Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 98
1.
Eur J Med Genet ; 66(10): 104825, 2023 Oct.
Article En | MEDLINE | ID: mdl-37659595

Cholesterol is essential in the brain from the earliest stages of embryonic development. Disruption of cholesterol synthesis pathways that leads to cholesterol deficiency underlies a few syndromes, including desmosterolosis and Smith-Lemli-Opitz syndrome. In both syndromes, brain anomalies can occur. The LSS gene encodes lanosterol synthase (LSS), an important enzyme in the cholesterol biosynthesis pathway. Biallelic pathogenic variants in this gene cause alopecia-intellectual disability type 4 syndrome (APMR4, MIM 618840), a rare autosomal recessive disorder. Here, we describe two new LSS variants (c.1016C > T; p. Ser339Leu and c.1522G > C; p. Gly508Arg) found in a compound heterozygous fetus diagnosed prenatally with brain abnormalities by ultrasound scanning. Two of his siblings from the same parents also harbored these variants. Both siblings had alopecia, mild intellectual disability, autism spectrum disorder, and cataracts. To the best of our knowledge, this case represents the first prenatal diagnosis of APMR4 first suspected by ultrasound. In addition, the phenotypic features of the siblings are extensive compared with those described in previous reports and include abnormal corpus callosum, cataracts, alopecia, and developmental delay.


Autism Spectrum Disorder , Cataract , Intellectual Disability , Pregnancy , Female , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Prenatal Diagnosis , Alopecia/genetics , Cholesterol/genetics , Cholesterol/metabolism
2.
Prenat Diagn ; 42(7): 881-889, 2022 06.
Article En | MEDLINE | ID: mdl-34132406

OBJECTIVE: To explain the importance of identifying an etiology for the pathological finding of nonimmune hydrops fetalis (NIHF) and to explore the impact of exome sequencing in recurrent NIHF. In addition, we present two cases of pregnancies affected with recurrent NIHF, in which genetic investigation was advantageous. METHODS: Our study aimed to investigate the genetic background, if available, of all fetuses with NIHF referred to our tertiary medical center from January 2013 to August 2020. We summarized the etiology of NIHF if known, sonographic findings, genetic investigation and the pregnancies' outcomes. RESULTS: We encountered 144 families with NIHF. Genetic investigation was performed by chromosomal microarray analysis (CMA) in 63 (63/144. 44%) fetuses. Seventeen of 63 (27%) had a positive CMA result. In the negative CMA group, 15 (15/46, 33%) opted for exome sequencing, of which seven exomes were positive (47%). Among these, there were four couples with recurrent pregnancies affected by hydrops. Among the remaining 11 exome investigations for non-recurrent hydrops, another three were diagnostic. CONCLUSION: As identifying the etiology of the NIHF is an invaluable tool for the prognosis of the pregnancy, exome sequencing can provide further elucidation of the underlying pathogenesis of NIHF. Thus, genetic investigation should be recommended for cases of NIHF.


Exome , Hydrops Fetalis , Female , Fetus , Humans , Hydrops Fetalis/diagnosis , Hydrops Fetalis/genetics , Pregnancy , Pregnancy Outcome , Exome Sequencing
3.
J Clin Endocrinol Metab ; 107(5): 1346-1356, 2022 04 19.
Article En | MEDLINE | ID: mdl-34971397

CONTEXT: The hyperinsulinism/hyperammonemia (HI/HA) syndrome, the second-most common form of congenital hyperinsulinism, has been associated with dominant mutations in GLUD1, coding for the mitochondrial enzyme glutamate dehydrogenase, that increase enzyme activity by reducing its sensitivity to allosteric inhibition by GTP. OBJECTIVE: To identify the underlying genetic etiology in 2 siblings who presented with the biochemical features of HI/HA syndrome but did not carry pathogenic variants in GLUD1, and to determine the functional impact of the newly identified mutation. METHODS: The patients were investigated by whole exome sequencing. Yeast complementation studies and biochemical assays on the recombinant mutated protein were performed. The consequences of stable slc25a36 silencing in HeLa cells were also investigated. RESULTS: A homozygous splice site variant was identified in solute carrier family 25, member 36 (SLC25A36), encoding the pyrimidine nucleotide carrier 2 (PNC2), a mitochondrial nucleotide carrier that transports pyrimidine as well as guanine nucleotides across the inner mitochondrial membrane. The mutation leads to a 26-aa in-frame deletion in the first repeat domain of the protein, which abolishes transport activity. Furthermore, knockdown of slc25a36 expression in HeLa cells caused a marked reduction in the mitochondrial GTP content, which likely leads to a hyperactivation of glutamate dehydrogenase in our patients. CONCLUSION: We report for the first time a mutation in PNC2/SLC25A36 leading to HI/HA and provide functional evidence of the molecular mechanism responsible for this phenotype. Our findings underscore the importance of mitochondrial nucleotide metabolism and expand the role of mitochondrial transporters in insulin secretion.


