Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6279, 2023 10 07.
Article in English | MEDLINE | ID: mdl-37805627

ABSTRACT

Hedgehog signaling mediates embryologic development of the central nervous system and other tissues and is frequently hijacked by neoplasia to facilitate uncontrolled cellular proliferation. Meningiomas, the most common primary brain tumor, exhibit Hedgehog signaling activation in 6.5% of cases, triggered by recurrent mutations in pathway mediators such as SMO. In this study, we find 35.6% of meningiomas that lack previously known drivers acquired various types of somatic structural variations affecting chromosomes 2q35 and 7q36.3. These cases exhibit ectopic expression of Hedgehog ligands, IHH and SHH, respectively, resulting in Hedgehog signaling activation. Recurrent tandem duplications involving IHH permit de novo chromatin interactions between super-enhancers within DIRC3 and a locus containing IHH. Our work expands the landscape of meningioma molecular drivers and demonstrates enhancer hijacking of Hedgehog ligands as a route to activate this pathway  in neoplasia.


Subject(s)
Meningeal Neoplasms , Meningioma , Humans , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Meningioma/genetics , Ligands , Signal Transduction , Meningeal Neoplasms/genetics
2.
Nat Med ; 27(12): 2165-2175, 2021 12.
Article in English | MEDLINE | ID: mdl-34887573

ABSTRACT

Intracranial aneurysm (IA) rupture leads to subarachnoid hemorrhage, a sudden-onset disease that often causes death or severe disability. Although genome-wide association studies have identified common genetic variants that increase IA risk moderately, the contribution of variants with large effect remains poorly defined. Using whole-exome sequencing, we identified significant enrichment of rare, deleterious mutations in PPIL4, encoding peptidyl-prolyl cis-trans isomerase-like 4, in both familial and index IA cases. Ppil4 depletion in vertebrate models causes intracerebral hemorrhage, defects in cerebrovascular morphology and impaired Wnt signaling. Wild-type, but not IA-mutant, PPIL4 potentiates Wnt signaling by binding JMJD6, a known angiogenesis regulator and Wnt activator. These findings identify a novel PPIL4-dependent Wnt signaling mechanism involved in brain-specific angiogenesis and maintenance of cerebrovascular integrity and implicate PPIL4 gene mutations in the pathogenesis of IA.


Subject(s)
Brain/blood supply , Cyclophilins/genetics , Intracranial Aneurysm/genetics , Neovascularization, Pathologic/genetics , RNA-Binding Proteins/genetics , Cyclophilins/physiology , Humans , Mutation , RNA-Binding Proteins/physiology , Exome Sequencing , Wnt Signaling Pathway/physiology
3.
J Hum Genet ; 66(2): 215-218, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32764695

ABSTRACT

Intellectual disability (ID) is a genetic and clinically heterogeneous common disease and underlying molecular pathogenesis can frequently not be identified by whole-exome/genome testing. Here, we report four siblings born to a consanguineous union who presented with intellectual disability and discuss the METAP1 pathway as a novel etiology of ID. Genomic analyses demonstrated that patients harbor a novel homozygous nonsense mutation in the gene METAP1. METAP1 codes for methionine aminopeptidase 1 (MetAP1) which oversees the co-translational excision of the first methionine remnants in eukaryotes. The loss-of-function mutations to this gene may result in a defect in the translation of many essential proteins within a cell. Improper neuronal function resulting from this loss of essential proteins could lead to neurologic impairment and ID.


Subject(s)
Aminopeptidases/genetics , Genes, Recessive , Intellectual Disability/genetics , Intellectual Disability/pathology , Mutation , Adolescent , Child , Female , Humans , Male , Pedigree , Siblings , Exome Sequencing
4.
Ann Neurol ; 84(5): 638-647, 2018 11.
Article in English | MEDLINE | ID: mdl-30178464

ABSTRACT

OBJECTIVE: To identify causes of the autosomal-recessive malformation, diencephalic-mesencephalic junction dysplasia (DMJD) syndrome. METHODS: Eight families with DMJD were studied by whole-exome or targeted sequencing, with detailed clinical and radiological characterization. Patient-derived induced pluripotent stem cells were derived into neural precursor and endothelial cells to study gene expression. RESULTS: All patients showed biallelic mutations in the nonclustered protocadherin-12 (PCDH12) gene. The characteristic clinical presentation included progressive microcephaly, craniofacial dysmorphism, psychomotor disability, epilepsy, and axial hypotonia with variable appendicular spasticity. Brain imaging showed brainstem malformations and with frequent thinned corpus callosum with punctate brain calcifications, reflecting expression of PCDH12 in neural and endothelial cells. These cells showed lack of PCDH12 expression and impaired neurite outgrowth. INTERPRETATION: DMJD patients have biallelic mutations in PCDH12 and lack of protein expression. These patients present with characteristic microcephaly and abnormalities of white matter tracts. Such pathogenic variants predict a poor outcome as a result of brainstem malformation and evidence of white matter tract defects, and should be added to the phenotypic spectrum associated with PCDH12-related conditions. Ann Neurol 2018;84:646-655.


Subject(s)
Brain Stem/abnormalities , Cadherins/genetics , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Mutation , Protocadherins
6.
Mol Genet Genomic Med ; 6(2): 230-248, 2018 03.
Article in English | MEDLINE | ID: mdl-29397575

ABSTRACT

BACKGROUND: The camptodactyly-arthropathy-coxa vara-pericarditis syndrome (CACP) is a rare autosomal recessive condition characterized by camptodactyly, noninflammatory arthropathy, coxa vara, and pericarditis. CACP is caused by mutations in the proteoglycan 4 (PRG4) gene, which encodes a lubricating glycoprotein present in the synovial fluid and at the surface of articular cartilage. METHODS: In the present study, we compared the clinical and molecular findings of CACP syndrome in 35 patients from 11 unrelated families. In 28 patients, whole exome sequencing was used to investigate genomic variations. RESULTS: We found that camptodactyly of hands was the first symptom presented by most patients. Swelling of wrists, knees, and elbows began before 4 years of age, while the age of joint involvement was variable. Patients reported an increased pain level after the age of 10, and severe hip involvement developed after 20 years old. All patients presented developmental coxa vara and seven patients (~22%) had pleural effusion, pericarditis, and/or ascites. We identified nine novel genomic alterations, including the first case of homozygous complete deletion of exon 1 in the PRG4 gene. CONCLUSION: With this study, we contribute to the catalog of CACP causing variants. We confirm that the skeletal component of this disease worsens with age, and presents the potential mechanisms for interfamily variability, by discussing the influence of a modifier gene and escape from nonsense-mediated mRNA decay. We believe that this report will increase awareness of this familial arthropathic condition and the characteristic clinical and radiological findings will facilitate the differentiation from the common childhood rheumatic diseases such as juvenile idiopathic arthritis.


Subject(s)
Arthropathy, Neurogenic/diagnosis , Arthropathy, Neurogenic/genetics , Coxa Vara/diagnosis , Coxa Vara/genetics , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Proteoglycans/genetics , Synovitis/diagnosis , Synovitis/genetics , Adolescent , Adult , Child , Child, Preschool , Exons/genetics , Female , Genetic Association Studies/methods , Humans , Male , Middle Aged , Mutation , Proteoglycans/metabolism , Retrospective Studies , Sequence Deletion , Exome Sequencing/methods
7.
Article in English | MEDLINE | ID: mdl-28630369

ABSTRACT

Primary cardiomyopathy is one of the most common inherited cardiac diseases and harbors significant phenotypic and genetic heterogeneity. Because of this, genetic testing has become standard in treatment of this disease group. Indeed, in recent years, next-generation DNA sequencing has found broad applications in medicine, both as a routine diagnostic tool for genetic disorders and as a high-throughput discovery tool for identifying novel disease-causing genes. We describe a male infant with primary dilated cardiomyopathy who was diagnosed using intrauterine echocardiography and found to progress to hypertrophic cardiomyopathy after birth. This proband was born to a nonconsanguineous family with a past history of a male fetus that died because of cardiac abnormalities at 30 wk of gestation. Using whole-exome sequencing, a novel homozygous frameshift mutation (c.2018delC; p.Gln675SerfsX30) in ALPK3 was identified and confirmed with Sanger sequencing. Heterozygous family members were normal with echocardiographic examination. To date, only two studies have reported homozygous pathogenic variants of ALPK3, with a total of seven affected individuals with cardiomyopathy from four unrelated consanguineous families. We include a discussion of the patient's phenotypic features and a review of relevant literature findings.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Muscle Proteins/genetics , Protein Kinases/genetics , Cardiomyopathies/genetics , Child, Preschool , Echocardiography , Exome , Frameshift Mutation , Genetic Predisposition to Disease , Genetic Testing , Heart/physiopathology , Homozygote , Humans , Male , Muscle Proteins/metabolism , Mutation , Pedigree , Protein Kinases/metabolism
8.
J Hepatol ; 67(1): 186-191, 2017 07.
Article in English | MEDLINE | ID: mdl-28323122

