Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Genet Med ; 25(7): 100861, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37087635

ABSTRACT

PURPOSE: This study aimed to establish variants in CBX1, encoding heterochromatin protein 1ß (HP1ß), as a cause of a novel syndromic neurodevelopmental disorder. METHODS: Patients with CBX1 variants were identified, and clinician researchers were connected using GeneMatcher and physician referrals. Clinical histories were collected from each patient. To investigate the pathogenicity of identified variants, we performed in vitro cellular assays and neurobehavioral and cytological analyses of neuronal cells obtained from newly generated Cbx1 mutant mouse lines. RESULTS: In 3 unrelated individuals with developmental delay, hypotonia, and autistic features, we identified heterozygous de novo variants in CBX1. The identified variants were in the chromodomain, the functional domain of HP1ß, which mediates interactions with chromatin. Cbx1 chromodomain mutant mice displayed increased latency-to-peak response, suggesting the possibility of synaptic delay or myelination deficits. Cytological and chromatin immunoprecipitation experiments confirmed the reduction of mutant HP1ß binding to heterochromatin, whereas HP1ß interactome analysis demonstrated that the majority of HP1ß-interacting proteins remained unchanged between the wild-type and mutant HP1ß. CONCLUSION: These collective findings confirm the role of CBX1 in developmental disabilities through the disruption of HP1ß chromatin binding during neurocognitive development. Because HP1ß forms homodimers and heterodimers, mutant HP1ß likely sequesters wild-type HP1ß and other HP1 proteins, exerting dominant-negative effects.


Subject(s)
Chromobox Protein Homolog 5 , Heterochromatin , Animals , Mice , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Histones/genetics , Histones/metabolism
2.
Muscle Nerve ; 68(6): 833-840, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37789688

ABSTRACT

INTRODUCTION/AIMS: Exome sequencing (ES) has proven to be a valuable diagnostic tool for neuromuscular disorders, which often pose a diagnostic challenge. The aims of this study were to investigate the clinical outcomes associated with utilization of ES in the pediatric neuromuscular clinic and to determine if specific phenotypic features or abnormal neurodiagnostic tests were predictive of a diagnostic result. METHODS: This was a retrospective medical record review of 76 pediatric neuromuscular clinic patients who underwent ES. Based upon clinical assessment prior to ES, patients were divided into two groups: affected by neuromuscular (n = 53) or non-neuromuscular (n = 23) syndromes. RESULTS: A diagnosis was made in 28/76 (36.8%), with 29 unique disorders identified. In the neuromuscular group, a neuromuscular condition was confirmed in 78% of those receiving a genetic diagnosis. Early age of symptom onset was associated with a significantly higher diagnostic yield. The most common reason neuromuscular diagnoses were not detected on prior testing was due to causative genes not being present on disease-specific panels. Changes to medical care were made in 57% of individuals receiving a diagnosis on ES. DISCUSSION: These data further support ES as a powerful diagnostic tool in the pediatric neuromuscular clinic and highlight the advantages of ES over gene panels, including the ability to identify diagnoses regardless of etiology, identify genes newly associated with disease, and identify multiple confounding diagnoses. Rapid and accurate diagnosis by ES can not only end the patient's diagnostic odyssey, but often impacts patients' medical management and genetic counseling of families.


Subject(s)
Genetic Counseling , Neuromuscular Diseases , Humans , Child , Exome Sequencing , Retrospective Studies , Neuromuscular Diseases/diagnosis , Neuromuscular Diseases/genetics , Genetic Testing
3.
J Genet Couns ; 32(6): 1213-1216, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37571913

ABSTRACT

As a result of the pandemic, the traditional in-person didactic lecture model was adapted to a virtual learning approach. Our Laboratory Genetics and Genomics fellowship program at Nationwide Children's hospital took advantage of this opportunity to organize a multi-institutional Fellow's Conference to educate fellows from different programs on a wide range of medical genetics topics. We describe our approach of developing this lecture series utilizing subject-matter experts across institutions. In addition, we discuss the value of such an approach in reducing the amount of time individual institutions spend creating and providing didactic content for their small number of learners. Our experience could serve as a model for other educators and program directors, including genetic counseling program directors, to develop multi-institutional collaborations for didactic learning.


Subject(s)
Fellowships and Scholarships , Learning , Child , Humans , Education, Medical, Graduate , Curriculum , Laboratories , Surveys and Questionnaires
4.
Genet Med ; 24(7): 1379-1391, 2022 07.
Article in English | MEDLINE | ID: mdl-35608568

ABSTRACT

PURPOSE: Noninvasive prenatal screening (NIPS) using cell-free DNA has been assimilated into prenatal care. Prior studies examined clinical validity and technical performance in high-risk populations. This systematic evidence review evaluates NIPS performance in a general-risk population. METHODS: Medline (PubMed) and Embase were used to identify studies examining detection of Down syndrome (T21), trisomy 18 (T18), trisomy 13 (T13), sex chromosome aneuploidies, rare autosomal trisomies, copy number variants, and maternal conditions, as well as studies assessing the psychological impact of NIPS and the rate of subsequent diagnostic testing. Random-effects meta-analyses were used to calculate pooled estimates of NIPS performance (P < .05). Heterogeneity was investigated through subgroup analyses. Risk of bias was assessed. RESULTS: A total of 87 studies met inclusion criteria. Diagnostic odds ratios were significant (P < .0001) for T21, T18, and T13 for singleton and twin pregnancies. NIPS was accurate (≥99.78%) in detecting sex chromosome aneuploidies. Performance for rare autosomal trisomies and copy number variants was variable. Use of NIPS reduced diagnostic tests by 31% to 79%. Conclusions regarding psychosocial outcomes could not be drawn owing to lack of data. Identification of maternal conditions was rare. CONCLUSION: NIPS is a highly accurate screening method for T21, T18, and T13 in both singleton and twin pregnancies.


Subject(s)
Cell-Free Nucleic Acids , Down Syndrome , Noninvasive Prenatal Testing , Trisomy 13 Syndrome , Trisomy 18 Syndrome , Cell-Free Nucleic Acids/genetics , Down Syndrome/diagnosis , Down Syndrome/genetics , Female , Humans , Noninvasive Prenatal Testing/methods , Pregnancy , Prenatal Diagnosis/methods , Sex Chromosome Aberrations , Trisomy/diagnosis , Trisomy/genetics , Trisomy 13 Syndrome/diagnosis , Trisomy 13 Syndrome/genetics , Trisomy 18 Syndrome/diagnosis , Trisomy 18 Syndrome/genetics
5.
Hematol Oncol ; 40(3): 475-478, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35613340

ABSTRACT

To investigate germline predisposition in lymphoma, we performed whole-exome sequencing and discovered a novel variant (c.817-1G>T) in programmed cell death 1 ligand 2 (PD-L2) in a family with early-onset lymphomas and other cancers. The variant was present in the proband with follicular lymphoma and his son with Hodgkin's lymphoma. It was in the terminal splice acceptor site of PD-L2 and embedded in a putative enhancer of Janus kinase 2 (JAK2) and programmed cell death 1 ligand (PD-L1). We also found that gene expression of PD-L2, PD-L1, and JAK2 was significantly increased. Using 3' rapid amplification of cDNA ends (3' RACE), we detected an abnormal PD-L2 transcript in the son. Thus, the c.817-1G>T variant may result in the elevated PD-L2 expression due to the abnormal PD-L2 transcript and the elevated PD-L1 and JAK2 expression due to increased enhancer activity of PD-L1 and JAK2. The PD-L2 novel variant likely underlies the genetic etiology of the lymphomas in the family. As PD-L2 plays critical roles in tumor immunity, identification of PD-L2 as a germline predisposition gene may inform personalized immunotherapy in lymphoma patients.


Subject(s)
B7-H1 Antigen , Lymphoma , Programmed Cell Death 1 Ligand 2 Protein , B7-H1 Antigen/genetics , Exome , Genetic Predisposition to Disease , Humans , Ligands , Lymphoma/genetics , Programmed Cell Death 1 Ligand 2 Protein/genetics , Exome Sequencing
6.
Genome Res ; 27(8): 1287-1299, 2017 08.
Article in English | MEDLINE | ID: mdl-28546418

ABSTRACT

Metastasis is a complex biological process that has been difficult to delineate in human colorectal cancer (CRC) patients. A major obstacle in understanding metastatic lineages is the extensive intra-tumor heterogeneity at the primary and metastatic tumor sites. To address this problem, we developed a highly multiplexed single-cell DNA sequencing approach to trace the metastatic lineages of two CRC patients with matched liver metastases. Single-cell copy number or mutational profiling was performed, in addition to bulk exome and targeted deep-sequencing. In the first patient, we observed monoclonal seeding, in which a single clone evolved a large number of mutations prior to migrating to the liver to establish the metastatic tumor. In the second patient, we observed polyclonal seeding, in which two independent clones seeded the metastatic liver tumor after having diverged at different time points from the primary tumor lineage. The single-cell data also revealed an unexpected independent tumor lineage that did not metastasize, and early progenitor clones with the "first hit" mutation in APC that subsequently gave rise to both the primary and metastatic tumors. Collectively, these data reveal a late-dissemination model of metastasis in two CRC patients and provide an unprecedented view of metastasis at single-cell genomic resolution.


Subject(s)
Adenocarcinoma/secondary , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , High-Throughput Nucleotide Sequencing/methods , Liver Neoplasms/secondary , Sequence Analysis, DNA/methods , Single-Cell Analysis/methods , Adenocarcinoma/genetics , Aged , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Exome , Genomics , Humans , Liver Neoplasms/genetics , Middle Aged , Mutation , Phylogeny , Tumor Cells, Cultured
7.
Nature ; 512(7513): 155-60, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25079324

ABSTRACT

Sequencing studies of breast tumour cohorts have identified many prevalent mutations, but provide limited insight into the genomic diversity within tumours. Here we developed a whole-genome and exome single cell sequencing approach called nuc-seq that uses G2/M nuclei to achieve 91% mean coverage breadth. We applied this method to sequence single normal and tumour nuclei from an oestrogen-receptor-positive (ER(+)) breast cancer and a triple-negative ductal carcinoma. In parallel, we performed single nuclei copy number profiling. Our data show that aneuploid rearrangements occurred early in tumour evolution and remained highly stable as the tumour masses clonally expanded. In contrast, point mutations evolved gradually, generating extensive clonal diversity. Using targeted single-molecule sequencing, many of the diverse mutations were shown to occur at low frequencies (<10%) in the tumour mass. Using mathematical modelling we found that the triple-negative tumour cells had an increased mutation rate (13.3×), whereas the ER(+) tumour cells did not. These findings have important implications for the diagnosis, therapeutic treatment and evolution of chemoresistance in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Clonal Evolution , Genome/genetics , Cell Line, Tumor , DNA Fingerprinting , Female , Genetic Variation , Humans , Models, Theoretical , Mutation/genetics , Sequence Analysis, DNA , Single-Cell Analysis , Triple Negative Breast Neoplasms/genetics
8.
Genet Med ; 20(4): 464-469, 2018 04.
Article in English | MEDLINE | ID: mdl-28914269

ABSTRACT

PurposeTo describe examples of missed pathogenic variants on whole-exome sequencing (WES) and the importance of deep phenotyping for further diagnostic testing.MethodsGuided by phenotypic information, three children with negative WES underwent targeted single-gene testing.ResultsIndividual 1 had a clinical diagnosis consistent with infantile systemic hyalinosis, although WES and a next-generation sequencing (NGS)-based ANTXR2 test were negative. Sanger sequencing of ANTXR2 revealed a homozygous single base pair insertion, previously missed by the WES variant caller software. Individual 2 had neurodevelopmental regression and cerebellar atrophy, with no diagnosis on WES. New clinical findings prompted Sanger sequencing and copy number testing of PLA2G6. A novel homozygous deletion of the noncoding exon 1 (not included in the WES capture kit) was detected, with extension into the promoter, confirming the clinical suspicion of infantile neuroaxonal dystrophy. Individual 3 had progressive ataxia, spasticity, and magnetic resonance image changes of vanishing white matter leukoencephalopathy. An NGS leukodystrophy gene panel and WES showed a heterozygous pathogenic variant in EIF2B5; no deletions/duplications were detected. Sanger sequencing of EIF2B5 showed a frameshift indel, probably missed owing to failure of alignment.ConclusionThese cases illustrate potential pitfalls of WES/NGS testing and the importance of phenotype-guided molecular testing in yielding diagnoses.


Subject(s)
Exome , Genetic Association Studies , Genetic Predisposition to Disease , Molecular Diagnostic Techniques , Alleles , Biopsy , Child , Child, Preschool , Female , Genetic Association Studies/methods , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genotype , Humans , Infant , Molecular Diagnostic Techniques/methods , Molecular Diagnostic Techniques/standards , Phenotype , Polymorphism, Single Nucleotide , Rare Diseases/diagnosis , Rare Diseases/genetics , Exome Sequencing , Whole Genome Sequencing
10.
Proc Natl Acad Sci U S A ; 111(5): E572-81, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449888

ABSTRACT

The roles of microRNAs (miRNAs) and the miRNA processing machinery in the regulation of stem cell biology are not well understood. Here, we show that the p53 family member and p63 isoform, ΔNp63, is a transcriptional activator of a cofactor critical for miRNA processing (DGCR8). This regulation gives rise to a unique miRNA signature resulting in reprogramming cells to multipotency. Strikingly, ΔNp63(-/-) epidermal cells display profound defects in terminal differentiation and express a subset of markers and miRNAs present in embryonic stem cells and fibroblasts induced to pluripotency using Yamanaka factors. Moreover, ΔNp63(-/-) epidermal cells transduced with an inducible DGCR8 plasmid can differentiate into multiple cell fates in vitro and in vivo. We found that human primary keratinocytes depleted of ΔNp63 or DGCR8 can be reprogrammed in 6 d and express a unique miRNA and gene expression signature that is similar but not identical to human induced pluripotent stem cells. Our data reveal a role for ΔNp63 in the transcriptional regulation of DGCR8 to reprogram adult somatic cells into multipotent stem cells.


Subject(s)
Down-Regulation/genetics , Keratinocytes/metabolism , Multipotent Stem Cells/cytology , Phosphoproteins/genetics , Proteins/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Chimera , Embryo, Mammalian/cytology , Epidermal Cells , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/cytology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Multipotent Stem Cells/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Phosphoproteins/deficiency , Phosphoproteins/metabolism , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Trans-Activators/deficiency , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
11.
Nature ; 467(7318): 986-90, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20962848

ABSTRACT

Aberrant expression of microRNAs (miRNAs) and the enzymes that control their processing have been reported in multiple biological processes including primary and metastatic tumours, but the mechanisms governing this are not clearly understood. Here we show that TAp63, a p53 family member, suppresses tumorigenesis and metastasis, and coordinately regulates Dicer and miR-130b to suppress metastasis. Metastatic mouse and human tumours deficient in TAp63 express Dicer at very low levels, and we found that modulation of expression of Dicer and miR-130b markedly affected the metastatic potential of cells lacking TAp63. TAp63 binds to and transactivates the Dicer promoter, demonstrating direct transcriptional regulation of Dicer by TAp63. These data provide a novel understanding of the roles of TAp63 in tumour and metastasis suppression through the coordinate transcriptional regulation of Dicer and miR-130b and may have implications for the many processes regulated by miRNAs.


Subject(s)
DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Neoplasm Metastasis/genetics , Phosphoproteins/metabolism , Ribonuclease III/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cellular Senescence , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Female , Genes, Tumor Suppressor/physiology , Genomic Instability , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Ribonuclease III/biosynthesis , Ribonuclease III/deficiency , Ribonuclease III/genetics , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
12.
J Mol Diagn ; 26(6): 447-455, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38378079

ABSTRACT

Microarray-based methylation profiling has emerged as a valuable tool for refining diagnoses and revealing novel tumor subtypes, particularly in central nervous system tumors. Despite the increasing adoption of this technique in clinical genomic laboratories, no technical standards have been published in establishing minimum criteria for test validation. A working group with experience and expertise in DNA-based methylation profiling tests on central nervous system tumors collaborated to develop practical discussion points and focus on important considerations for validating this test in clinical laboratory settings. The experience in validating this methodology in a clinical setting is summarized. Specifically, the advantages and challenges associated with utilizing an in-house classifier compared with a third-party classifier are highlighted. Additionally, experiences in demonstrating the assay's sensitivity and specificity, establishing minimum sample criteria, and implementing quality control metrics are described. As methylation profiling for tumor classification expands to other tumor types and continues to evolve for various other applications, the critical considerations described here are expected to serve as a guidance for future efforts in establishing professional guidelines for this assay.


Subject(s)
DNA Methylation , Oligonucleotide Array Sequence Analysis , Humans , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Array Sequence Analysis/standards , Reproducibility of Results , Sensitivity and Specificity , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/diagnosis , Gene Expression Profiling/methods
13.
J Appl Lab Med ; 9(1): 61-75, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38167757

ABSTRACT

BACKGROUND: Throughout history, the field of cytogenetics has witnessed significant changes due to the constant evolution of technologies used to assess chromosome number and structure. Similar to the evolution of single nucleotide variant detection from Sanger sequencing to next-generation sequencing, the identification of chromosome alterations has progressed from banding to fluorescence in situ hybridization (FISH) to chromosomal microarrays. More recently, emerging technologies such as optical genome mapping and genome sequencing have made noteworthy contributions to clinical laboratory testing in the field of cytogenetics. CONTENT: In this review, we journey through some of the most pivotal discoveries that have shaped the development of clinical cytogenetics testing. We also explore the current test offerings, their uses and limitations, and future directions in technology advancements. SUMMARY: Cytogenetics methods, including banding and targeted assessments like FISH, continue to hold crucial roles in cytogenetic testing. These methods offer a rapid turnaround time, especially for conditions with a known etiology involving recognized cytogenetic aberrations. Additionally, laboratories have the flexibility to now employ higher-throughput methodologies to enhance resolution for cases with greater complexity.


Subject(s)
Chromosome Aberrations , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence/methods , Cytogenetics/methods , Chromosome Mapping , High-Throughput Nucleotide Sequencing/methods
14.
Mol Genet Genomic Med ; 12(3): e2349, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38263869

ABSTRACT

BACKGROUND: Chromosomal microarray (CMA) is commonly utilized in the obstetrics setting. CMA is recommended when one or more fetal structural abnormalities is identified. CMA is also commonly used to determine genetic etiologies for miscarriages, fetal demise, and confirming positive prenatal cell-free DNA screening results. METHODS: In this study, we retrospectively examined 523 prenatal and 319 products-of-conception (POC) CMA cases tested at Nationwide Children's Hospital from 2011 to 2020. We reviewed the referral indications, the diagnostic yield, and the reported copy number variants (CNV) findings. RESULTS: In our cohort, the diagnostic yield of clinically significant CNV findings for prenatal testing was 7.8% (n = 41/523) compared to POC testing (16.3%, n = 52/319). Abnormal ultrasound findings were the most common indication present in 81% of prenatal samples. Intrauterine fetal demise was the common indication identified in POC samples. The most common pathogenic finding observed in all samples was isolated trisomy 21, detected in seven samples. CONCLUSION: Our CMA study supports the clinical utility of prenatal CMA for clinical management and identifying genetic etiology in POC arrays. In addition, it provides insight to the spectrum of prenatal and POC CMA results as detected in an academic hospital clinical laboratory setting that serves as a reference laboratory.


Subject(s)
Chromosome Disorders , Down Syndrome , Female , Humans , Pregnancy , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Fetal Death , Prenatal Diagnosis/methods , Retrospective Studies
15.
Eur J Med Genet ; 66(4): 104731, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36775013

ABSTRACT

Aicardi-Goutières syndrome (AGS) is a progressive multisystem disorder including encephalopathy with significant impacts on intellectual and physical abilities. An early diagnosis is becoming ever more crucial, as targeted therapies are emerging. A deep understanding of the molecular heterogeneity of AGS can help guide the early diagnosis and clinical management of patients, and inform recurrence risks. Here, we detail the diagnostic odyssey of a patient with an early presentation of AGS. Exome and genome sequencing detected an intronic RNASEH2B variant missed in a conventional leukodystrophy NGS gene panel. RNA studies demonstrated that a c.322-17 A > G variant affected splicing and caused 16-nucleotide intronic retention in the RNASEH2B transcript, introducing an out-of-frame early termination codon. RNASEH2B expression in the patient's blood was reduced when compared to controls. Our study highlights the pathogenicity of this intronic variant and the importance of its inclusion in variant assessment.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Humans , Mutation , Autoimmune Diseases of the Nervous System/genetics , Nervous System Malformations/genetics , Exome
16.
Front Genet ; 14: 1298574, 2023.
Article in English | MEDLINE | ID: mdl-38304066

ABSTRACT

Background: Leigh syndrome is a rare, genetic, and severe mitochondrial disorder characterized by neuromuscular issues (ataxia, seizure, hypotonia, developmental delay, dystonia) and ocular abnormalities (nystagmus, atrophy, strabismus, ptosis). It is caused by pathogenic variants in either mitochondrial or nuclear DNA genes, with an estimated incidence rate of 1 per 40,000 live births. Case presentation: Herein, we present an infant male with nystagmus, hypotonia, and developmental delay who carried a clinical diagnosis of Leigh-like syndrome. Cerebral magnetic resonance imaging changes further supported the clinical evidence of an underlying mitochondrial disorder, but extensive diagnostic testing was negative. Trio exome sequencing under a research protocol uncovered compound-heterozygous missense variants in the HTRA2 gene (MIM: #606441): NM_013247.5:c.1037A>T:(p.Glu346Val) (maternal) and NM_013247.5:c.1172T>A:(p.Val391Glu) (paternal). Both variants are absent from public databases, making them extremely rare in the population. The maternal variant is adjacent to an exon-intron boundary and predicted to disrupt splicing, while the paternal variant alters a highly conserved amino acid and is predicted to be damaging by nearly all in silico tools. Biallelic variants in HTRA2 cause 3-methylglutaconic aciduria, type VIII (MGCA8), an extremely rare autosomal recessive disorder with fewer than ten families reported to date. Variant interpretation is challenging given the paucity of known disease-causing variants, and indeed we assess both paternal and maternal variants as Variants of Uncertain Significance under current American College of Medical Genetics guidelines. However, based on the inheritance pattern, suggestive evidence of pathogenicity, and significant clinical correlation with other reported MGCA8 patients, the clinical care team considers this a diagnostic result. Conclusion: Our findings ended the diagnostic odyssey for this family and provide further insights into the genetic and clinical spectrum of this critically under-studied disorder.

17.
J Mol Diagn ; 24(2): 177-188, 2022 02.
Article in English | MEDLINE | ID: mdl-35074075

ABSTRACT

Exome reanalysis is useful for providing molecular diagnoses for previously uninformative samples. However, challenges exist in implementing a practical solution for clinicians and laboratories. This study complements the current literature by providing practical considerations for patient-level and cohort-level reanalyses. The Clinical and Laboratory Standards Institute assembled the Document Development Committee and an interpretation working group that developed the framework for reevaluation of exome-based data. We describe two distinct but complementary approaches toward exome reanalyses: clinician-initiated patient-level reanalysis, and laboratory-initiated cohort-level reanalysis. We highlight the advantages and constraints for both approaches, and provide a high-level conceptual guide for ordering clinicians and laboratories through the critical decision pathways. Because clinical exome sequencing continues to be the standard of care in genetics, exome reanalysis would be critical in increasing the overall diagnostic yield. A systematic guide will facilitate the efficient adoption of reevaluation of exome data for laboratories, health care professionals, genetic counselors, and clinicians.


Subject(s)
Clinical Laboratory Services , Exome , Exome/genetics , Humans , Laboratories , Laboratories, Clinical , Exome Sequencing
18.
J Mol Diagn ; 24(9): 1031-1040, 2022 09.
Article in English | MEDLINE | ID: mdl-35718094

ABSTRACT

Chromosomal microarray (CMA) is a testing modality frequently used in pediatric patients; however, published data on its utilization are limited to the genetic setting. We performed a database search for all CMA testing performed from 2010 to 2020, and delineated the diagnostic yield based on patient characteristics, including sex, age, clinical specialty of providers, indication of testing, and pathogenic finding. The indications for testing were further categorized into Human Phenotype Ontology categories for analysis. This study included a cohort of 14,541 patients from 29 different medical specialties, of whom 30% were from the genetics clinic. The clinical indications for testing suggested that neonatology patients demonstrated the greatest involvement of multiorgan systems, involving the most Human Phenotype Ontology categories, compared with developmental behavioral pediatrics and neurology patients being the least. The top pathogenic findings for each specialty differed, likely due to the varying clinical features and indications for testing. Deletions involving the 22q11.21 locus were the top pathogenic findings for patients presenting to genetics, neonatology, cardiology, and surgery. Our data represent the largest pediatric cohort published to date. This study is the first to demonstrate the diagnostic utility of this assay for patients seen in the setting of different specialties, and it provides normative data of CMA results among a general pediatric population referred for testing because of variable clinical presentations.


Subject(s)
Pediatrics , Child , Cohort Studies , Humans , Microarray Analysis/methods
19.
Mol Diagn Ther ; 25(5): 529-536, 2021 09.
Article in English | MEDLINE | ID: mdl-34283395

ABSTRACT

Novel gene-disease discoveries, rapid advancements in technology, and improved bioinformatics tools all have the potential to yield additional molecular diagnoses through the reanalysis of exome sequencing data. Collaborations between clinical laboratories, ordering physicians, and researchers are also driving factors that can contribute to these new insights. Automation in ongoing natural history collection, evolving phenotype updates, advancements in processing next-generation sequencing data, and up-to-date variant-gene-disease databases are increasingly needed for systematic exome reanalysis. Here, we review some of the advantages and challenges for clinician-initiated and laboratory-initiated exome reanalysis, and we propose a model for the future that could potentially maximize the clinical utility of exome reanalysis by integrating information from electronic medical records and knowledge databases into routine clinical workflows.


Subject(s)
Exome , Laboratories, Clinical , Computational Biology , Exome/genetics , Humans , Phenotype , Exome Sequencing
20.
J Mol Diagn ; 23(1): 91-102, 2021 01.
Article in English | MEDLINE | ID: mdl-33349347

ABSTRACT

Carrier screening involves detection of carrier status for genes associated with recessive conditions. A negative carrier screening test result bears a nonzero residual risk (RR) for the individual to have an affected child. The RR depends on the prevalence of specific conditions and the detection rate (DR) of the test itself. Herein, we provide a detailed approach for calculating DR and RR. DR was calculated on the basis of the sum of disease allele frequencies (DAFs) of pathogenic variants found in published literature. As a proof of concept, DAF data for cystic fibrosis were compared with society guidelines. The DAF data calculated by this method were consistent with the published cystic fibrosis guideline. In addition, we compared DAF for four genes (ABCC8, ASPA, GAA, and MMUT) across three laboratories, and outlined the likely reasons for discrepancies between these laboratories. The utility of carrier screening is to support couples with information while making reproductive choices. Accurate development of DR and RR is therefore critical. The method described herein provides an unbiased and transparent process to collect, calculate, and report these data.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/prevention & control , Gene Frequency , Genetic Carrier Screening/methods , Amidohydrolases/genetics , Consanguinity , Family , Genetic Counseling/methods , Humans , Mass Screening/methods , Sulfonylurea Receptors/genetics , alpha-Glucosidases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL