Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Appl Crystallogr ; 54(Pt 4): 1034-1046, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34429718

ABSTRACT

A novel capillary-based microfluidic strategy to accelerate the process of small-molecule-compound screening by room-temperature X-ray crystallography using protein crystals is reported. The ultra-thin microfluidic devices are composed of a UV-curable polymer, patterned by cleanroom photolithography, and have nine capillary channels per chip. The chip was designed for ease of sample manipulation, sample stability and minimal X-ray background. 3D-printed frames and cassettes conforming to SBS standards are used to house the capillary chips, providing additional mechanical stability and compatibility with automated liquid- and sample-handling robotics. These devices enable an innovative in situ crystal-soaking screening workflow, akin to high-throughput compound screening, such that quantitative electron density maps sufficient to determine weak binding events are efficiently obtained. This work paves the way for adopting a room-temperature microfluidics-based sample delivery method at synchrotron sources to facilitate high-throughput protein-crystallography-based screening of compounds at high concentration with the aim of discovering novel binding events in an automated manner.

2.
Hum Mol Genet ; 27(16): 2830-2839, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29771329

ABSTRACT

Peripheral nerve myelination is adversely affected in the most common form of the hereditary peripheral neuropathy called Charcot-Marie-Tooth Disease. This form, classified as CMT1A, is caused by a 1.4 Mb duplication on chromosome 17, which includes the abundantly expressed Schwann cell myelin gene, Peripheral Myelin Protein 22 (PMP22). This is one of the most common copy number variants causing neurological disease. Overexpression of Pmp22 in rodent models recapitulates several aspects of neuropathy, and reduction of Pmp22 in such models results in amelioration of the neuropathy phenotype. Recently we identified a potential super-enhancer approximately 90-130 kb upstream of the Pmp22 transcription start sites. This super-enhancer encompasses a cluster of individual enhancers that have the acetylated histone H3K27 active enhancer mark, and coincides with smaller duplications identified in patients with milder CMT1A-like symptoms, where the PMP22 coding region itself was not part of the duplication. In this study, we have utilized genome editing to create a deletion of this super-enhancer to determine its role in Pmp22 regulation. Our data show a significant decrease in Pmp22 transcript expression using allele-specific internal controls. Moreover, the P2 promoter of the Pmp22 gene, which is used in other cell types, is affected, but we find that the Schwann cell-specific P1 promoter is disproportionately more sensitive to loss of the super-enhancer. These data show for the first time the requirement of these upstream enhancers for full Pmp22 expression.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , DNA Copy Number Variations/genetics , Enhancer Elements, Genetic/genetics , Myelin Proteins/genetics , Animals , Cell Line , Charcot-Marie-Tooth Disease/pathology , Chromosome Duplication/genetics , Chromosomes, Human, Pair 17/genetics , Gene Editing , Gene Expression Regulation/genetics , Humans , Promoter Regions, Genetic , Rats , Schwann Cells/pathology
3.
J Neurosci ; 36(35): 9135-47, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27581455

ABSTRACT

UNLABELLED: The rapid and dynamic transcriptional changes of Schwann cells in response to injury are critical to peripheral nerve repair, yet the epigenomic reprograming that leads to the induction of injury-activated genes has not been characterized. Polycomb Repressive Complex 2 (PRC2) catalyzes the trimethylation of lysine 27 of histone H3 (H3K27me3), which produces a transcriptionally repressive chromatin environment. We find that many promoters and/or gene bodies of injury-activated genes of mature rat nerves are occupied with H3K27me3. In contrast, the majority of distal enhancers that gain H3K27 acetylation after injury are not repressed by H3K27 methylation before injury, which is normally observed in developmentally poised enhancers. Injury induces demethylation of H3K27 in many genes, such as Sonic hedgehog (Shh), which is silenced throughout Schwann cell development before injury. In addition, experiments using a Schwann cell-specific mouse knock-out of the Eed subunit of PRC2 indicate that demethylation is a rate-limiting step in the activation of such genes. We also show that some transcription start sites of H3K27me3-repressed injury genes of uninjured nerves are bound with a mark of active promoters H3K4me3, for example, Shh and Gdnf, and the reduction of H3K27me3 results in increased trimethylation of H3K4. Our findings identify reversal of polycomb repression as a key step in gene activation after injury. SIGNIFICANCE STATEMENT: Peripheral nerve regeneration after injury is dependent upon implementation of a novel genetic program in Schwann cells that supports axonal survival and regeneration. Identifying means to enhance Schwann cell reprogramming after nerve injury could be used to foster effective remyelination in the treatment of demyelinating disorders and in identifying pathways involved in regenerative process of myelination. Although recent progress has identified transcriptional determinants of successful reprogramming of the Schwann cell transcriptome after nerve injury, our results have highlighted a novel epigenomic pathway in which reversal of the Polycomb pathway of repressive histone methylation is required for activation of a significant number of injury-induced genes.