ABSTRACT

Hepatocellular adenoma (HCA) is a rare benign liver tumor, predominantly seen in young women. Its major complications are malignant transformation, spontaneous hemorrhage, and rupture. We describe a case of a young female with no underlying liver disease who presented with acute abdominal pain and was found to have a 17cm heterogeneous mass in the left lobe of the liver. She underwent left hepatectomy and pathology revealed a 14cm moderately differentiated hepatocellular carcinoma (HCC) arising in a shell of a HCA. At that time, vascular invasion was already present. She rapidly developed recurrent multifocal hepatic lesions and subsequent spread to the brain, leading to her death 18months after surgery. To investigate the underlying genetic events occurring during hepatocellular adenoma-carcinoma transition and extra-hepatic dissemination, we performed whole exome sequencing of DNA isolated from peripheral blood leucocytes, HCA, HCC, tumor thrombus and brain metastasis. Our data show a step-wise addition of somatic mutations and copy number variations with disease progression, suggesting a linear tumor evolution, which is supported by clonality analysis. Specifically, using a model based clustering of somatic mutations, one single founding clone arising in the HCA, which included catenin beta 1 (CTNNB1) and IL6ST driver mutations, was identified and displayed an increasing clonality rate in HCC, tumor thrombus and brain metastasis. Our data highlight the feasibility of performing whole exome capture, sequencing and analysis using formalin-fixed paraffin-embedded (FFPE) samples, and we describe the first genomic longitudinal study of hepatocellular adenoma-carcinoma transition, vascular invasion and brain metastasis with detailed clinicopathologic annotation.


Subject(s)
Adenoma/pathology , Carcinoma, Hepatocellular/pathology , Exome , Liver Neoplasms/pathology , Adult , Carcinoma, Hepatocellular/genetics , DNA Copy Number Variations , Female , Humans , Liver Neoplasms/genetics , Mutation , Neoplasm Invasiveness
9.
Genome Med ; 9(1): 12, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28153049

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) constitutes nearly half of all malignant brain tumors and has a median survival of 15 months. The standard treatment for these lesions includes maximal resection, radiotherapy, and chemotherapy; however, individual tumors display immense variability in their response to these approaches. Genomic techniques such as whole-exome sequencing (WES) provide an opportunity to understand the molecular basis of this variability. METHODS: Here, we report WES-guided treatment of a patient with a primary GBM and two subsequent recurrences, demonstrating the dynamic nature of treatment-induced molecular changes and their implications for clinical decision-making. We also analyze the Yale-Glioma cohort, composed of 110 whole exome- or whole genome-sequenced tumor-normal pairs, to assess the frequency of genomic events found in the presented case. RESULTS: Our longitudinal analysis revealed how the genomic profile evolved under the pressure of therapy. Specifically targeted approaches eradicated treatment-sensitive clones while enriching for resistant ones, generated due to chromothripsis, which we show to be a frequent event in GBMs based on our extended analysis of 110 gliomas in the Yale-Glioma cohort. Despite chromothripsis and the later acquired mismatch-repair deficiency, genomics-guided personalized treatment extended survival to over 5 years. Interestingly, the case displayed a favorable response to immune checkpoint inhibition after acquiring mismatch repair deficiency. CONCLUSIONS: Our study demonstrates the importance of longitudinal genomic profiling to adjust to the dynamic nature of treatment-induced molecular changes to improve the outcomes of precision therapies.


Subject(s)
Chromosome Aberrations , Genomics , Glioblastoma/therapy , Neoplasm Recurrence, Local , Precision Medicine , Antineoplastic Agents/therapeutic use , Combined Modality Therapy , DNA Mismatch Repair , DNA Mutational Analysis , DNA, Neoplasm , Disease Progression , Exome , Female , General Surgery , Genome, Human , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunotherapy , Longitudinal Studies , Middle Aged , Mutation , Radiotherapy , Treatment Outcome
10.
Nat Commun ; 8: 14433, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195122

ABSTRACT

Meningiomas are mostly benign brain tumours, with a potential for becoming atypical or malignant. On the basis of comprehensive genomic, transcriptomic and epigenomic analyses, we compared benign meningiomas to atypical ones. Here, we show that the majority of primary (de novo) atypical meningiomas display loss of NF2, which co-occurs either with genomic instability or recurrent SMARCB1 mutations. These tumours harbour increased H3K27me3 signal and a hypermethylated phenotype, mainly occupying the polycomb repressive complex 2 (PRC2) binding sites in human embryonic stem cells, thereby phenocopying a more primitive cellular state. Consistent with this observation, atypical meningiomas exhibit upregulation of EZH2, the catalytic subunit of the PRC2 complex, as well as the E2F2 and FOXM1 transcriptional networks. Importantly, these primary atypical meningiomas do not harbour TERT promoter mutations, which have been reported in atypical tumours that progressed from benign ones. Our results establish the genomic landscape of primary atypical meningiomas and potential therapeutic targets.


Subject(s)
Gene Regulatory Networks/genetics , Gene Regulatory Networks/physiology , Genome , Genomics/methods , Meningeal Neoplasms/genetics , Meningeal Neoplasms/metabolism , Meningioma/genetics , Meningioma/metabolism , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Chromosomal Instability , Cluster Analysis , DNA Methylation , E2F2 Transcription Factor/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenomics/methods , Exome/genetics , Forkhead Box Protein M1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Neurofibromatosis 2 , Genotyping Techniques , Human Embryonic Stem Cells/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Molecular Probe Techniques , Mutation , Phenotype , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , SMARCB1 Protein/genetics , Sequence Analysis , Signal Transduction/genetics , Transcriptome
11.
Nat Genet ; 48(10): 1253-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27548314

ABSTRACT

RNA polymerase II mediates the transcription of all protein-coding genes in eukaryotic cells, a process that is fundamental to life. Genomic mutations altering this enzyme have not previously been linked to any pathology in humans, which is a testament to its indispensable role in cell biology. On the basis of a combination of next-generation genomic analyses of 775 meningiomas, we report that recurrent somatic p.Gln403Lys or p.Leu438_His439del mutations in POLR2A, which encodes the catalytic subunit of RNA polymerase II (ref. 1), hijack this essential enzyme and drive neoplasia. POLR2A mutant tumors show dysregulation of key meningeal identity genes, including WNT6 and ZIC1/ZIC4. In addition to mutations in POLR2A, NF2, SMARCB1, TRAF7, KLF4, AKT1, PIK3CA, and SMO, we also report somatic mutations in AKT3, PIK3R1, PRKAR1A, and SUFU in meningiomas. Our results identify a role for essential transcriptional machinery in driving tumorigenesis and define mutually exclusive meningioma subgroups with distinct clinical and pathological features.


Subject(s)
Meningeal Neoplasms/genetics , Meningioma/genetics , Mutation , RNA Polymerase II/genetics , Catalytic Domain/genetics , Chromosomes, Human, Pair 22 , Cohort Studies , DNA Mutational Analysis , Enhancer Elements, Genetic , Exome , Gene Expression Regulation, Neoplastic , Genotype , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Meningeal Neoplasms/classification , Meningioma/classification , Neurofibromin 2/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics
12.
Am J Hum Genet ; 99(2): 501-10, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27453578

ABSTRACT

Cell division terminates with cytokinesis and cellular separation. Autosomal-recessive primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by a reduction in brain and head size at birth in addition to non-progressive intellectual disability. MCPH is genetically heterogeneous, and 16 loci are known to be associated with loss-of-function mutations predominantly affecting centrosomal-associated proteins, but the multiple roles of centrosomes in cellular function has left questions about etiology. Here, we identified three families affected by homozygous missense mutations in CIT, encoding citron rho-interacting kinase (CIT), which has established roles in cytokinesis. All mutations caused substitution of conserved amino acid residues in the kinase domain and impaired kinase activity. Neural progenitors that were differentiated from induced pluripotent stem cells (iPSCs) derived from individuals with these mutations exhibited abnormal cytokinesis with delayed mitosis, multipolar spindles, and increased apoptosis, rescued by CRISPR/Cas9 genome editing. Our results highlight the importance of cytokinesis in the pathology of primary microcephaly.


