Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neurology ; 103(3): e209585, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38959435

ABSTRACT

BACKGROUND AND OBJECTIVES: Identification of fluid biomarkers for progressive supranuclear palsy (PSP) is critical to enhance therapeutic development. We implemented unbiased DNA aptamer (SOMAmer) proteomics to identify novel CSF PSP biomarkers. METHODS: This is a cross-sectional study in original (18 clinically diagnosed PSP-Richardson syndrome [PSP-RS], 28 cognitively healthy controls]), validation (23 PSP-RS, 26 healthy controls), and neuropathology-confirmed (21 PSP, 52 non-PSP frontotemporal lobar degeneration) cohorts. Participants were recruited through the University of California, San Francisco, and the 4-Repeat Neuroimaging Initiative. The original and neuropathology cohorts were analyzed with the SomaScan platform version 3.0 (5026-plex) and the validation cohort with version 4.1 (7595-plex). Clinical severity was measured with the PSP Rating Scale (PSPRS). CSF proteomic data were analyzed to identify differentially expressed targets, implicated biological pathways using enrichment and weighted consensus gene coexpression analyses, diagnostic value of top targets with receiver-operating characteristic curves, and associations with disease severity with linear regressions. RESULTS: A total of 136 participants were included (median age 70.6 ± 8 years, 68 [50%] women). One hundred fifty-five of 5,026 (3.1%), 959 of 7,595 (12.6%), and 321 of 5,026 (6.3%) SOMAmers were differentially expressed in PSP compared with controls in original, validation, and neuropathology-confirmed cohorts, with most of the SOMAmers showing reduced signal (83.1%, 95.1%, and 73.2%, respectively). Three coexpression modules were associated with PSP across cohorts: (1) synaptic function/JAK-STAT (ß = -0.044, corrected p = 0.002), (2) vesicle cytoskeletal trafficking (ß = 0.039, p = 0.007), and (3) cytokine-cytokine receptor interaction (ß = -0.032, p = 0.035) pathways. Axon guidance was the top dysregulated pathway in PSP in original (strength = 1.71, p < 0.001), validation (strength = 0.84, p < 0.001), and neuropathology-confirmed (strength = 0.78, p < 0.001) cohorts. A panel of axon guidance pathway proteins discriminated between PSP and controls in original (area under the curve [AUC] = 0.924), validation (AUC = 0.815), and neuropathology-confirmed (AUC = 0.932) cohorts. Two inflammatory proteins, galectin-10 and cytotoxic T lymphocyte-associated protein-4, correlated with PSPRS scores across cohorts. DISCUSSION: Axon guidance pathway proteins and several other molecular pathways are downregulated in PSP, compared with controls. Proteins in these pathways may be useful targets for biomarker or therapeutic development.


Subject(s)
Biomarkers , Proteomics , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/cerebrospinal fluid , Supranuclear Palsy, Progressive/diagnosis , Female , Male , Aged , Proteomics/methods , Biomarkers/cerebrospinal fluid , Cross-Sectional Studies , Middle Aged , Cohort Studies , Aged, 80 and over
2.
Res Sq ; 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38585969

ABSTRACT

The pathophysiological mechanisms driving disease progression of frontotemporal lobar degeneration (FTLD) and corresponding biomarkers are not fully understood. We leveraged aptamer-based proteomics (> 4,000 proteins) to identify dysregulated communities of co-expressed cerebrospinal fluid proteins in 116 adults carrying autosomal dominant FTLD mutations (C9orf72, GRN, MAPT) compared to 39 noncarrier controls. Network analysis identified 31 protein co-expression modules. Proteomic signatures of genetic FTLD clinical severity included increased abundance of RNA splicing (particularly in C9orf72 and GRN) and extracellular matrix (particularly in MAPT) modules, as well as decreased abundance of synaptic/neuronal and autophagy modules. The generalizability of genetic FTLD proteomic signatures was tested and confirmed in independent cohorts of 1) sporadic progressive supranuclear palsy-Richardson syndrome and 2) frontotemporal dementia spectrum syndromes. Network-based proteomics hold promise for identifying replicable molecular pathways in adults living with FTLD. 'Hub' proteins driving co-expression of affected modules warrant further attention as candidate biomarkers and therapeutic targets.