Subject(s)
Cellular Reprogramming/physiology , Epigenomics , Gene Expression Regulation/physiology , Schwann Cells/metabolism , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Animals , Benzazepines/pharmacology , Cellular Reprogramming/genetics , Chromatin Immunoprecipitation , Computational Biology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Histones/genetics , Histones/metabolism , In Vitro Techniques , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Regeneration/genetics , Schwann Cells/drug effects , Schwann Cells/physiology , Signal Transduction/genetics
4.
Mol Cell Endocrinol ; 421: 82-97, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26678830

ABSTRACT

The molecular basis of the organogenesis, homeostasis, and tumorigenesis of the adrenal cortex has been the subject of intense study for many decades. Specifically, characterization of tumor predisposition syndromes with adrenocortical manifestations and molecular profiling of sporadic adrenocortical tumors have led to the discovery of key molecular pathways that promote pathological adrenal growth. However, given the observational nature of such studies, several important questions regarding the molecular pathogenesis of adrenocortical tumors have remained. This review will summarize naturally occurring and genetically engineered mouse models that have provided novel tools to explore the molecular and cellular underpinnings of adrenocortical tumors. New paradigms of cancer initiation, maintenance, and progression that have emerged from this work will be discussed.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Disease Models, Animal , Adrenal Cortex Neoplasms/metabolism , Animals , Humans , Insulin-Like Growth Factor II/genetics , Mice , Tumor Suppressor Protein p53/genetics , Wnt Signaling Pathway
5.
J Neurosci ; 35(22): 8640-52, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041929

ABSTRACT

Myelination of peripheral nerves by Schwann cells requires coordinate regulation of gene repression as well as gene activation. Several chromatin remodeling pathways critical for peripheral nerve myelination have been identified, but the functions of histone methylation in the peripheral nerve have not been elucidated. To determine the role of histone H3 Lys27 methylation, we have generated mice with a Schwann cell-specific knock-out of Eed, which is an essential subunit of the polycomb repressive complex 2 (PRC2) that catalyzes methylation of histone H3 Lys27. Analysis of this mutant revealed no significant effects on early postnatal development of myelin. However, its loss eventually causes progressive hypermyelination of small-diameter axons and apparent fragmentation of Remak bundles. These data identify the PRC2 complex as an epigenomic modulator of mature myelin thickness, which is associated with changes in Akt phosphorylation. Interestingly, we found that Eed inactivation causes derepression of several genes, e.g., Sonic hedgehog (Shh) and Insulin-like growth factor-binding protein 2 (Igfbp2), that become activated after nerve injury, but without activation of a primary regulator of the injury program, c-Jun. Analysis of the activated genes in cultured Schwann cells showed that Igfbp2 regulates Akt activation. Our results identify an epigenomic pathway required for establishing thickness of mature myelin and repressing genes that respond to nerve injury.


Subject(s)
Gene Expression Regulation/physiology , Myelin Sheath/metabolism , Polycomb Repressive Complex 2/metabolism , Schwann Cells/metabolism , Sciatic Nerve/cytology , Animals , Animals, Newborn , Cells, Cultured , Chromatin Immunoprecipitation , Hedgehog Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Myelin P0 Protein/genetics , Oncogene Protein v-akt/metabolism , Polycomb Repressive Complex 2/genetics , Rats , Sciatic Nerve/ultrastructure , Signal Transduction/physiology , Transfection
6.
Glia ; 63(11): 1897-1914, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25974668

ABSTRACT

Myelin is formed by specialized myelinating glia: oligodendrocytes and Schwann cells in the central and peripheral nervous systems, respectively. While there are distinct developmental aspects and regulatory pathways in these two cell types, myelination in both systems requires the transcriptional activator Sox10. Sox10 interacts with cell type-specific transcription factors at some loci to induce myelin gene expression, but it is largely unknown how Sox10 transcriptional networks globally compare between oligodendrocytes and Schwann cells. We used in vivo ChIP-Seq analysis of spinal cord and peripheral nerve (sciatic nerve) to identify unique and shared Sox10 binding sites and assess their correlation with active enhancers and transcriptional profiles in oligodendrocytes and Schwann cells. Sox10 binding sites overlap with active enhancers and critical cell type-specific regulators of myelination, such as Olig2 and Myrf in oligodendrocytes, and Egr2/Krox20 in Schwann cells. Sox10 sites also associate with genes critical for myelination in both oligodendrocytes and Schwann cells and are found within super-enhancers previously defined in brain. In Schwann cells, Sox10 sites contain binding motifs of putative partners in the Sp/Klf, Tead, and nuclear receptor protein families. Specifically, siRNA analysis of nuclear receptors Nr2f1 and Nr2f2 revealed downregulation of myelin genes Mbp and Ndrg1 in primary Schwann cells. Our analysis highlights different mechanisms that establish cell type-specific genomic occupancy of Sox10, which reflects the unique characteristics of oligodendrocyte and Schwann cell differentiation. GLIA 2015;63:1897-1914.

7.
J Biol Chem ; 290(11): 6937-50, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25614629

ABSTRACT

Myelination of the peripheral nervous system is required for axonal function and long term stability. After peripheral nerve injury, Schwann cells transition from axon myelination to a demyelinated state that supports neuronal survival and ultimately remyelination of axons. Reprogramming of gene expression patterns during development and injury responses is shaped by the actions of distal regulatory elements that integrate the actions of multiple transcription factors. We used ChIP-seq to measure changes in histone H3K27 acetylation, a mark of active enhancers, to identify enhancers in myelinating rat peripheral nerve and their dynamics after demyelinating nerve injury. Analysis of injury-induced enhancers identified enriched motifs for c-Jun, a transcription factor required for Schwann cells to support nerve regeneration. We identify a c-Jun-bound enhancer in the gene for Runx2, a transcription factor induced after nerve injury, and we show that Runx2 is required for activation of other induced genes. In contrast, enhancers that lose H3K27ac after nerve injury are enriched for binding sites of the Sox10 and early growth response 2 (Egr2/Krox20) transcription factors, which are critical determinants of Schwann cell differentiation. Egr2 expression is lost after nerve injury, and many Egr2-binding sites lose H3K27ac after nerve injury. However, the majority of Egr2-bound enhancers retain H3K27ac, indicating that other transcription factors maintain active enhancer status after nerve injury. The global epigenomic changes in H3K27ac deposition pinpoint dynamic changes in enhancers that mediate the effects of transcription factors that control Schwann cell myelination and peripheral nervous system responses to nerve injury.


Subject(s)
Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Peripheral Nerve Injuries/metabolism , Schwann Cells/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Demyelinating Diseases/complications , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Male , Mice , Myelin Sheath/genetics , Myelin Sheath/pathology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction , Transcription Factors/genetics
8.
PLoS One ; 9(11): e109691, 2014.
Article in English | MEDLINE | ID: mdl-25380244

ABSTRACT

As next generation sequencing technologies are becoming more economical, large-scale ChIP-seq studies are enabling the investigation of the roles of transcription factor binding and epigenome on phenotypic variation. Studying such variation requires individual level ChIP-seq experiments. Standard designs for ChIP-seq experiments employ a paired control per ChIP-seq sample. Genomic coverage for control experiments is often sacrificed to increase the resources for ChIP samples. However, the quality of ChIP-enriched regions identifiable from a ChIP-seq experiment depends on the quality and the coverage of the control experiments. Insufficient coverage leads to loss of power in detecting enrichment. We investigate the effect of in silico pooling of control samples within multiple biological replicates, multiple treatment conditions, and multiple cell lines and tissues across multiple datasets with varying levels of genomic coverage. Our computational studies suggest guidelines for performing in silico pooling of control experiments. Using vast amounts of ENCODE data, we show that pairwise correlations between control samples originating from multiple biological replicates, treatments, and cell lines/tissues can be grouped into two classes representing whether or not in silico pooling leads to power gain in detecting enrichment between the ChIP and the control samples. Our findings have important implications for multiplexing samples.


Subject(s)
Chromatin Immunoprecipitation , Computational Biology/methods , Computer Simulation , High-Throughput Nucleotide Sequencing , Animals , CCCTC-Binding Factor , Cluster Analysis , Histone Deacetylases/genetics , Histones/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , K562 Cells , Rats , Repressor Proteins/genetics
9.
J Neurosci ; 32(5): 1517-27, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302795

ABSTRACT

Several key transcription factors and coregulators important to peripheral nerve myelination have been identified, but the contributions of specific chromatin remodeling complexes to peripheral nerve myelination have not been analyzed. Chromodomain helicase DNA-binding protein 4 (Chd4) is the core catalytic subunit of the nucleosome remodeling and deacetylase (NuRD) chromatin remodeling complex. Previous studies have shown Chd4 interacts with Nab (NGFI-A/Egr-binding) corepressors, which are required for early growth response 2 (Egr2/Krox20), to direct peripheral nerve myelination by Schwann cells. In this study, we examined the developmental importance of the NuRD complex in peripheral nerve myelination through the generation of conditional Chd4 knock-out mice in Schwann cells (Chd4(loxP/loxP); P0-cre). Chd4 conditional null mice were found to have delayed myelination, radial sorting defects, hypomyelination, and the persistence of promyelinating Schwann cells. Loss of Chd4 leads to elevated expression of immature Schwann cell genes (Id2, c-Jun, and p75), and sustained expression of the promyelinating Schwann cell gene, Oct6/Scip, without affecting the levels of Egr2/Krox20. Furthermore, Schwann cell proliferation is upregulated in Chd4-null sciatic nerve. In vivo chromatin immunoprecipitation studies reveal recruitment of Chd4 and another NuRD component, Mta2, to genes that are positively and negatively regulated by Egr2 during myelination. Together, these results underscore the necessity of Chd4 function to guide proper terminal differentiation of Schwann cells and implicate the NuRD chromatin remodeling complex as a requisite factor in timely and stable peripheral nerve myelination.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/physiology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Myelin Sheath/enzymology , Nerve Fibers, Myelinated/enzymology , Peripheral Nerves/enzymology , Animals , Animals, Newborn , Cell Differentiation/genetics , Chromatin Assembly and Disassembly/genetics , DNA Helicases/deficiency , Mi-2 Nucleosome Remodeling and Deacetylase Complex/deficiency , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Nerve Fibers, Myelinated/physiology , Nucleosomes/enzymology , Nucleosomes/genetics , Nucleosomes/ultrastructure , Peripheral Nerves/ultrastructure , Schwann Cells/enzymology , Schwann Cells/ultrastructure
10.
J Biol Chem ; 287(2): 1235-41, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22110129

ABSTRACT

Jarid2/Jumonji, the founding member of the Jmj factor family, critically regulates various developmental processes, including cardiovascular development. The Jmj family was identified as histone demethylases, indicating epigenetic regulation by Jmj proteins. Deletion of Jarid2 in mice resulted in cardiac malformation and increased endocardial Notch1 expression during development. Although Jarid2 has been shown to occupy the Notch1 locus in the developing heart, the precise molecular role of Jarid2 remains unknown. Here we show that deletion of Jarid2 results in reduced methylation of lysine 9 on histone H3 (H3K9) at the Notch1 genomic locus in embryonic hearts. Interestingly, SETDB1, a histone H3K9 methyltransferase, was identified as a putative cofactor of Jarid2 by yeast two-hybrid screening, and the physical interaction between Jarid2 and SETDB1 was confirmed by coimmunoprecipitation experiments. Concurrently, accumulation of SETDB1 at the site of Jarid2 occupancy was significantly reduced in Jarid2 knock out (KO) hearts. Employing genome-wide approaches, putative Jarid2 target genes regulated by SETDB1 via H3K9 methylation were identified in the developing heart by ChIP-chip. These targets are involved in biological processes that, when dysregulated, could manifest in the phenotypic defects observed in Jarid2 KO mice. Our data demonstrate that Jarid2 functions as a transcriptional repressor of target genes, including Notch1, through a novel process involving the modification of H3K9 methylation via specific interaction with SETDB1 during heart development. Therefore, our study provides new mechanistic insights into epigenetic regulation by Jarid2, which will enhance our understanding of the molecular basis of other organ development and biological processes.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart/embryology , Histones/metabolism , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/biosynthesis , Repressor Proteins/metabolism , Animals , Epigenesis, Genetic/physiology , Female , Genome-Wide Association Study , Histone-Lysine N-Methyltransferase , Histones/genetics , Male , Methylation , Mice , Mice, Knockout , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Polycomb Repressive Complex 2 , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational/physiology , Receptor, Notch1/genetics , Repressor Proteins/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...