Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Hum Genet ; 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36952035

ABSTRACT

The multidisciplinary Epigenetics and Chromatin Clinic at Johns Hopkins provides comprehensive medical care for individuals with rare disorders that involve disrupted epigenetics. Initially centered on classical imprinting disorders, the focus shifted to the rapidly emerging group of genetic disorders resulting from pathogenic germline variants in epigenetic machinery genes. These are collectively called the Mendelian disorders of the epigenetic machinery (MDEMs), or more broadly, Chromatinopathies. In five years, 741 clinic visits have been completed for 432 individual patients, with 153 having confirmed epigenetic diagnoses. Of these, 115 individuals have one of 26 MDEMs with every single one exhibiting global developmental delay and/or intellectual disability. This supports prior observations that intellectual disability is the most common phenotypic feature of MDEMs. Additional common phenotypes in our clinic include growth abnormalities and neurodevelopmental issues, particularly hypotonia, attention-deficit/hyperactivity disorder (ADHD), and anxiety, with seizures and autism being less common. Overall, our patient population is representative of the broader group of MDEMs and includes mostly autosomal dominant disorders impacting writers more so than erasers, readers, and remodelers of chromatin marks. There is an increased representation of dual function components with a reader and an enzymatic domain. As expected, diagnoses were made mostly by sequencing but were aided in some cases by DNA methylation profiling. Our clinic has helped to facilitate the discovery of two new disorders, and our providers are actively developing and implementing novel therapeutic strategies for MDEMs. These data and our high follow-up rate of over 60% suggest that we are achieving our mission to diagnose, learn from, and provide optimal care for our patients with disrupted epigenetics.

2.
Am J Hum Genet ; 105(1): 48-64, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31178128

ABSTRACT

We report biallelic missense and frameshift pathogenic variants in the gene encoding human nucleoporin NUP214 causing acute febrile encephalopathy. Clinical symptoms include neurodevelopmental regression, seizures, myoclonic jerks, progressive microcephaly, and cerebellar atrophy. NUP214 and NUP88 protein levels were reduced in primary skin fibroblasts derived from affected individuals, while the total number and density of nuclear pore complexes remained normal. Nuclear transport assays exhibited defects in the classical protein import and mRNA export pathways in affected cells. Direct surface imaging of fibroblast nuclei by scanning electron microscopy revealed a large increase in the presence of central particles (known as "plugs") in the nuclear pore channels of affected cells. This observation suggests that large transport cargoes may be delayed in passage through the nuclear pore channel, affecting its selective barrier function. Exposure of fibroblasts from affected individuals to heat shock resulted in a marked delay in their stress response, followed by a surge in apoptotic cell death. This suggests a mechanistic link between decreased cell survival in cell culture and severe fever-induced brain damage in affected individuals. Our study provides evidence by direct imaging at the single nuclear pore level of functional changes linked to a human disease.


Subject(s)
Acute Febrile Encephalopathy/etiology , Fibroblasts/pathology , Frameshift Mutation , Ion Channels/physiology , Mutation, Missense , Nuclear Pore Complex Proteins/genetics , Nuclear Pore/pathology , Active Transport, Cell Nucleus , Acute Febrile Encephalopathy/metabolism , Acute Febrile Encephalopathy/pathology , Apoptosis , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/metabolism , Humans , Infant , Male , Nuclear Pore/genetics , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Pedigree , Protein Conformation
3.
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
4.
Brain ; 143(11): 3242-3261, 2020 12 05.
Article in English | MEDLINE | ID: mdl-33150406

ABSTRACT

Heterozygous mutations in KMT2B are associated with an early-onset, progressive and often complex dystonia (DYT28). Key characteristics of typical disease include focal motor features at disease presentation, evolving through a caudocranial pattern into generalized dystonia, with prominent oromandibular, laryngeal and cervical involvement. Although KMT2B-related disease is emerging as one of the most common causes of early-onset genetic dystonia, much remains to be understood about the full spectrum of the disease. We describe a cohort of 53 patients with KMT2B mutations, with detailed delineation of their clinical phenotype and molecular genetic features. We report new disease presentations, including atypical patterns of dystonia evolution and a subgroup of patients with a non-dystonic neurodevelopmental phenotype. In addition to the previously reported systemic features, our study has identified co-morbidities, including the risk of status dystonicus, intrauterine growth retardation, and endocrinopathies. Analysis of this study cohort (n = 53) in tandem with published cases (n = 80) revealed that patients with chromosomal deletions and protein truncating variants had a significantly higher burden of systemic disease (with earlier onset of dystonia) than those with missense variants. Eighteen individuals had detailed longitudinal data available after insertion of deep brain stimulation for medically refractory dystonia. Median age at deep brain stimulation was 11.5 years (range: 4.5-37.0 years). Follow-up after deep brain stimulation ranged from 0.25 to 22 years. Significant improvement of motor function and disability (as assessed by the Burke Fahn Marsden's Dystonia Rating Scales, BFMDRS-M and BFMDRS-D) was evident at 6 months, 1 year and last follow-up (motor, P = 0.001, P = 0.004, and P = 0.012; disability, P = 0.009, P = 0.002 and P = 0.012). At 1 year post-deep brain stimulation, >50% of subjects showed BFMDRS-M and BFMDRS-D improvements of >30%. In the long-term deep brain stimulation cohort (deep brain stimulation inserted for >5 years, n = 8), improvement of >30% was maintained in 5/8 and 3/8 subjects for the BFMDRS-M and BFMDRS-D, respectively. The greatest BFMDRS-M improvements were observed for trunk (53.2%) and cervical (50.5%) dystonia, with less clinical impact on laryngeal dystonia. Improvements in gait dystonia decreased from 20.9% at 1 year to 16.2% at last assessment; no patient maintained a fully independent gait. Reduction of BFMDRS-D was maintained for swallowing (52.9%). Five patients developed mild parkinsonism following deep brain stimulation. KMT2B-related disease comprises an expanding continuum from infancy to adulthood, with early evidence of genotype-phenotype correlations. Except for laryngeal dysphonia, deep brain stimulation provides a significant improvement in quality of life and function with sustained clinical benefit depending on symptoms distribution.


Subject(s)
Dystonic Disorders/genetics , Histone-Lysine N-Methyltransferase/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosome Deletion , Cohort Studies , Computer Simulation , Deep Brain Stimulation , Disease Progression , Dystonic Disorders/therapy , Endocrine System Diseases/complications , Endocrine System Diseases/genetics , Female , Fetal Growth Retardation/genetics , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/therapy , Humans , Laryngeal Diseases/etiology , Laryngeal Diseases/therapy , Male , Mutation , Mutation, Missense , Phenotype , Quality of Life , Treatment Outcome , Young Adult
5.
Proc Natl Acad Sci U S A ; 115(10): E2358-E2365, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463756

ABSTRACT

Telomere length (TL) predicts the onset of cellular senescence in vitro but the diagnostic utility of TL measurement in clinical settings is not fully known. We tested the value of TL measurement by flow cytometry and FISH (flowFISH) in patients with mutations in telomerase and telomere maintenance genes. TL had a discrete and reproducible normal range with definable upper and lower boundaries. While TL above the 50th age-adjusted percentile had a 100% negative predictive value for clinically relevant mutations, the lower threshold in mutation carriers was age-dependent, and adult mutation carriers often overlapped with the lowest decile of controls. The extent of telomere shortening correlated with the age at diagnosis as well as the short telomere syndrome phenotype. Extremely short TL caused bone marrow failure and immunodeficiency in children and young adults, while milder defects manifested as pulmonary fibrosis-emphysema in adults. We prospectively examined whether TL altered treatment decisions for newly diagnosed idiopathic bone marrow failure patients and found abnormally short TL enriched for patients with mutations in some inherited bone marrow failure genes, such as RUNX1, in addition to telomerase and telomere maintenance genes. The result was actionable, altering the choice of treatment regimen and/or hematopoietic stem cell donor in one-fourth of the cases (9 of 38, 24%). We conclude that TL measurement by flowFISH, when used for targeted clinical indications and in limited settings, can influence treatment decisions in ways that improve outcome.


Subject(s)
Pulmonary Emphysema/metabolism , Pulmonary Fibrosis/metabolism , Telomere Shortening , Telomere/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Hospitals/statistics & numerical data , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Middle Aged , Mutation , Pulmonary Emphysema/diagnosis , Pulmonary Emphysema/genetics , Pulmonary Fibrosis/diagnosis , Pulmonary Fibrosis/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/chemistry , Young Adult
6.
J Med Genet ; 55(8): 561-566, 2018 08.
Article in English | MEDLINE | ID: mdl-28866611

ABSTRACT

BACKGROUND: The list of Mendelian disorders of the epigenetic machinery has expanded rapidly during the last 5 years. A few missense variants in the chromatin remodeler CHD1 have been found in several large-scale sequencing efforts focused on uncovering the genetic aetiology of autism. OBJECTIVES: To explore whether variants in CHD1 are associated with a human phenotype. METHODS: We used GeneMatcher to identify other physicians caring for patients with variants in CHD1. We also explored the epigenetic consequences of one of these variants in cultured fibroblasts. RESULTS: Here we describe six CHD1 heterozygous missense variants in a cohort of patients with autism, speech apraxia, developmental delay and facial dysmorphic features. Importantly, three of these variants occurred de novo. We also report on a subject with a de novo deletion covering a large fraction of the CHD1 gene without any obvious neurological phenotype. Finally, we demonstrate increased levels of the closed chromatin modification H3K27me3 in fibroblasts from a subject carrying a de novo variant in CHD1. CONCLUSIONS: Our results suggest that variants in CHD1 can lead to diverse phenotypic outcomes; however, the neurodevelopmental phenotype appears to be limited to patients with missense variants, which is compatible with a dominant negative mechanism of disease.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation, Missense , Child , Child, Preschool , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Developmental Disabilities/diagnosis , Facies , Female , Fibroblasts/metabolism , Genetic Association Studies/methods , Histones/metabolism , Humans , Infant , Models, Molecular , Phenotype , Protein Conformation , Structure-Activity Relationship
7.
J Genet Couns ; 27(1): 263-273, 2018 02.
Article in English | MEDLINE | ID: mdl-28932961

ABSTRACT

Genetic counselors working in a clinical setting may find themselves recruiting, enrolling, and returning results for genomic research, and existing clinical relationships with study participants may impact these research interactions. We present a qualitative study using semi-structured interviews of participants enrolled in a genome sequencing/exome sequencing (GS/ES) study at the same institution where they receive clinical care. Interviews were coded for motivations to participate and expectations of this research. The interviews revealed common motivations for participation, including altruism and hope for benefit for themselves, family members, and/or others with their condition. Additionally, themes emerged related to unintentional influence based on trust of the clinical provider that recruited them to the study. Participant trust in the enrolling provider at times appeared to extend to the study team to decide which research results to return and to do so in an appropriate format. Participants also based expectations for research results return on previous clinical genetic testing experiences, which may or may not be realistic depending on study design. It is imperative that genetic counselors enrolling patients into research studies be aware of the potential influence of their clinical relationship on potential subjects, be transparent about their role on the study team, and help set expectations about the study process, including results return.


Subject(s)
Genetic Testing , Informed Consent , Trust , Base Sequence , Decision Making , Family , Female , Humans , Male , Motivation , Qualitative Research , Research Design
8.
Genet Med ; 19(4): 412-420, 2017 04.
Article in English | MEDLINE | ID: mdl-27657687

ABSTRACT

PURPOSE: To investigate the utility of whole-exome sequencing (WES) to define a molecular diagnosis for patients clinically diagnosed with congenital anomalies of kidney and urinary tract (CAKUT). METHODS: WES was performed in 62 families with CAKUT. WES data were analyzed for single-nucleotide variants (SNVs) in 35 known CAKUT genes, putatively deleterious sequence changes in new candidate genes, and potentially disease-associated copy-number variants (CNVs). RESULTS: In approximately 5% of families, pathogenic SNVs were identified in PAX2, HNF1B, and EYA1. Observed phenotypes in these families expand the current understanding about the role of these genes in CAKUT. Four pathogenic CNVs were also identified using two CNV detection tools. In addition, we found one deleterious de novo SNV in FOXP1 among the 62 families with CAKUT. The clinical database of the Baylor Miraca Genetics laboratory was queried and seven additional unrelated individuals with novel de novo SNVs in FOXP1 were identified. Six of these eight individuals with FOXP1 SNVs have syndromic urinary tract defects, implicating this gene in urinary tract development. CONCLUSION: We conclude that WES can be used to identify molecular etiology (SNVs, CNVs) in a subset of individuals with CAKUT. WES can also help identify novel CAKUT genes.Genet Med 19 4, 412-420.


Subject(s)
DNA Copy Number Variations , Exome Sequencing/methods , Genetic Predisposition to Disease/genetics , Urogenital Abnormalities/diagnosis , Vesico-Ureteral Reflux/diagnosis , Adolescent , Child , Child, Preschool , Female , Forkhead Transcription Factors/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Infant , Intracellular Signaling Peptides and Proteins/genetics , Male , Nuclear Proteins/genetics , PAX2 Transcription Factor/genetics , Pedigree , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatases/genetics , Repressor Proteins/genetics , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Young Adult
9.
Genet Med ; 16(9): 657-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24625444

ABSTRACT

PURPOSE: Chromosomal microarray has been widely adopted as the first-tier clinical test for individuals with multiple congenital anomalies, developmental delay, intellectual disability, and autism spectrum disorders. Although chromosomal microarray has been extensively shown to provide a higher diagnostic yield than conventional cytogenetic methods, some health insurers refuse to provide coverage for this test, claiming that it is experimental and does not affect patients' clinical management. METHODS: We retrospectively reviewed the electronic medical records of all patients who had abnormal chromosomal microarray findings reported by our laboratory over a 3-year period and quantified the management recommendations made in response to these results. RESULTS: Abnormal chromosomal microarray findings were reported for 12.7% of patients (227/1,780). For patients with clinical follow-up notes available, these results had management implications for 54.5% of patients in the entire abnormal cohort (102/187) and for 42.1% of patients referred for isolated neurodevelopmental disorders (16/38). Recommendations included pharmacological treatment, cancer-related screening or exclusion of screening, contraindications, and referrals for further evaluation. CONCLUSION: These results empirically demonstrate the clinical utility of chromosomal microarray by providing evidence that management was directly affected for the majority of patients in our cohort with abnormal chromosomal microarray findings.


Subject(s)
Chromosomes, Human , Disease Management , Genetic Testing , Oligonucleotide Array Sequence Analysis , Child , Child, Preschool , Early Detection of Cancer , Female , Follow-Up Studies , Genetic Testing/methods , Humans , Infant , Male , Polymorphism, Single Nucleotide , Referral and Consultation , Retrospective Studies
10.
Am J Med Genet A ; 158A(6): 1414-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22585553

ABSTRACT

The RAS-MAPK pathway is critical for human growth and development. Abnormalities at different steps of this signaling cascade result in neuro-cardio-facial-cutaneous syndromes, or the RASopathies, a group of disorders with overlapping yet distinct phenotypes. RASopathy patients have variable degrees of intellectual disability, poor growth, relative macrocephaly, ectodermal abnormalities, dysmorphic features, and increased risk for certain malignancies. Congenital heart disease, particularly hypertrophic cardiomyopathy (HCM) and pulmonic stenosis, are prominent features in these disorders. Significant locus heterogeneity exists for many of the RASopathies. Traditionally, these diseases were thought to be inherited in an autosomal dominant manner. However, recently patients with defects in two components of this pathway and overlapping features of various forms of Noonan syndrome and neurofibromatosis 1 and have been reported. Here we present a patient with severe, progressive neonatal HCM, elevated urinary catecholamine metabolites, and dysmorphic features in whom we identified a known LEOPARD syndrome-associated PTPN11 mutation (c.1403 C > T; p.T468M) and a novel, potentially pathogenic missense SOS1 variant (c.1018 C > T; p.P340S) replacing a rigid nonpolar imino acid with a polar amino acid at a highly conserved position. We describe detailed clinical manifestations, cardiac histopathology, and the molecular genetic findings. Oligogenic models of inheritance with potential synergistic effects should be considered in the RASopathies.


Subject(s)
Cardiomyopathy, Hypertrophic/congenital , Cardiomyopathy, Hypertrophic/genetics , Mutation , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , SOS1 Protein/genetics , Amino Acid Sequence , Cardiomyopathy, Hypertrophic/diagnosis , Female , Humans , Infant , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/chemistry , Signal Transduction
11.
Curr Protoc ; 2(9): e530, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36130039

ABSTRACT

Structural variation in genomes, such as copy number variants (CNVs), is under scrutiny for its contribution to phenotypic expression and evolution. Regions of homozygosity (ROH) are ripe for phenotype-gene discovery. Determining the genes and related phenotypes within genomic regions is key to studying potential functional and phenotypic consequences. Because individuals have multiple CNVs and ROHs in their genome, identifying genomic regions that are phenotypically significant is challenging. GeneScout is a web-based tool that can be used to search genomic regions to display and filter the genes and their associated phenotypes within regions of interest. Phenotypes and their associated gene(s) can then be filtered to show only the genes with phenotypes that have a particular inheritance pattern and/or specific clinical feature(s). Phenotypes can then be selected to compare the clinical synopses side-by-side in Online Mendelian Inheritance in Man (OMIM® ). Additionally, two coordinate sets can be compared to determine either the regions of overlap or the unique regions (subtraction). The resulting coordinate ranges are displayed on the results page, and the results table displays only the genes and phenotypes present within the coordinate ranges. The interactive table includes gene-specific links to external resources such as ClinVar, ClinGen validity, ClinGen dosage, and gnomAD, and a diamond symbol next to the gene name indicates a gene that spans the start or end of a coordinate range. Searches and comparisons may be performed for coordinates in assemblies GRCh37 (hg19) and GRCh38 (hg38). The results page offers the option to liftover coordinates entered in GRCh37 to GRCh38 and updates the results table to display the gene content based on assembly GRCh38. The search coordinates and results table can be downloaded in a tab-delimited or Excel file. © 2022 Wiley Periodicals LLC. Basic Protocol: Searching GeneScout.


Subject(s)
Databases, Genetic , Genome , DNA Copy Number Variations/genetics , Genomics , Humans , Phenotype
12.
Mol Genet Genomic Med ; 8(2): e1072, 2020 02.
Article in English | MEDLINE | ID: mdl-31814321

ABSTRACT

We present the first patient described with haploinsufficency of KMT2D leading to Kabuki syndrome. Deletion of KMT2D has been thought to be lethal, but here we describe a patient with KMT2D deletion and classical Kabuki syndrome phenotype.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Face/abnormalities , Hematologic Diseases/genetics , Neoplasm Proteins/genetics , Phenotype , Vestibular Diseases/genetics , Abnormalities, Multiple/pathology , Face/pathology , Female , Gene Deletion , Haploinsufficiency , Hematologic Diseases/pathology , Humans , Infant , Vestibular Diseases/pathology
14.
J Clin Invest ; 128(12): 5222-5234, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30179220

ABSTRACT

The mechanisms that drive T cell aging are not understood. We report that children and adult telomerase mutation carriers with short telomere length (TL) develop a T cell immunodeficiency that can manifest in the absence of bone marrow failure and causes life-threatening opportunistic infections. Mutation carriers shared T cell-aging phenotypes seen in adults 5 decades older, including depleted naive T cells, increased apoptosis, and restricted T cell repertoire. T cell receptor excision circles (TRECs) were also undetectable or low, suggesting that newborn screening may identify individuals with germline telomere maintenance defects. Telomerase-null mice with short TL showed defects throughout T cell development, including increased apoptosis of stimulated thymocytes, their intrathymic precursors, in addition to depleted hematopoietic reserves. When we examined the transcriptional programs of T cells from telomerase mutation carriers, we found they diverged from older adults with normal TL. Short telomere T cells upregulated DNA damage and intrinsic apoptosis pathways, while older adult T cells upregulated extrinsic apoptosis pathways and programmed cell death 1 (PD-1) expression. T cells from mice with short TL also showed an active DNA-damage response, in contrast with old WT mice, despite their shared propensity to apoptosis. Our data suggest there are TL-dependent and TL-independent mechanisms that differentially contribute to distinct molecular programs of T cell apoptosis with aging.


Subject(s)
Apoptosis/immunology , Growth Disorders , Hypercalcemia , Immunologic Deficiency Syndromes , Metabolic Diseases , Mutation , Nephrocalcinosis , Telomerase , Telomere Homeostasis/immunology , Adult , Aging/genetics , Aging/immunology , Aging/pathology , Animals , Apoptosis/genetics , DNA Damage/immunology , Female , Growth Disorders/complications , Growth Disorders/genetics , Growth Disorders/immunology , Growth Disorders/pathology , Humans , Hypercalcemia/complications , Hypercalcemia/genetics , Hypercalcemia/immunology , Hypercalcemia/pathology , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/genetics , Male , Metabolic Diseases/complications , Metabolic Diseases/genetics , Metabolic Diseases/immunology , Metabolic Diseases/pathology , Mice , Mice, Knockout , Nephrocalcinosis/complications , Nephrocalcinosis/genetics , Nephrocalcinosis/immunology , Nephrocalcinosis/pathology , Primary Immunodeficiency Diseases , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Telomerase/genetics , Telomerase/immunology
15.
Sci Transl Med ; 8(351): 351ra107, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27510903

ABSTRACT

Chronic obstructive pulmonary disease and pulmonary fibrosis have been hypothesized to represent premature aging phenotypes. At times, they cluster in families, but the genetic basis is not understood. We identified rare, frameshift mutations in the gene for nuclear assembly factor 1, NAF1, a box H/ACA RNA biogenesis factor, in pulmonary fibrosis-emphysema patients. The mutations segregated with short telomere length, low telomerase RNA levels, and extrapulmonary manifestations including myelodysplastic syndrome and liver disease. A truncated NAF1 was detected in cells derived from patients, and, in cells in which the frameshift mutation was introduced by genome editing, telomerase RNA levels were reduced. The mutant NAF1 lacked a conserved carboxyl-terminal motif, which we show is required for nuclear localization. To understand the disease mechanism, we used CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein-9 nuclease) to generate Naf1(+/-) mice and found that they had half the levels of telomerase RNA. Other box H/ACA RNA levels were also decreased, but rRNA pseudouridylation, which is guided by snoRNAs, was intact. Moreover, first-generation Naf1(+/-) mice showed no evidence of ribosomal pathology. Our data indicate that disease in NAF1 mutation carriers is telomere-mediated; they show that NAF1 haploinsufficiency selectively disturbs telomere length homeostasis by decreasing the levels of telomerase RNA while sparing rRNA pseudouridylation.


Subject(s)
Emphysema/genetics , Pulmonary Fibrosis/genetics , RNA/genetics , Animals , Autophagy-Related Proteins , Carrier Proteins/genetics , Genetic Predisposition to Disease/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/genetics , Ribonucleoproteins/genetics , Telomerase/genetics , Telomere/genetics
SELECTION OF CITATIONS
SEARCH DETAIL