3.
ACS Chem Neurosci ; 14(22): 3993-4012, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37903506

ABSTRACT

Copy number variants (CNVs) that delete or duplicate 30 genes within the 16p11.2 genomic region give rise to a range of neurodevelopmental phenotypes with high penetrance in humans. Despite the identification of this small region, the mechanisms by which 16p11.2 CNVs lead to disease are unclear. Relevant models, such as human cortical organoids (hCOs), are needed to understand the human-specific mechanisms of neurodevelopmental disease. We generated hCOs from 17 patients and controls, profiling 167,958 cells with single-cell RNA-sequencing analysis, which revealed neuronal-specific differential expression of genes outside the 16p11.2 region that are related to cell-cell adhesion, neuronal projection growth, and neurodevelopmental disorders. Furthermore, 16p11.2 deletion syndrome organoids exhibited reduced mRNA and protein levels of RBFOX1, a gene that can also harbor CNVs linked to neurodevelopmental phenotypes. We found that the genes previously shown to be regulated by RBFOX1 are also perturbed in organoids from patients with the 16p11.2 deletion syndrome and thus identified a novel link between independent CNVs associated with neuronal development and autism. Overall, this work suggests convergent signaling, which indicates the possibility of a common therapeutic mechanism across multiple rare neuronal diseases.


Subject(s)
Chromosome Deletion , DNA Copy Number Variations , Humans , DNA Copy Number Variations/genetics , Brain , Phenotype , Organoids , RNA Splicing Factors/genetics
4.
Cell Stem Cell ; 30(3): 312-332.e13, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36796362

ABSTRACT

Human genome variation contributes to diversity in neurodevelopmental outcomes and vulnerabilities; recognizing the underlying molecular and cellular mechanisms will require scalable approaches. Here, we describe a "cell village" experimental platform we used to analyze genetic, molecular, and phenotypic heterogeneity across neural progenitor cells from 44 human donors cultured in a shared in vitro environment using algorithms (Dropulation and Census-seq) to assign cells and phenotypes to individual donors. Through rapid induction of human stem cell-derived neural progenitor cells, measurements of natural genetic variation, and CRISPR-Cas9 genetic perturbations, we identified a common variant that regulates antiviral IFITM3 expression and explains most inter-individual variation in susceptibility to the Zika virus. We also detected expression QTLs corresponding to GWAS loci for brain traits and discovered novel disease-relevant regulators of progenitor proliferation and differentiation such as CACHD1. This approach provides scalable ways to elucidate the effects of genes and genetic variation on cellular phenotypes.


Subject(s)
Neural Stem Cells , Zika Virus Infection , Zika Virus , Humans , Neural Stem Cells/metabolism , Cell Differentiation/genetics , Brain/metabolism , Zika Virus/metabolism , Gene Expression , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism
5.
ACS Chem Biol ; 17(6): 1401-1414, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35508359

ABSTRACT

Unbiased transcriptomic RNA-seq data has provided deep insights into biological processes. However, its impact in drug discovery has been narrow given high costs and low throughput. Proof-of-concept studies with Digital RNA with pertUrbation of Genes (DRUG)-seq demonstrated the potential to address this gap. We extended the DRUG-seq platform by subjecting it to rigorous testing and by adding an open-source analysis pipeline. The results demonstrate high reproducibility and ability to resolve the mechanism(s) of action for a diverse set of compounds. Furthermore, we demonstrate how this data can be incorporated into a drug discovery project aiming to develop therapeutics for schizophrenia using human stem cell-derived neurons. We identified both an on-target activation signature, induced by a set of chemically distinct positive allosteric modulators of the N-methyl-d-aspartate (NMDA) receptor, and independent off-target effects. Overall, the protocol and open-source analysis pipeline are a step toward industrializing RNA-seq for high-complexity transcriptomics studies performed at a saturating scale.


Subject(s)
Drug Discovery , Transcriptome , Drug Discovery/methods , Humans , RNA , Reproducibility of Results , Sequence Analysis, RNA/methods
6.
Cell Rep ; 27(2): 616-630.e6, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970262

ABSTRACT

Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Testing/methods , Genome/genetics , Pluripotent Stem Cells/metabolism , Humans
7.
Nat Neurosci ; 22(3): 374-385, 2019 03.
Article in English | MEDLINE | ID: mdl-30718903

ABSTRACT

Synapse density is reduced in postmortem cortical tissue from schizophrenia patients, which is suggestive of increased synapse elimination. Using a reprogrammed in vitro model of microglia-mediated synapse engulfment, we demonstrate increased synapse elimination in patient-derived neural cultures and isolated synaptosomes. This excessive synaptic pruning reflects abnormalities in both microglia-like cells and synaptic structures. Further, we find that schizophrenia risk-associated variants within the human complement component 4 locus are associated with increased neuronal complement deposition and synapse uptake; however, they do not fully explain the observed increase in synapse uptake. Finally, we demonstrate that the antibiotic minocycline reduces microglia-mediated synapse uptake in vitro and its use is associated with a modest decrease in incident schizophrenia risk compared to other antibiotics in a cohort of young adults drawn from electronic health records. These findings point to excessive pruning as a potential target for delaying or preventing the onset of schizophrenia in high-risk individuals.


Subject(s)
Microglia/physiology , Neuronal Plasticity , Schizophrenia/physiopathology , Synapses/physiology , Adolescent , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology , Male , Microglia/drug effects , Middle Aged , Minocycline/administration & dosage , Neural Stem Cells/physiology , Neuronal Plasticity/drug effects , Risk Factors , Synapses/drug effects , Young Adult
8.
Nat Med ; 24(7): 939-946, 2018 07.
Article in English | MEDLINE | ID: mdl-29892062

ABSTRACT

CRISPR/Cas9 has revolutionized our ability to engineer genomes and conduct genome-wide screens in human cells1-3. Whereas some cell types are amenable to genome engineering, genomes of human pluripotent stem cells (hPSCs) have been difficult to engineer, with reduced efficiencies relative to tumour cell lines or mouse embryonic stem cells3-13. Here, using hPSC lines with stable integration of Cas9 or transient delivery of Cas9-ribonucleoproteins (RNPs), we achieved an average insertion or deletion (indel) efficiency greater than 80%. This high efficiency of indel generation revealed that double-strand breaks (DSBs) induced by Cas9 are toxic and kill most hPSCs. In previous studies, the toxicity of Cas9 in hPSCs was less apparent because of low transfection efficiency and subsequently low DSB induction3. The toxic response to DSBs was P53/TP53-dependent, such that the efficiency of precise genome engineering in hPSCs with a wild-type P53 gene was severely reduced. Our results indicate that Cas9 toxicity creates an obstacle to the high-throughput use of CRISPR/Cas9 for genome engineering and screening in hPSCs. Moreover, as hPSCs can acquire P53 mutations14, cell replacement therapies using CRISPR/Cas9-enginereed hPSCs should proceed with caution, and such engineered hPSCs should be monitored for P53 function.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Genetic Engineering , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Breaks, Double-Stranded , Gene Deletion , Humans , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , fas Receptor/genetics , fas Receptor/metabolism
9.
Cell Rep ; 23(8): 2509-2523, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791859

ABSTRACT

Transcription factor programming of pluripotent stem cells (PSCs) has emerged as an approach to generate human neurons for disease modeling. However, programming schemes produce a variety of cell types, and those neurons that are made often retain an immature phenotype, which limits their utility in modeling neuronal processes, including synaptic transmission. We report that combining NGN2 programming with SMAD and WNT inhibition generates human patterned induced neurons (hpiNs). Single-cell analyses showed that hpiN cultures contained cells along a developmental continuum, ranging from poorly differentiated neuronal progenitors to well-differentiated, excitatory glutamatergic neurons. The most differentiated neurons could be identified using a CAMK2A::GFP reporter gene and exhibited greater functionality, including NMDAR-mediated synaptic transmission. We conclude that utilizing single-cell and reporter gene approaches for selecting successfully programmed cells for study will greatly enhance the utility of hpiNs and other programmed neuronal populations in the modeling of nervous system disorders.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Adult , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Differentiation , Cells, Cultured , Fetus/cytology , Gene Expression Regulation , Humans , Neurons/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Smad Proteins/metabolism , Synapses/metabolism , Time Factors , Transcription, Genetic , Wnt Proteins/metabolism
10.
Cell Stem Cell ; 19(6): 703-708, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912091

ABSTRACT

Zika virus (ZIKV) can cross the placental barrier, resulting in infection of the fetal brain and neurological defects including microcephaly. The cellular tropism of ZIKV and the identity of attachment factors used by the virus to gain access to key cell types involved in pathogenesis are under intense investigation. Initial studies suggested that ZIKV preferentially targets neural progenitor cells (NPCs), providing an explanation for the developmental phenotypes observed in some pregnancies. The AXL protein has been nominated as a key attachment factor for ZIKV in several cell types including NPCs. However, here we show that genetic ablation of AXL has no effect on ZIKV entry or ZIKV-mediated cell death in human induced pluripotent stem cell (iPSC)-derived NPCs or cerebral organoids. These findings call into question the utility of AXL inhibitors for preventing birth defects after infection and suggest that further studies of viral attachment factors in NPCs are needed.


Subject(s)
Cerebrum/pathology , Gene Deletion , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Neuroprotection , Organoids/virology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Zika Virus Infection/prevention & control , Cell Death , Gene Knockout Techniques , Humans , Neural Stem Cells/pathology , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Zika Virus Infection/pathology , Axl Receptor Tyrosine Kinase
11.
Elife ; 52016 07 26.
Article in English | MEDLINE | ID: mdl-27458797

ABSTRACT

Dravet Syndrome is an intractable form of childhood epilepsy associated with deleterious mutations in SCN1A, the gene encoding neuronal sodium channel Nav1.1. Earlier studies using human induced pluripotent stem cells (iPSCs) have produced mixed results regarding the importance of Nav1.1 in human inhibitory versus excitatory neurons. We studied a Nav1.1 mutation (p.S1328P) identified in a pair of twins with Dravet Syndrome and generated iPSC-derived neurons from these patients. Characterization of the mutant channel revealed a decrease in current amplitude and hypersensitivity to steady-state inactivation. We then differentiated Dravet-Syndrome and control iPSCs into telencephalic excitatory neurons or medial ganglionic eminence (MGE)-like inhibitory neurons. Dravet inhibitory neurons showed deficits in sodium currents and action potential firing, which were rescued by a Nav1.1 transgene, whereas Dravet excitatory neurons were normal. Our study identifies biophysical impairments underlying a deleterious Nav1.1 mutation and supports the hypothesis that Dravet Syndrome arises from defective inhibitory neurons.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Mutation , NAV1.1 Voltage-Gated Sodium Channel/deficiency , Neurons/physiology , Telencephalon/physiology , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology
12.
Cell Stem Cell ; 14(1): 40-52, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24239284

ABSTRACT

Reprogramming differentiated cells into induced pluripotent stem cells (iPSCs) promotes a broad array of cellular changes. Here we show that the let-7 family of microRNAs acts as an inhibitory influence on the reprogramming process through a regulatory pathway involving prodifferentiation factors, including EGR1. Inhibiting let-7 in human cells promotes reprogramming to a comparable extent to c-MYC when combined with OCT4, SOX2, and KLF4, and persistence of let-7 inhibits reprogramming. Inhibiting let-7 during reprogramming leads to an increase in the level of the let-7 target LIN-41/TRIM71, which in turn promotes reprogramming and is important for overcoming the let-7 barrier to reprogramming. Mechanistic studies revealed that LIN-41 regulates a broad array of differentiation genes, and more specifically, inhibits translation of EGR1 through binding its cognate mRNA. Together our findings outline a let-7-based pathway that counteracts the activity of reprogramming factors through promoting the expression of prodifferentiation genes.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Early Growth Response Protein 1/metabolism , Induced Pluripotent Stem Cells/cytology , MicroRNAs/genetics , Ubiquitin-Protein Ligases/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Early Growth Response Protein 1/genetics , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
13.
BMC Mol Biol ; 10: 33, 2009 Apr 22.
Article in English | MEDLINE | ID: mdl-19386123

ABSTRACT

BACKGROUND: The Male Specific Lethal (MSL) complex is enriched on the single X chromosome in male Drosophila cells and functions to upregulate X-linked gene expression and equalize X-linked gene dosage with XX females. The zinc finger protein Zn72D is required for productive splicing of the maleless (mle) transcript, which encodes an essential subunit of the MSL complex. In the absence of Zn72D, MLE levels are decreased, and as a result, the MSL complex no longer localizes to the X chromosome and dosage compensation is disrupted. To understand the molecular basis of Zn72D function, we identified proteins that interact with Zn72D. RESULTS: Among several proteins that associate with Zn72D, we found the DEAD box helicase Belle (Bel). Simultaneous knockdown of Zn72D and bel restored MSL complex localization to the X chromosome and dosage compensation. MLE protein was restored to 70% of wild-type levels, although the level of productively spliced mle transcript was still four-fold lower than in wild-type cells. The increase in production of MLE protein relative to the amount of correctly spliced mle mRNA could not be attributed to an alteration in MLE stability. CONCLUSION: These data indicate that Zn72D and Bel work together to control mle splicing and protein levels. Thus Zn72D and Bel may be factors that coordinate splicing and translational regulation.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Gene Expression Regulation , RNA, Messenger/metabolism , Animals , Cell Line , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Protein Synthesis Inhibitors/pharmacology , RNA Splicing/genetics , X Chromosome/genetics
14.
Mol Cell Biol ; 27(24): 8760-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923683

ABSTRACT

In organisms with sex chromosomes, dosage compensation equalizes gene expression between the sexes. In Drosophila melanogaster males, the male-specific lethal (MSL) complex of proteins and two noncoding roX RNAs coat the X chromosome, resulting in a twofold transcriptional upregulation to equalize gene expression with that of females. How MSL complex enrichment on the X chromosome is regulated is not well understood. We performed an RNA interference screen to identify new factors required for dosage compensation. Using a Drosophila Schneider S2 cell line in which green fluorescent protein (GFP)-tagged MSL2 localizes to the X chromosome, we assayed approximately 7,200 knockdowns for their effects on GFP-MSL2 distribution. One factor identified is the zinc finger protein Zn72D. In its absence, the MSL complex no longer coats the X chromosome. We demonstrate that Zn72D is required for productive splicing of the transcript for the MSL protein Maleless, explaining the dosage compensation defect. However, Zn72D is required for the viability of both sexes, indicating its functions are not sex specific. Consistent with this, Zn72D colocalizes with elongating RNA polymerase II, implicating it as a more general factor involved in RNA metabolism.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , RNA Splicing/genetics , Transcription Factors/genetics , Zinc Fingers , Animals , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA, Complementary/metabolism , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Female , Genes, X-Linked , Introns , Male , Multiprotein Complexes/metabolism , Protein Transport , RNA Interference , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transgenes , X Chromosome/metabolism
15.
Chromosoma ; 114(3): 183-92, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15986205

ABSTRACT

Chromatin modifications are among the epigenetic alterations essential for genetic reprogramming during development. The Polycomb group (PcG) gene family mediates chromatin modifications that contribute to developmentally regulated transcriptional silencing. Trimethylation of histone H3 on lysine 27, mediated by a PcG protein complex consisting of Eed, Ezh2, and Suz12, is integral in differentiation, stem cell self-renewal, and tumorigenesis. Eed and Ezh2 are also implicated in the developmentally regulated silencing of the inactive X chromosome, as they are transiently enriched on the inactive X chromosome when X chromosome silencing is initiated. Here we analyze the dynamic localization of Suz12 during cellular differentiation and X-inactivation. Though Suz12 is a requisite member of the Eed/Ezh2 complexes, we found that Suz12 exhibits a notable difference from Ezh2 and Eed: while Ezh2 and Eed levels decrease during stem cell differentiation, Suz12 levels remain constant. Despite the differential regulation in abundance of Suz12 and Eed/Ezh2, Suz12 is also transiently enriched on the Xi during early stages of X-inactivation, and this accumulation is Xist RNA dependent. These results suggest that Suz12 may have a function that is not mediated by its association with Eed and Ezh2, and that this additional function is not involved in the regulation of X-inactivation.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , X Chromosome Inactivation , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Neoplasm Proteins , Nuclear Proteins , Polycomb Repressive Complex 2 , Protein Methyltransferases , RNA, Long Noncoding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Transfection , Trophoblasts/cytology , Trophoblasts/metabolism
16.
Prog Mol Subcell Biol ; 38: 91-122, 2005.
Article in English | MEDLINE | ID: mdl-15881892

ABSTRACT

In female mammals, one X chromosome is transcriptionally silenced to achieve dosage compensation between XX females and XY males. This process, known as X-inactivation, occurs early in development, such that one X chromosome is silenced in every cell. Once X-inactivation has occurred, the inactive X chromosome is marked by a unique set of epigenetic features that distinguishes it from the active X chromosome and autosomes. These modifications appear sequentially during the transition from a transcriptionally active to an inactive state and, once established, act redundantly to maintain transcriptional silencing. In this review, we survey the unique epigenetic features that characterize the inactive X chromosome, describe the mechanisms by which these marks are established and maintained, and discuss how each contributes to silencing the inactive X chromosome.


