Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cytotherapy ; 26(1): 88-95, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37747395

ABSTRACT

BACKGROUND AND AIMS: As cell and gene therapy (CGT) has grown in availability and scope, more unapproved regenerative medicine is being marketed to the public. It is essential that health care providers have sufficient knowledge and comfort to determine whether treatments are properly regulated and address these topics with patients. Due to the applicability of CGT to genetic disease, genetic counselors could be key in providing education and answering patients' questions about these topics. However, previous studies have focused only on physicians' knowledge and comfort with CGT and unapproved regenerative medicine. The purpose of this study was to assess genetic counselors' self-reported knowledge and comfort discussing these topics with patients and to explore what factors predict increased knowledge and comfort. METHODS: The authors designed an online survey distributed to genetic counselors who were part of the National Society of Genetic Counselors Student Research Program e-mail list. The survey addressed genetic counselors' demographics, practice experience with CGT, education about CGT, knowledge and comfort. RESULTS: The survey was completed by 144 genetic counselors. The best predictor of increased knowledge and comfort was experience discussing CGT in practice. In addition, those who worked at an institution at which CGT trials were offered had greater knowledge and comfort. However, most genetic counselors reported their knowledge was not sufficient to address questions from patients, and most had little-to-no knowledge or comfort determining whether a trial was properly regulated. There was no correlation between education and either knowledge or comfort; however, most participants desired more education about these topics. CONCLUSIONS: This study suggests that genetic counselors who (i) have experience with CGT in practice or (ii) work at institutions at which CGT trials are offered may have better knowledge regarding CGT. These results may help identify individuals and/or institutions in whom increasing knowledge regarding CGT could be beneficial. This is crucial as CGT becomes mainstream, leading to more widely marketed unapproved regenerative medicine. Several gaps in knowledge and comfort were identified, including participants' ability to determine whether a treatment is properly regulated. Further research is needed to better characterize the educational needs of genetic counselors surrounding these topics to address these gaps.


Subject(s)
Counselors , Genetic Counseling , Humans , Genetic Counseling/methods , Surveys and Questionnaires , Cell- and Tissue-Based Therapy , Regenerative Medicine
2.
Am J Hum Genet ; 106(1): 121-128, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31883643

ABSTRACT

In two independent ongoing next-generation sequencing projects for individuals with holoprosencephaly and individuals with disorders of sex development, and through international research collaboration, we identified twelve individuals with de novo loss-of-function (LoF) variants in protein phosphatase 1, regulatory subunit 12a (PPP1R12A), an important developmental gene involved in cell migration, adhesion, and morphogenesis. This gene has not been previously reported in association with human disease, and it has intolerance to LoF as illustrated by a very low observed-to-expected ratio of LoF variants in gnomAD. Of the twelve individuals, midline brain malformations were found in five, urogenital anomalies in nine, and a combination of both phenotypes in two. Other congenital anomalies identified included omphalocele, jejunal, and ileal atresia with aberrant mesenteric blood supply, and syndactyly. Six individuals had stop gain variants, five had a deletion or duplication resulting in a frameshift, and one had a canonical splice acceptor site loss. Murine and human in situ hybridization and immunostaining revealed PPP1R12A expression in the prosencephalic neural folds and protein localization in the lower urinary tract at critical periods for forebrain division and urogenital development. Based on these clinical and molecular findings, we propose the association of PPP1R12A pathogenic variants with a congenital malformations syndrome affecting the embryogenesis of the brain and genitourinary systems and including disorders of sex development.


Subject(s)
Abnormalities, Multiple/pathology , Disorders of Sex Development/pathology , Holoprosencephaly/pathology , Mutation , Myosin-Light-Chain Phosphatase/genetics , Urogenital Abnormalities/pathology , Abnormalities, Multiple/genetics , Adolescent , Child , Child, Preschool , Disorders of Sex Development/genetics , Female , Gestational Age , Holoprosencephaly/genetics , Humans , Male , Phenotype , Pregnancy , Urogenital Abnormalities/genetics
3.
Am J Hum Genet ; 107(3): 499-513, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32721402

ABSTRACT

Signal transduction through the RAF-MEK-ERK pathway, the first described mitogen-associated protein kinase (MAPK) cascade, mediates multiple cellular processes and participates in early and late developmental programs. Aberrant signaling through this cascade contributes to oncogenesis and underlies the RASopathies, a family of cancer-prone disorders. Here, we report that de novo missense variants in MAPK1, encoding the mitogen-activated protein kinase 1 (i.e., extracellular signal-regulated protein kinase 2, ERK2), cause a neurodevelopmental disease within the RASopathy phenotypic spectrum, reminiscent of Noonan syndrome in some subjects. Pathogenic variants promote increased phosphorylation of the kinase, which enhances translocation to the nucleus and boosts MAPK signaling in vitro and in vivo. Two variant classes are identified, one of which directly disrupts binding to MKP3, a dual-specificity protein phosphatase negatively regulating ERK function. Importantly, signal dysregulation driven by pathogenic MAPK1 variants is stimulus reliant and retains dependence on MEK activity. Our data support a model in which the identified pathogenic variants operate with counteracting effects on MAPK1 function by differentially impacting the ability of the kinase to interact with regulators and substrates, which likely explains the minor role of these variants as driver events contributing to oncogenesis. After nearly 20 years from the discovery of the first gene implicated in Noonan syndrome, PTPN11, the last tier of the MAPK cascade joins the group of genes mutated in RASopathies.


Subject(s)
Carcinogenesis/genetics , Mitogen-Activated Protein Kinase 1/genetics , Neurodevelopmental Disorders/genetics , Noonan Syndrome/genetics , Child, Preschool , Female , Humans , MAP Kinase Signaling System/genetics , Male , Mutation, Missense/genetics , Neurodevelopmental Disorders/pathology , Noonan Syndrome/physiopathology , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction , Exome Sequencing , ras Proteins/genetics
4.
Semin Neurol ; 42(6): 716-722, 2022 12.
Article in English | MEDLINE | ID: mdl-36417990

ABSTRACT

The diagnosis of neuromuscular disorders requires a thorough history including family history and examination, with the next steps broadened now beyond electromyography and neuropathology to include genetic testing. The challenge in diagnosis can often be putting all the information together. With advances in genetic testing, some diagnoses that adult patients may have received as children deserve a second look and may result in diagnoses better defined or alternative diagnoses made. Clearly defining or redefining a diagnosis can result in understanding of potential other systems involved, prognosis, or potential treatments. This article presents several cases and approach to diagnosis as well as potential treatment and prognostic concerns, including seipinopathy, congenital myasthenic syndrome, central core myopathy, and myotonic dystrophy type 2.


Subject(s)
Myotonic Dystrophy , Neuromuscular Diseases , Child , Adult , Humans , Neuromuscular Diseases/diagnosis , Neuromuscular Diseases/therapy , Neuromuscular Diseases/genetics , Electromyography , Genetic Testing , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy
5.
Am J Hum Genet ; 102(2): 309-320, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29394990

ABSTRACT

Exome sequencing has markedly enhanced the discovery of genes implicated in Mendelian disorders, particularly for individuals in whom a known clinical entity could not be assigned. This has led to the recognition that phenotypic heterogeneity resulting from allelic mutations occurs more commonly than previously appreciated. Here, we report that missense variants in CDC42, a gene encoding a small GTPase functioning as an intracellular signaling node, underlie a clinically heterogeneous group of phenotypes characterized by variable growth dysregulation, facial dysmorphism, and neurodevelopmental, immunological, and hematological anomalies, including a phenotype resembling Noonan syndrome, a developmental disorder caused by dysregulated RAS signaling. In silico, in vitro, and in vivo analyses demonstrate that mutations variably perturb CDC42 function by altering the switch between the active and inactive states of the GTPase and/or affecting CDC42 interaction with effectors, and differentially disturb cellular and developmental processes. These findings reveal the remarkably variable impact that dominantly acting CDC42 mutations have on cell function and development, creating challenges in syndrome definition, and exemplify the importance of functional profiling for syndrome recognition and delineation.


Subject(s)
Abnormalities, Multiple/genetics , Craniofacial Abnormalities/genetics , Genetic Heterogeneity , Muscular Atrophy/genetics , Mutation, Missense , Neurodevelopmental Disorders/genetics , Noonan Syndrome/genetics , cdc42 GTP-Binding Protein/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Adolescent , Adult , Child , Child, Preschool , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Female , Gene Expression , Humans , Infant , Male , Models, Molecular , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Noonan Syndrome/metabolism , Noonan Syndrome/pathology , Phenotype , Protein Structure, Secondary , Severity of Illness Index , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism
6.
Am J Hum Genet ; 102(6): 1195-1203, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861108

ABSTRACT

Next-generation sequencing is a powerful tool for the discovery of genes related to neurodevelopmental disorders (NDDs). Here, we report the identification of a distinct syndrome due to de novo or inherited heterozygous mutations in Tousled-like kinase 2 (TLK2) in 38 unrelated individuals and two affected mothers, using whole-exome and whole-genome sequencing technologies, matchmaker databases, and international collaborations. Affected individuals had a consistent phenotype, characterized by mild-borderline neurodevelopmental delay (86%), behavioral disorders (68%), severe gastro-intestinal problems (63%), and facial dysmorphism including blepharophimosis (82%), telecanthus (74%), prominent nasal bridge (68%), broad nasal tip (66%), thin vermilion of the upper lip (62%), and upslanting palpebral fissures (55%). Analysis of cell lines from three affected individuals showed that mutations act through a loss-of-function mechanism in at least two case subjects. Genotype-phenotype analysis and comparison of computationally modeled faces showed that phenotypes of these and other individuals with loss-of-function variants significantly overlapped with phenotypes of individuals with other variant types (missense and C-terminal truncating). This suggests that haploinsufficiency of TLK2 is the most likely underlying disease mechanism, leading to a consistent neurodevelopmental phenotype. This work illustrates the power of international data sharing, by the identification of 40 individuals from 26 different centers in 7 different countries, allowing the identification, clinical delineation, and genotype-phenotype evaluation of a distinct NDD caused by mutations in TLK2.


Subject(s)
Genetic Association Studies , Inheritance Patterns/genetics , Loss of Function Mutation/genetics , Neurodevelopmental Disorders/genetics , Protein Kinases/genetics , Adolescent , Adult , Base Sequence , Cell Line , Child , Child, Preschool , Facies , Female , Humans , Infant , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Translocation, Genetic , Young Adult
7.
Genet Med ; 23(6): 1028-1040, 2021 06.
Article in English | MEDLINE | ID: mdl-33658631

ABSTRACT

PURPOSE: We describe a novel neurobehavioral phenotype of autism spectrum disorder (ASD), intellectual disability, and/or attention-deficit/hyperactivity disorder (ADHD) associated with de novo or inherited deleterious variants in members of the RFX family of genes. RFX genes are evolutionarily conserved transcription factors that act as master regulators of central nervous system development and ciliogenesis. METHODS: We assembled a cohort of 38 individuals (from 33 unrelated families) with de novo variants in RFX3, RFX4, and RFX7. We describe their common clinical phenotypes and present bioinformatic analyses of expression patterns and downstream targets of these genes as they relate to other neurodevelopmental risk genes. RESULTS: These individuals share neurobehavioral features including ASD, intellectual disability, and/or ADHD; other frequent features include hypersensitivity to sensory stimuli and sleep problems. RFX3, RFX4, and RFX7 are strongly expressed in developing and adult human brain, and X-box binding motifs as well as RFX ChIP-seq peaks are enriched in the cis-regulatory regions of known ASD risk genes. CONCLUSION: These results establish a likely role of deleterious variation in RFX3, RFX4, and RFX7 in cases of monogenic intellectual disability, ADHD and ASD, and position these genes as potentially critical transcriptional regulators of neurobiological pathways associated with neurodevelopmental disease pathogenesis.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Autistic Disorder , Intellectual Disability , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Humans , Intellectual Disability/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics
8.
Am J Hum Genet ; 101(2): 267-273, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28777933