Subject(s)
Alleles , Cytokinesis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microcephaly/genetics , Microcephaly/pathology , Mitosis/genetics , Mutation, Missense/genetics , Protein Serine-Threonine Kinases/genetics , Apoptosis/genetics , Centrosome/metabolism , Child , Child, Preschool , Female , Genes, Recessive , Humans , Infant, Newborn , Male , Pedigree
13.
J Hum Genet ; 61(5): 395-403, 2016 May.
Article in English | MEDLINE | ID: mdl-26740239

ABSTRACT

The fat mass and obesity associated (FTO) gene has previously been associated with a variety of diseases and conditions, notably obesity, acute coronary syndrome and metabolic syndrome. Reports describing mutations in FTO as well as in FTO animal models have further demonstrated a role for FTO in the development of the brain and other organs. Here, we describe a patient born of consanguineous union who presented with microcephaly, developmental delay, behavioral abnormalities, dysmorphic facial features, hypotonia and other various phenotypic abnormalities. Whole-exome sequencing revealed a novel homozygous missense mutation in FTO and a nonsense mutation in the cholesteryl ester transfer protein (CETP). Exome copy number variation analysis revealed no disease-causing large duplications or deletions within coding regions. Patient's, her parents' and non-related control' fibroblasts were analyzed for morphologic defects, abnormal proliferation, apoptosis and transcriptome profile. We have shown that FTO is located in the nucleus of cells from each tested sample. Western blot analysis demonstrated no changes in patient FTO. Quantitative (qPCR) analysis revealed slightly decreased levels of FTO expression in patient cells compared with controls. No morphological or proliferation differences between the patient and control fibroblasts were observed. There is still much to be learned about the molecular mechanisms by which mutations in FTO contribute to such severe phenotypes.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Cholesterol Ester Transfer Proteins/genetics , Genetic Association Studies , Homozygote , Mutation, Missense , Apoptosis/genetics , Biopsy , Child, Preschool , Computational Biology/methods , Consanguinity , DNA Copy Number Variations , DNA Mutational Analysis , Exome , Female , Gene Expression , Gene Expression Profiling , Genotype , High-Throughput Nucleotide Sequencing , Humans , Phenotype , Transcriptome
14.
Nat Genet ; 48(1): 59-66, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26618343

ABSTRACT

Gliomas represent approximately 30% of all central nervous system tumors and 80% of malignant brain tumors. To understand the molecular mechanisms underlying the malignant progression of low-grade gliomas with mutations in IDH1 (encoding isocitrate dehydrogenase 1), we studied paired tumor samples from 41 patients, comparing higher-grade, progressed samples to their lower-grade counterparts. Integrated genomic analyses, including whole-exome sequencing and copy number, gene expression and DNA methylation profiling, demonstrated nonlinear clonal expansion of the original tumors and identified oncogenic pathways driving progression. These include activation of the MYC and RTK-RAS-PI3K pathways and upregulation of the FOXM1- and E2F2-mediated cell cycle transitions, as well as epigenetic silencing of developmental transcription factor genes bound by Polycomb repressive complex 2 in human embryonic stem cells. Our results not only provide mechanistic insight into the genetic and epigenetic mechanisms driving glioma progression but also identify inhibition of the bromodomain and extraterminal (BET) family as a potential therapeutic approach.


Subject(s)
Central Nervous System Neoplasms/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Central Nervous System Neoplasms/pathology , DNA Methylation , Embryonic Stem Cells/metabolism , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Dosage , Gene Expression Regulation, Neoplastic , Genes, myc , Glioma/pathology , Humans , Isocitrate Dehydrogenase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
15.
Genes Chromosomes Cancer ; 54(9): 542-54, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26032282

ABSTRACT

As subsets of pheochromocytomas (PCCs) lack a defined molecular etiology, we sought to characterize the mutational landscape of PCCs to identify novel gene candidates involved in disease development. A discovery cohort of 15 PCCs wild type for mutations in PCC susceptibility genes underwent whole-exome sequencing, and an additional 83 PCCs served as a verification cohort for targeted sequencing of candidate mutations. A low rate of nonsilent single nucleotide variants (SNVs) was detected (6.1/sample). Somatic HRAS and EPAS1 mutations were observed in one case each, whereas the remaining 13 cases did not exhibit variants in established PCC genes. SNVs aggregated in apoptosis-related pathways, and mutations in COSMIC genes not previously reported in PCCs included ZAN, MITF, WDTC1, and CAMTA1. Two somatic mutations and one constitutional variant in the well-established cancer gene lysine (K)-specific methyltransferase 2D (KMT2D, MLL2) were discovered in one sample each, prompting KMT2D screening using focused exome-sequencing in the verification cohort. An additional 11 PCCs displayed KMT2D variants, of which two were recurrent. In total, missense KMT2D variants were found in 14 (11 somatic, two constitutional, one undetermined) of 99 PCCs (14%). Five cases displayed somatic mutations in the functional FYR/SET domains of KMT2D, constituting 36% of all KMT2D-mutated PCCs. KMT2D expression was upregulated in PCCs compared to normal adrenals, and KMT2D overexpression positively affected cell migration in a PCC cell line. We conclude that KMT2D represents a recurrently mutated gene with potential implication for PCC development.


Subject(s)
Adrenal Gland Neoplasms/genetics , DNA-Binding Proteins/genetics , Exome , Mutation , Neoplasm Proteins/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/etiology , Cell Line, Tumor , Cohort Studies , DNA-Binding Proteins/metabolism , Female , Gene Dosage , Humans , Male , Neoplasm Proteins/metabolism , Pheochromocytoma/etiology , Pheochromocytoma/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Analysis, Protein , Transcriptome
16.
Neuro Oncol ; 17(10): 1356-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25740784

ABSTRACT

BACKGROUND: Malignant high-grade gliomas (HGGs), including the most aggressive form, glioblastoma multiforme, show significant clinical and genomic heterogeneity. Despite recent advances, the overall survival of HGGs and their response to treatment remain poor. In order to gain further insight into disease pathophysiology by correlating genomic landscape with clinical behavior, thereby identifying distinct HGG molecular subgroups associated with improved prognosis, we performed a comprehensive genomic analysis. METHODS: We analyzed and compared 720 exome-sequenced gliomas (136 from Yale, 584 from The Cancer Genome Atlas) based on their genomic, histological, and clinical features. RESULTS: We identified a subgroup of HGGs (6 total, 4 adults and 2 children) that harbored a statistically significantly increased number of somatic mutations (mean = 9257.3 vs 76.2, P = .002). All of these "ultramutated" tumors harbored somatic mutations in the exonuclease domain of the polymerase epsilon gene (POLE), displaying a distinctive genetic profile, characterized by genomic stability and increased C-to-A transversions. Histologically, they all harbored multinucleated giant or bizarre cells, some with predominant infiltrating immune cells. One adult and both pediatric patients carried homozygous germline mutations in the mutS homolog 6 (MSH6) gene. In adults, POLE mutations were observed in patients younger than 40 years and were associated with a longer progression-free survival. CONCLUSIONS: We identified a genomically, histologically, and clinically distinct subgroup of HGGs that harbored somatic POLE mutations and carried an improved prognosis. Identification of distinctive molecular and pathological HGG phenotypes has implications not only for improved classification but also for potential targeted treatments.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Polymerase II/genetics , Glioma/genetics , Glioma/pathology , Mutation , Adult , Brain Neoplasms/classification , Brain Neoplasms/diagnosis , Child , Child, Preschool , DNA Copy Number Variations , DNA Mutational Analysis , Disease-Free Survival , Glioma/classification , Glioma/diagnosis , Humans , Phenotype , Poly-ADP-Ribose Binding Proteins , Prognosis , Young Adult
18.
Eur J Med Genet ; 58(1): 39-43, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25220016

ABSTRACT

N-glycanase 1 (NGLY1) is a conserved enzyme that is responsible for the deglycosylation of misfolded N-glycosylated proteins in the cytoplasm prior to their proteasome-mediated degradation. Disruption of this degradation process has been associated with various neurologic diseases including amyotrophic lateral sclerosis and Parkinson's disease. Here, we describe two siblings with neuromotor impairment, apparent intellectual disability, corneal opacities, and neuropathy who were found to possess a novel homozygous frame-shift mutation due to a 4 base pair deletion in NGLY1 (c.1533_1536delTCAA, p.Asn511LysfsX51). We hypothesize that this mutation likely limits the capability of neuronal cells to respond to stress due to accumulation of misfolded proteins, thereby impairing their survival and resulting in progressive loss of neurological function.


Subject(s)
Developmental Disabilities/genetics , Intellectual Disability/genetics , Movement Disorders/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peripheral Nervous System Diseases/genetics , Abnormalities, Multiple/genetics , Adolescent , Child , Corneal Opacity/genetics , Female , Frameshift Mutation , Genotype , Humans , Male
19.
Neuron ; 84(6): 1226-39, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25521378

ABSTRACT

Exome sequencing analysis of over 2,000 children with complex malformations of cortical development identified five independent (four homozygous and one compound heterozygous) deleterious mutations in KATNB1, encoding the regulatory subunit of the microtubule-severing enzyme Katanin. Mitotic spindle formation is defective in patient-derived fibroblasts, a consequence of disrupted interactions of mutant KATNB1 with KATNA1, the catalytic subunit of Katanin, and other microtubule-associated proteins. Loss of KATNB1 orthologs in zebrafish (katnb1) and flies (kat80) results in microcephaly, recapitulating the human phenotype. In the developing Drosophila optic lobe, kat80 loss specifically affects the asymmetrically dividing neuroblasts, which display supernumerary centrosomes and spindle abnormalities during mitosis, leading to cell cycle progression delays and reduced cell numbers. Furthermore, kat80 depletion results in dendritic arborization defects in sensory and motor neurons, affecting neural architecture. Taken together, we provide insight into the mechanisms by which KATNB1 mutations cause human cerebral cortical malformations, demonstrating its fundamental role during brain development.


Subject(s)
Adenosine Triphosphatases/genetics , Brain/abnormalities , Brain/pathology , Microcephaly/genetics , Neural Stem Cells/pathology , Neurogenesis/genetics , Optic Lobe, Nonmammalian/abnormalities , Animals , Brain/growth & development , Cell Count , Cell Division/genetics , Dendrites/genetics , Drosophila , Drosophila Proteins/genetics , Humans , Katanin , Mice , Microcephaly/pathology , Microtubule-Associated Proteins/genetics , Mutation , Spindle Apparatus/genetics , Zebrafish
20.
Pediatr Neurol ; 51(6): 806-813.e8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456301

ABSTRACT

BACKGROUND: Knobloch syndrome is a rare, autosomal recessive, developmental disorder characterized by stereotyped ocular abnormalities with or without occipital skull deformities (encephalocele, bone defects, and cutis aplasia). Although there is clear heterogeneity in clinical presentation, central nervous system malformations, aside from the characteristic encephalocele, have not typically been considered a component of the disease phenotype. METHODS: Four patients originally presented for genetic evaluation of symptomatic structural brain malformations. Whole-genome genotyping, whole-exome sequencing, and confirmatory Sanger sequencing were performed. Using immunohistochemical analysis, we investigated the protein expression pattern of COL18A1 in the mid-fetal and adult human cerebral cortex and then analyzed the spatial and temporal changes in the expression pattern of COL18A1 during human cortical development using the Human Brain Transcriptome database. RESULTS: We identified two novel homozygous deleterious frame-shift mutations in the COL18A1 gene. On further investigation of these patients and their families, we found that many exhibited certain characteristics of Knobloch syndrome, including pronounced ocular defects. Our data strongly support an important role for COL18A1 in brain development, and this report contributes to an enhanced characterization of the brain malformations that can result from deficiencies of collagen XVIII. CONCLUSIONS: This case series highlights the diagnostic power and clinical utility of whole-exome sequencing technology-allowing clinicians and physician scientists to better understand the pathophysiology and presentations of rare diseases. We suggest that patients who are clinically diagnosed with Knobloch syndrome and/or found to have COL18A1 mutations via genetic screening should be investigated for potential structural brain abnormalities even in the absence of an encephalocele.


Subject(s)
Cerebral Cortex/metabolism , Collagen Type XVIII/genetics , Encephalocele/genetics , Encephalocele/pathology , Exome/genetics , Retinal Detachment/congenital , Adolescent , Adult , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Consanguinity , Female , Fetus , Humans , Male , Mutation , Retinal Degeneration , Retinal Detachment/genetics , Retinal Detachment/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL