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










Publication year range
1.
Transl Vis Sci Technol ; 13(7): 7, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980261

ABSTRACT

Purpose: Lipid nanoparticles (LNPs) show promise in their ability to introduce mRNA to drive protein expression in specific cell types of the mammalian eye. Here, we examined the ability of mRNA encapsulated in LNPs with two distinct formulations to drive gene expression in mouse and human retina and other ocular tissues. Methods: We introduced mRNA-carrying LNPs into two biological systems. Intravitreal injections were tested to deliver LNPs into the mouse eye. Human retinal pigment epithelium (RPE) and retinal explants were used to assess mRNA expression in human tissue. We analyzed specificity of expression using histology, immunofluorescence, and imaging. Results: In mice, mRNAs encoding GFP and ciliary neurotrophic factor (CNTF) were specifically expressed by Müller glia and RPE. Acute inflammatory changes measured by microglia distribution (Iba-1) or interleukin-6 (IL-6) expression were not observed 6 hours post-injection. Human RPE also expressed high levels of GFP. Human retinal explants expressed GFP in cells with apical and basal processes consistent with Müller glia and in perivascular cells consistent with macrophages. Conclusions: We demonstrated the ability to reliably transfect subpopulations of retinal cells in mouse eye tissues in vivo and in human ocular tissues. Of significance, intravitreal injections were sufficient to transfect the RPE in mice. To our knowledge, we demonstrate delivery of mRNA using LNPs in human ocular tissues for the first time. Translational Relevance: Ocular gene-replacement therapies using non-viral vector methods are a promising alternative to adeno-associated virus (AAV) vectors. Our studies show that mRNA LNP delivery can be used to transfect retinal cells in both mouse and human tissues without inducing significant inflammation. This methodology could be used to transfect retinal cell lines, tissue explants, mice, or potentially as gene-replacement therapy in a clinical setting in the future.


Subject(s)
Intravitreal Injections , Nanoparticles , RNA, Messenger , Retinal Pigment Epithelium , Animals , Humans , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Mice , Retinal Pigment Epithelium/metabolism , Nanoparticles/chemistry , Mice, Inbred C57BL , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/administration & dosage , Retina/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ependymoglial Cells/metabolism , Gene Transfer Techniques , Liposomes
2.
Mol Vis ; 30: 58-66, 2024.
Article in English | MEDLINE | ID: mdl-38601016

ABSTRACT

Purpose: Pathogenic variants in North Carolina macular dystrophy (NCMD) have rarely been reported in the East Asian population. Herein, we reported novel variants of NCMD in 2 Korean families. Methods: The regions associated with NCMD were analyzed with genome sequencing, and variants were filtered based on the minor allele frequency (0.5%) and heterozygosity. Non-coding variants were functionally annotated using multiple computational tools. Results: We identified two rare novel variants, chr6:g.99,598,914T>C (hg38; V17) and chr6:g.99,598,926G>A (hg38; V18) upstream of PRDM13 in families A and B, respectively. In Family 1, Grade 2 NCMD and a best-corrected visual acuity of 20/25 and 20/200 in the right and left eyes, respectively, were observed. In Family B, all affected individuals had Grade 1 NCMD with characteristic confluent drusen at the fovea and a best-corrected visual acuity of 20/20 in both eyes. These two variants are 10-22 bp downstream of the reported V10 variant within the DNase1 hypersensitivity site. This site is associated with progressive bifocal chorioretinal atrophy and congenital posterior polar chorioretinal hypertrophy and lies in the putative enhancer site of PRDM13. Conclusion: We identified two novel NCMD variants in the Korean population and further validated the regulatory role of the DNase1 hypersensitivity site upstream of PRDM13.


Subject(s)
Corneal Dystrophies, Hereditary , Humans , Corneal Dystrophies, Hereditary/genetics , Fovea Centralis , Nucleotides , Pedigree , Republic of Korea
3.
bioRxiv ; 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37502987

ABSTRACT

Purpose: Lipid nanoparticles (LNPs) show promise in their ability to introduce mRNA to drive protein expression in specific cell types of the mammalian eye. Here, we examined the ability of mRNA encapsulated in lipid nanoparticles (LNPs) with two distinct formulations to drive gene expression in mouse and human retina and other ocular tissues. Methods: We introduced mRNA carrying LNPs into two biological systems. Intravitreal injections were tested to deliver LNPs into the mouse eye. Human retinal pigment epithelium (RPE) and retinal explants were used to assess mRNA expression in human tissue. We analyzed specificity of expression using histology, immunofluorescence, and imaging. Results: In mice, mRNAs encoding GFP and ciliary neurotrophic factor (CNTF) were specifically expressed by Müller glia and retinal pigment epithelium (RPE). Acute inflammatory changes measured by microglia distribution (Iba-1) or interleukin-6 (IL-6) expression were not observed 6 hours post-injection. Human RPE also expressed high levels of GFP. Human retinal explants expressed GFP in cells with apical and basal processes consistent with Müller glia and in perivascular cells consistent with macrophages. Conclusions: We demonstrated the ability to reliably transfect subpopulations of retinal cells in mice eye tissues in vivo and in human ocular tissues. Of significance, intravitreal injections were sufficient to transfect the RPE in mice. To our knowledge we demonstrate delivery of mRNA using LNPs in human ocular tissues for the first time.

4.
Transl Vis Sci Technol ; 11(4): 16, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35435921

ABSTRACT

Purpose: Prior studies have demonstrated the significance of specific cis-regulatory variants in retinal disease; however, determining the functional impact of regulatory variants remains a major challenge. In this study, we utilized a machine learning approach, trained on epigenomic data from the adult human retina, to systematically quantify the predicted impact of cis-regulatory variants. Methods: We used human retinal DNA accessibility data (ATAC-seq) to determine a set of 18.9k high-confidence, putative cis-regulatory elements. Eighty percent of these elements were used to train a machine learning model utilizing a gapped k-mer support vector machine-based approach. In silico saturation mutagenesis and variant scoring was applied to predict the functional impact of all potential single nucleotide variants within cis-regulatory elements. Impact scores were tested in a 20% hold-out dataset and compared to allele population frequency, phylogenetic conservation, transcription factor (TF) binding motifs, and existing massively parallel reporter assay data. Results: We generated a model that distinguishes between human retinal regulatory elements and negative test sequences with 95% accuracy. Among a hold-out test set of 3.7k human retinal CREs, all possible single nucleotide variants were scored. Variants with negative impact scores correlated with higher phylogenetic conservation of the reference allele, disruption of predicted TF binding motifs, and massively parallel reporter expression. Conclusions: We demonstrated the utility of human retinal epigenomic data to train a machine learning model for the purpose of predicting the impact of non-coding regulatory sequence variants. Our model accurately scored sequences and predicted putative transcription factor binding motifs. This approach has the potential to expedite the characterization of pathogenic non-coding sequence variants in the context of unexplained retinal disease. Translational Relevance: This workflow and resulting dataset serve as a promising genomic tool to facilitate the clinical prioritization of functionally disruptive non-coding mutations in the retina.


Subject(s)
Machine Learning , Retinal Diseases , Humans , Nucleotides , Phylogeny , Retina , Retinal Diseases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Invest Ophthalmol Vis Sci ; 63(4): 1, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35363247

ABSTRACT

Purpose: Succinate is exported by the retina and imported by eyecup tissue. The transporters mediating this process have not yet been identified. Recent studies showed that monocarboxylate transporter 1 (MCT1) can transport succinate across plasma membranes in cardiac and skeletal muscle. Retina and retinal pigment epithelium (RPE) both express multiple MCT isoforms including MCT1. We tested the hypothesis that MCTs facilitate retinal succinate export and RPE succinate import. Methods: We assessed retinal succinate export and eyecup succinate import in short-term ex vivo culture using gas chromatography-mass spectrometry. We tested the dependence of succinate export and import on pH, proton ionophores, conventional MCT substrates, and the MCT inhibitors AZD3965, AR-C155858, and diclofenac. Results: Succinate exits retinal tissue through MCT1 but does not enter the RPE through MCT1 or any other MCT. Intracellular succinate levels are a contributing factor that determines if an MCT1-expressing tissue will export succinate. Conclusions: MCT1 facilitates export of succinate from retinas. An unidentified, non-MCT transporter facilitates import of succinate into RPE.


Subject(s)
Succinates , Succinic Acid , Membrane Transport Proteins , Retina , Retinal Pigment Epithelium
6.
Dev Cell ; 57(6): 820-836.e6, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35303433

ABSTRACT

Cis-regulatory elements (CREs) play a critical role in the development and disease-states of all human cell types. In the retina, CREs have been implicated in several inherited disorders. To better characterize human retinal CREs, we performed single-nucleus assay for transposase-accessible chromatin sequencing (snATAC-seq) and single-nucleus RNA sequencing (snRNA-seq) on the developing and adult human retina and on induced pluripotent stem cell (iPSC)-derived retinal organoids. These analyses identified developmentally dynamic, cell-class-specific CREs, enriched transcription-factor-binding motifs, and putative target genes. CREs in the retina and organoids are highly correlated at the single-cell level, and this supports the use of organoids as a model for studying disease-associated CREs. As a proof of concept, we disrupted a disease-associated CRE at 5q14.3, confirming its principal target gene as the miR-9-2 primary transcript and demonstrating its role in neurogenesis and gene regulation in mature glia. This study provides a resource for characterizing human retinal CREs and showcases organoids as a model to study the function of CREs that influence development and disease.


Subject(s)
Organoids , Retina , Adult , Chromatin/genetics , Humans , Regulatory Sequences, Nucleic Acid , Sequence Analysis, RNA
7.
J Cereb Blood Flow Metab ; 42(6): 966-978, 2022 06.
Article in English | MEDLINE | ID: mdl-34929105

ABSTRACT

Perivascular fibroblasts (PVFs) are recognized for their pro-fibrotic role in many central nervous system disorders. Like mural cells, PVFs surround blood vessels and express Pdgfrß. However, these shared attributes hinder the ability to distinguish PVFs from mural cells. We used in vivo two-photon imaging and transgenic mice with PVF-targeting promoters (Col1a1 or Col1a2) to compare the structure and distribution of PVFs and mural cells in cerebral cortex of healthy, adult mice. We show that PVFs localize to all cortical penetrating arterioles and their offshoots (arteriole-capillary transition zone), as well as the main trunk of only larger ascending venules. However, the capillary zone is devoid of PVF coverage. PVFs display short-range mobility along the vessel wall and exhibit distinct structural features (flattened somata and thin ruffled processes) not seen with smooth muscle cells or pericytes. These findings clarify that PVFs and mural cells are distinct cell types coexisting in a similar perivascular niche.


Subject(s)
Capillaries , Pericytes , Animals , Brain/blood supply , Capillaries/diagnostic imaging , Fibroblasts/metabolism , Mice , Mice, Transgenic , Pericytes/metabolism
8.
Cell Rep ; 37(7): 109994, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34788628

ABSTRACT

Gene regulatory networks (GRNs), consisting of transcription factors and their target sites, control neurogenesis and cell-fate specification in the developing central nervous system. In this study, we use integrated single-cell RNA and single-cell ATAC sequencing (scATAC-seq) analysis in developing mouse and human retina to identify multiple interconnected, evolutionarily conserved GRNs composed of cell-type-specific transcription factors that both activate genes within their own network and inhibit genes in other networks. These GRNs control temporal patterning in primary progenitors, regulate transition from primary to neurogenic progenitors, and drive specification of each major retinal cell type. We confirm that NFI transcription factors selectively activate expression of genes promoting late-stage temporal identity in primary retinal progenitors and identify other transcription factors that regulate rod photoreceptor specification in postnatal retina. This study inventories cis- and trans-acting factors that control retinal development and can guide cell-based therapies aimed at replacing retinal neurons lost to disease.


Subject(s)
Body Patterning/genetics , Cell Lineage/genetics , Neurogenesis/genetics , Retina/embryology , Animals , Cell Differentiation/genetics , Eye Proteins/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice/embryology , NFI Transcription Factors/metabolism , Retinal Neurons/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Trans-Activators/metabolism
9.
Proc Natl Acad Sci U S A ; 117(16): 9001-9012, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32265282

ABSTRACT

The interplay of transcription factors and cis-regulatory elements (CREs) orchestrates the dynamic and diverse genetic programs that assemble the human central nervous system (CNS) during development and maintain its function throughout life. Genetic variation within CREs plays a central role in phenotypic variation in complex traits including the risk of developing disease. We took advantage of the retina, a well-characterized region of the CNS known to be affected by pathogenic variants in CREs, to establish a roadmap for characterizing regulatory variation in the human CNS. This comprehensive analysis of tissue-specific regulatory elements, transcription factor binding, and gene expression programs in three regions of the human visual system (retina, macula, and retinal pigment epithelium/choroid) reveals features of regulatory element evolution that shape tissue-specific gene expression programs and defines regulatory elements with the potential to contribute to Mendelian and complex disorders of human vision.


Subject(s)
Evolution, Molecular , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid/genetics , Retina/pathology , Retinal Diseases/genetics , Adult , Animals , DNA Mutational Analysis , Epigenomics , Female , Genetic Variation , Humans , Male , Mice , Middle Aged , Mutation , RNA-Seq , Retina/growth & development , Retinal Diseases/pathology , Species Specificity
10.
Cell Rep ; 29(7): 2001-2015.e5, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722213

ABSTRACT

Compensation among paralogous transcription factors (TFs) confers genetic robustness of cellular processes, but how TFs dynamically respond to paralog depletion on a genome-wide scale in vivo remains incompletely understood. Using single and double conditional knockout of myocyte enhancer factor 2 (MEF2) family TFs in granule neurons of the mouse cerebellum, we find that MEF2A and MEF2D play functionally redundant roles in cerebellar-dependent motor learning. Although both TFs are highly expressed in granule neurons, transcriptomic analyses show MEF2D is the predominant genomic regulator of gene expression in vivo. Strikingly, genome-wide occupancy analyses reveal upon depletion of MEF2D, MEF2A occupancy robustly increases at a subset of sites normally bound to MEF2D. Importantly, sites experiencing compensatory MEF2A occupancy are concentrated within open chromatin and undergo functional compensation for genomic activation and gene expression. Finally, motor activity induces a switch from non-compensatory to compensatory MEF2-dependent gene regulation. These studies uncover genome-wide functional interdependency between paralogous TFs in the brain.


Subject(s)
Cerebellum/metabolism , Chromatin/metabolism , Gene Expression Regulation , Neurons/metabolism , Animals , Cerebellum/cytology , Chromatin/genetics , Genome-Wide Association Study , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Neurons/cytology
11.
Genet Med ; 21(3): 694-704, 2019 03.
Article in English | MEDLINE | ID: mdl-30072743

ABSTRACT

PURPOSE: With the advent of gene therapies for inherited retinal degenerations (IRDs), genetic diagnostics will have an increasing role in clinical decision-making. Yet the genetic cause of disease cannot be identified using exon-based sequencing for a significant portion of patients. We hypothesized that noncoding pathogenic variants contribute significantly to the genetic causality of IRDs and evaluated patients with single coding pathogenic variants in RPGRIP1 to test this hypothesis. METHODS: IRD families underwent targeted panel sequencing. Unsolved cases were explored by exome and genome sequencing looking for additional pathogenic variants. Candidate pathogenic variants were then validated by Sanger sequencing, quantitative polymerase chain reaction, and in vitro splicing assays in two cell lines analyzed through amplicon sequencing. RESULTS: Among 1722 families, 3 had biallelic loss-of-function pathogenic variants in RPGRIP1 while 7 had a single disruptive coding pathogenic variants. Exome and genome sequencing revealed potential noncoding pathogenic variants in these 7 families. In 6, the noncoding pathogenic variants were shown to lead to loss of function in vitro. CONCLUSION: Noncoding pathogenic variants were identified in 6 of 7 families with single coding pathogenic variants in RPGRIP1. The results suggest that noncoding pathogenic variants contribute significantly to the genetic causality of IRDs and RPGRIP1-mediated IRDs are more common than previously thought.


Subject(s)
DNA, Intergenic/genetics , Proteins/genetics , Retinal Degeneration/genetics , Adult , Chromosome Mapping , Cytoskeletal Proteins , DNA Mutational Analysis/methods , DNA, Intergenic/physiology , Female , HEK293 Cells , Humans , Male , Mutation , Pedigree , Proteins/physiology , Retinal Degeneration/etiology , Exome Sequencing/methods , Whole Genome Sequencing/methods
12.
Genet Med ; 21(8): 1998, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30297699

ABSTRACT

The original version of this Article contained an error in the spelling of the author Anja K. Mayer, which was incorrectly given as Anja Kathrin Mayer. This has now been corrected in both the PDF and HTML versions of the Article.

13.
Genet Med ; 20(2): 202-213, 2018 02.
Article in English | MEDLINE | ID: mdl-28749477

ABSTRACT

PurposePart of the hidden genetic variation in heterogeneous genetic conditions such as inherited retinal diseases (IRDs) can be explained by copy-number variations (CNVs). Here, we explored the genomic landscape of IRD genes listed in RetNet to identify and prioritize those genes susceptible to CNV formation.MethodsRetNet genes underwent an assessment of genomic features and of CNV occurrence in the Database of Genomic Variants and literature. CNVs identified in an IRD cohort were characterized using targeted locus amplification (TLA) on extracted genomic DNA.ResultsExhaustive literature mining revealed 1,345 reported CNVs in 81 different IRD genes. Correlation analysis between rankings of genomic features and CNV occurrence demonstrated the strongest correlation between gene size and CNV occurrence of IRD genes. Moreover, we identified and delineated 30 new CNVs in IRD cases, 13 of which are novel and three of which affect noncoding, putative cis-regulatory regions. Finally, the breakpoints of six complex CNVs were determined using TLA in a hypothesis-neutral manner.ConclusionWe propose a ranking of CNV-prone IRD genes and demonstrate the efficacy of TLA for the characterization of CNVs on extracted DNA. Finally, this IRD-oriented CNV study can serve as a paradigm for other genetically heterogeneous Mendelian diseases with hidden genetic variation.


Subject(s)
Chromosome Mapping , DNA Copy Number Variations , Genome, Human , Genomics , Open Reading Frames , RNA, Untranslated , Retinal Diseases/genetics , Alleles , Cadherin Related Proteins , Cadherins/genetics , Databases, Genetic , Eye Proteins/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics/methods , Humans , Regulatory Sequences, Nucleic Acid , Retinal Diseases/diagnosis , Sequence Analysis, DNA , Sequence Deletion
14.
Neuron ; 86(1): 247-63, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25801704

ABSTRACT

Organismal development requires the precise coordination of genetic programs to regulate cell fate and function. MEF2 transcription factors (TFs) play essential roles in this process but how these broadly expressed factors contribute to the generation of specific cell types during development is poorly understood. Here we show that despite being expressed in virtually all mammalian tissues, in the retina MEF2D binds to retina-specific enhancers and controls photoreceptor cell development. MEF2D achieves specificity by cooperating with a retina-specific factor CRX, which recruits MEF2D away from canonical MEF2 binding sites and redirects it to retina-specific enhancers that lack the consensus MEF2-binding sequence. Once bound to retina-specific enhancers, MEF2D and CRX co-activate the expression of photoreceptor-specific genes that are critical for retinal function. These findings demonstrate that broadly expressed TFs acquire specific functions through competitive recruitment to enhancers by tissue-specific TFs and through selective activation of these enhancers to regulate tissue-specific genes.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Photoreceptor Cells/physiology , Retina/cytology , Trans-Activators/metabolism , Adaptation, Ocular/genetics , Age Factors , Animals , Animals, Newborn , Chromatin Immunoprecipitation , Electroretinography , Embryo, Mammalian , Eye Proteins/metabolism , Genome , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Retina/growth & development
15.
J Neurosci ; 31(20): 7365-79, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21593321

ABSTRACT

Members of the basic helix-loop-helix (bHLH) family of transcription factors have been shown to control critical aspects of development in many tissues. To identify bHLH genes that might regulate specific aspects of retinal cell development, we investigated the expression of bHLH genes in single, developing mouse retinal cells, with particular emphasis on the NeuroD family. Two of these factors, NeuroD2 and NeuroD6/NEX, had not been previously reported as expressed in the retina. A series of loss- and gain-of-function experiments was performed, which suggested that NeuroD genes have both similarities and differences in their activities. Notably, misexpression of NeuroD genes can direct amacrine cell processes to two to three specific sublaminae in the inner plexiform layer. This effect is specific to cell type and NeuroD gene, as the AII amacrine cell type is refractory to the effects of NeuroD1 and NeuroD6, but uniquely sensitive to the effect of NeuroD2 on neurite targeting. Additionally, NeuroD2 is endogenously expressed in AII amacrine cells, among others, and loss of NeuroD2 function results in a partial loss of AII amacrine cells. The effects of misexpressing NeuroD genes on retinal cell fate determination also suggested shared and divergent functions. Remarkably, NeuroD2 misexpression induced ganglion cell production even after the normal developmental window of ganglion cell genesis. Together, these data suggest that members of the NeuroD family are important for neuronal cell type identity and may be involved in several cell type-specific aspects of retinal development, including fate determination, differentiation, morphological development, and circuit formation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Neurites/metabolism , Neuropeptides/physiology , Retina/growth & development , Retina/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , Female , Gene Knock-In Techniques , Glycine/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neurogenesis/physiology , Retina/cytology , Stem Cells/cytology , Stem Cells/metabolism , gamma-Aminobutyric Acid/physiology
16.
Proc Natl Acad Sci U S A ; 106(23): 9495-500, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19470466

ABSTRACT

The vertebrate retina uses diverse neuronal cell types arrayed into complex neural circuits to extract, process, and relay information from the visual scene to the higher order processing centers of the brain. Amacrine cells, a class of interneurons, are thought to mediate much of the processing of the visual signal that occurs within the retina. Although amacrine cells display extensive morphological diversity, the molecular nature of this diversity is largely unknown. Furthermore, it is not known how this diversity arises during development. Here, we have combined in vivo genetic labeling, single cell genome-wide expression profiling, and classical birthdating to (i) identify specific molecular types of amacrine cells, (ii) demonstrate the molecular diversity of the amacrine cell class, and (iii) show that amacrine cell diversity arises at least in part through temporal patterning.


Subject(s)
Amacrine Cells/cytology , Retina/embryology , Animals , Gene Expression Profiling , Mice , Neurogenesis , Retina/cytology
17.
Science ; 308(5729): 1794-8, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15894532

ABSTRACT

The human left and right cerebral hemispheres are anatomically and functionally asymmetric. To test whether human cortical asymmetry has a molecular basis, we studied gene expression levels between the left and right embryonic hemispheres using serial analysis of gene expression (SAGE). We identified and verified 27 differentially expressed genes, which suggests that human cortical asymmetry is accompanied by early, marked transcriptional asymmetries. LMO4 is consistently more highly expressed in the right perisylvian human cerebral cortex than in the left and is essential for cortical development in mice, suggesting that human left-right specialization reflects asymmetric cortical development at early stages.


Subject(s)
Cerebral Cortex/embryology , Functional Laterality , Gene Expression , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing , Animals , Brain Mapping , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Humans , In Situ Hybridization , LIM Domain Proteins , Mice , Reverse Transcriptase Polymerase Chain Reaction
18.
Brain Dev ; 26(5): 326-34, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15165674

ABSTRACT

Periventricular heterotopia (PH) represents a neuronal migration disorder that results in gray matter nodules along the lateral ventricles beneath an otherwise normal appearing cortex. While prior reports have shown that mutations in the filamin A (FLNA) gene can cause X-linked dominant PH, an increasing number of studies suggest the existence of additional PH syndromes. Further classification of these cortical malformation syndromes associated with PH allows for determination of the causal genes. Here we report three familial cases of PH with hydrocephalus. One pedigree has a known FLNA mutation with hydrocephalus occurring in the setting of valproic acid exposure. Another pedigree demonstrated possible linkage to the Xq28 locus including FLNA, although uncharacteristically a male was affected and sequencing of the FLNA gene in this individual revealed no mutation. However, in the third family with an autosomal mode of inheritance, microsatellite analysis ruled out linkage with the FLNA gene. Routine karyotyping and fluorescent in situ hybridization using BAC probes localized to FLNA also showed no evidence of genomic rearrangement. Western blot analysis of one of the affected individuals demonstrated normal expression of the FLNA protein. Lastly, sequencing of greater than 95% of the FLNA gene in an affected member failed to demonstrate a mutation. In conclusion, these findings demonstrate the etiological heterogeneity of PH with hydrocephalus. Furthermore, there likely exists an autosomal PH gene, distinct from the previously described X-linked and autosomal recessive forms. Affected individuals have severe developmental delay and may have radiographic findings of hydrocephalus.


Subject(s)
Brain/pathology , Choristoma/genetics , Hydrocephalus/genetics , Adult , Anticonvulsants/adverse effects , Blotting, Western , Child , Child, Preschool , Choristoma/pathology , Contractile Proteins/genetics , Epilepsy, Complex Partial/genetics , Female , Filamins , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genetic Linkage/genetics , Humans , Hydrocephalus/pathology , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Male , Microfilament Proteins/genetics , Mutation/drug effects , Mutation/genetics , Pedigree , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Valproic Acid/adverse effects , Ventriculoperitoneal Shunt
19.
J Comp Neurol ; 460(2): 266-79, 2003 May 26.
Article in English | MEDLINE | ID: mdl-12687690

ABSTRACT

Foxp2 and Foxp1 are recently identified members of the Fox family of winged-helix/forkhead transcription factor genes. A recent study has found that mutations in human FOXP2 produce a severe language disorder. Since Foxp2 appears to be important in language, we wanted to explore the expression of this gene and a homologous gene, Foxp1, in the developing brain. In the present study, we investigated the time course and localization of Foxp2 and Foxp1 mRNA and protein expression in the developing and adult mouse using in situ hybridization and immunohistochemistry. Foxp2 and Foxp1 are expressed as early as E12.5 and persist into adulthood. Foxp2 and Foxp1 were most highly expressed in the developing and mature basal ganglia. Expression of Foxp2 was also observed in the cerebral cortex (layer 6), cerebellum (Purkinje neurons), and thalamus. Foxp1 expression was observed in the cerebral cortex (layers 3-5), hippocampus (CA1), and thalamus. Very little ventricular zone expression was observed for Foxp2 and Foxp1 and the expression of both of these genes occurred following neuronal migration, suggesting a role for these genes in postmigratory neuronal differentiation. Furthermore, we demonstrated the expression of FOXP2 in human fetal brain by RT-PCR, in the perisylvian area of the left and right cerebral hemispheres, as well as in the frontal and occipital cortices. Overall, the widespread expression of Foxp2 in the developing brain makes it difficult to draw specific conclusions about which areas of Foxp2 expression are critical to human language function.


Subject(s)
Brain/metabolism , RNA, Messenger/biosynthesis , Repressor Proteins/biosynthesis , Transcription Factors , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Embryo, Mammalian , Forkhead Transcription Factors , Gene Expression Regulation, Developmental/physiology , Humans , Mice , Mice, Neurologic Mutants , RNA, Messenger/analysis , Repressor Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...