Congenital Hyperinsulinism , Hyperammonemia , Hyperinsulinism , Congenital Hyperinsulinism/genetics , Glutamate Dehydrogenase/genetics , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Hyperammonemia/genetics , Hyperinsulinism/genetics , Hypoglycemia , Mutation , Nucleotides
4.
Metab Brain Dis ; 36(4): 581-588, 2021 04.
Article En | MEDLINE | ID: mdl-33471299

Isolated defects in the mitochondrial respiratory chain complex II (CII; succinate-ubiquinone oxidoreductase) are extremely rare and mainly result from bi-allelic mutations in one of the nuclear encoded subunits: SDHA, SDHB and SDHD, which comprise CII and the assembly CII factor SDHAF1. We report an adolescent female who presented with global developmental delay, intellectual disability and childhood onset progressive bilateral optic atrophy. Whole exome sequencing of the patient and her unaffected parents identified the novel heterozygous de novo variant c.1984C > T [NM_004168.4] in the SDHA gene. Biochemical assessment of CII in the patient's derived fibroblasts and lymphocytes displayed considerably decreased CII residual activity compared with normal controls, when normalized to the integral mitochondrial enzyme citrate synthase. Protein modeling of the consequent p.Arg662Cys variant [NP-004159.2] suggested that this substitution will compromise the structural integrity of the FAD-binding protein at the C-terminus that will ultimately impair the FAD binding to SDHA, thus decreasing the entire CII activity. Our study emphasizes the role of certain heterozygous SDHA mutations in a distinct clinical phenotype dominated by optic atrophy and neurological impairment. This is the second mutation that has been reported to cause this phenotype. Furthermore, it adds developmental delay and cognitive disability to the expanding spectrum of the disorder. We propose to add SDHA to next generation sequencing gene panels of optic atrophy.


Cognitive Dysfunction/genetics , Electron Transport Complex II/genetics , Genetic Variation/genetics , Heterozygote , Optic Atrophy/genetics , Adolescent , Amino Acid Sequence , Cognitive Dysfunction/complications , Cognitive Dysfunction/diagnostic imaging , Electron Transport Complex II/chemistry , Female , Humans , Optic Atrophy/complications , Optic Atrophy/diagnostic imaging , Protein Structure, Secondary
5.
J Inherit Metab Dis ; 44(3): 606-617, 2021 05.
Article En | MEDLINE | ID: mdl-33190319

Urea cycle disorders (UCDs), including OTC deficiency (OTCD), are life-threatening diseases with a broad clinical spectrum. Early diagnosis and initiation of treatment based on a newborn screening (NBS) test for OTCD with high specificity and sensitivity may contribute to reduction of the significant complications and high mortality. The efficacy of incorporating orotic acid determination into routine NBS was evaluated. Combined measurement of orotic acid and citrulline in archived dried blood spots from newborns with urea cycle disorders and normal controls was used to develop an algorithm for routine NBS for OTCD in Israel. Clinical information and genetic confirmation results were obtained from the follow-up care providers. About 1147986 newborns underwent routine NBS including orotic acid determination, 25 of whom were ultimately diagnosed with a UCD. Of 11 newborns with OTCD, orotate was elevated in seven but normal in two males with early-onset and two males with late-onset disease. Orotate was also elevated in archived dried blood spots of all seven retrospectively tested historical OTCD patients, only three of whom had originally been identified by NBS with low citrulline and elevated glutamine. Among the other UCDs emerge, three CPS1D cases and additional three retrospective CPS1D cases otherwise reported as a very rare condition. Combined levels of orotic acid and citrulline in routine NBS can enhance the detection of UCD, especially increasing the screening sensitivity for OTCD and differentiate it from CPS1D. Our data and the negligible extra cost for orotic acid determination might contribute to the discussion on screening for proximal UCDs in routine NBS.


Citrulline/blood , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Orotic Acid/blood , Urea Cycle Disorders, Inborn/diagnosis , Dried Blood Spot Testing , Female , Humans , Infant, Newborn , Israel/epidemiology , Male , Neonatal Screening , Ornithine Carbamoyltransferase Deficiency Disease/epidemiology , Retrospective Studies , Urea Cycle Disorders, Inborn/epidemiology
6.
J Clin Immunol ; 41(1): 147-162, 2021 01.
Article En | MEDLINE | ID: mdl-33111199

PURPOSE: Bacillus Calmette-Guerin (BCG) is a live attenuated vaccine with the potential of causing severe iatrogenic complications in patients with primary immunodeficiency diseases (PID) before and after hematopoietic stem cell transplantation (HSCT). We aim to investigate risk factors of post-HSCT BCG-related complications in PID patients. METHODS: A retrospective analysis of pediatric PID patients who had received the BCG vaccine and underwent HSCT at Hadassah-Hebrew University Medical Center, between 2007 and 2019. RESULTS: We found 15/36 (41.67%) patients who developed post-HSCT BCG-related complications. The most significant risk factor for developing BCG-related complications was T cell deficiency (47.6% of the non-complicated vs 83.3% of the BCGitis and 100% of the BCGosis groups had T cell lymphopenia, p = 0.013). None of the chronic granulomatous patients developed BCG-related manifestation post-transplant. Among T cell-deficient patients, lower NK (127 vs 698 cells/µl, p = 0.04) cell counts and NK-SCID were risk factors for ongoing post-HSCT BCGosis, as was pretransplant disseminated BCGosis (33.3% of patients with BCGosis vs none of the non-BCGosis patients, p = 0.04). Immune reconstitution inflammatory syndrome (IRIS) was observed in 3/5 patients with Omenn syndrome. Prophylactic antimycobacterial treatment was not proven effective. CONCLUSION: BCG vaccination can cause significant morbidity and mortality in the post-transplant T cell-deficient patient, especially in the presence of pre-transplant disease. Taking a detailed medical history prior to administering, the BCG vaccine is crucial for prevention of this complication.


BCG Vaccine/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Immunocompromised Host , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/immunology , BCG Vaccine/immunology , Biomarkers , Child, Preschool , Diagnosis, Differential , Disease Susceptibility , Female , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Infant, Newborn , Male , Patient Outcome Assessment , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/therapy , Retrospective Studies , Vaccination/adverse effects
7.
Nat Commun ; 11(1): 5518, 2020 11 02.
Article En | MEDLINE | ID: mdl-33139704

Full genome sequences are increasingly used to track the geographic spread and transmission dynamics of viral pathogens. Here, with a focus on Israel, we sequence 212 SARS-CoV-2 sequences and use them to perform a comprehensive analysis to trace the origins and spread of the virus. We find that travelers returning from the United States of America significantly contributed to viral spread in Israel, more than their proportion in incoming infected travelers. Using phylodynamic analysis, we estimate that the basic reproduction number of the virus was initially around 2.5, dropping by more than two-thirds following the implementation of social distancing measures. We further report high levels of transmission heterogeneity in SARS-CoV-2 spread, with between 2-10% of infected individuals resulting in 80% of secondary infections. Overall, our findings demonstrate the effectiveness of social distancing measures for reducing viral spread.


Betacoronavirus/genetics , Communicable Diseases, Imported/virology , Coronavirus Infections/transmission , Genome, Viral/genetics , Pneumonia, Viral/transmission , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Basic Reproduction Number/statistics & numerical data , COVID-19 , Child , Child, Preschool , Communicable Diseases, Imported/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/prevention & control , Female , Humans , Infant , Infant, Newborn , Israel/epidemiology , Male , Middle Aged , Pandemics/prevention & control , Phylogeny , Pneumonia, Viral/epidemiology , Pneumonia, Viral/prevention & control , Psychological Distance , RNA, Viral/genetics , SARS-CoV-2 , Sequence Analysis, RNA , United States , Young Adult
8.
Proc Natl Acad Sci U S A ; 117(36): 22113-22121, 2020 09 08.
Article En | MEDLINE | ID: mdl-32843346

RNA polymerase (Pol) III has a noncanonical role of viral DNA sensing in the innate immune system. This polymerase transcribes viral genomes to produce RNAs that lead to induction of type I interferons (IFNs). However, the genetic and functional links of Pol III to innate immunity in humans remain largely unknown. Here, we describe a rare homozygous mutation (D40H) in the POLR3E gene, coding for a protein subunit of Pol III, in a child with recurrent and systemic viral infections and Langerhans cell histiocytosis. Fibroblasts derived from the patient exhibit impaired induction of type I IFN and increased susceptibility to human cytomegalovirus (HCMV) infection. Cultured cell lines infected with HCMV show induction of POLR3E expression. However, induction is not restricted to DNA virus, as sindbis virus, an RNA virus, enhances the expression of this protein. Likewise, foreign nonviral DNA elevates the steady-state level of POLR3E and elicits promoter-dependent and -independent transcription by Pol III. Remarkably, the molecular mechanism underlying the D40H mutation of POLR3E involves the assembly of defective initiation complexes of Pol III. Our study links mutated POLR3E and Pol III to an innate immune deficiency state in humans.


Cytomegalovirus/physiology , Fibroblasts/immunology , Fibroblasts/virology , RNA Polymerase III/metabolism , Animals , Chlorocebus aethiops , Cytomegalovirus/immunology , Dendritic Cells , Gene Expression Regulation, Enzymologic , Humans , Mutation , RNA Polymerase III/genetics , Vero Cells
9.
Eur J Med Genet ; 63(6): 103901, 2020 Jun.
Article En | MEDLINE | ID: mdl-32151765

BACKGROUND: Maple syrup urine disease is a rare autosomal-recessive aminoacidopathy, caused by deficient branched-chain 2-keto acid dehydrogenase (BCKD), with subsequent accumulation of branched-chain amino acids (BCAAs): leucine, isoleucine and valine. While most cases of MSUD are classic, some 20% of cases are non-classic variants, designated as intermediate- or intermittent-types. Patients with the latter form usually develop normally and are cognitively intact, with normal BCAA levels when asymptomatic. However, intercurrent febrile illness and catabolism may cause metabolic derailment with life-threatening neurological sequelae. Thus, early detection and dietary intervention are warranted in intermittent MSUD. PATIENTS AND METHODS: We describe eight patients from four unrelated families, diagnosed with intermittent MSUD. Their presenting symptoms during metabolic crises varied from confusion and decreased consciousness, to ataxia, and acute psychosis. Molecular confirmation of MSUD was pursued via sequencing of the BCKDHA, BCKDHB and DBT genes. RESULTS: All affected individuals were found to harbor bi-allelic pathogenic variants in either BCKDHB or DBT. Of the seven variants, four variants in BCKDHB (p.G101D, p. V103A, p. A221D, p. Y195C) and one variant in DBT (p.K427E) were not previously described. CONCLUSIONS: While newborn screening programs allow for early detection of classic MSUD, cases of the intermittent form might go undetected, and present later in childhood following metabolic derailment, with an array of non-specific symptoms. Our experience with the families reported herein adds to the current knowledge regarding the phenotype and mutational spectrum of this unique inborn error of branched-chain amino acid metabolism, and underscore the high index of suspicion required for its diagnosis.


Genetic Testing/methods , Maple Syrup Urine Disease/diagnosis , Mutation , Phenotype , Adolescent , Child , Child, Preschool , Disease Progression , Female , Genetic Testing/standards , Humans , Male , Maple Syrup Urine Disease/genetics , Protein Kinases/genetics
10.
Am J Med Genet A ; 182(4): 689-696, 2020 04.
Article En | MEDLINE | ID: mdl-32027463

Trio exome sequencing is a powerful tool in the molecular investigation of monogenic disorders and provides an incremental diagnostic yield over proband-only sequencing, mainly due to the rapid identification of de novo disease-causing variants. However, heterozygous variants inherited from unaffected parents may be inadvertently dismissed, although multiple explanations are available for such scenarios including mosaicism in the parent, incomplete penetrance, imprinting, or skewed X-inactivation. We report three probands, in which a pathogenic or likely pathogenic variant was identified upon exome sequencing, yet was inherited from an unaffected parent. Segregation of the variants (in NOTCH1, PHF6, and SOX10) in the grandparent generation revealed that the variant was de novo in each case. Additionally, one proband had skewed X-inactivation. We discuss the possible genetic mechanism in each case, and urge caution in data interpretation of exome sequencing data. We illustrate the utility of expanding segregation studies to the grandparent generation and demonstrate the impact on exome interpretation strategies, by showing that objective genotype data can overcome subjective parental report of lack of symptoms.


Epilepsy/genetics , Exome/genetics , Genetic Predisposition to Disease , Grandparents , Hypoplastic Left Heart Syndrome/genetics , Mutation , Adult , Child , Epilepsy/pathology , Female , Genetic Testing , Genotype , Humans , Hypoplastic Left Heart Syndrome/pathology , Infant, Newborn , Male , Mosaicism , Parents , Receptor, Notch1/genetics , Repressor Proteins/genetics , SOXE Transcription Factors/genetics , Exome Sequencing
11.
Am J Hum Genet ; 105(1): 48-64, 2019 07 03.
Article En | MEDLINE | ID: mdl-31178128

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.


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
12.
Nat Commun ; 10(1): 605, 2019 02 05.
Article En | MEDLINE | ID: mdl-30723199

Nuclear envelopathies comprise a heterogeneous group of diseases caused by mutations in genes encoding nuclear envelope proteins. Mutations affecting lamina-associated polypeptide 1 (LAP1) result in two discrete phenotypes of muscular dystrophy and progressive dystonia with cerebellar atrophy. We report 7 patients presenting at birth with severe progressive neurological impairment, bilateral cataract, growth retardation and early lethality. All the patients are homozygous for a nonsense mutation in the TOR1AIP1 gene resulting in the loss of both protein isoforms LAP1B and LAP1C. Patient-derived fibroblasts exhibit changes in nuclear envelope morphology and large nuclear-spanning channels containing trapped cytoplasmic organelles. Decreased and inefficient cellular motility is also observed in these fibroblasts. Our study describes the complete absence of both major human LAP1 isoforms, underscoring their crucial role in early development and organogenesis. LAP1-associated defects may thus comprise a broad clinical spectrum depending on the availability of both isoforms in the nuclear envelope throughout life.


Abnormalities, Multiple/genetics , Genetic Predisposition to Disease/genetics , Membrane Proteins/genetics , Mutation , Nuclear Envelope/genetics , Nuclear Proteins/genetics , Abnormalities, Multiple/metabolism , Base Sequence , Child , Child, Preschool , Cytoskeletal Proteins , DNA Mutational Analysis , Fatal Outcome , Female , Humans , Male , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
13.
Metab Brain Dis ; 34(2): 557-563, 2019 04.
Article En | MEDLINE | ID: mdl-30637540

D-glycerate 2 kinase (DGK) is an enzyme that mediates the conversion of D-glycerate, an intermediate metabolite of serine and fructose metabolism, to 2-phosphoglycerate. Deficiency of DGK leads to accumulation of D-glycerate in various tissues and its massive excretion in urine. D-glyceric aciduria (DGA) is an autosomal recessive metabolic disorder caused by mutations in the GLYCTK gene. The clinical spectrum of DGA is highly variable, ranging from severe progressive infantile encephalopathy to a practically asymptomatic condition. We describe a male patient from a consanguineous Arab family with infantile onset of DGA, characterized by profound psychomotor retardation, progressive microcephaly, intractable seizures, cortical blindness and deafness. Consecutive brain MR imaging showed an evolving brain atrophy, thinning of the corpus callosum and diffuse abnormal white matter signals. Whole exome sequencing identified the homozygous missense variant in the GLYCTK gene [c.455 T > C, NM_145262.3], which affected a highly conserved leucine residue located at a domain of yet unknown function of the enzyme [p.Leu152Pro, NP_660305]. In silico analysis of the variant supported its pathogenicity. A review of the 15 previously reported patients, together with the current one, confirms a clear association between DGA and severe neurological impairment. Yet, future studies of additional patients with DGA are required to better understand the clinical phenotype and pathogenesis.


Brain Diseases/metabolism , Epilepsy/metabolism , Hyperoxaluria, Primary/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Brain Diseases/genetics , Child , Epilepsy/diagnosis , Epilepsy/genetics , Glyceric Acids/metabolism , Humans , Hyperoxaluria, Primary/genetics , Infant , Male , Mutation/genetics , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Spasms, Infantile/genetics , Spasms, Infantile/metabolism
14.
Am J Hum Genet ; 104(1): 179-185, 2019 01 03.
Article En | MEDLINE | ID: mdl-30595371

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) initiates a stress response mechanism to clear out the unfolded proteins by either facilitating their re-folding or inducing their degradation. When this fails, an apoptotic cascade is initiated so that the affected cell is eliminated. IRE1α is a critical sensor of the unfolded-protein response, essential for initiating the apoptotic signaling. Here, we report an infantile neurodegenerative disorder associated with enhanced activation of IRE1α and increased apoptosis. Three unrelated affected individuals with congenital microcephaly, infantile epileptic encephalopathy, and profound developmental delay were found to carry heterozygous variants (c.932T>C [p.Leu311Ser] or c.935T>C [p.Leu312Pro]) in RNF13, which codes for an IRE1α-interacting protein. Structural modeling predicted that the variants, located on the surface of the protein, would not alter overall protein folding. Accordingly, the abundance of RNF13 and IRE1α was not altered in affected individuals' cells. However, both IRE1α-mediated stress signaling and stress-induced apoptosis were increased in affected individuals' cells. These results indicate that the RNF13 variants confer gain of function to the encoded protein and thereby lead to altered signaling of the ER stress response associated with severe neurodegeneration in infancy.


Blindness/congenital , Blindness/genetics , Failure to Thrive/genetics , Gain of Function Mutation , Heterozygote , Microcephaly/genetics , Spasms, Infantile/genetics , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Apoptosis , Child , Child, Preschool , Developmental Disabilities/genetics , Endoplasmic Reticulum Stress , Humans , Infant , Male , Models, Molecular , Spasms, Infantile/congenital , Ubiquitin-Protein Ligases/chemistry , Unfolded Protein Response
15.
PLoS Genet ; 14(8): e1007602, 2018 08.
Article En | MEDLINE | ID: mdl-30148830

The clinical spectrum of ciliopathies affecting motile cilia spans impaired mucociliary clearance in the respiratory system, laterality defects including heart malformations, infertility and hydrocephalus. Using linkage analysis and whole exome sequencing, we identified two recessive loss-of-function MNS1 mutations in five individuals from four consanguineous families: 1) a homozygous nonsense mutation p.Arg242* in four males with laterality defects and infertility and 2) a homozygous nonsense mutation p.Gln203* in one female with laterality defects and recurrent respiratory infections additionally carrying homozygous mutations in DNAH5. Consistent with the laterality defects observed in these individuals, we found Mns1 to be expressed in mouse embryonic ventral node. Immunofluorescence analysis further revealed that MNS1 localizes to the axonemes of respiratory cilia as well as sperm flagella in human. In-depth ultrastructural analyses confirmed a subtle outer dynein arm (ODA) defect in the axonemes of respiratory epithelial cells resembling findings reported in Mns1-deficient mice. Ultrastructural analyses in the female carrying combined mutations in MNS1 and DNAH5 indicated a role for MNS1 in the process of ODA docking (ODA-DC) in the distal respiratory axonemes. Furthermore, co-immunoprecipitation and yeast two hybrid analyses demonstrated that MNS1 dimerizes and interacts with the ODA docking complex component CCDC114. Overall, we demonstrate that MNS1 deficiency in humans causes laterality defects (situs inversus) and likely male infertility and that MNS1 plays a role in the ODA-DC assembly.


