Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(22): 4868-4884.e12, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37863056

ABSTRACT

Single-cell analysis in living humans is essential for understanding disease mechanisms, but it is impractical in non-regenerative organs, such as the eye and brain, because tissue biopsies would cause serious damage. We resolve this problem by integrating proteomics of liquid biopsies with single-cell transcriptomics from all known ocular cell types to trace the cellular origin of 5,953 proteins detected in the aqueous humor. We identified hundreds of cell-specific protein markers, including for individual retinal cell types. Surprisingly, our results reveal that retinal degeneration occurs in Parkinson's disease, and the cells driving diabetic retinopathy switch with disease stage. Finally, we developed artificial intelligence (AI) models to assess individual cellular aging and found that many eye diseases not associated with chronological age undergo accelerated molecular aging of disease-specific cell types. Our approach, which can be applied to other organ systems, has the potential to transform molecular diagnostics and prognostics while uncovering new cellular disease and aging mechanisms.


Subject(s)
Aging , Aqueous Humor , Artificial Intelligence , Liquid Biopsy , Proteomics , Humans , Aging/metabolism , Aqueous Humor/chemistry , Biopsy , Parkinson Disease/diagnosis
2.
J Neurosci ; 43(2): 221-239, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36442999

ABSTRACT

Lesion localization is the basis for understanding neurologic disease, which is predicated on neuroanatomical knowledge carefully cataloged from histology and imaging atlases. However, it is often difficult to correlate clinical images of brainstem injury obtained by MRI scans with the details of human brainstem neuroanatomy represented in atlases, which are mostly based on cytoarchitecture using Nissl stain or a single histochemical stain, and usually do not include the cerebellum. Here, we report a high-resolution (200 µm) 7T MRI of a cadaveric male human brainstem and cerebellum paired with detailed, coregistered histology (at 2 µm single-cell resolution) of the immunohistochemically stained cholinergic, serotonergic, and catecholaminergic (dopaminergic, noradrenergic, and adrenergic) neurons, in relationship to each other and to the cerebellum. These immunohistochemical findings provide novel insights into the spatial relationships of brainstem cell types and nuclei, including subpopulations of melanin and TH+ neurons, and allows for more informed structural annotation of cell groups. Moreover, the coregistered MRI-paired histology helps validate imaging findings. This is useful for interpreting both scans and histology, and to understand the cell types affected by lesions. Our detailed chemoarchitecture and cytoarchitecture with corresponding high-resolution MRI builds on previous atlases of the human brainstem and cerebellum, and makes precise identification of brainstem and cerebellar cell groups involved in clinical lesions accessible for both laboratory scientists and clinicians alike.SIGNIFICANCE STATEMENT Clinicians and neuroscientists frequently use cross-sectional anatomy of the human brainstem from MRI scans for both clinical and laboratory investigations, but they must rely on brain atlases to neuroanatomical structures. Such atlases generally lack both detail of brainstem chemical cell types, and the cerebellum, which provides an important spatial reference. Our current atlas maps the distribution of key brainstem cell types (cholinergic, serotonergic, and catecholaminergic neurons) in relationship to each other and the cerebellum, and pairs this histology with 7T MR images from the identical brain. This atlas allows correlation of the chemoarchitecture with corresponding MRI, and makes the identification of cell groups that are often discussed, but rarely identifiable on MRI scan, accessible to clinicians and clinical researchers.


Subject(s)
Cerebellum , Magnetic Resonance Imaging , Humans , Male , Brain Stem/diagnostic imaging , Brain/metabolism , Neurons
3.
J Proteome Res ; 23(2): 511-522, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38171013

ABSTRACT

Minimally invasive liquid biopsies from the eye capture locally enriched fluids that contain thousands of proteins from highly specialized ocular cell types, presenting a promising alternative to solid tissue biopsies. The advantages of liquid biopsies include sampling the eye without causing irreversible functional damage, potentially better reflecting tissue heterogeneity, collecting samples in an outpatient setting, monitoring therapeutic response with sequential sampling, and even allowing examination of disease mechanisms at the cell level in living humans, an approach that we refer to as TEMPO (Tracing Expression of Multiple Protein Origins). Liquid biopsy proteomics has the potential to transform molecular diagnostics and prognostics and to assess disease mechanisms and personalized therapeutic strategies in individual patients. This review addresses opportunities, challenges, and future directions of high-resolution liquid biopsy proteomics in ophthalmology, with particular emphasis on the large-scale collection of high-quality samples, cutting edge proteomics technology, and artificial intelligence-supported data analysis.


Subject(s)
Ophthalmology , Humans , Proteomics , Artificial Intelligence , Liquid Biopsy , Proteins , Biopsy
4.
J Autoimmun ; 144: 103183, 2024 04.
Article in English | MEDLINE | ID: mdl-38401466

ABSTRACT

Chronic nonbacterial osteomyelitis (CNO), an autoinflammatory bone disease primarily affecting children, can cause pain, hyperostosis and fractures, affecting quality-of-life and psychomotor development. This study investigated CNO-associated variants in P2RX7, encoding for the ATP-dependent trans-membrane K+ channel P2X7, and their effects on NLRP3 inflammasome assembly. Whole exome sequencing in two related transgenerational CNO patients, and target sequencing of P2RX7 in a large CNO cohort (N = 190) were conducted. Results were compared with publicly available datasets and regional controls (N = 1873). Findings were integrated with demographic and clinical data. Patient-derived monocytes and genetically modified THP-1 cells were used to investigate potassium flux, inflammasome assembly, pyroptosis, and cytokine release. Rare presumably damaging P2RX7 variants were identified in two related CNO patients. Targeted P2RX7 sequencing identified 62 CNO patients with rare variants (32.4%), 11 of which (5.8%) carried presumably damaging variants (MAF <1%, SIFT "deleterious", Polyphen "probably damaging", CADD >20). This compared to 83 of 1873 controls (4.4%), 36 with rare and presumably damaging variants (1.9%). Across the CNO cohort, rare variants unique to one (Median: 42 versus 3.7) or more (≤11 patients) participants were over-represented when compared to 190 randomly selected controls. Patients with rare damaging variants more frequently experienced gastrointestinal symptoms and lymphadenopathy while having less spinal, joint and skin involvement (psoriasis). Monocyte-derived macrophages from patients, and genetically modified THP-1-derived macrophages reconstituted with CNO-associated P2RX7 variants exhibited altered potassium flux, inflammasome assembly, IL-1ß and IL-18 release, and pyroptosis. Damaging P2RX7 variants occur in a small subset of CNO patients, and rare P2RX7 variants may represent a CNO risk factor. Observations argue for inflammasome inhibition and/or cytokine blockade and may allow future patient stratification and individualized care.


Subject(s)
Inflammasomes , Osteomyelitis , Humans , Cytokines , Inflammasomes/genetics , Inflammasomes/metabolism , Osteomyelitis/genetics , Potassium , Pyroptosis , Receptors, Purinergic P2X7/genetics
5.
Epilepsia ; 63(12): 3051-3065, 2022 12.
Article in English | MEDLINE | ID: mdl-36168798

ABSTRACT

OBJECTIVE: Many seizing neonates fail to respond to first-line anticonvulsant medications. Phenobarbital, an allosteric modulator of γ-aminobutyric acid type A (GABAA ) receptors, has low efficacy in treating neonatal seizures and causes neuronal apoptosis. Nonetheless, it is one of the most used anticonvulsants in this age group. In neonatal mice, phenobarbital's poor effectiveness is due in part to high intraneuronal chloride concentration, which causes GABA to exert depolarizing actions. Therefore, another approach to treat neonatal seizures could be to use anticonvulsants that do not rely on GABAergic modulation. We evaluated whether lacosamide decreases seizures in neonatal mice and whether it increases apoptosis in vitro and in vivo. METHODS: In vitro, we measured the effect of different lacosamide concentrations on seizure-like activity induced by the pro-convulsant drug 4-aminopyridine in neocortical brain slices (layer IV/V) from neonatal (postnatal day 8-11) and adult (1-1.6 months old) C57BL/6J mice. In vivo, we recorded the effect of different lacosamide concentrations on neonatal behavioral seizures induced by kainic acid. We studied neocortical apoptosis in vitro and in vivo, measuring terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling signal and cleaved-caspase 3. RESULTS: Lacosamide reduced epileptiform activity in neocortical brain slices of neonates and adults in a concentration-dependent manner. In vivo, lacosamide reduced the duration and number of behavioral seizures. Lacosamide did not increase total or neuronal apoptosis in the neocortex in vitro or in vivo. SIGNIFICANCE: Lacosamide reduces neocortical seizure-like activity in neonatal mice in vitro and in vivo without an acute increase in apoptosis. Our results support the use of lacosamide to treat neonatal seizures, with the advantage of not increasing apoptosis acutely.


Subject(s)
Apoptosis , Seizures , Animals , Mice , Lacosamide/therapeutic use , Mice, Inbred C57BL , Seizures/chemically induced , Seizures/drug therapy , gamma-Aminobutyric Acid
6.
Proc Natl Acad Sci U S A ; 116(24): 11872-11877, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31138708

ABSTRACT

Autoinflammatory syndromes are characterized by dysregulation of the innate immune response with subsequent episodes of acute spontaneous inflammation. Chronic recurrent multifocal osteomyelitis (CRMO) is an autoinflammatory bone disorder that presents with bone pain and localized swelling. Ali18 mice, isolated from a mutagenesis screen, exhibit a spontaneous inflammatory paw phenotype that includes sterile osteomyelitis and systemic reduced bone mineral density. To elucidate the molecular basis of the disease, positional cloning of the causative gene for Ali18 was attempted. Using a candidate gene approach, a missense mutation in the C-terminal region of Fgr, a member of Src family tyrosine kinases (SFKs), was identified. For functional confirmation, additional mutations at the N terminus of Fgr were introduced in Ali18 mice by CRISPR/Cas9-mediated genome editing. N-terminal deleterious mutations of Fgr abolished the inflammatory phenotype in Ali18 mice, but in-frame and missense mutations in the same region continue to exhibit the phenotype. The fact that Fgr null mutant mice are morphologically normal suggests that the inflammation in this model depends on Fgr products. Furthermore, the levels of C-terminal negative regulatory phosphorylation of Fgr Ali18 are distinctly reduced compared with that of wild-type Fgr. In addition, whole-exome sequencing of 99 CRMO patients including 88 trios (proband and parents) identified 13 patients with heterozygous coding sequence variants in FGR, including two missense mutant proteins that affect kinase activity. Our results strongly indicate that gain-of-function mutations in Fgr are involved in sterile osteomyelitis, and thus targeting SFKs using specific inhibitors may allow for efficient treatment of the disease.


Subject(s)
Bone Diseases/genetics , Gain of Function Mutation/genetics , Inflammation/genetics , src-Family Kinases/genetics , Amino Acid Sequence , Animals , Humans , Immunity, Innate/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteomyelitis/genetics , Phosphorylation/genetics
7.
Hum Mol Genet ; 28(20): 3475-3485, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31518400

ABSTRACT

Hypoxia associated with the high metabolic demand of rods has been implicated in the pathology of age-related macular degeneration (AMD), the most common cause of adult blindness in the developed world. The majority of AMD-associated severe vision loss cases are due to exudative AMD, characterized by neovascularization. To further investigate the causes and histopathology of exudative AMD, we conditionally induced hypoxia in a novel preclinical AMD model (Pde6gcreERT2/+;Vhl-/-) by targeting Vhl and used multimodal imaging and immunohistochemistry to track the development of hypoxia-induced neovascularization. In addition to developing a preclinical model that phenocopies exudative AMD, our studies revealed that the photoreceptor hypoxic response initiates and drives type 3 neovascularization, mainly in the outer retina. Activation of the VHL-HIF1a-VEGF-EPO pathway in the adult retina led to long-term neovascularization, retinal hemorrhages and compromised retinal layers. Our novel preclinical model would accelerate the testing of therapies that use metabolomic approaches to ameliorate AMD.


Subject(s)
Hypoxia/metabolism , Hypoxia/pathology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Animals , Erythropoietin/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Vascular Endothelial Growth Factor A/metabolism
8.
Hum Mol Genet ; 28(10): 1726-1737, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30689861

ABSTRACT

Mutations in IRF6, TFAP2A and GRHL3 cause orofacial clefting syndromes in humans. However, Tfap2a and Grhl3 are also required for neurulation in mice. Here, we found that homeostasis of Irf6 is also required for development of the neural tube and associated structures. Over-expression of Irf6 caused exencephaly, a rostral neural tube defect, through suppression of Tfap2a and Grhl3 expression. Conversely, loss of Irf6 function caused a curly tail and coincided with a reduction of Tfap2a and Grhl3 expression in tail tissues. To test whether Irf6 function in neurulation was conserved, we sequenced samples obtained from human cases of spina bifida and anencephaly. We found two likely disease-causing variants in two samples from patients with spina bifida. Overall, these data suggest that the Tfap2a-Irf6-Grhl3 genetic pathway is shared by two embryologically distinct morphogenetic events that previously were considered independent during mammalian development. In addition, these data suggest new candidates to delineate the genetic architecture of neural tube defects and new therapeutic targets to prevent this common birth defect.


Subject(s)
DNA-Binding Proteins/genetics , Interferon Regulatory Factors/genetics , Neurulation/genetics , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Animals , Conserved Sequence/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mutation , Neural Tube/growth & development , Neural Tube/pathology , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Signal Transduction/genetics , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology
9.
J Neuroinflammation ; 18(1): 151, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225752

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a leading cause of death and disability that lacks neuroprotective therapies. Following a TBI, secondary injury response pathways are activated and contribute to ongoing neurodegeneration. Microglia and astrocytes are critical neuroimmune modulators with early and persistent reactivity following a TBI. Although histologic glial reactivity is well established, a precise understanding of microglia and astrocyte function following trauma remains unknown. METHODS: Adult male C57BL/6J mice underwent either fluid percussion or sham injury. RNA sequencing of concurrently isolated microglia and astrocytes was conducted 7 days post-injury to evaluate cell-type-specific transcriptional responses to TBI. Dual in situ hybridization and immunofluorescence were used to validate the TBI-induced gene expression changes in microglia and astrocytes and to identify spatial orientation of cells expressing these genes. Comparative analysis was performed between our glial transcriptomes and those from prior reports in mild TBI and other neurologic diseases to determine if severe TBI induces unique states of microglial and astrocyte activation. RESULTS: Our findings revealed sustained, lineage-specific transcriptional changes in both microglia and astrocytes, with microglia showing a greater transcriptional response than astrocytes at this subacute time point. Microglia and astrocytes showed overlapping enrichment for genes related to type I interferon signaling and MHC class I antigen presentation. The microglia and astrocyte transcriptional response to severe TBI was distinct from prior reports in mild TBI and other neurodegenerative and neuroinflammatory diseases. CONCLUSION: Concurrent lineage-specific analysis revealed novel TBI-specific transcriptional changes; these findings highlight the importance of cell-type-specific analysis of glial reactivity following TBI and may assist with the identification of novel, targeted therapies.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Interferon Type I/biosynthesis , Microglia/metabolism , Transcriptome/physiology , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Interferon Type I/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/pathology
10.
J Neurogenet ; 35(2): 74-83, 2021.
Article in English | MEDLINE | ID: mdl-33970744

ABSTRACT

KCTD7 is a member of the potassium channel tetramerization domain-containing protein family and has been associated with progressive myoclonic epilepsy (PME), characterized by myoclonus, epilepsy, and neurological deterioration. Here we report four affected individuals from two unrelated families in which we identified KCTD7 compound heterozygous single nucleotide variants through exome sequencing. RNAseq was used to detect a non-annotated splicing junction created by a synonymous variant in the second family. Whole-cell patch-clamp analysis of neuroblastoma cells overexpressing the patients' variant alleles demonstrated aberrant potassium regulation. While all four patients experienced many of the common clinical features of PME, they also showed variable phenotypes not previously reported, including dysautonomia, brain pathology findings including a significantly reduced thalamus, and the lack of myoclonic seizures. To gain further insight into the pathogenesis of the disorder, zinc finger nucleases were used to generate kctd7 knockout zebrafish. Kctd7 homozygous mutants showed global dysregulation of gene expression and increased transcription of c-fos, which has previously been correlated with seizure activity in animal models. Together these findings expand the known phenotypic spectrum of KCTD7-associated PME, report a new animal model for future studies, and contribute valuable insights into the disease.


Subject(s)
Myoclonic Epilepsies, Progressive/genetics , Potassium Channels/genetics , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Mutation , Myoclonic Epilepsies, Progressive/physiopathology , Pedigree , Phenotype , Zebrafish
11.
Cell Mol Life Sci ; 76(18): 3657-3665, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30976840

ABSTRACT

D190N, a missense mutation in rhodopsin, causes photoreceptor degeneration in patients with autosomal dominant retinitis pigmentosa (adRP). Two competing hypotheses have been developed to explain why D190N rod photoreceptors degenerate: (a) defective rhodopsin trafficking prevents proteins from correctly exiting the endoplasmic reticulum, leading to their accumulation, with deleterious effects or (b) elevated mutant rhodopsin expression and unabated signaling causes excitotoxicity. A knock-in D190N mouse model was engineered to delineate the mechanism of pathogenesis. Wild type (wt) and mutant rhodopsin appeared correctly localized in rod outer segments of D190N heterozygotes. Moreover, the rhodopsin glycosylation state in the mutants appeared similar to that in wt mice. Thus, it seems plausible that the injurious effect of the heterozygous mutation is not related to mistrafficking of the protein, but rather from constitutive rhodopsin activity and a greater propensity for chromophore isomerization even in the absence of light.


Subject(s)
Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/genetics , Amino Acid Sequence , Animals , Disease Models, Animal , Electroretinography , Gene Knock-In Techniques , Glycosylation , Mice , Mice, Inbred C57BL , Mutation, Missense , Protein Structure, Tertiary , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Sequence Alignment
12.
Hum Mutat ; 40(12): 2377-2392, 2019 12.
Article in English | MEDLINE | ID: mdl-31403230

ABSTRACT

Small molecule pharmacological inhibition of dominant human genetic disease is a feasible treatment that does not rely on the development of individual, patient-specific gene therapy vectors. However, the consequences of protein inhibition as a clinical therapeutic are not well-studied. In advance of human therapeutic trials for CAPN5 vitreoretinopathy, genetic inactivation can be used to infer the effect of protein inhibition in vivo. We created a photoreceptor-specific knockout (KO) mouse for Capn5 and compared the retinal phenotype to both wild-type and an existing Capn5 KO mouse model. In humans, CAPN5 loss-of-function (LOF) gene variants were ascertained in large exome databases from 60,706 unrelated subjects without severe disease phenotypes. Ocular examination of the retina of Capn5 KO mice by histology and electroretinography showed no significant abnormalities. In humans, there were 22 LOF CAPN5 variants located throughout the gene and in all major protein domains. Structural modeling of coding variants showed these LOF variants were nearby known disease-causing variants within the proteolytic core and in regions of high homology between human CAPN5 and 150 homologs, yet the LOF of CAPN5 was tolerated as opposed to gain-of-function disease-causing variants. These results indicate that localized inhibition of CAPN5 is a viable strategy for hyperactivating disease alleles.


Subject(s)
Calpain/genetics , Choroid Diseases/genetics , Eye Diseases, Hereditary/genetics , Mutation , Retinal Degeneration/genetics , Tamoxifen/pharmacology , Animals , Calpain/chemistry , Calpain/metabolism , Disease Models, Animal , Female , Gene Knockout Techniques , Gene Silencing , Humans , Male , Mice , Models, Molecular , Photoreceptor Cells, Vertebrate/metabolism
13.
Genet Res (Camb) ; 101: e8, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31190668

ABSTRACT

Compound heterozygotes occur when different variants at the same locus on both maternal and paternal chromosomes produce a recessive trait. Here we present the tool VarCount for the quantification of variants at the individual level. We used VarCount to characterize compound heterozygous coding variants in patients with epileptic encephalopathy and in the 1000 Genomes Project participants. The Epi4k data contains variants identified by whole exome sequencing in patients with either Lennox-Gastaut Syndrome (LGS) or infantile spasms (IS), as well as their parents. We queried the Epi4k dataset (264 trios) and the phased 1000 Genomes Project data (2504 participants) for recessive variants. To assess enrichment, transcript counts were compared between the Epi4k and 1000 Genomes Project participants using minor allele frequency (MAF) cutoffs of 0.5 and 1.0%, and including all ancestries or only probands of European ancestry. In the Epi4k participants, we found enrichment for rare, compound heterozygous variants in six genes, including three involved in neuronal growth and development - PRTG (p = 0.00086, 1% MAF, combined ancestries), TNC (p = 0.022, 1% MAF, combined ancestries) and MACF1 (p = 0.0245, 0.5% MAF, EU ancestry). Due to the total number of transcripts considered in these analyses, the enrichment detected was not significant after correction for multiple testing and higher powered or prospective studies are necessary to validate the candidacy of these genes. However, PRTG, TNC and MACF1 are potential novel recessive epilepsy genes and our results highlight that compound heterozygous variants should be considered in sporadic epilepsy.


Subject(s)
Epilepsy/genetics , Epilepsy/metabolism , Sequence Analysis, DNA/methods , Adult , Alleles , Exome , Female , Gene Frequency/genetics , Genes, Recessive/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Heterozygote , Humans , Infant , Infant, Newborn , Lennox Gastaut Syndrome/genetics , Lennox Gastaut Syndrome/metabolism , Male , Membrane Proteins/genetics , Microfilament Proteins/genetics , Mutation , Phenotype , Prospective Studies , Spasms, Infantile/genetics , Spasms, Infantile/metabolism , Tenascin/genetics
14.
Hum Mol Genet ; 25(13): 2672-2680, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27193166

ABSTRACT

Bestrophin1 (BEST1) is expressed in human retinal pigment epithelium (RPE) and mutations in the BEST1 gene commonly cause retinal dysfunction and macular degeneration. BEST1 is presumed to assemble into a calcium-activated chloride channel and be involved in chloride transport but there is no direct evidence in live human RPE cells to support this idea. To test whether BEST1 functions as a chloride channel in living tissue, BEST1-mutant RPE (R218H, L234P, A243T) were generated from patient-derived induced pluripotent stem cells and compared with wild-type RPE in a retinal environment, using a biosensor that visualizes calcium-induced chloride ion flux in the cell. Calcium stimulation elicited chloride ion export in normal RPE but not in RPE derived from three patients with BEST1 mutations. These data, along with three-dimensional modeling, provide evidence that BEST1 assembles into a key calcium-sensing chloride channel in human RPE.


Subject(s)
Chloride Channels/genetics , Chloride Channels/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Bestrophins , Calcium Signaling , Chlorides , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Retinal Pigment Epithelium/metabolism , Vitelliform Macular Dystrophy/genetics
15.
Hum Mol Genet ; 25(19): 4244-4255, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27516389

ABSTRACT

Retinitis pigmentosa (RP) is an incurable neurodegenerative condition featuring photoreceptor death that leads to blindness. Currently, there is no approved therapeutic for photoreceptor degenerative conditions like RP and atrophic age-related macular degeneration (AMD). Although there are promising results in human gene therapy, RP is a genetically diverse disorder, such that gene-specific therapies would be practical in a small fraction of patients with RP. Here, we explore a non-gene-specific strategy that entails reprogramming photoreceptors towards anabolism by upregulating the mechanistic target of rapamycin (mTOR) pathway. We conditionally ablated the tuberous sclerosis complex 1 (Tsc1) gene, an mTOR inhibitor, in the rods of the Pde6bH620Q/H620Q preclinical RP mouse model and observed, functionally and morphologically, an improvement in the survival of rods and cones at early and late disease stages. These results elucidate the ability of reprogramming the metabolome to slow photoreceptor degeneration. This strategy may also be applicable to a wider range of neurodegenerative diseases, as enhancement of nutrient uptake is not gene-specific and is implicated in multiple pathologies. Enhancing anabolism promoted neuronal survival and function and could potentially benefit a number of photoreceptor and other degenerative conditions.


Subject(s)
Cone-Rod Dystrophies/genetics , Macular Degeneration/genetics , Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Death/genetics , Cellular Reprogramming/genetics , Cone-Rod Dystrophies/pathology , Disease Models, Animal , Humans , Macular Degeneration/pathology , Metabolism/genetics , Mice , Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Tuberous Sclerosis Complex 1 Protein
16.
Hum Mol Genet ; 25(19): 4201-4210, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27516388

ABSTRACT

Inactivating mutations of the TSC1/TSC2 complex (TSC1/2) cause tuberous sclerosis (TSC), a hereditary syndrome with neurological symptoms and benign hamartoma tumours in the brain. Since TSC effectors are largely unknown in the human brain, TSC patient cortical tubers were used to uncover hyperphosphorylation unique to TSC primary astrocytes, the cell type affected in the brain. We found abnormal hyperphosphorylation of catenin delta-1 S268, which was reversible by mTOR-specific inhibitors. In contrast, in three metastatic astrocytoma cell lines, S268 was under phosphorylated, suggesting S268 phosphorylation controls metastasis. TSC astrocytes appeared epithelial (i.e. tightly adherent, less motile, and epithelial (E)-cadherin positive), whereas wild-type astrocytes were mesenchymal (i.e. E-cadherin negative and highly motile). Despite their epithelial phenotype, TSC astrocytes outgrew contact inhibition, and monolayers sporadically generated tuberous foci, a phenotype blocked by the mTOR inhibitor, Torin1. Also, mTOR-regulated phosphokinase C epsilon (PKCe) activity induced phosphorylation of catenin delta-1 S268, which in turn mediated cell-cell adhesion in astrocytes. The mTOR-dependent, epithelial phenotype of TSC astrocytes suggests TSC1/2 and mTOR tune the phosphorylation level of catenin delta-1 by controlling PKCe activity, thereby regulating the mesenchymal-epithelial-transition (MET). Thus, some forms of TSC could be treated with PKCe inhibitors, while metastasis of astrocytomas might be blocked by PKCe stimulators.


Subject(s)
Catenins/genetics , Hamartoma/genetics , Protein Kinase C-epsilon/genetics , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Astrocytes/drug effects , Astrocytes/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Contact Inhibition/drug effects , Epithelial-Mesenchymal Transition/genetics , Hamartoma/pathology , Humans , Naphthyridines/administration & dosage , Neoplasm Metastasis , Phosphorylation/drug effects , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Delta Catenin
17.
J Neurogenet ; 32(4): 313-315, 2018 12.
Article in English | MEDLINE | ID: mdl-29790814

ABSTRACT

Homozygous recessive mutations in the PRICKLE1 gene were first described in three consanguineous families with myoclonic epilepsy. Subsequent studies have identified neurological abnormalities in humans and animal models with both heterozygous and homozygous mutations in PRICKLE1 orthologs. We describe a 7-year-old with a novel de novo missense mutation in PRICKLE1 associated with epilepsy, autism spectrum disorder and global developmental delay.


Subject(s)
Autism Spectrum Disorder/genetics , Epilepsies, Myoclonic/genetics , LIM Domain Proteins/genetics , Tumor Suppressor Proteins/genetics , Child , Developmental Disabilities/genetics , Humans , Intellectual Disability/genetics , Male , Mutation, Missense
18.
Doc Ophthalmol ; 137(1): 15-23, 2018 08.
Article in English | MEDLINE | ID: mdl-29770904

ABSTRACT

PURPOSE: To report a case of deferoxamine-induced retinopathy characterized by electroretinography (ERG), optical coherence tomography angiography (OCT-A), and other multimodal imaging. METHODS: This is an observational case report of one patient. Full-field ERG was performed. OCT-A, spectral-domain optical coherence tomography (SD-OCT), color fundus photography, and fundus autofluorescence were used to characterize the retinopathy induced by deferoxamine use. RESULTS: A 64-year-old man with a history of ß-thalassemia intermedia presented with worsening visual acuity, nyctalopia, and electronegative ERG. OCT-A revealed atrophy of the choriocapillaris in areas of hypoautofluorescence, corresponding to regions of retinal atrophy. SD-OCT showed disruption of the ellipsoid zone, granular hyperreflective deposits within the retinal pigment epithelium, thinning of the retinal layers, and extensive choroidal sclerosis and atrophy of the retinal pigment epithelium. CONCLUSION: Deferoxamine-induced retinopathy can manifest with electronegative maximal ERG responses, and OCT-A can be used to detect deferoxamine toxicity.


Subject(s)
Deferoxamine/toxicity , Electroretinography/drug effects , Retinal Diseases/chemically induced , Siderophores/toxicity , Atrophy , Fluorescein Angiography , Humans , Iron Overload/prevention & control , Male , Middle Aged , Multimodal Imaging , Night Blindness/chemically induced , Night Blindness/diagnosis , Night Blindness/physiopathology , Retinal Diseases/diagnosis , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence/methods , Visual Acuity/drug effects
19.
Retina ; 38(8): 1443-1455, 2018 08.
Article in English | MEDLINE | ID: mdl-29746416

ABSTRACT

PURPOSE: Recent concerns regarding the clinical utilization of clustered regularly interspaced short palindromic repeats (CRISPR) involve uncertainties about the potential detrimental effects that many arise due to unintended genetic changes, as in off-target mutagenesis, during CRISPR genome surgery. This review gives an overview of off-targeting detection methods and CRISPR's place in the clinical setting, specifically in the field of ophthalmology. RESULTS: As CRISPR utilization in the laboratory setting has increased, knowledge regarding CRISPR mechanisms including its off-target effects has also increased. Although a perfect method for achieving 100% specificity is yet to be determined, the past few years have seen many developments in off-targeting detection and in increasing efficacy of CRISPR tools. CONCLUSION: The CRISPR system has high potential to be an invaluable therapeutic tool as it has the ability to modify and repair pathogenic retinal lesions. Although it is not yet a perfect system, with further efforts to improve its specificity and efficacy along with careful screening of off-target mutations, CRISPR-mediated genome surgery potential can become maximized and applied to patients.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Genetic Therapy/methods , Retinal Diseases/therapy , Genetic Therapy/adverse effects , Humans , Ophthalmology , Retinal Diseases/genetics
20.
PLoS Genet ; 11(3): e1005022, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25763846

ABSTRACT

Epilepsy is a common disabling disease with complex, multifactorial genetic and environmental etiology. The small fraction of epilepsies subject to Mendelian inheritance offers key insight into epilepsy disease mechanisms; and pathologies brought on by mutations in a single gene can point the way to generalizable therapeutic strategies. Mutations in the PRICKLE genes can cause seizures in humans, zebrafish, mice, and flies, suggesting the seizure-suppression pathway is evolutionarily conserved. This pathway has never been targeted for novel anti-seizure treatments. Here, the mammalian PRICKLE-interactome was defined, identifying prickle-interacting proteins that localize to synapses and a novel interacting partner, USP9X, a substrate-specific de-ubiquitinase. PRICKLE and USP9X interact through their carboxy-termini; and USP9X de-ubiquitinates PRICKLE, protecting it from proteasomal degradation. In forebrain neurons of mice, USP9X deficiency reduced levels of Prickle2 protein. Genetic analysis suggests the same pathway regulates Prickle-mediated seizures. The seizure phenotype was suppressed in prickle mutant flies by the small-molecule USP9X inhibitor, Degrasyn/WP1130, or by reducing the dose of fat facets a USP9X orthologue. USP9X mutations were identified by resequencing a cohort of patients with epileptic encephalopathy, one patient harbored a de novo missense mutation and another a novel coding mutation. Both USP9X variants were outside the PRICKLE-interacting domain. These findings demonstrate that USP9X inhibition can suppress prickle-mediated seizure activity, and that USP9X variants may predispose to seizures. These studies point to a new target for anti-seizure therapy and illustrate the translational power of studying diseases in species across the evolutionary spectrum.


Subject(s)
Seizures/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Drosophila melanogaster , Humans , Mass Spectrometry , Mice , Seizures/drug therapy , Ubiquitin Thiolesterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL