mutation , APOBEC Deaminases/genetics , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase, Mitochondrial/genetics , Brazil/epidemiology , China/epidemiology , Female , Humans , Incidence , Iran/epidemiology , Male , Middle Aged , Tumor Suppressor Protein p53/genetics , United Kingdom/epidemiology , Whole Genome Sequencing
3.
Cancers (Basel) ; 13(9)2021 May 07.
Article in English | MEDLINE | ID: mdl-34067022

ABSTRACT

Background: Malignant melanoma and RCC have different embryonic origins, no common lifestyle risk factors but intriguingly share biological properties such as immune regulation and radioresistance. An excess risk of malignant melanoma is observed in RCC patients and vice versa. This bidirectional association is poorly understood, and hypothetic genetic co-susceptibility remains largely unexplored. Results: We hereby provide a clinical and genetic description of a series of 125 cases affected by both malignant melanoma and RCC. Clinical germline mutation testing identified a pathogenic variant in a melanoma and/or RCC predisposing gene in 17/125 cases (13.6%). This included mutually exclusive variants in MITF (p.E318K locus, N = 9 cases), BAP1 (N = 3), CDKN2A (N = 2), FLCN (N = 2), and PTEN (N = 1). A subset of 46 early-onset cases, without underlying germline variation, was whole-exome sequenced. In this series, thirteen genes were significantly enriched in mostly exclusive rare variants predicted to be deleterious, compared to 19,751 controls of similar ancestry. The observed variation mainly consisted of novel or low-frequency variants (<0.01%) within genes displaying strong evolutionary mutational constraints along the PI3K/mTOR pathway, including PIK3CD, NFRKB, EP300, MTOR, and related epigenetic modifier SETD2. The screening of independently processed germline exomes from The Cancer Genome Atlas confirmed an association with melanoma and RCC but not with cancers of established differing etiology such as lung cancers. Conclusions: Our study highlights that an exome-wide case-control enrichment approach may better characterize the rare variant-based missing heritability of multiple primary cancers. In our series, the co-occurrence of malignant melanoma and RCC was associated with germline variation in the PI3K/mTOR signaling cascade, with potential relevance for early diagnostic and clinical management.

4.
Hum Mol Genet ; 30(5): 343-355, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33527138

ABSTRACT

Sexual dimorphism in cancer incidence and outcome is widespread. Understanding the underlying mechanisms is fundamental to improve cancer prevention and clinical management. Sex disparities are particularly striking in kidney cancer: across diverse populations, men consistently show unexplained 2-fold increased incidence and worse prognosis. We have characterized genome-wide expression and regulatory networks of 609 renal tumors and 256 non-tumor renal tissues. Normal kidney displayed sex-specific transcriptional signatures, including higher expression of X-linked tumor suppressor genes in women. Sex-dependent genotype-phenotype associations unraveled women-specific immune regulation. Sex differences were markedly expanded in tumors, with male-biased expression of key genes implicated in metabolism, non-malignant diseases with male predominance and carcinogenesis, including markers of tumor infiltrating leukocytes. Analysis of sex-dependent RCC progression and survival uncovered prognostic markers involved in immune response and oxygen homeostasis. In summary, human kidney tissues display remarkable sexual dimorphism at the molecular level. Sex-specific transcriptional signatures further shape renal cancer, with relevance for clinical management.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Kidney Neoplasms/genetics , Sex Characteristics , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/metabolism , Disease Progression , Female , Genes, Tumor Suppressor , Genes, X-Linked , Genetic Association Studies , Genome-Wide Association Study , Humans , Kidney Neoplasms/metabolism , Male , Middle Aged , Prognosis
5.
Virchows Arch ; 478(6): 1099-1107, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33403511

ABSTRACT

There are unexplained geographical variations in the incidence of kidney cancer with the high rates reported in Baltic countries, as well as eastern and central Europe. Having access to a large and well-annotated collection of "tumor/non-tumor" pairs of kidney cancer patients from the Czech Republic, Romania, Serbia, UK, and Russia, we aimed to analyze the morphology of non-neoplastic renal tissue in nephrectomy specimens. By applying digital pathology, we performed a microscopic examination of 1012 frozen non-neoplastic kidney tissues from patients with renal cell carcinoma. Four components of renal parenchyma were evaluated and scored for the intensity of interstitial inflammation and fibrosis, tubular atrophy, glomerulosclerosis, and arterial wall thickening, globally called chronic renal parenchymal changes. Moderate or severe changes were observed in 54 (5.3%) of patients with predominance of occurrence in Romania (OR = 2.67, CI 1.07-6.67) and Serbia (OR = 4.37, CI 1.20-15.96) in reference to those from Russia. Further adjustment for comorbidities, tumor characteristics, and stage did not change risk estimates. In multinomial regression model, relative probability of non-glomerular changes was 5.22 times higher for Romania and Serbia compared to Russia. Our findings show that the frequency of chronic renal parenchymal changes, with the predominance of chronic interstitial nephritis pattern, in kidney cancer patients varies by country, significantly more frequent in countries located in central and southeastern Europe where the incidence of kidney cancer has been reported to be moderate to high. The observed association between these pathological features and living in certain geographic areas requires a larger population-based study to confirm this association on a large scale.


Subject(s)
Carcinoma, Renal Cell/pathology , Fibrosis/pathology , Kidney Neoplasms/pathology , Kidney/pathology , Adult , Aged , Europe , Female , Glomerular Filtration Rate/physiology , Humans , Male , Middle Aged , Nephrectomy/methods , Russia
6.
Hum Mol Genet ; 28(20): 3466-3474, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31504499

ABSTRACT

Recurrent pregnancy loss (RPL) is defined as two or more consecutive miscarriages and affects an estimated 1.5% of couples trying to conceive. RPL has been attributed to genetic, endocrine, immune and thrombophilic disorders, but many cases remain unexplained. We investigated a Bangladeshi family where the proband experienced 29 consecutive pregnancy losses with no successful pregnancies from three different marriages. Whole exome sequencing identified rare genetic variants in several candidate genes. These were further investigated in Asian and white European RPL cohorts, and in Bangladeshi controls. FKBP4, encoding the immunophilin FK506-binding protein 4, was identified as a plausible candidate, with three further novel variants identified in Asian patients. None were found in European patients or controls. In silico structural studies predicted damaging effects of the variants in the structure-function properties of the FKBP52 protein. These were located within domains reported to be involved in Hsp90 binding and peptidyl-prolyl cis-trans isomerase (PPIase) activity. Profound effects on PPIase activity were demonstrated in transiently transfected HEK293 cells comparing wild-type and mutant FKBP4 constructs. Mice lacking FKBP4 have been previously reported as infertile through implantation failure. This study therefore strongly implicates FKBP4 as associated with fetal losses in humans, particularly in the Asian population.


Subject(s)
Abortion, Habitual/genetics , Exome Sequencing/methods , Tacrolimus Binding Proteins/genetics , Exome/genetics , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mutation, Missense/genetics , Pedigree , Pregnancy , Protein Structure, Secondary
7.
Eur Urol Focus ; 4(4): 586-590, 2018 07.
Article in English | MEDLINE | ID: mdl-28753845

ABSTRACT

It is well established that men are at higher risk of most non-sex-specific cancers than women, but there has been surprisingly little research investigating these differences. This is possibly because differences in exposure to established risk factors and hypothesised protection by female sex hormones are thought to account for the totality of the sex differences. These explanations remain somewhat speculative, as the magnitude of the sex ratio in cancer incidence has not been systematically studied, with no quantitative estimate of the variability of the sex ratio across countries, age groups, and periods of diagnosis. We analysed worldwide cancer incidence data for the years 1978-2007 in terms of sex disparities, and explicitly quantified the variability in sex disparities by age, year, and geographical region. Our analysis highlights several cancer types for which suspected and commonly accepted factors are unlikely to fully explain the observed sex disparity. In particular, kidney cancer showed a 2:1 male/female case incidence ratio that was constant by age, year, and region, suggesting that factors other than sociocultural habits and health behaviours are responsible for this sex disparity. PATIENT SUMMARY: We quantified the difference in the incidence of various cancer types between men and women across the world over 30 years. While the trends for some cancers such as lung cancer are clearly correlated with known variations in lifestyle habits, we found that the sex disparity observed for others such as kidney cancer is unlikely to be explained by known risk factors.


Subject(s)
Kidney Neoplasms/epidemiology , Sex Distribution , Adult , Age Distribution , Aged , Analysis of Variance , Female , Health Status Disparities , Humans , Incidence , Male , Middle Aged , Risk Factors
8.
Int J Cancer ; 141(9): 1730-1740, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28639257

ABSTRACT

Circulating miRNAs have shown great promises as noninvasive diagnostic and predictive biomarkers in several solid tumors. While the miRNA profiles of renal tumors have been extensively explored, knowledge of their circulating counterparts is limited. Our study aimed to provide a large-scale genome-wide profiling of plasma circulating miRNA in clear-cell renal cell carcinoma (ccRCC). Plasma samples from 94 ccRCC cases and 100 controls were screened for 754 circulating micro-RNAs (miRNA) by TaqMan arrays. Analyses including known risk factors for renal cancer-namely, age, sex, hypertension, obesity, diabetes, tobacco smoking and alcohol consumption-highlighted that circulating miRNA profiles were tightly correlated with the stage of the disease. Advanced tumors, characterized as stage III and IV, were associated with specific miRNA signatures that significantly differ from both controls and earlier stage ccRCC cases. Molecular pathway enrichment analyses of their gene targets showed high similarities with alterations observed in renal tumors. Plasma circulating levels of miR-150 were significantly associated with RCC-specific survival and could marginally improve the predictive accuracy of clinical parameters in our series, including age at diagnosis, sex and conventional staging. In summary, our results suggest that circulating miRNAs may provide insights into renal cell carcinoma progression.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Renal Cell/blood , MicroRNAs/blood , Adult , Aged , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Disease Progression , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Staging , Prognosis
9.
EBioMedicine ; 10: 117-23, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27377626

ABSTRACT

Circulating tumor DNA (ctDNA) is emerging as a key potential biomarker for post-diagnosis surveillance but it may also play a crucial role in the detection of pre-clinical cancer. Small-cell lung cancer (SCLC) is an excellent candidate for early detection given there are no successful therapeutic options for late-stage disease, and it displays almost universal inactivation of TP53. We assessed the presence of TP53 mutations in the cell-free DNA (cfDNA) extracted from the plasma of 51 SCLC cases and 123 non-cancer controls. We identified mutations using a pipeline specifically designed to accurately detect variants at very low fractions. We detected TP53 mutations in the cfDNA of 49% SCLC patients and 11.4% of non-cancer controls. When stratifying the 51 initial SCLC cases by stage, TP53 mutations were detected in the cfDNA of 35.7% early-stage and 54.1% late-stage SCLC patients. The results in the controls were further replicated in 10.8% of an independent series of 102 non-cancer controls. The detection of TP53 mutations in 11% of the 225 non-cancer controls suggests that somatic mutations in cfDNA among individuals without any cancer diagnosis is a common occurrence, and poses serious challenges for the development of ctDNA screening tests.


Subject(s)
Biomarkers, Tumor , DNA, Neoplasm , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/genetics , Case-Control Studies , DNA, Neoplasm/blood , Early Detection of Cancer , Female , Humans , Leukocytes/metabolism , Lung Neoplasms/blood , Male , Mutation , Neoplasm Staging , Small Cell Lung Carcinoma/blood , Tumor Suppressor Protein p53/genetics
10.
Eur J Hum Genet ; 24(2): 298-301, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26059842

ABSTRACT

The success of whole-exome sequencing to identify mutations causing single-gene disorders has been well documented. In contrast whole-exome sequencing has so far had limited success in the identification of variants causing more complex phenotypes that seem unlikely to be due to the disruption of a single gene. We describe a family where two male offspring of healthy first cousin parents present a complex phenotype consisting of peripheral neuropathy and bronchiectasis that has not been described previously in the literature. Due to the fact that both children had the same problems in the context of parental consanguinity we hypothesised illness resulted from either X-linked or autosomal recessive inheritance. Through the use of whole-exome sequencing we were able to simplify this complex phenotype and identified a causative mutation (p.R1070*) in the gene periaxin (PRX), a gene previously shown to cause peripheral neuropathy (Dejerine-Sottas syndrome) when this mutation is present. For the bronchiectasis phenotype we were unable to identify a causal single mutation or compound heterozygote, reflecting the heterogeneous nature of this phenotype. In conclusion, in this study we show that whole-exome sequencing has the power to disentangle complex phenotypes through the identification of causative genetic mutations for distinct clinical disorders that were previously masked.


Subject(s)
Exome/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Membrane Proteins/genetics , Peripheral Nervous System Diseases/genetics , Female , Hereditary Sensory and Motor Neuropathy/pathology , Heterozygote , Humans , Male , Mutation/genetics , Pedigree , Peripheral Nervous System Diseases/pathology , Phenotype , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
12.
J Invest Dermatol ; 135(8): 2093-2101, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25815427

ABSTRACT

Congenital melanocytic nevus (CMN) syndrome is the association of pigmented melanocytic nevi with extra-cutaneous features, classically melanotic cells within the central nervous system, most frequently caused by a mutation of NRAS codon 61. This condition is currently untreatable and carries a significant risk of melanoma within the skin, brain, or leptomeninges. We have previously proposed a key role for Wnt signaling in the formation of melanocytic nevi, suggesting that activated Wnt signaling may be synergistic with activated NRAS in the pathogenesis of CMN syndrome. Some familial pre-disposition suggests a germ-line contribution to CMN syndrome, as does variability of neurological phenotypes in individuals with similar cutaneous phenotypes. Accordingly, we performed exome sequencing of germ-line DNA from patients with CMN to reveal rare or undescribed Wnt-signaling alterations. A murine model harboring activated NRAS(Q61K) and Wnt signaling in melanocytes exhibited striking features of CMN syndrome, in particular neurological involvement. In the first model of treatment for this condition, these congenital, and previously assumed permanent, features were profoundly suppressed by acute post-natal treatment with a MEK inhibitor. These data suggest that activated NRAS and aberrant Wnt signaling conspire to drive CMN syndrome. Post-natal MEK inhibition is a potential candidate therapy for patients with this debilitating condition.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Membrane Proteins/physiology , Nevus, Pigmented/congenital , Signal Transduction/physiology , Skin Neoplasms/congenital , Wnt Proteins/physiology , Animals , Child , DNA/genetics , Disease Models, Animal , Female , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/physiology , Mutation/genetics , Nevus, Pigmented/metabolism , Nevus, Pigmented/physiopathology , Sequence Analysis, DNA , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology
13.
J Clin Immunol ; 35(2): 147-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25663137

ABSTRACT

PURPOSE: Mutations in RMRP primarily give rise to Cartilage Hair Hypoplasia (CHH), a highly diverse skeletal disorder which can be associated with severe immunodeficiency. Increased availability of RMRP mutation screening has uncovered a number of infants with significant immunodeficiency but only mild or absent skeletal features. We surveyed the clinical and immunological phenotype of children who have undergone allogeneic haematopoietic stem cell transplantation for this condition in the UK. METHODS: Thirteen patients with confirmed RMRP mutations underwent allogeneic stem cell transplantation (SCT) at two nationally commissioned centres using a variety of donors and conditioning regimens. Records were retrospectively reviewed. RESULTS: Median time from clinical presentation to diagnosis was 12 months (range 1 to 276 months), with three infants diagnosed with severe combined immunodeficiency (SCID) without radiographical manifestations of CHH. A total of 17 allogeneic procedures were performed on 13 patients including two stem-cell top-ups. The median age at transplant was 32.4 months (range 1.5 to 125 months). Of the eleven surviving patients, median follow-up was 50 months (range 21.6 to 168 months). CONCLUSIONS: RMRP mutations can cause short stature and significant immunodeficiency which can be corrected by allogeneic SCT and the diagnosis should be considered even in the absence of skeletal manifestations.


Subject(s)
Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Mutation , Phenotype , RNA, Long Noncoding/genetics , Alleles , Child , Child, Preschool , Female , Follow-Up Studies , Genotype , Graft vs Host Disease/etiology , Hair/abnormalities , Hematopoietic Stem Cell Transplantation/adverse effects , Hirschsprung Disease/diagnosis , Hirschsprung Disease/genetics , Humans , Immunologic Deficiency Syndromes/therapy , Immunophenotyping , Infant , Infant, Newborn , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Osteochondrodysplasias/congenital , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Primary Immunodeficiency Diseases , Severity of Illness Index , Siblings , Transplantation Chimera , Treatment Outcome
14.
J Med Genet ; 52(3): 147-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25564561

ABSTRACT

BACKGROUND: Mutations in microtubule-regulating genes are associated with disorders of neuronal migration and microcephaly. Regulation of centriole length has been shown to underlie the pathogenesis of certain ciliopathy phenotypes. Using a next-generation sequencing approach, we identified mutations in a novel centriolar disease gene in a kindred with an embryonic lethal ciliopathy phenotype and in a patient with primary microcephaly. METHODS AND RESULTS: Whole exome sequencing data from a non-consanguineous Caucasian kindred exhibiting mid-gestation lethality and ciliopathic malformations revealed two novel non-synonymous variants in CENPF, a microtubule-regulating gene. All four affected fetuses showed segregation for two mutated alleles [IVS5-2A>C, predicted to abolish the consensus splice-acceptor site from exon 6; c.1744G>T, p.E582X]. In a second unrelated patient exhibiting microcephaly, we identified two CENPF mutations [c.1744G>T, p.E582X; c.8692 C>T, p.R2898X] by whole exome sequencing. We found that CENP-F colocalised with Ninein at the subdistal appendages of the mother centriole in mouse inner medullary collecting duct cells. Intraflagellar transport protein-88 (IFT-88) colocalised with CENP-F along the ciliary axonemes of renal epithelial cells in age-matched control human fetuses but did not in truncated cilia of mutant CENPF kidneys. Pairwise co-immunoprecipitation assays of mitotic and serum-starved HEKT293 cells confirmed that IFT88 precipitates with endogenous CENP-F. CONCLUSIONS: Our data identify CENPF as a new centriolar disease gene implicated in severe human ciliopathy and microcephaly related phenotypes. CENP-F has a novel putative function in ciliogenesis and cortical neurogenesis.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Cilia/genetics , Genetics, Medical , Microcephaly/genetics , Microfilament Proteins/genetics , Animals , Centrioles/genetics , Cilia/pathology , Exome/genetics , Female , Fetus , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Microcephaly/pathology , Mutation , NIH 3T3 Cells , Pedigree , Pregnancy , Zebrafish
15.
Am J Hum Genet ; 95(5): 611-21, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25439728

ABSTRACT

Intellectual disability and cerebellar atrophy occur together in a large number of genetic conditions and are frequently associated with microcephaly and/or epilepsy. Here we report the identification of causal mutations in Sorting Nexin 14 (SNX14) found in seven affected individuals from three unrelated consanguineous families who presented with recessively inherited moderate-severe intellectual disability, cerebellar ataxia, early-onset cerebellar atrophy, sensorineural hearing loss, and the distinctive association of progressively coarsening facial features, relative macrocephaly, and the absence of seizures. We used homozygosity mapping and whole-exome sequencing to identify a homozygous nonsense mutation and an in-frame multiexon deletion in two families. A homozygous splice site mutation was identified by Sanger sequencing of SNX14 in a third family, selected purely by phenotypic similarity. This discovery confirms that these characteristic features represent a distinct and recognizable syndrome. SNX14 encodes a cellular protein containing Phox (PX) and regulator of G protein signaling (RGS) domains. Weighted gene coexpression network analysis predicts that SNX14 is highly coexpressed with genes involved in cellular protein metabolism and vesicle-mediated transport. All three mutations either directly affected the PX domain or diminished SNX14 levels, implicating a loss of normal cellular function. This manifested as increased cytoplasmic vacuolation as observed in cultured fibroblasts. Our findings indicate an essential role for SNX14 in neural development and function, particularly in development and maturation of the cerebellum.


Subject(s)
Cerebellar Ataxia/genetics , Intellectual Disability/genetics , Sorting Nexins/genetics , Base Sequence , Cerebellar Ataxia/pathology , Chromosome Mapping , Codon, Nonsense/genetics , Female , Fibroblasts/ultrastructure , Gene Regulatory Networks/genetics , Genes, Recessive/genetics , Humans , Intellectual Disability/pathology , Male , Microscopy, Electron , Molecular Sequence Data , Pedigree , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
16.
Hum Mol Genet ; 23(10): 2511-26, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24412933

ABSTRACT

Ocular coloboma is a congenital defect resulting from failure of normal closure of the optic fissure during embryonic eye development. This birth defect causes childhood blindness worldwide, yet the genetic etiology is poorly understood. Here, we identified a novel homozygous mutation in the SALL2 gene in members of a consanguineous family affected with non-syndromic ocular coloboma variably affecting the iris and retina. This mutation, c.85G>T, introduces a premature termination codon (p.Glu29*) predicted to truncate the SALL2 protein so that it lacks three clusters of zinc-finger motifs that are essential for DNA-binding activity. This discovery identifies SALL2 as the third member of the Drosophila homeotic Spalt-like family of developmental transcription factor genes implicated in human disease. SALL2 is expressed in the developing human retina at the time of, and subsequent to, optic fissure closure. Analysis of Sall2-deficient mouse embryos revealed delayed apposition of the optic fissure margins and the persistence of an anterior retinal coloboma phenotype after birth. Sall2-deficient embryos displayed correct posterior closure toward the optic nerve head, and upon contact of the fissure margins, dissolution of the basal lamina occurred and PAX2, known to be critical for this process, was expressed normally. Anterior closure was disrupted with the fissure margins failing to meet, or in some cases misaligning leading to a retinal lesion. These observations demonstrate, for the first time, a role for SALL2 in eye morphogenesis and that loss of function of the gene causes ocular coloboma in humans and mice.


Subject(s)
Codon, Nonsense , Coloboma/genetics , Transcription Factors/genetics , Adolescent , Animals , Child , Consanguinity , DNA Mutational Analysis , DNA-Binding Proteins , Eye/embryology , Eye/pathology , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homozygote , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pedigree , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism
17.
Nat Genet ; 46(1): 70-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24241535

ABSTRACT

Lenz-Majewski syndrome (LMS) is a syndrome of intellectual disability and multiple congenital anomalies that features generalized craniotubular hyperostosis. By using whole-exome sequencing and selecting variants consistent with the predicted dominant de novo etiology of LMS, we identified causative heterozygous missense mutations in PTDSS1, which encodes phosphatidylserine synthase 1 (PSS1). PSS1 is one of two enzymes involved in the production of phosphatidylserine. Phosphatidylserine synthesis was increased in intact fibroblasts from affected individuals, and end-product inhibition of PSS1 by phosphatidylserine was markedly reduced. Therefore, these mutations cause a gain-of-function effect associated with regulatory dysfunction of PSS1. We have identified LMS as the first human disease, to our knowledge, caused by disrupted phosphatidylserine metabolism. Our results point to an unexplored link between phosphatidylserine synthesis and bone metabolism.


Subject(s)
Abnormalities, Multiple/genetics , Mutation , Nitrogenous Group Transferases/genetics , Adolescent , Animals , Cells, Cultured , Child , Dwarfism , Embryo, Nonmammalian , Female , Fibroblasts/metabolism , Humans , Hyperostosis , Male , Molecular Sequence Data , Nitrogenous Group Transferases/metabolism , Phosphatidylserines/biosynthesis , Phosphatidylserines/genetics , Syndrome , Zebrafish/embryology , Zebrafish/genetics
18.
Brain ; 136(Pt 10): 3096-105, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24022475

ABSTRACT

We describe a previously unreported syndrome characterized by secondary (post-natal) microcephaly with fronto-temporal lobe hypoplasia, multiple pituitary hormone deficiency, seizures, severe visual impairment and abnormalities of the kidneys and urinary tract in a highly consanguineous family with six affected children. Homozygosity mapping and exome sequencing revealed a novel homozygous frameshift mutation in the basic helix-loop-helix transcription factor gene ARNT2 (c.1373_1374dupTC) in affected individuals. This mutation results in absence of detectable levels of ARNT2 transcript and protein from patient fibroblasts compared with controls, consistent with nonsense-mediated decay of the mutant transcript and loss of ARNT2 function. We also show expression of ARNT2 within the central nervous system, including the hypothalamus, as well as the renal tract during human embryonic development. The progressive neurological abnormalities, congenital hypopituitarism and post-retinal visual pathway dysfunction in affected individuals demonstrates for the first time the essential role of ARNT2 in the development of the hypothalamo-pituitary axis, post-natal brain growth, and visual and renal function in humans.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypopituitarism/genetics , Kidney/abnormalities , Microcephaly/genetics , Mutation/genetics , Pituitary Hormones/metabolism , Visual Perception , Child , Child, Preschool , Female , Humans , Hypopituitarism/diagnosis , Hypothalamus/metabolism , Kidney/metabolism , Male , Microcephaly/diagnosis , Pituitary Hormones/genetics , Syndrome , Transcription Factors
19.
Am J Med Genet A ; 161A(10): 2401-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23949889

ABSTRACT

Here we report on a Portuguese family with three sisters who shared moderate intellectual disability, unusual facial morphology (short palpebral fissures; broad nasal tip; thin upper and lower vermillion; broad and pointed chin) and hand anomalies in two of them (short left third and fifth right metacarpals in one case; marked syndactyly between the third and fourth fingers in another). One of the sisters had microcephaly and short stature, and the other two were obese. Obesity and somewhat similar facial features were also present in the otherwise healthy mother. Despite the overlap with several known syndromes (Albright osteodystrophy; Filippi syndrome; Rubinstein-Taybi syndrome; microdeletion 2q37), we suggest this condition is previously unreported, and most likely displays an autosomal recessive pattern of inheritance. © 2013 Wiley Periodicals, Inc.


Subject(s)
Abnormalities, Multiple/diagnosis , Hand Deformities, Congenital/diagnosis , Intellectual Disability/diagnosis , Abnormalities, Multiple/genetics , Adult , Aged , Child , Chromosome Aberrations , Facies , Female , Genotype , Hand Deformities, Congenital/genetics , Humans , Intellectual Disability/genetics , Male , Middle Aged , Siblings , Syndrome , X Chromosome Inactivation
20.
J Invest Dermatol ; 133(9): 2229-36, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23392294

ABSTRACT

Congenital melanocytic nevi (CMN) can be associated with neurological abnormalities and an increased risk of melanoma. Mutations in NRAS, BRAF, and Tp53 have been described in individual CMN samples; however, their role in the pathogenesis of multiple CMN within the same subject and development of associated features has not been clear. We hypothesized that a single postzygotic mutation in NRAS could be responsible for multiple CMN in the same individual, as well as for melanocytic and nonmelanocytic central nervous system (CNS) lesions. From 15 patients, 55 samples with multiple CMN were sequenced after site-directed mutagenesis and enzymatic digestion of the wild-type allele. Oncogenic missense mutations in codon 61 of NRAS were found in affected neurological and cutaneous tissues of 12 out of 15 patients, but were absent from unaffected tissues and blood, consistent with NRAS mutation mosaicism. In 10 patients, the mutation was consistently c.181C>A, p.Q61K, and in 2 patients c.182A>G, p.Q61R. All 11 non-melanocytic and melanocytic CNS samples from 5 patients were mutation positive, despite NRAS rarely being reported as mutated in CNS tumors. Loss of heterozygosity was associated with the onset of melanoma in two cases, implying a multistep progression to malignancy. These results suggest that single postzygotic NRAS mutations are responsible for multiple CMN and associated neurological lesions in the majority of cases.


Subject(s)
GTP Phosphohydrolases/genetics , Melanosis/genetics , Membrane Proteins/genetics , Neurocutaneous Syndromes/genetics , Nevus, Pigmented/genetics , Skin Neoplasms/genetics , Adolescent , Central Nervous System Neoplasms/epidemiology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Hamartoma/epidemiology , Hamartoma/genetics , Hamartoma/pathology , Humans , Loss of Heterozygosity/genetics , Magnetic Resonance Imaging , Male , Melanosis/congenital , Melanosis/epidemiology , Meningeal Neoplasms/epidemiology , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/epidemiology , Meningioma/genetics , Meningioma/pathology , Mosaicism , Mutation, Missense/genetics , Neurocutaneous Syndromes/congenital , Neurocutaneous Syndromes/epidemiology , Nevus, Pigmented/congenital , Nevus, Pigmented/epidemiology , Prevalence , Risk Factors , Skin Neoplasms/congenital , Skin Neoplasms/epidemiology , Young Adult , Zygote
SELECTION OF CITATIONS
SEARCH DETAIL
...