Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
medRxiv ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38370739

ABSTRACT

Background and aims: Inflammatory Bowel Diseases (IBD) are chronic inflammatory conditions influenced heavily by environmental factors. DNA methylation is a form of epigenetic regulation linking environmental stimuli to gene expression changes and inflammation. Here, we investigated how DNA methylation of the TNF promoter differs between inflamed and uninflamed mucosa of IBD patients, including anti-TNF responders and non-responders. Methods: We obtained mucosal biopsies from 200 participants (133 IBD and 67 controls) and analyzed TNF promoter methylation using bisulfite sequencing, comparing inflamed with uninflamed segments, in addition to paired inflamed/uninflamed samples from individual patients. We conducted similar analyses on purified intestinal epithelial cells from bowel resections. We also compared TNF methylation levels of inflamed and uninflamed mucosa from a separate cohort of 15 anti-TNF responders and 17 non-responders. Finally, we sequenced DNA methyltransferase genes to identify rare variants in IBD patients and functionally tested them using rescue experiments in a zebrafish genetic model of DNA methylation deficiency. Results: TNF promoter methylation levels were decreased in inflamed mucosa of IBD patients and correlated with disease severity. Isolated IECs from inflamed tissue showed proportional decreases in TNF methylation. Anti-TNF non-responders showed lower levels of TNF methylation than responders in uninflamed mucosa. Our sequencing analysis revealed two missense variants in DNMT1, one of which had reduced function in vivo. Conclusions: Our study reveals an association of TNF promoter hypomethylation with mucosal inflammation, suggesting that IBD patients may be particularly sensitive to inflammatory environmental insults affecting DNA methylation. Together, our analyses indicate that TNF promoter methylation analysis may aid in the characterization of IBD status and evaluation of anti-TNF therapy response.

2.
Nat Commun ; 14(1): 5671, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37704658

ABSTRACT

The primary cilium is a signaling organelle with a unique membrane composition maintained by a diffusional barrier residing at the transition zone. Many transition zone proteins, such as the tectonic complex, are linked to preserving ciliary composition but the mechanism remains unknown. To understand tectonic's role, we generate a photoreceptor-specific Tctn1 knockout mouse. Loss of Tctn1 results in the absence of the entire tectonic complex and associated MKS proteins yet has minimal effects on the transition zone structure of rod photoreceptors. We find that the protein composition of the photoreceptor cilium is disrupted as non-resident membrane proteins accumulate in the cilium over time, ultimately resulting in photoreceptor degeneration. We further show that fluorescent rhodopsin moves faster through the transition zone in photoreceptors lacking tectonic, which suggests that the tectonic complex acts as a physical barrier to slow down membrane protein diffusion in the photoreceptor transition zone to ensure proper removal of non-resident membrane proteins.


Subject(s)
Cilia , Membrane Proteins , Animals , Mice , Membrane Proteins/genetics , Rhodopsin/genetics , Neurites , Coloring Agents , Mice, Knockout
3.
J Clin Invest ; 133(8)2023 04 17.
Article in English | MEDLINE | ID: mdl-36862503

ABSTRACT

Defects in primary or motile cilia result in a variety of human pathologies, and retinal degeneration is frequently associated with these so-called ciliopathies. We found that homozygosity for a truncating variant in CEP162, a centrosome and microtubule-associated protein required for transition zone assembly during ciliogenesis and neuronal differentiation in the retina, caused late-onset retinitis pigmentosa in 2 unrelated families. The mutant CEP162-E646R*5 protein was expressed and properly localized to the mitotic spindle, but it was missing from the basal body in primary and photoreceptor cilia. This impaired recruitment of transition zone components to the basal body and corresponded to complete loss of CEP162 function at the ciliary compartment, reflected by delayed formation of dysmorphic cilia. In contrast, shRNA knockdown of Cep162 in the developing mouse retina increased cell death, which was rescued by expression of CEP162-E646R*5, indicating that the mutant retains its role for retinal neurogenesis. Human retinal degeneration thus resulted from specific loss of the ciliary function of CEP162.


Subject(s)
Retinal Degeneration , Animals , Humans , Mice , Centrosome/metabolism , Cilia/metabolism , Microtubule-Associated Proteins/genetics , Neurogenesis/genetics , Retina/metabolism , Retinal Degeneration/metabolism
4.
Elife ; 122023 01 04.
Article in English | MEDLINE | ID: mdl-36598133

ABSTRACT

The small GTPase Arl3 is important for the enrichment of lipidated proteins to primary cilia, including the outer segment of photoreceptors. Human mutations in the small GTPase Arl3 cause both autosomal recessive and dominant inherited retinal dystrophies. We discovered that dominant mutations result in increased active G-protein-Arl3-D67V has constitutive activity and Arl3-Y90C is fast cycling-and their expression in mouse rods resulted in a displaced nuclear phenotype due to an aberrant Arl3-GTP gradient. Using multiple strategies, we go on to show that removing or restoring the Arl3-GTP gradient within the cilium is sufficient to rescue the nuclear migration defect. Together, our results reveal that an Arl3 ciliary gradient is involved in proper positioning of photoreceptor nuclei during retinal development.


Subject(s)
ADP-Ribosylation Factors , Membrane Proteins , Retinal Rod Photoreceptor Cells , Animals , Humans , Mice , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Cilia/metabolism , Guanosine Triphosphate/metabolism , Membrane Proteins/metabolism , Protein Transport , Retinal Rod Photoreceptor Cells/metabolism
5.
J Neurosci ; 41(14): 3094-3104, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33637563

ABSTRACT

Vision begins when light is captured by the outer segment organelle of photoreceptor cells in the retina. Outer segments are modified cilia filled with hundreds of flattened disk-shaped membranes. Disk membranes are separated from the surrounding plasma membrane, and each membrane type has unique protein components. The mechanisms underlying this protein sorting remain entirely unknown. In this study, we investigated the outer segment delivery of the rod cyclic nucleotide-gated (CNG) channel, which is located in the outer segment plasma membrane, where it mediates the electrical response to light. Using Xenopus and mouse models of both sexes, we now show that the targeted delivery of the CNG channel to the outer segment uses the conventional secretory pathway, including protein processing in both ER and Golgi, and requires preassembly of its constituent α1 and ß1 subunits. We further demonstrate that the N-terminal glutamic acid-rich protein (GARP) domain of CNGß1 contains two distinct functional regions. The glutamic acid-rich region encodes specific information targeting the channel to rod outer segments. The adjacent proline-enriched region connects the CNG channel to photoreceptor disk rims, likely through an interaction with peripherin-2. These data reveal fine functional specializations within the structural domains of the CNG channel and suggest that its sequestration to the outer segment plasma membrane requires an interaction with peripherin-2.SIGNIFICANCE STATEMENT Neurons and other differentiated cells have a remarkable ability to deliver and organize signaling proteins at precise subcellular locations. We now report that the CNG channel, mediating the electrical response to light in rod photoreceptors, contains two specialized regions within the N terminus of its ß-subunit: one responsible for delivery of this channel to the ciliary outer segment organelle and another for subsequent channel sequestration into the outer segment plasma membrane. These findings expand our understanding of the molecular specializations used by neurons to populate their critical functional compartments.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Domains/physiology , Rod Cell Outer Segment/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Binding Sites/physiology , Cyclic Nucleotide-Gated Cation Channels/chemistry , Female , Male , Membrane Proteins/chemistry , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Rod Cell Outer Segment/chemistry , Xenopus
6.
Hum Genomics ; 13(1): 19, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992063

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis [1] is a genetically heterogeneous neurodegenerative disorder, characterized by late-onset degeneration of motor neurons leading to progressive limb and bulbar weakness, as well as of the respiratory muscles, which is the primary cause of disease fatality. To date, over 25 genes have been implicated as causative in ALS with C9orf72, SOD1, FUS, and TARDBP accounting for the majority of genetically positive cases. RESULTS: We identified two patients of Italian and French ancestry with a clinical diagnosis of juvenile-onset ALS who were mutation-negative in any of the known ALS causative genes. Starting with the index case, a consanguineous family of Italian origin, we performed whole-exome sequencing and identified candidate pathogenic mutations in 35 genes, 27 of which were homozygous. We next parsed all candidates against a cohort of 3641 ALS cases; only ATP13A2 was found to harbor recessive changes, in a patient with juvenile-onset ALS, similar to the index case. In vivo complementation of ATP13A2 using a zebrafish surrogate model that focused on the assessment of motor neuron morphology and cerebellar integrity confirmed the role of this gene in central and peripheral nervous system maintenance and corroborated the damaging direction of effect of the change detected in the index case of this study. CONCLUSIONS: We here expand the phenotypic spectrum associated with genetic variants in ATP13A2 that previously comprised Kufor-Rakeb syndrome, spastic paraplegia 78, and neuronal ceroid lipofuscinosis type 12 (CLN12), to also include juvenile-onset ALS, as supported by both genetic and functional data. Our findings highlight the importance of establishing a complete genetic profile towards obtaining an accurate clinical diagnosis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genetic Predisposition to Disease , Proton-Translocating ATPases/genetics , Adult , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Female , Humans , Male , Middle Aged , Motor Neurons/pathology , Mutation/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Pedigree , Phenotype , Exome Sequencing , Zebrafish
7.
Hum Genomics ; 11(1): 16, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724397

ABSTRACT

BACKGROUND: The ciliopathies represent an umbrella group of >50 clinical entities that share both clinical features and molecular etiology underscored by structural and functional defects of the primary cilium. Despite the advances in gene discovery, this group of entities continues to pose a diagnostic challenge, in part due to significant genetic and phenotypic heterogeneity and variability. We consulted a pediatric case from asymptomatic, non-consanguineous parents who presented as a suspected ciliopathy due to a constellation of retinal, renal, and skeletal findings. RESULTS: Although clinical panel sequencing of genes implicated in nephrotic syndromes yielded no likely causal mutation, an oligo-SNP microarray identified a ~20-Mb region of homozygosity, with no altered gene dosage, on chromosome 16p13. Intersection of the proband's phenotypes with known disease genes within the homozygous region yielded a single candidate, IFT140, encoding a retrograde intraflagellar transport protein implicated previously in several ciliopathies, including the phenotypically overlapping Mainzer-Saldino syndrome (MZSDS). Sanger sequencing yielded a maternally inherited homozygous c.634G>A; p.Gly212Arg mutation altering the exon 6 splice donor site. Functional studies in cells from the proband showed that the locus produced two transcripts: a majority message containing a mis-splicing event that caused a premature termination codon and a minority message homozygous for the p.Gly212Arg allele. Zebrafish in vivo complementation studies of the latter transcript demonstrated a loss of function effect. Finally, we conducted post-hoc trio-based whole exome sequencing studies to (a) test the possibility of other causal loci in the proband and (b) explain the Mendelian error of segregation for the IFT140 mutation. We show that the proband harbors a chromosome 16 maternal heterodisomy, with segmental isodisomy at 16p13, likely due to a meiosis I error in the maternal gamete. CONCLUSIONS: Using clinical phenotyping combined with research-based genetic and functional studies, we have characterized a recurrent IFT140 mutation in the proband; together, these data are consistent with MZSDS. Additionally, we report a rare instance of a uniparental isodisomy unmasking a deleterious mutation to cause a ciliary disorder.


Subject(s)
B-Lymphocytes/pathology , Carrier Proteins/genetics , Cerebellar Ataxia/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Animals , B-Lymphocytes/metabolism , Cells, Cultured , Cerebellar Ataxia/pathology , Child, Preschool , Chromosomes, Human, Pair 16 , Exons , Female , Homozygote , Humans , Male , Pedigree , Phenotype , Retinitis Pigmentosa/pathology , Uniparental Disomy , Zebrafish/metabolism
9.
Am J Hum Genet ; 100(4): 666-675, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28318500

ABSTRACT

Despite the accelerated discovery of genes associated with syndromic traits, the majority of families affected by such conditions remain undiagnosed. Here, we employed whole-exome sequencing in two unrelated consanguineous kindreds with central nervous system (CNS), cardiac, renal, and digit abnormalities. We identified homozygous truncating mutations in TMEM260, a locus predicted to encode numerous splice isoforms. Systematic expression analyses across tissues and developmental stages validated two such isoforms, which differ in the utilization of an internal exon. The mutations in both families map uniquely to the long isoform, raising the possibility of an isoform-specific disorder. Consistent with this notion, RT-PCR of lymphocyte cell lines from one of the kindreds showed reduced levels of only the long isoform, which could be ameliorated by emetine, suggesting that the mutation induces nonsense-mediated decay. Subsequent in vivo testing supported this hypothesis. First, either transient suppression or CRISPR/Cas9 genome editing of zebrafish tmem260 recapitulated key neurological phenotypes. Second, co-injection of morphants with the long human TMEM260 mRNA rescued CNS pathology, whereas the short isoform was significantly less efficient. Finally, immunocytochemical and biochemical studies showed preferential enrichment of the long TMEM260 isoform to the plasma membrane. Together, our data suggest that there is overall reduced, but not ablated, functionality of TMEM260 and that attenuation of the membrane-associated functions of this protein is a principal driver of pathology. These observations contribute to an appreciation of the roles of splice isoforms in genetic disorders and suggest that dissection of the functions of these transcripts will most likely inform pathomechanism.


Subject(s)
Abnormalities, Multiple/genetics , Cardio-Renal Syndrome/genetics , Membrane Proteins/genetics , Neurodevelopmental Disorders/genetics , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Pedigree , Point Mutation , Protein Isoforms/genetics
10.
Invest Ophthalmol Vis Sci ; 58(3): 1570-1576, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28282489

ABSTRACT

Purpose: Genome-wide association (GWAS) and sequencing studies for AMD have highlighted the importance of coding variants at loci that encode components of the complement pathway. However, assessing the contribution of such alleles to AMD, especially when they are rare, remains coarse, in part because of the persistent challenge in establishing their functional relevance. Others and we have shown previously that rare alleles in complement factor I (CFI) can be tested functionally using a surrogate in vivo assay of retinal vascularization in zebrafish embryos. Here, we have implemented and scaled these tools to assess the overall contribution of rare alleles in CFI to AMD. Methods: We performed targeted sequencing of CFI in 731 AMD patients, followed by replication in a second patient cohort of 511 older healthy individuals. Systematic functional testing of all alleles and post-hoc statistical analysis of functional variants was also performed. Results: We discovered 20 rare coding nonsynonymous variants, including the previously reported G119R allele. In vivo testing led to the identification of nine variants that alter CFI; six of which are associated with hypoactive complement factor I (FI). Post-hoc analysis in ethnically matched, population controls showed six of these to be present exclusively in cases. Conclusions: Taken together, our data argue that multiple rare and ultra-rare alleles in CFI contribute to AMD pathogenesis; they improve the precision of the assessment of the contribution of CFI to AMD; and they offer a rational route to establishing both causality and direction of allele effect for genes associated with this disorder.


Subject(s)
Complement Factor I/genetics , DNA/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Macular Degeneration/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Alleles , Animals , Complement Factor I/metabolism , Female , Genotype , Humans , Macular Degeneration/diagnosis , Macular Degeneration/metabolism , Male , Middle Aged , Zebrafish/embryology
11.
Nat Genet ; 49(2): 238-248, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067909

ABSTRACT

Arhinia, or absence of the nose, is a rare malformation of unknown etiology that is often accompanied by ocular and reproductive defects. Sequencing of 40 people with arhinia revealed that 84% of probands harbor a missense mutation localized to a constrained region of SMCHD1 encompassing the ATPase domain. SMCHD1 mutations cause facioscapulohumeral muscular dystrophy type 2 (FSHD2) via a trans-acting loss-of-function epigenetic mechanism. We discovered shared mutations and comparable DNA hypomethylation patterning between these distinct disorders. CRISPR/Cas9-mediated alteration of smchd1 in zebrafish yielded arhinia-relevant phenotypes. Transcriptome and protein analyses in arhinia probands and controls showed no differences in SMCHD1 mRNA or protein abundance but revealed regulatory changes in genes and pathways associated with craniofacial patterning. Mutations in SMCHD1 thus contribute to distinct phenotypic spectra, from craniofacial malformation and reproductive disorders to muscular dystrophy, which we speculate to be consistent with oligogenic mechanisms resulting in pleiotropic outcomes.


Subject(s)
Choanal Atresia/genetics , Chromosomal Proteins, Non-Histone/genetics , Genetic Predisposition to Disease/genetics , Microphthalmos/genetics , Muscular Dystrophies/genetics , Mutation/genetics , Nose/abnormalities , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Phenotype
12.
Am J Hum Genet ; 99(2): 318-36, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27486776

ABSTRACT

Bardet-Biedl syndrome (BBS) is a defining ciliopathy, notable for extensive allelic and genetic heterogeneity, almost all of which has been identified through sequencing. Recent data have suggested that copy-number variants (CNVs) also contribute to BBS. We used a custom oligonucleotide array comparative genomic hybridization (aCGH) covering 20 genes that encode intraflagellar transport (IFT) components and 74 ciliopathy loci to screen 92 unrelated individuals with BBS, irrespective of their known mutational burden. We identified 17 individuals with exon-disruptive CNVs (18.5%), including 13 different deletions in eight BBS genes (BBS1, BBS2, ARL6/BBS3, BBS4, BBS5, BBS7, BBS9, and NPHP1) and a deletion and a duplication in other ciliopathy-associated genes (ALMS1 and NPHP4, respectively). By contrast, we found a single heterozygous exon-disruptive event in a BBS-associated gene (BBS9) in 229 control subjects. Superimposing these data with resequencing revealed CNVs to (1) be sufficient to cause disease, (2) Mendelize heterozygous deleterious alleles, and (3) contribute oligogenic alleles by combining point mutations and exonic CNVs in multiple genes. Finally, we report a deletion and a splice site mutation in IFT74, inherited under a recessive paradigm, defining a candidate BBS locus. Our data suggest that CNVs contribute pathogenic alleles to a substantial fraction of BBS-affected individuals and highlight how either deletions or point mutations in discrete splice isoforms can induce hypomorphic mutations in genes otherwise intolerant to deleterious variation. Our data also suggest that CNV analyses and resequencing studies unbiased for previous mutational burden is necessary to delineate the complexity of disease architecture.


Subject(s)
Bardet-Biedl Syndrome/genetics , DNA Copy Number Variations/genetics , Mutation , Adolescent , Adult , Alleles , Animals , Child , Child, Preschool , Chromosome Breakpoints , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Exons/genetics , Female , Gastrulation/genetics , Genes, Recessive , Humans , Infant , Male , Pedigree , Young Adult , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Nat Commun ; 7: 11491, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27173435

ABSTRACT

Cellular organelles provide opportunities to relate biological mechanisms to disease. Here we use affinity proteomics, genetics and cell biology to interrogate cilia: poorly understood organelles, where defects cause genetic diseases. Two hundred and seventeen tagged human ciliary proteins create a final landscape of 1,319 proteins, 4,905 interactions and 52 complexes. Reverse tagging, repetition of purifications and statistical analyses, produce a high-resolution network that reveals organelle-specific interactions and complexes not apparent in larger studies, and links vesicle transport, the cytoskeleton, signalling and ubiquitination to ciliary signalling and proteostasis. We observe sub-complexes in exocyst and intraflagellar transport complexes, which we validate biochemically, and by probing structurally predicted, disruptive, genetic variants from ciliary disease patients. The landscape suggests other genetic diseases could be ciliary including 3M syndrome. We show that 3M genes are involved in ciliogenesis, and that patient fibroblasts lack cilia. Overall, this organelle-specific targeting strategy shows considerable promise for Systems Medicine.


Subject(s)
Cilia/metabolism , Ciliopathies/genetics , Dwarfism/genetics , Muscle Hypotonia/genetics , Protein Interaction Maps , Proteins/metabolism , Spine/abnormalities , Biological Transport/physiology , Chromatography, Affinity/methods , Ciliopathies/pathology , Ciliopathies/therapy , DNA Mutational Analysis , Datasets as Topic , Dwarfism/pathology , Dwarfism/therapy , Fibroblasts , HEK293 Cells , Humans , Mass Spectrometry , Molecular Targeted Therapy/methods , Muscle Hypotonia/pathology , Muscle Hypotonia/therapy , Protein Interaction Mapping/methods , Proteins/genetics , Proteins/isolation & purification , Proteomics/methods , Spine/pathology , Systems Analysis
14.
Am J Hum Genet ; 97(6): 904-13, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637980

ABSTRACT

Meier-Gorlin syndrome (MGS) is a genetically heterogeneous primordial dwarfism syndrome known to be caused by biallelic loss-of-function mutations in one of five genes encoding pre-replication complex proteins: ORC1, ORC4, ORC6, CDT1, and CDC6. Mutations in these genes cause disruption of the origin of DNA replication initiation. To date, only an autosomal-recessive inheritance pattern has been described in individuals with this disorder, with a molecular etiology established in about three-fourths of cases. Here, we report three subjects with MGS and de novo heterozygous mutations in the 5' end of GMNN, encoding the DNA replication inhibitor geminin. We identified two truncating mutations in exon 2 (the 1(st) coding exon), c.16A>T (p.Lys6(∗)) and c.35_38delTCAA (p.Ile12Lysfs(∗)4), and one missense mutation, c.50A>G (p.Lys17Arg), affecting the second-to-last nucleotide of exon 2 and possibly RNA splicing. Geminin is present during the S, G2, and M phases of the cell cycle and is degraded during the metaphase-anaphase transition by the anaphase-promoting complex (APC), which recognizes the destruction box sequence near the 5' end of the geminin protein. All three GMNN mutations identified alter sites 5' to residue Met28 of the protein, which is located within the destruction box. We present data supporting a gain-of-function mechanism, in which the GMNN mutations result in proteins lacking the destruction box and hence increased protein stability and prolonged inhibition of replication leading to autosomal-dominant MGS.


Subject(s)
Congenital Microtia/genetics , Dwarfism/genetics , Geminin/genetics , Growth Disorders/genetics , Micrognathism/genetics , Mutation , Patella/abnormalities , Adolescent , Amino Acid Sequence , Base Sequence , Cell Cycle/genetics , Child, Preschool , Congenital Microtia/metabolism , Dwarfism/metabolism , Dwarfism/pathology , Exons , Female , Geminin/metabolism , Gene Expression , Genes, Dominant , Growth Disorders/metabolism , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Inheritance Patterns , Male , Micrognathism/metabolism , Molecular Sequence Data , Patella/metabolism , Pedigree , Protein Stability , Proteolysis , RNA Splicing , Sequence Alignment
15.
Genetics ; 198(2): 723-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25316788

ABSTRACT

Neurodevelopmental defects in humans represent a clinically heterogeneous group of disorders. Here, we report the genetic and functional dissection of a multigenerational pedigree with an X-linked syndromic disorder hallmarked by microcephaly, growth retardation, and seizures. Using an X-linked intellectual disability (XLID) next-generation sequencing diagnostic panel, we identified a novel missense mutation in the gene encoding 60S ribosomal protein L10 (RPL10), a locus associated previously with autism spectrum disorders (ASD); the p.K78E change segregated with disease under an X-linked recessive paradigm while, consistent with causality, carrier females exhibited skewed X inactivation. To examine the functional consequences of the p.K78E change, we modeled RPL10 dysfunction in zebrafish. We show that endogenous rpl10 expression is augmented in anterior structures, and that suppression decreases head size in developing morphant embryos, concomitant with reduced bulk translation and increased apoptosis in the brain. Subsequently, using in vivo complementation, we demonstrate that p.K78E is a loss-of-function variant. Together, our findings suggest that a mutation within the conserved N-terminal end of RPL10, a protein in close proximity to the peptidyl transferase active site of the 60S ribosomal subunit, causes severe defects in brain formation and function.


Subject(s)
Microcephaly/genetics , Ribosomal Proteins/genetics , Adult , Animals , Apoptosis , Brain/pathology , Cell Proliferation , Child, Preschool , Female , Genes, X-Linked , Genetic Association Studies , Humans , Male , Mutation, Missense , Pedigree , Ribosomal Protein L10 , Young Adult , Zebrafish
16.
Am J Hum Genet ; 94(5): 745-54, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24746959

ABSTRACT

Homozygosity for a recurrent 290 kb deletion of NPHP1 is the most frequent cause of isolated nephronophthisis (NPHP) in humans. A deletion of the same genomic interval has also been detected in individuals with Joubert syndrome (JBTS), and in the mouse, Nphp1 interacts genetically with Ahi1, a known JBTS locus. Given these observations, we investigated the contribution of NPHP1 in Bardet-Biedl syndrome (BBS), a ciliopathy of intermediate severity. By using a combination of array-comparative genomic hybridization, TaqMan copy number assays, and sequencing, we studied 200 families affected by BBS. We report a homozygous NPHP1 deletion CNV in a family with classical BBS that is transmitted with autosomal-recessive inheritance. Further, we identified heterozygous NPHP1 deletions in two more unrelated persons with BBS who bear primary mutations at another BBS locus. In parallel, we identified five families harboring an SNV in NPHP1 resulting in a conserved missense change, c.14G>T (p.Arg5Leu), that is enriched in our Hispanic pedigrees; in each case, affected individuals carried additional bona fide pathogenic alleles in another BBS gene. In vivo functional modeling in zebrafish embryos demonstrated that c.14G>T is a loss-of-function variant, and suppression of nphp1 in concert with each of the primary BBS loci found in our NPHP1-positive pedigrees exacerbated the severity of the phenotype. These results suggest that NPHP1 mutations are probably rare primary causes of BBS that contribute to the mutational burden of the disorder.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Bardet-Biedl Syndrome/genetics , DNA Copy Number Variations , Membrane Proteins/genetics , Alleles , Animals , Cytoskeletal Proteins , Gastrulation/genetics , Genetic Loci , Heterozygote , Homozygote , Humans , Kidney/abnormalities , Mice , Pedigree , Sequence Deletion , Zebrafish/abnormalities , Zebrafish/genetics
17.
Invest Ophthalmol Vis Sci ; 55(4): 2121-9, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24595387

ABSTRACT

PURPOSE: Mutations at some retinitis pigmentosa (RP) loci are associated with variable penetrance and expressivity, exacerbating diagnostic challenges. The purpose of this study was to dissect the genetic underpinnings of nonsyndromic RP with variable age of onset in a large Mexican family. METHODS: We ascertained members of a large, multigenerational pedigree using a complete ophthalmic examination. We performed whole exome sequencing on two affected first cousins, an obligate carrier, and a married-in spouse. Confirmatory sequencing of candidate variants was performed in the entire pedigree, as well as genotyping and mRNA studies to investigate expression changes in the causal locus. RESULTS: We identified a 14-base pair (bp) deletion in PRPF31, a gene implicated previously in autosomal dominant (ad) RP. The mutation segregated with the phenotype of all 10 affected females, but also was present in six asymptomatics (two females and four males). Studies in patient cells showed that the penetrance/expressivity of the PRPF31 deletion allele was concordant with the expression levels of wild-type message. However, neither the known PRPF31 modulators nor cis-eQTLs within 1 Mb of the locus could account for the variable expression of message or the clinical phenotype. CONCLUSIONS: We have identified a novel 14-bp deletion in PRPF31 as the genetic driver of adRP in a large Mexican family that exhibits nonpenetrance and variable expressivity, known properties of this locus. However, our studies intimate the presence of additional loci that can modify PRPF31 expression.


Subject(s)
Exome/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , RNA/genetics , Retinitis Pigmentosa/genetics , Sequence Deletion , Adolescent , Adult , Alleles , Child , Eye Proteins/metabolism , Female , Genes, Dominant , Genotype , Humans , Male , Middle Aged , Pedigree , Phenotype , Polymerase Chain Reaction , RNA Splicing , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/metabolism , Young Adult
18.
Am J Hum Genet ; 93(2): 197-210, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23810381

ABSTRACT

White matter hyperintensities (WMHs) of the brain are important markers of aging and small-vessel disease. WMHs are rare in healthy children and, when observed, often occur with comorbid neuroinflammatory or vasculitic processes. Here, we describe a complex 4 kb deletion in 2q36.3 that segregates with early childhood communication disorders and WMH in 15 unrelated families predominantly from Southeast Asia. The premature brain aging phenotype with punctate and multifocal WMHs was observed in ~70% of young carrier parents who underwent brain MRI. The complex deletion removes the penultimate exon 3 of TM4SF20, a gene encoding a transmembrane protein of unknown function. Minigene analysis showed that the resultant net loss of an exon introduces a premature stop codon, which, in turn, leads to the generation of a stable protein that fails to target to the plasma membrane and accumulates in the cytoplasm. Finally, we report this deletion to be enriched in individuals of Vietnamese Kinh descent, with an allele frequency of about 1%, embedded in an ancestral haplotype. Our data point to a constellation of early language delay and WMH phenotypes, driven by a likely toxic mechanism of TM4SF20 truncation, and highlight the importance of understanding and managing population-specific low-frequency pathogenic alleles.


Subject(s)
Aging, Premature/genetics , Base Sequence , Genetic Predisposition to Disease , Language Development Disorders/genetics , Leukoencephalopathies/genetics , Sequence Deletion , Tetraspanins/genetics , Age of Onset , Aging, Premature/complications , Aging, Premature/ethnology , Aging, Premature/pathology , Asian People , Brain/metabolism , Brain/pathology , Child , Child, Preschool , Chromosomes, Human, Pair 2 , Exons , Female , Humans , Language Development Disorders/complications , Language Development Disorders/ethnology , Language Development Disorders/pathology , Leukoencephalopathies/complications , Leukoencephalopathies/ethnology , Leukoencephalopathies/pathology , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA
19.
Methods Mol Biol ; 1003: 133-47, 2013.
Article in English | MEDLINE | ID: mdl-23585039

ABSTRACT

Humans have approximately 400 intact olfactory receptors (ORs). Among this set there are a large number of variations between individuals, a subset of which affects receptor function and can lead to interindividual variation in olfactory perception. Technological progress and cost erosion in next-generation sequencing have given us the opportunity to determine the sequence of the entire OR gene set with high fidelity and to measure the extent of variation in this functional module across many individuals. Given that whole genome sequencing remains prohibitively expensive for this purpose, especially since the OR sub-genome represents only ~0.0125 % of the human genome, we have designed a targeted capture method to enrich the OR for next-generation sequencing, which we describe here. Using this method we have been able to sequence an individual's OR sub-genome with high coverage, enabling us to identify variation with high sensitivity and specificity. This method can be used to accurate assess the amount of variability in this module and to identify the functional role of individual ORs in olfactory perception.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Receptors, Odorant/genetics , Sequence Analysis, DNA/methods , Gene Library , Humans , Nucleic Acid Hybridization , Polymerase Chain Reaction
20.
Am J Hum Genet ; 89(6): 713-30, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152675

ABSTRACT

Joubert syndrome related disorders (JSRDs) have broad but variable phenotypic overlap with other ciliopathies. The molecular etiology of this overlap is unclear but probably arises from disrupting common functional module components within primary cilia. To identify additional module elements associated with JSRDs, we performed homozygosity mapping followed by next-generation sequencing (NGS) and uncovered mutations in TMEM237 (previously known as ALS2CR4). We show that loss of the mammalian TMEM237, which localizes to the ciliary transition zone (TZ), results in defective ciliogenesis and deregulation of Wnt signaling. Furthermore, disruption of Danio rerio (zebrafish) tmem237 expression produces gastrulation defects consistent with ciliary dysfunction, and Caenorhabditis elegans jbts-14 genetically interacts with nphp-4, encoding another TZ protein, to control basal body-TZ anchoring to the membrane and ciliogenesis. Both mammalian and C. elegans TMEM237/JBTS-14 require RPGRIP1L/MKS5 for proper TZ localization, and we demonstrate additional functional interactions between C. elegans JBTS-14 and MKS-2/TMEM216, MKSR-1/B9D1, and MKSR-2/B9D2. Collectively, our findings integrate TMEM237/JBTS-14 in a complex interaction network of TZ-associated proteins and reveal a growing contribution of a TZ functional module to the spectrum of ciliopathy phenotypes.


Subject(s)
Cerebellar Diseases/genetics , Cilia/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Mutation , Abnormalities, Multiple , Adult , Animals , Bardet-Biedl Syndrome/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/ultrastructure , Case-Control Studies , Cell Line , Cerebellum/abnormalities , Child , Child, Preschool , Chromosome Mapping , Cilia/metabolism , Female , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Genetic Association Studies , Haplotypes , Humans , Infant , Infant, Newborn , Male , Membrane Proteins/metabolism , Mice , Microscopy, Electron, Transmission , Multiprotein Complexes/metabolism , Polymorphism, Single Nucleotide , Retina/abnormalities , Sequence Analysis, DNA , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/embryology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...