Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(5): 1373-1386, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38504517

ABSTRACT

Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aß plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.


Subject(s)
Alzheimer Disease , Apolipoprotein E2 , Disease Models, Animal , Genetic Therapy , Mice, Transgenic , Microglia , Plaque, Amyloid , Animals , Alzheimer Disease/therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Mice , Genetic Therapy/methods , Humans , Apolipoprotein E2/genetics , Apolipoprotein E2/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Microglia/metabolism , Brain/metabolism , Brain/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/therapy , Neuroinflammatory Diseases/metabolism , Amyloid beta-Peptides/metabolism , Biomarkers
2.
Mol Ther Nucleic Acids ; 33: 296-304, 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37547292

ABSTRACT

Recombinant adeno-associated viral vectors (rAAVs) are a promising strategy to treat neurodegenerative diseases because of their ability to infect non-dividing cells and confer long-term transgene expression. Despite an ever-growing library of capsid variants, widespread delivery of AAVs in the adult central nervous system remains a challenge. We have previously demonstrated successful distribution of secreted proteins by infection of the ependyma, a layer of post-mitotic epithelial cells lining the ventricles of the brain and central column of the spinal cord, and subsequent protein delivery via the cerebrospinal fluid (CSF). Here we define a functional ependyma promoter to enhance expression from this cell type. Using RNA sequencing on human autopsy samples, we identified disease- and age-independent ependyma gene signatures. Associated promoters were cloned and screened as libraries in mouse and rhesus macaque to reveal cross-species function of a human DNA-derived von Willebrand factor domain containing 3A (VWA3A) promoter. When tested in mice, our VWA3A promoter drove strong, ependyma-localized expression of eGFP and increased secreted ApoE protein levels in the CSF by 2-12× over the ubiquitous iCAG promoter.

3.
bioRxiv ; 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37645718

ABSTRACT

Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do they have a later age of onset, milder clinical course, and less severe neuropathological findings than others with Alzheimer disease. The contrast is especially stark in comparison to the phenotype associated with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, as well as a more aggressive clinical course and notably more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Even one APOE ε2 allele improves phenotype, but it is uncertain if that is due to the replacement of a more toxic allele by APOE ε2, or if APOE ε2 has a protective, neuro-modulatory effect. Here, we demonstrate that brain exposure to APOE2 via a gene therapy approach which bathes the entire cortical mantle in the gene product after transduction of the ependyma, rapidly ameliorates established Aß plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE4. This result suggests a promising protective effect of exogenous APOE2, revealing a cell non-autonomous effect of the protein on microglial activation. We also show that plaque associated microglia in the brain of patients who inherit APOE2 similarly have less microglial reactivity to plaques. These data raise the potential that an APOE2 therapeutic could be effective in Alzheimer disease even in individuals born with the risk ε4 allele. One Sentence Summary: Introduction of ApoE2 using an AAV that transduces the ependymal cells of the ventricle causes a reduction in amyloid load and plaque associated synapse loss, and reduces neuroinflammation by modulating microglial responsiveness to plaques.

4.
BMC Med Genomics ; 16(1): 133, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37322474

ABSTRACT

BACKGROUND: The primary pathological alterations of Pendred syndrome are endolymphatic pH acidification and luminal enlargement of the inner ear. However, the molecular contributions of specific cell types remain poorly characterized. Therefore, we aimed to identify pH regulators in pendrin-expressing cells that may contribute to the homeostasis of endolymph pH and define the cellular pathogenic mechanisms that contribute to the dysregulation of cochlear endolymph pH in Slc26a4-/- mice. METHODS: We used single-cell RNA sequencing to identify both Slc26a4-expressing cells and Kcnj10-expressing cells in wild-type (WT, Slc26a4+/+) and Slc26a4-/- mice. Bioinformatic analysis of expression data confirmed marker genes defining the different cell types of the stria vascularis. In addition, specific findings were confirmed at the protein level by immunofluorescence. RESULTS: We found that spindle cells, which express pendrin, contain extrinsic cellular components, a factor that enables cell-to-cell communication. In addition, the gene expression profile informed the pH of the spindle cells. Compared to WT, the transcriptional profiles in Slc26a4-/- mice showed downregulation of extracellular exosome-related genes in spindle cells. Immunofluorescence studies in spindle cells of Slc26a4-/- mice validated the increased expression of the exosome-related protein, annexin A1, and the clathrin-mediated endocytosis-related protein, adaptor protein 2. CONCLUSION: Overall, cell isolation of stria vascularis from WT and Slc26a4-/- samples combined with cell type-specific transcriptomic analyses revealed pH-dependent alternations in spindle cells and intermediate cells, inspiring further studies into the dysfunctional role of stria vascularis cells in SLC26A4-related hearing loss.


Subject(s)
Deafness , Stria Vascularis , Mice , Animals , Stria Vascularis/metabolism , Stria Vascularis/pathology , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cochlea/metabolism , Cochlea/pathology , Deafness/genetics , Sulfate Transporters/genetics , RNA/metabolism
5.
Nat Commun ; 14(1): 2628, 2023 05 06.
Article in English | MEDLINE | ID: mdl-37149717

ABSTRACT

Alternative splicing of neuronal genes is controlled partly by the coordinated action of polypyrimidine tract binding proteins (PTBPs). While PTBP1 is ubiquitously expressed, PTBP2 is predominantly neuronal. Here, we define the PTBP2 footprint in the human transcriptome using brain tissue and human induced pluripotent stem cell-derived neurons (iPSC-neurons). We map PTBP2 binding sites, characterize PTBP2-dependent alternative splicing events, and identify novel PTBP2 targets including SYNGAP1, a synaptic gene whose loss-of-function leads to a complex neurodevelopmental disorder. We find that PTBP2 binding to SYNGAP1 mRNA promotes alternative splicing and nonsense-mediated decay, and that antisense oligonucleotides (ASOs) that disrupt PTBP binding redirect splicing and increase SYNGAP1 mRNA and protein expression. In SYNGAP1 haploinsufficient iPSC-neurons generated from two patients, we show that PTBP2-targeting ASOs partially restore SYNGAP1 expression. Our data comprehensively map PTBP2-dependent alternative splicing in human neurons and cerebral cortex, guiding development of novel therapeutic tools to benefit neurodevelopmental disorders.


Subject(s)
Induced Pluripotent Stem Cells , Nerve Tissue Proteins , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , RNA Splicing , Alternative Splicing/genetics , Brain/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , ras GTPase-Activating Proteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism
7.
Life Sci Alliance ; 6(3)2023 03.
Article in English | MEDLINE | ID: mdl-36574989

ABSTRACT

Hearing loss is the most common sensory deficit, of which genetic etiologies are a frequent cause. Dominant and recessive mutations in TMC1, a gene encoding the pore-forming subunit of the hair cell mechanotransduction channel, cause DFNA36 and DFNB7/11, respectively, accounting for ∼2% of genetic hearing loss. Previous work has established the efficacy of mutation-targeted RNAi in treatment of murine models of autosomal dominant non-syndromic deafness. However, application of such approaches is limited by the infeasibility of development and validation of novel constructs for each variant. We developed an allele-non-specific approach consisting of mutation-agnostic RNAi suppression of both mutant and WT alleles, co-delivered with a knockdown-resistant engineered WT allele with or without the use of woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) to augment transgene expression. This therapeutic construct was delivered into the mature murine model of DFNA36 with an AAV vector and achieved robust hair cell and auditory brainstem response preservation. However, WPRE-enhanced Tmc1 expression resulted in inferior outcomes, suggesting a role for gene dosage optimization in future TMC1 gene therapy development.


Subject(s)
Hearing Loss , Mechanotransduction, Cellular , Mice , Animals , RNA Interference , Hearing Loss/genetics , Hearing Loss/therapy , Mutation/genetics , Membrane Proteins/genetics
9.
Nat Med ; 27(11): 1982-1989, 2021 11.
Article in English | MEDLINE | ID: mdl-34663988

ABSTRACT

RNA interference (RNAi) for spinocerebellar ataxia type 1 can prevent and reverse behavioral deficits and neuropathological readouts in mouse models, with safety and benefit lasting over many months. The RNAi trigger, expressed from adeno-associated virus vectors (AAV.miS1), also corrected misregulated microRNAs (miRNA) such as miR150. Subsequently, we showed that the delivery method was scalable, and that AAV.miS1 was safe in short-term pilot nonhuman primate (NHP) studies. To advance the technology to patients, investigational new drug (IND)-enabling studies in NHPs were initiated. After AAV.miS1 delivery to deep cerebellar nuclei, we unexpectedly observed cerebellar toxicity. Both small-RNA-seq and studies using AAVs devoid of miRNAs showed that this was not a result of saturation of the endogenous miRNA processing machinery. RNA-seq together with sequencing of the AAV product showed that, despite limited amounts of cross-packaged material, there was substantial inverted terminal repeat (ITR) promoter activity that correlated with neuropathologies. ITR promoter activity was reduced by altering the miS1 expression context. The surprising contrast between our rodent and NHP findings highlight the need for extended safety studies in multiple species when assessing new therapeutics for human application.


Subject(s)
Dependovirus/genetics , Drug Carriers/administration & dosage , Genetic Therapy/methods , MicroRNAs/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/therapy , Animals , Animals, Genetically Modified , Brain Stem/pathology , Cerebellum/pathology , Female , Macaca mulatta , Male , Mice , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA-Seq , Terminal Repeat Sequences/genetics
10.
Nature ; 596(7871): 291-295, 2021 08.
Article in English | MEDLINE | ID: mdl-34321659

ABSTRACT

So far, gene therapies have relied on complex constructs that cannot be finely controlled1,2. Here we report a universal switch element that enables precise control of gene replacement or gene editing after exposure to a small molecule. The small-molecule inducers are currently in human use, are orally bioavailable when given to animals or humans and can reach both peripheral tissues and the brain. Moreover, the switch system, which we denote Xon, does not require the co-expression of any regulatory proteins. Using Xon, the translation of the desired elements for controlled gene replacement or gene editing machinery occurs after a single oral dose of the inducer, and the robustness of expression can be controlled by the drug dose, protein stability and redosing. The ability of Xon to provide temporal control of protein expression can be adapted for cell-biology applications and animal studies. Additionally, owing to the oral bioavailability and safety of the drugs used, the Xon switch system provides an unprecedented opportunity to refine and tailor the application of gene therapies in humans.


Subject(s)
Alternative Splicing/drug effects , Gene Editing/methods , Genetic Therapy/methods , Protein Biosynthesis/drug effects , Animals , Brain/drug effects , Brain/metabolism , CRISPR-Associated Protein 9/metabolism , Drug Delivery Systems/methods , Erythropoietin/biosynthesis , Erythropoietin/genetics , Erythropoietin/metabolism , Exons/genetics , Female , Frontotemporal Dementia/metabolism , HEK293 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscular Atrophy, Spinal/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Progranulins/biosynthesis , Progranulins/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism
11.
Hum Mol Genet ; 30(24): 2469-2487, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34296279

ABSTRACT

We have previously established induced pluripotent stem cell (iPSC) models of Huntington's disease (HD), demonstrating CAG-repeat-expansion-dependent cell biological changes and toxicity. However, the current differentiation protocols are cumbersome and time consuming, making preparation of large quantities of cells for biochemical or screening assays difficult. Here, we report the generation of immortalized striatal precursor neurons (ISPNs) with normal (33) and expanded (180) CAG repeats from HD iPSCs, differentiated to a phenotype resembling medium spiny neurons (MSN), as a proof of principle for a more tractable patient-derived cell model. For immortalization, we used co-expression of the enzymatic component of telomerase hTERT and conditional expression of c-Myc. ISPNs can be propagated as stable adherent cell lines, and rapidly differentiated into highly homogeneous MSN-like cultures within 2 weeks, as demonstrated by immunocytochemical criteria. Differentiated ISPNs recapitulate major HD-related phenotypes of the parental iPSC model, including brain-derived neurotrophic factor (BDNF)-withdrawal-induced cell death that can be rescued by small molecules previously validated in the parental iPSC model. Proteome and RNA-seq analyses demonstrate separation of HD versus control samples by principal component analysis. We identified several networks, pathways, and upstream regulators, also found altered in HD iPSCs, other HD models, and HD patient samples. HD ISPN lines may be useful for studying HD-related cellular pathogenesis, and for use as a platform for HD target identification and screening experimental therapeutics. The described approach for generation of ISPNs from differentiated patient-derived iPSCs could be applied to a larger allelic series of HD cell lines, and to comparable modeling of other genetic disorders.


Subject(s)
Huntington Disease , Induced Pluripotent Stem Cells , Cell Differentiation/genetics , Cell Line , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/therapy , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism
12.
Cell Rep ; 26(11): 3160-3171.e3, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865901

ABSTRACT

Single-cell RNA sequencing is a powerful tool by which to characterize the transcriptional profile of low-abundance cell types, but its application to the inner ear has been hampered by the bony labyrinth, tissue sparsity, and difficulty dissociating the ultra-rare cells of the membranous cochlea. Herein, we present a method to isolate individual inner hair cells (IHCs), outer hair cells (OHCs), and Deiters' cells (DCs) from the murine cochlea at any post-natal time point. We harvested more than 200 murine IHCs, OHCs, and DCs from post-natal days 15 (p15) to 228 (p228) and leveraged both short- and long-read single-cell RNA sequencing to profile transcript abundance and structure. Our results provide insights into the expression profiles of these cells and document an unappreciated complexity in isoform variety in deafness-associated genes. This refined view of transcription in the organ of Corti improves our understanding of the biology of hearing and deafness.


Subject(s)
Deafness/genetics , Organ of Corti/metabolism , Transcriptome , Animals , Female , Gene Expression Profiling , Male , Mice , Organ of Corti/growth & development , Single-Cell Analysis
13.
Mol Ther ; 27(3): 681-690, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30686588

ABSTRACT

Hearing loss is the most common human sensory deficit. Its correction has been the goal of several gene-therapy based studies exploring a variety of interventions. Although these studies report varying degrees of success, all treatments have targeted developing inner ears in neonatal mice, a time point in the structural maturation of the cochlea prior to 26 weeks gestational age in humans. It is unclear whether cochlear gene therapy can salvage hearing in the mature organ of Corti. Herein, we report the first study to test gene therapy in an adult murine model of human deafness. Using a single intracochlear injection of an artificial microRNA carried in an AAV vector, we show that RNAi-mediated gene silencing can slow progression of hearing loss, improve inner hair cell survival, and prevent stereocilia bundle degeneration in the mature Beethoven mouse, a model of human TMC1 deafness. The ability to study gene therapy in mature murine ears constitutes a significant step toward its translation to human subjects.


Subject(s)
Deafness/genetics , Genetic Vectors/genetics , Membrane Proteins/metabolism , Animals , Cochlea/metabolism , Cochlea/physiopathology , Cochlea/ultrastructure , Deafness/physiopathology , Dependovirus/genetics , Disease Models, Animal , Ear, Inner/metabolism , Ear, Inner/physiopathology , Ear, Inner/ultrastructure , Genetic Therapy , Hearing Loss/genetics , Hearing Loss/physiopathology , Humans , Immunohistochemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Microscopy, Electron, Scanning , RNA Interference , Vestibule, Labyrinth/metabolism , Vestibule, Labyrinth/physiopathology , Vestibule, Labyrinth/ultrastructure
14.
Sci Rep ; 8(1): 2980, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445157

ABSTRACT

Cochlear gene therapy holds promise for the treatment of genetic deafness. Assessing its impact in adult murine models of hearing loss, however, has been hampered by technical challenges that have made it difficult to establish a robust method to deliver transgenes to the mature murine inner ear. Here in we demonstrate the feasibility of a combined round window membrane injection and semi-circular canal fenestration technique in the adult cochlea. Injection of both AAV2/9 and AAV2/Anc80L65 via this approach in P15-16 and P56-60 mice permits robust eGFP transduction of virtually all inner hair cells throughout the cochlea with variable transduction of vestibular hair cells. Auditory thresholds are not compromised. Transduction rate and cell tropism is primarily influenced by viral titer and AAV serotype but not age at injection. This approach is safe, versatile and efficient. Its use will facilitate studies using cochlear gene therapy in murine models of hearing loss over a wide range of time points.


Subject(s)
Cochlea/physiology , Deafness/therapy , Fenestration, Labyrinth/methods , Gene Transfer Techniques , Genetic Therapy/methods , Hair Cells, Auditory, Inner/physiology , Hearing Loss/therapy , Adenoviridae/genetics , Animals , Cochlea/surgery , Cochlear Implantation , Deafness/genetics , Disease Models, Animal , Ear Canal/surgery , Female , Genetic Vectors , Hearing Loss/genetics , Humans , Male , Mice , Mice, Inbred C3H , Round Window, Ear/surgery , Tympanic Membrane/surgery
15.
Sci Rep ; 7(1): 9609, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852025

ABSTRACT

Gene therapy for genetic deafness is a promising approach by which to prevent hearing loss or to restore hearing after loss has occurred. Although a variety of direct approaches to introduce viral particles into the inner ear have been described, presumed physiological barriers have heretofore precluded investigation of systemic gene delivery to the cochlea. In this study, we sought to characterize systemic delivery of a rAAV2/9 vector as a non-invasive means of cochlear transduction. In wild-type neonatal mice (postnatal day 0-1), we show that intravenous injection of rAAV2/9 carrying an eGFP-reporter gene results in binaural transduction of inner hair cells, spiral ganglion neurons and vestibular hair cells. Transduction efficiency increases in a dose-dependent manner. Inner hair cells are transduced in an apex-to-base gradient, with transduction reaching 96% in the apical turn. Hearing acuity in treated animals is unaltered at postnatal day 30. Transduction is influenced by viral serotype and age at injection, with less efficient cochlear transduction observed with systemic delivery of rAAV2/1 and in juvenile mice with rAAV2/9. Collectively, these data validate intravenous delivery of rAAV2/9 as a novel and atraumatic technique for inner ear transgene delivery in early postnatal mice.


Subject(s)
Cochlea/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Administration, Intravenous , Animals , Animals, Newborn , Dependovirus/classification , Gene Expression , Genes, Reporter , Genetic Vectors/administration & dosage , Immunohistochemistry , Mice , Spiral Ganglion/metabolism , Transduction, Genetic , Transgenes
16.
Am J Hum Genet ; 98(6): 1101-1113, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27236922

ABSTRACT

Hearing impairment is the most common sensory deficit. It is frequently caused by the expression of an allele carrying a single dominant missense mutation. Herein, we show that a single intracochlear injection of an artificial microRNA carried in a viral vector can slow progression of hearing loss for up to 35 weeks in the Beethoven mouse, a murine model of non-syndromic human deafness caused by a dominant gain-of-function mutation in Tmc1 (transmembrane channel-like 1). This outcome is noteworthy because it demonstrates the feasibility of RNA-interference-mediated suppression of an endogenous deafness-causing allele to slow progression of hearing loss. Given that most autosomal-dominant non-syndromic hearing loss in humans is caused by this mechanism of action, microRNA-based therapeutics might be broadly applicable as a therapy for this type of deafness.


Subject(s)
Auditory Pathways , Hearing Loss/prevention & control , Membrane Proteins/physiology , MicroRNAs/genetics , Mutation, Missense/genetics , Animals , Dependovirus/genetics , Hearing Loss/etiology , Hearing Loss/pathology , Humans , Mechanotransduction, Cellular , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C3H , Mice, Knockout , MicroRNAs/administration & dosage , RNA Interference
17.
Hum Genet ; 135(4): 441-450, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26969326

ABSTRACT

Hearing loss is the most common sensory deficit in humans, affecting 1 in 500 newborns. Due to its genetic heterogeneity, comprehensive diagnostic testing has not previously been completed in a large multiethnic cohort. To determine the aggregate contribution inheritance makes to non-syndromic hearing loss, we performed comprehensive clinical genetic testing with targeted genomic enrichment and massively parallel sequencing on 1119 sequentially accrued patients. No patient was excluded based on phenotype, inheritance or previous testing. Testing resulted in identification of the underlying genetic cause for hearing loss in 440 patients (39%). Pathogenic variants were found in 49 genes and included missense variants (49%), large copy number changes (18%), small insertions and deletions (18%), nonsense variants (8%), splice-site alterations (6%), and promoter variants (<1%). The diagnostic rate varied considerably based on phenotype and was highest for patients with a positive family history of hearing loss or when the loss was congenital and symmetric. The spectrum of implicated genes showed wide ethnic variability. These findings support the more efficient utilization of medical resources through the development of evidence-based algorithms for the diagnosis of hearing loss.


Subject(s)
Genetic Testing , Hearing Loss/genetics , Adolescent , Child , Child, Preschool , Female , Genetic Heterogeneity , Hearing Loss/diagnosis , Humans , Infant , Male
18.
PLoS Genet ; 11(3): e1005137, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25816005

ABSTRACT

Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action.


Subject(s)
Carrier Proteins/genetics , Ear, Inner/metabolism , Exome/genetics , Hearing Loss, Sensorineural/genetics , Animals , Carrier Proteins/biosynthesis , Cochlea/metabolism , Cochlea/pathology , Ear, Inner/pathology , Gene Expression Regulation , Hearing Loss, Sensorineural/pathology , High-Throughput Nucleotide Sequencing , Homer Scaffolding Proteins , Humans , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stereocilia/genetics , Stereocilia/pathology , Zebrafish , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
19.
Mol Biol Cell ; 24(5): 588-600, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283985

ABSTRACT

The length of Chlamydomonas flagella is tightly regulated. Mutations in four genes-LF1, LF2, LF3, and LF4-cause cells to assemble flagella up to three times wild-type length. LF2 and LF4 encode protein kinases. Here we describe a new gene, LF5, in which null mutations cause cells to assemble flagella of excess length. The LF5 gene encodes a protein kinase very similar in sequence to the protein kinase CDKL5. In humans, mutations in this kinase cause a severe form of juvenile epilepsy. The LF5 protein localizes to a unique location: the proximal 1 µm of the flagella. The proximal localization of the LF5 protein is lost when genes that make up the proteins in the cytoplasmic length regulatory complex (LRC)-LF1, LF2, and LF3-are mutated. In these mutants LF5p becomes localized either at the distal tip of the flagella or along the flagellar length, indicating that length regulation involves, at least in part, control of LF5p localization by the LRC.


Subject(s)
Chlamydomonas/genetics , Flagella/genetics , Protein Serine-Threonine Kinases/genetics , Chlamydomonas/growth & development , Epilepsy, Absence/genetics , Flagella/ultrastructure , Humans , Mutation , Phenotype , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...