ABSTRACT

Ribosomal RNA (rRNA) is transcribed from rDNA by RNA polymerase I (Pol I) to produce the 45S precursor of the 28S, 5.8S, and 18S rRNA components of the ribosome. Two transcription factors have been defined for Pol I in mammals, the selectivity factor SL1, and the upstream binding transcription factor (UBF), which interacts with the upstream control element to facilitate the assembly of the transcription initiation complex including SL1 and Pol I. In seven unrelated affected individuals, all suffering from developmental regression starting at 2.5-7 years, we identified a heterozygous variant, c.628G>A in UBTF, encoding p.Glu210Lys in UBF, which occurred de novo in all cases. While the levels of UBF, Ser388 phosphorylated UBF, and other Pol I-related components (POLR1E, TAF1A, and TAF1C) remained unchanged in cells of an affected individual, the variant conferred gain of function to UBF, manifesting by markedly increased UBF binding to the rDNA promoter and to the 5'- external transcribed spacer. This was associated with significantly increased 18S expression, and enlarged nucleoli which were reduced in number per cell. The data link neurodegeneration in childhood with altered rDNA chromatin status and rRNA metabolism.


Subject(s)
Brain Diseases/genetics , Cell Nucleolus/pathology , Neurodegenerative Diseases/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , RNA, Ribosomal, 18S/biosynthesis , Adolescent , Adult , Atrophy/genetics , Brain/pathology , Brain Diseases/pathology , Child , Chromatin/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Male , Neurodegenerative Diseases/pathology , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Young Adult
9.
Am J Hum Genet ; 101(1): 139-148, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28686853

ABSTRACT

We report 15 individuals with de novo pathogenic variants in WDR26. Eleven of the individuals carry loss-of-function mutations, and four harbor missense substitutions. These 15 individuals comprise ten females and five males, and all have intellectual disability with delayed speech, a history of febrile and/or non-febrile seizures, and a wide-based, spastic, and/or stiff-legged gait. These subjects share a set of common facial features that include a prominent maxilla and upper lip that readily reveal the upper gingiva, widely spaced teeth, and a broad nasal tip. Together, these features comprise a recognizable facial phenotype. We compared these features with those of chromosome 1q41q42 microdeletion syndrome, which typically contains WDR26, and noted that clinical features are consistent between the two subsets, suggesting that haploinsufficiency of WDR26 contributes to the pathology of 1q41q42 microdeletion syndrome. Consistent with this, WDR26 loss-of-function single-nucleotide mutations identified in these subjects lead to nonsense-mediated decay with subsequent reduction of RNA expression and protein levels. We derived a structural model of WDR26 and note that missense variants identified in these individuals localize to highly conserved residues of this WD-40-repeat-containing protein. Given that WDR26 mutations have been identified in ∼1 in 2,000 of subjects in our clinical cohorts and that WDR26 might be poorly annotated in exome variant-interpretation pipelines, we would anticipate that this disorder could be more common than currently appreciated.


Subject(s)
Facies , Gait/genetics , Haploinsufficiency/genetics , Intellectual Disability/genetics , Proteins/genetics , Seizures/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Base Sequence , Child, Preschool , Chromosome Deletion , Female , Growth and Development/genetics , Humans , Intellectual Disability/complications , Male , Mutation/genetics , Proteins/chemistry , RNA Stability/genetics , Seizures/complications , Syndrome
10.
Genet Med ; 22(3): 538-546, 2020 03.
Article in English | MEDLINE | ID: mdl-31723249

ABSTRACT

PURPOSE: Intellectual disability (ID) and autism spectrum disorder (ASD) are genetically heterogeneous neurodevelopmental disorders. We sought to delineate the clinical, molecular, and neuroimaging spectrum of a novel neurodevelopmental disorder caused by variants in the zinc finger protein 292 gene (ZNF292). METHODS: We ascertained a cohort of 28 families with ID due to putatively pathogenic ZNF292 variants that were identified via targeted and exome sequencing. Available data were analyzed to characterize the canonical phenotype and examine genotype-phenotype relationships. RESULTS: Probands presented with ID as well as a spectrum of neurodevelopmental features including ASD, among others. All ZNF292 variants were de novo, except in one family with dominant inheritance. ZNF292 encodes a highly conserved zinc finger protein that acts as a transcription factor and is highly expressed in the developing human brain supporting its critical role in neurodevelopment. CONCLUSION: De novo and dominantly inherited variants in ZNF292 are associated with a range of neurodevelopmental features including ID and ASD. The clinical spectrum is broad, and most individuals present with mild to moderate ID with or without other syndromic features. Our results suggest that variants in ZNF292 are likely a recurrent cause of a neurodevelopmental disorder manifesting as ID with or without ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Carrier Proteins/genetics , Genetic Predisposition to Disease , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Adolescent , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/pathology , Neuroimaging/methods , Exome Sequencing/methods
11.
Am J Med Genet A ; 182(1): 38-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31782611

ABSTRACT

White-Sutton syndrome (WHSUS) is a recently-identified genetic disorder resulting from de novo heterozygous pathogenic variants in POGZ. Thus far, over 50 individuals have been reported worldwide, however phenotypic characterization and data regarding the natural history are still incomplete. Here we report the clinical features of 22 individuals with 21 unique loss of function POGZ variants. We observed a broad spectrum of intellectual disability and/or developmental delay with or without autism, and speech delay in all individuals. Other common problems included ocular abnormalities, hearing loss and gait abnormalities. A validated sleep disordered breathing questionnaire identified symptoms of obstructive sleep apnea in 4/12 (33%) individuals. A higher-than-expected proportion of cases also had gastrointestinal phenotypes, both functional and anatomical, as well as genitourinary anomalies. In line with previous publications, we observed an increased body mass index (BMI) z-score compared to the general population (mean 0.59, median 0.9; p 0.0253). Common facial features included microcephaly, broad forehead, midface hypoplasia, triangular mouth, broad nasal root and flat nasal bridge. Analysis of the Baylor Genetics clinical laboratory database revealed that POGZ variants were implicated in approximately 0.14% of cases who underwent clinical exome sequencing for neurological indications with or without involvement of other body systems. This study describes a greater allelic series and expands the phenotypic spectrum of this new syndromic form of intellectual disability and autism.


Subject(s)
Autistic Disorder/genetics , Intellectual Disability/genetics , Language Development Disorders/genetics , Transposases/genetics , Adolescent , Adult , Autistic Disorder/pathology , Child , Child, Preschool , Exome/genetics , Female , Heterozygote , Humans , Infant , Intellectual Disability/epidemiology , Intellectual Disability/pathology , Language Development Disorders/pathology , Male , Microcephaly/genetics , Microcephaly/pathology , Middle Aged , Mutation/genetics , Phenotype , Exome Sequencing , Young Adult
12.
Brain ; 142(10): 2948-2964, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31501903

ABSTRACT

Axon pathfinding and synapse formation are essential processes for nervous system development and function. The assembly of myelinated fibres and nodes of Ranvier is mediated by a number of cell adhesion molecules of the immunoglobulin superfamily including neurofascin, encoded by the NFASC gene, and its alternative isoforms Nfasc186 and Nfasc140 (located in the axonal membrane at the node of Ranvier) and Nfasc155 (a glial component of the paranodal axoglial junction). We identified 10 individuals from six unrelated families, exhibiting a neurodevelopmental disorder characterized with a spectrum of central (intellectual disability, developmental delay, motor impairment, speech difficulties) and peripheral (early onset demyelinating neuropathy) neurological involvement, who were found by exome or genome sequencing to carry one frameshift and four different homozygous non-synonymous variants in NFASC. Expression studies using immunostaining-based techniques identified absent expression of the Nfasc155 isoform as a consequence of the frameshift variant and a significant reduction of expression was also observed in association with two non-synonymous variants affecting the fibronectin type III domain. Cell aggregation studies revealed a severely impaired Nfasc155-CNTN1/CASPR1 complex interaction as a result of the identified variants. Immunofluorescence staining of myelinated fibres from two affected individuals showed a severe loss of myelinated fibres and abnormalities in the paranodal junction morphology. Our results establish that recessive variants affecting the Nfasc155 isoform can affect the formation of paranodal axoglial junctions at the nodes of Ranvier. The genetic disease caused by biallelic NFASC variants includes neurodevelopmental impairment and a spectrum of central and peripheral demyelination as part of its core clinical phenotype. Our findings support possible overlapping molecular mechanisms of paranodal damage at peripheral nerves in both the immune-mediated and the genetic disease, but the observation of prominent central neurological involvement in NFASC biallelic variant carriers highlights the importance of this gene in human brain development and function.


Subject(s)
Cell Adhesion Molecules/genetics , Demyelinating Diseases/genetics , Nerve Growth Factors/genetics , Neurodevelopmental Disorders/genetics , Adolescent , Adult , Alleles , Axons/metabolism , Cell Adhesion Molecules/metabolism , Child , Child, Preschool , Demyelinating Diseases/metabolism , Female , Gene Frequency/genetics , Humans , Infant , Male , Mutation , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/physiology , Nerve Growth Factors/metabolism , Nervous System Malformations , Neurodevelopmental Disorders/metabolism , Neuroglia/metabolism , Pedigree , Peripheral Nerves , Protein Isoforms/metabolism , Ranvier's Nodes/genetics , Ranvier's Nodes/metabolism
13.
Brain ; 142(9): 2617-2630, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31327001

ABSTRACT

The underpinnings of mild to moderate neurodevelopmental delay remain elusive, often leading to late diagnosis and interventions. Here, we present data on exome and genome sequencing as well as array analysis of 13 individuals that point to pathogenic, heterozygous, mostly de novo variants in WDFY3 (significant de novo enrichment P = 0.003) as a monogenic cause of mild and non-specific neurodevelopmental delay. Nine variants were protein-truncating and four missense. Overlapping symptoms included neurodevelopmental delay, intellectual disability, macrocephaly, and psychiatric disorders (autism spectrum disorders/attention deficit hyperactivity disorder). One proband presented with an opposing phenotype of microcephaly and the only missense-variant located in the PH-domain of WDFY3. Findings of this case are supported by previously published data, demonstrating that pathogenic PH-domain variants can lead to microcephaly via canonical Wnt-pathway upregulation. In a separate study, we reported that the autophagy scaffolding protein WDFY3 is required for cerebral cortical size regulation in mice, by controlling proper division of neural progenitors. Here, we show that proliferating cortical neural progenitors of human embryonic brains highly express WDFY3, further supporting a role for this molecule in the regulation of prenatal neurogenesis. We present data on Wnt-pathway dysregulation in Wdfy3-haploinsufficient mice, which display macrocephaly and deficits in motor coordination and associative learning, recapitulating the human phenotype. Consequently, we propose that in humans WDFY3 loss-of-function variants lead to macrocephaly via downregulation of the Wnt pathway. In summary, we present WDFY3 as a novel gene linked to mild to moderate neurodevelopmental delay and intellectual disability and conclude that variants putatively causing haploinsufficiency lead to macrocephaly, while an opposing pathomechanism due to variants in the PH-domain of WDFY3 leads to microcephaly.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy-Related Proteins/genetics , Brain/embryology , Brain/pathology , Genetic Variation/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Adaptor Proteins, Signal Transducing/chemistry , Adolescent , Animals , Autophagy-Related Proteins/chemistry , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Transgenic , Organ Size , Protein Structure, Secondary
14.
Hum Mutat ; 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31646703

ABSTRACT

We recently described a new neurodevelopmental syndrome (TAF1/MRXS33 intellectual disability syndrome) (MIM# 300966) caused by pathogenic variants involving the X-linked gene TAF1, which participates in RNA polymerase II transcription. The initial study reported eleven families, and the syndrome was defined as presenting early in life with hypotonia, facial dysmorphia, and developmental delay that evolved into intellectual disability (ID) and/or autism spectrum disorder (ASD). We have now identified an additional 27 families through a genotype-first approach. Familial segregation analysis, clinical phenotyping, and bioinformatics were capitalized on to assess potential variant pathogenicity, and molecular modelling was performed for those variants falling within structurally characterized domains of TAF1. A novel phenotypic clustering approach was also applied, in which the phenotypes of affected individuals were classified using 51 standardized Human Phenotype Ontology (HPO) terms. Phenotypes associated with TAF1 variants show considerable pleiotropy and clinical variability, but prominent among previously unreported effects were brain morphological abnormalities, seizures, hearing loss, and heart malformations. Our allelic series broadens the phenotypic spectrum of TAF1/MRXS33 intellectual disability syndrome and the range of TAF1 molecular defects in humans. It also illustrates the challenges for determining the pathogenicity of inherited missense variants, particularly for genes mapping to chromosome X. This article is protected by copyright. All rights reserved.

15.
Genet Med ; 21(9): 2036-2042, 2019 09.
Article in English | MEDLINE | ID: mdl-30739909

ABSTRACT

PURPOSE: To define the clinical characteristics of patients with variants in TCF20, we describe 27 patients, 26 of whom were identified via exome sequencing. We compare detailed clinical data with 17 previously reported patients. METHODS: Patients were ascertained through molecular testing laboratories performing exome sequencing (and other testing) with orthogonal confirmation; collaborating referring clinicians provided detailed clinical information. RESULTS: The cohort of 27 patients all had novel variants, and ranged in age from 2 to 68 years. All had developmental delay/intellectual disability. Autism spectrum disorders/autistic features were reported in 69%, attention disorders or hyperactivity in 67%, craniofacial features (no recognizable facial gestalt) in 67%, structural brain anomalies in 24%, and seizures in 12%. Additional features affecting various organ systems were described in 93%. In a majority of patients, we did not observe previously reported findings of postnatal overgrowth or craniosynostosis, in comparison with earlier reports. CONCLUSION: We provide valuable data regarding the prognosis and clinical manifestations of patients with variants in TCF20.


Subject(s)
Autism Spectrum Disorder/genetics , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Adolescent , Adult , Aged , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Exome/genetics , Female , Humans , Intellectual Disability/epidemiology , Intellectual Disability/pathology , Male , Middle Aged , Mutation , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/pathology , Exome Sequencing , Young Adult
16.
Am J Med Genet A ; 179(2): 300-305, 2019 02.
Article in English | MEDLINE | ID: mdl-30549396

ABSTRACT

Wiedemann-Steiner syndrome is a genetic condition associated with dysmorphic facies, hypertrichosis, short stature, developmental delay, and intellectual disability. Congenital malformations of the cerebral, cardiac, renal, and optic structures have also been reported. Because the majority of reported individuals with this condition have been under age 20, the long-term prognosis is not well defined. Here we report on two further unrelated individuals diagnosed with Wiedemann-Steiner syndrome, one of whom is in her third decade of life. In addition, both individuals have novel KMT2A mutations. The information provided below about the outcome in Wiedemann-Steiner syndrome is important for families of affected individuals.


Subject(s)
Abnormalities, Multiple/genetics , Contracture/genetics , Developmental Disabilities/genetics , Growth Disorders/genetics , Histone-Lysine N-Methyltransferase/genetics , Hypertrichosis/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , Contracture/physiopathology , Developmental Disabilities/physiopathology , Disease Progression , Facies , Female , Growth Disorders/physiopathology , Humans , Hypertrichosis/physiopathology , Infant , Intellectual Disability/physiopathology , Male , Microcephaly/physiopathology , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/physiopathology , Phenotype , Young Adult
18.
Genet Med ; 18(11): 1143-1150, 2016 11.
Article in English | MEDLINE | ID: mdl-26986877

ABSTRACT

PURPOSE: Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is an autosomal-dominant disorder characterized by optic atrophy and intellectual disability caused by loss-of-function mutations in NR2F1. We report 20 new individuals with BBSOAS, exploring the spectrum of clinical phenotypes and assessing potential genotype-phenotype correlations. METHODS: Clinical features of individuals with pathogenic NR2F1 variants were evaluated by review of medical records. The functional relevance of coding nonsynonymous NR2F1 variants was assessed with a luciferase assay measuring the impact on transcriptional activity. The effects of two start codon variants on protein expression were evaluated by western blot analysis. RESULTS: We recruited 20 individuals with novel pathogenic NR2F1 variants (seven missense variants, five translation initiation variants, two frameshifting insertions/deletions, one nonframeshifting insertion/deletion, and five whole-gene deletions). All the missense variants were found to impair transcriptional activity. In addition to visual and cognitive deficits, individuals with BBSOAS manifested hypotonia (75%), seizures (40%), autism spectrum disorder (35%), oromotor dysfunction (60%), thinning of the corpus callosum (53%), and hearing defects (20%). CONCLUSION: BBSOAS encompasses a broad range of clinical phenotypes. Functional studies help determine the severity of novel NR2F1 variants. Some genotype-phenotype correlations seem to exist, with missense mutations in the DNA-binding domain causing the most severe phenotypes.Genet Med 18 11, 1143-1150.


Subject(s)
Autism Spectrum Disorder/genetics , COUP Transcription Factor I/genetics , Genetic Association Studies , Optic Atrophy/genetics , Adolescent , Adult , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/physiopathology , Child , Child, Preschool , Female , Gene Deletion , Humans , Male , Mutation, Missense , Optic Atrophy/complications , Optic Atrophy/physiopathology , Pedigree
19.
Nat Commun ; 14(1): 6878, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898601

ABSTRACT

Wastewater is a discarded human by-product, but its analysis may help us understand the health of populations. Epidemiologists first analyzed wastewater to track outbreaks of poliovirus decades ago, but so-called wastewater-based epidemiology was reinvigorated to monitor SARS-CoV-2 levels while bypassing the difficulties and pit falls of individual testing. Current approaches overlook the activity of most human viruses and preclude a deeper understanding of human virome community dynamics. Here, we conduct a comprehensive sequencing-based analysis of 363 longitudinal wastewater samples from ten distinct sites in two major cities. Critical to detection is the use of a viral probe capture set targeting thousands of viral species or variants. Over 450 distinct pathogenic viruses from 28 viral families are observed, most of which have never been detected in such samples. Sequencing reads of established pathogens and emerging viruses correlate to clinical data sets of SARS-CoV-2, influenza virus, and monkeypox viruses, outlining the public health utility of this approach. Viral communities are tightly organized by space and time. Finally, the most abundant human viruses yield sequence variant information consistent with regional spread and evolution. We reveal the viral landscape of human wastewater and its potential to improve our understanding of outbreaks, transmission, and its effects on overall population health.


Subject(s)
Poliovirus , Virome , Humans , Virome/genetics , Wastewater , Cities , Disease Outbreaks , SARS-CoV-2/genetics
20.
Neurol Clin Pract ; 11(1): 69-77, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33968475

ABSTRACT

PURPOSE OF REVIEW: With the advent of precision medicine and demand for genomic testing information, we may question whether it is time to offer genetic testing to our patients with Parkinson disease (PD). This review updates the current genetic landscape of PD, describes what genetic testing may offer, provides strategies for evaluating whom to test, and provides resources for the busy clinician. RECENT FINDINGS: Patients with PD and their relatives, in various settings, have expressed an interest in learning their PD genetic status; however, physicians may be hesitant to widely offer testing due to the perceived low clinical utility of PD genetic test results. The rise of clinical trials available for patients with gene-specific PD and emerging information on genotype-phenotype correlations are starting to shift this discussion about testing. SUMMARY: By learning more about the various genetic testing options for PD and utility of results for patients and their care, clinicians may become more comfortable with widespread PD genetic testing in the research and clinical setting.

SELECTION OF CITATIONS
SEARCH DETAIL