Subject(s)
Chromatin/metabolism , Dosage Compensation, Genetic , X Chromosome/genetics , X Chromosome/metabolism , Animals , Chromatin/genetics , DNA/chemistry , DNA/metabolism , DNA Replication , Female , Histones/metabolism , Humans , Male , Methylation , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Stem Cells/physiology
17.
Science ; 300(5616): 131-5, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12649488

ABSTRACT

The Polycomb group (PcG) protein Eed is implicated in regulation of imprinted X-chromosome inactivation in extraembryonic cells but not of random X inactivation in embryonic cells. The Drosophila homolog of the Eed-Ezh2 PcG protein complex achieves gene silencing through methylation of histone H3 on lysine 27 (H3-K27), which suggests a role for H3-K27 methylation in imprinted X inactivation. Here we demonstrate that transient recruitment of the Eed-Ezh2 complex to the inactive X chromosome (Xi) occurs during initiation of X inactivation in both extraembryonic and embryonic cells and is accompanied by H3-K27 methylation. Recruitment of the complex and methylation on the Xi depend on Xist RNA but are independent of its silencing function. Together, our results suggest a role for Eed-Ezh2-mediated H3-K27 methylation during initiation of both imprinted and random X inactivation and demonstrate that H3-K27 methylation is not sufficient for silencing of the Xi.


Subject(s)
Blastocyst/physiology , Dosage Compensation, Genetic , Histones/metabolism , Stem Cells/physiology , Trophoblasts/physiology , X Chromosome/metabolism , Animals , Blastocyst/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Female , Fluorescent Antibody Technique , Genomic Imprinting , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Lysine/metabolism , Male , Methylation , Mice , Mutation , Polycomb Repressive Complex 2 , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Transgenes
18.
J Virol ; 76(22): 11551-60, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388716

ABSTRACT

The latent membrane protein-1 (LMP-1) of Epstein-Barr virus (EBV) contributes to the proliferation of infected B lymphocytes by signaling through its binding to cellular signaling molecules. It apparently mimics members of the tumor necrosis factor receptor family, in particular, CD40, by binding a similar set of cellular molecules as does CD40. LMP-1 differs dramatically in its structure from CD40. LMP-1 has six membrane-spanning domains as opposed to CD40's one. LMP-1 also differs from CD40 in its apparent independence of a ligand for its signaling. We have examined the role of LMP-1's membrane-spanning domains in its signaling. Their substitution with six membrane-spanning domains from the LMP-2A protein of EBV yields a derivative which neither coimmunoprecipitates with LMP-1 nor signals to increase the activity of NF-kappaB as does wild-type LMP-1. These observations indicate that LMP-1 has specific sequences in its membrane-spanning domains required for these activities. LMP-1's first and sixth membrane-spanning domains have multiple leucine residues potentially similar to leucine-heptad motifs that can mediate protein-protein interactions in membranes (Gurezka et al., J. Biol. Chem. 274:9265-9270, 1999). Substitution of seven leucines in LMP-1's sixth membrane-spanning domain has no effect on its function, whereas similar substitutions in its first membrane-spanning domain yielded a derivative which aggregates as does wild-type LMP-1 but has only 3% of wild-type's ability to signal through NF-kappaB. Importantly, this derivative complements a mutant of LMP-1 with wild-type membrane-spanning domains but no carboxy-terminal signaling domain. These findings together indicate that the membrane-spanning domains of LMP-1 contribute multiple functions to its signaling.


Subject(s)
Herpesvirus 4, Human/metabolism , Signal Transduction , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Amino Acid Sequence , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cell Line , Cell Membrane/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Structure-Activity Relationship , Transcriptional Activation , Transfection , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...