Codon, Nonsense , Functional Laterality/genetics , Homozygote , Infertility, Male/genetics , Nuclear Proteins/metabolism , Adolescent , Adult , Animals , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Axoneme/metabolism , Cell Cycle Proteins , Child , Child, Preschool , Cilia/ultrastructure , Female , Gene Expression Regulation , Genetic Linkage , Humans , Infant , Male , Mice , Mice, Knockout , Middle Aged , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Pedigree , Polymorphism, Single Nucleotide , Sperm Tail , Exome Sequencing , Young Adult
16.
Neurogenetics ; 19(4): 227-235, 2018 12.
Article En | MEDLINE | ID: mdl-30043326

The major facilitator superfamily domain-containing protein 2A (MFSD2A) is a constituent of the blood-brain barrier and functions to transport lysophosphatidylcholines (LPCs) into the central nervous system. LPCs such as that derived from docosahexanoic acid (DHA) are indispensable to neurogenesis and maintenance of neurons, yet cannot be synthesized within the brain and are dependent on MFSD2A for brain uptake. Recent studies have implicated MFSD2A mutations in lethal and non-lethal microcephaly syndromes, with the severity correlating to the residual activity of the transporter. We describe two siblings with shared parental ancestry, in whom we identified a homozygous missense mutation (c.1205C > A; p.Pro402His) in MFSD2A. Both affected individuals had microcephaly, hypotonia, appendicular spasticity, dystonia, strabismus, and global developmental delay. Neuroimaging revealed paucity of white matter with enlarged lateral ventricles. Plasma lysophosphatidylcholine (LPC) levels were elevated, reflecting reduced brain transport. Cell-based studies of the p.Pro402His mutant protein indicated complete loss of activity of the transporter despite the non-lethal, attenuated phenotype. The aggregate data of MFSD2A-associated genotypes and phenotypes suggest that additional factors, such as nutritional supplementation or modifying genetic factors, may modulate the severity of disease and call for consideration of treatment options for affected individuals.


Demyelinating Diseases/genetics , Docosahexaenoic Acids/metabolism , Microcephaly/genetics , Mutation, Missense , Tumor Suppressor Proteins/genetics , Amino Acid Substitution , Animals , Biological Transport/genetics , Blood-Brain Barrier/metabolism , Child , Child, Preschool , Demyelinating Diseases/metabolism , Developmental Disabilities/genetics , Female , HEK293 Cells , Homozygote , Humans , Lipid Metabolism/genetics , Lysophosphatidylcholines/metabolism , Male , Mice , Mice, Knockout , Microcephaly/metabolism , Models, Molecular , Myelin Sheath/metabolism , Pedigree , Siblings , Symporters , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
17.
Eur J Med Genet ; 61(10): 616-620, 2018 Oct.
Article En | MEDLINE | ID: mdl-29655802

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed enzymes responsible for charging tRNA with cognate amino acids during protein translation. Non-canonical functions are increasingly recognized, and include transcription and translation control and extracellular signaling. Monoallelic mutations in genes encoding several ARSs have been identified in axonal Charcot-Marie-Tooth (CMT2) disease, whereas biallelic mutations in ARS loci have been associated with multi-tissue syndromes, variably involving the central nervous system, lung, and liver. We report a male infant of non-consanguineous origin, presenting with successive onset of transfusion-dependent anemia, hypothyroidism, cholestasis, interstitial lung disease, and developmental delay. Whole-exome sequencing (WES) revealed compound heterozygosity for two variants (p.Tyr307Cys and p.Arg618Cys) in MARS, encoding methionyl-tRNA synthetase. Biallelic MARS mutations are associated with interstitial lung and liver disease (ILLD). Interestingly, the p.Arg618Cys variant, inherited from an unaffected father, was previously reported in a family with autosomal dominant late-onset CMT2. Yeast complementation assays confirmed pathogenicity of p.Arg618Cys, yet suggested retained function of p.Tyr307Cys. Our findings underscore the phenotypic variability associated with ARS mutations, and suggest genetic or environmental modifying factors in the onset of monoallelic MARS-associated CMT2.


Charcot-Marie-Tooth Disease/genetics , Cholestasis/genetics , Developmental Disabilities/genetics , Lung Diseases, Interstitial/genetics , Methionine-tRNA Ligase/genetics , Mutation, Missense , Phenotype , Cholestasis/diagnosis , Developmental Disabilities/pathology , Genes, Dominant , Genes, Recessive , Humans , Infant , Lung Diseases, Interstitial/diagnosis , Male , Pedigree , Saccharomyces cerevisiae/genetics , Syndrome
18.
Am J Med Genet A ; 176(1): 92-98, 2018 01.
Article En | MEDLINE | ID: mdl-29130579

The primary cilium is a key organelle in numerous physiological and developmental processes. Genetic defects in the formation of this non-motile structure, in its maintenance and function, underlie a wide array of ciliopathies in human, including craniofacial, brain and heart malformations, and retinal and hearing defects. We used exome sequencing to study the molecular basis of disease in an 11-year-old female patient who suffered from growth retardation, global developmental delay with absent speech acquisition, agenesis of corpus callosum and paucity of white matter, sensorineural deafness, retinitis pigmentosa, vertebral anomalies, patent ductus arteriosus, and facial dysmorphism reminiscent of STAR syndrome, a suspected ciliopathy. A homozygous variant, c.870_871del, was identified in the CDK10 gene, predicted to cause a frameshift, p.Trp291Alafs*18, in the cyclin-dependent kinase 10 protein. CDK10 mRNAs were detected in patient cells and do not seem to undergo non-sense mediated decay. CDK10 is the binding partner of Cyclin M (CycM) and CDK10/CycM protein kinase regulates ciliogenesis and primary cilium elongation. Notably, CycM gene is mutated in patients with STAR syndrome. Following incubation, the patient cells appeared less elongated and more densely populated than the control cells suggesting that the CDK10 mutation affects the cytoskeleton. Upon starvation and staining with acetylated-tubulin, γ-tubulin, and Arl13b, the patient cells exhibited fewer and shorter cilia than control cells. These findings underscore the importance of CDK10 for the regulation of ciliogenesis. CDK10 defect is likely associated with a new form of ciliopathy phenotype; additional patients may further validate this association.


Agenesis of Corpus Callosum/genetics , Cyclin-Dependent Kinases , Deafness/genetics , Genetic Association Studies , Homozygote , Mutation , Retinal Dysplasia/genetics , Agenesis of Corpus Callosum/diagnosis , Alleles , Brain/abnormalities , Brain/diagnostic imaging , Child , DNA Mutational Analysis , Deafness/diagnosis , Exome , Facies , Female , Gene Expression , Humans , Pedigree , Phenotype , RNA, Messenger/genetics , Retinal Dysplasia/diagnosis
19.
Am J Hum Genet ; 101(2): 267-273, 2017 Aug 03.
Article En | MEDLINE | ID: mdl-28777933

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.


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
20.
Am J Hum Genet ; 101(2): 291-299, 2017 Aug 03.
Article En | MEDLINE | ID: mdl-28777934

Progressive childhood encephalopathy is an etiologically heterogeneous condition characterized by progressive central nervous system dysfunction in association with a broad range of morbidity and mortality. The causes of encephalopathy can be either non-genetic or genetic. Identifying the genetic causes and dissecting the underlying mechanisms are critical to understanding brain development and improving treatments. Here, we report that variants in TRAPPC12 result in progressive childhood encephalopathy. Three individuals from two unrelated families have either a homozygous deleterious variant (c.145delG [p.Glu49Argfs∗14]) or compound-heterozygous variants (c.360dupC [p.Glu121Argfs∗7] and c.1880C>T [p. Ala627Val]). The clinical phenotypes of the three individuals are strikingly similar: severe disability, microcephaly, hearing loss, spasticity, and characteristic brain imaging findings. Fibroblasts derived from all three individuals showed a fragmented Golgi that could be rescued by expression of wild-type TRAPPC12. Protein transport from the endoplasmic reticulum to and through the Golgi was delayed. TRAPPC12 is a member of the TRAPP protein complex, which functions in membrane trafficking. Variants in several other genes encoding members of the TRAPP complex have been associated with overlapping clinical presentations, indicating shared and distinct functions for each complex member. Detailed understanding of the TRAPP-opathies will illuminate the role of membrane protein transport in human disease.


Brain Diseases/genetics , Endoplasmic Reticulum/metabolism , Golgi Apparatus/pathology , Membrane Transport Proteins/genetics , Protein Transport/genetics , Transcription Factors/genetics , Atrophy/pathology , Base Sequence , Brain/pathology , Brain Diseases/pathology , Cells, Cultured , Child, Preschool , Exome/genetics , Female , Genetic Predisposition to Disease , Humans , Infant , Magnetic Resonance Imaging , Male , Protein Transport/physiology , Sequence Analysis, DNA
...