Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Hum Mol Genet ; 32(2): 177-191, 2023 01 06.
Article in English | MEDLINE | ID: mdl-35925868

ABSTRACT

Mutations in LMNA, the gene encoding A-type lamins, cause laminopathies-diseases of striated muscle and other tissues. The aetiology of laminopathies has been attributed to perturbation of chromatin organization or structural weakening of the nuclear envelope (NE) such that the nucleus becomes more prone to mechanical damage. The latter model requires a conduit for force transmission to the nucleus. NE-associated Linker of Nucleoskeleton and Cytoskeleton (LINC) complexes are one such pathway. Using clustered regularly interspaced short palindromic repeats to disrupt the Nesprin-1 KASH (Klarsicht, ANC-1, Syne Homology) domain, we identified this LINC complex protein as the predominant NE anchor for microtubule cytoskeleton components, including nucleation activities and motor complexes, in mouse cardiomyocytes. Loss of Nesprin-1 LINC complexes resulted in loss of microtubule cytoskeleton proteins at the nucleus and changes in nuclear morphology and positioning in striated muscle cells, but with no overt physiological defects. Disrupting the KASH domain of Nesprin-1 suppresses Lmna-linked cardiac pathology, likely by reducing microtubule cytoskeleton activities at the nucleus. Nesprin-1 LINC complexes thus represent a potential therapeutic target for striated muscle laminopathies.


Subject(s)
Laminopathies , Muscle, Striated , Animals , Mice , Microtubule Proteins/metabolism , Nuclear Proteins/metabolism , Membrane Proteins/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Nuclear Matrix/genetics , Microtubules/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Intermediate Filament Proteins/metabolism , Muscle, Striated/metabolism , Laminopathies/metabolism
2.
BMC Genomics ; 23(1): 725, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36284304

ABSTRACT

BACKGROUND: Eukaryotic genome is compartmentalized into structural and functional domains. One of the concepts of higher order organization of chromatin posits that the DNA is organized in constrained loops that behave as independent functional domains. Nuclear Matrix (NuMat), a ribo-proteinaceous nucleoskeleton, provides the structural basis for this organization. DNA sequences located at base of the loops are known as the Matrix Attachment Regions (MARs). NuMat relates to multiple nuclear processes and is partly cell type specific in composition. It is a biochemically defined structure and several protocols have been used to isolate the NuMat where some of the steps have been critically evaluated. These sequences play an important role in genomic organization it is imperative to know their dynamics during development and differentiation. RESULTS: Here we look into the dynamics of MARs when the preparation process is varied and during embryonic development of D. melanogaster. A subset of MARs termed as "Core-MARs" present abundantly in pericentromeric heterochromatin, are constant unalterable anchor points as they associate with NuMat through embryonic development and are independent of the isolation procedure. Euchromatic MARs are dynamic and reflect the transcriptomic profile of the cell. New MARs are generated by nuclear stabilization, and during development, mostly at paused RNA polymerase II promoters. Paused Pol II MARs depend on RNA transcripts for NuMat association. CONCLUSIONS: Our data reveals the role of MARs in functionally dynamic nucleus and contributes to the current understanding of nuclear architecture in genomic context.


Subject(s)
Drosophila melanogaster , Heterochromatin , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Heterochromatin/metabolism , RNA Polymerase II/metabolism , Nuclear Matrix/genetics , Nuclear Matrix/chemistry , Nuclear Matrix/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA/metabolism , RNA/metabolism
3.
Curr Opin Genet Dev ; 75: 101940, 2022 08.
Article in English | MEDLINE | ID: mdl-35777349

ABSTRACT

There is a long experimental history supporting the principle that RNA is essential for normal nuclear and chromatin architecture. Most of the genome is transcribed into RNA but only 2% of the sequence codes for proteins. In the nucleus, most non-coding RNA, packaged in proteins, is bound into structures including chromatin and a non-chromatin scaffolding, the nuclear matrix, which was first observed by electron microscopy. Removing nuclear RNA or inhibiting its transcription causes the condensation of chromatin, showing the importance of RNA in spatially and functionally organizing the genome. Today, powerful techniques for the molecular characterization of RNA and for mapping its spatial organization in the nucleus have provided molecular detail to these principles.


Subject(s)
Cell Nucleus , Ribonucleoproteins , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Nuclear Matrix/chemistry , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , RNA/metabolism , Ribonucleoproteins/analysis , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
4.
STAR Protoc ; 3(2): 101394, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35600936

ABSTRACT

Nuclear Matrix (NuMat) is a biochemically defined entity that provides us with a snapshot of the features of the nuclear architecture. Here, we present a protocol to isolate and visualize NuMat in situ in the intact embryo or tissues of Drosophila melanogaster and its applications. We remove the chromatin to reveal underlying nuclear architectural components in organismal context. This protocol couples the power of Drosophila genetics with cell biological observation of the nuclear architecture. For complete details on the use and execution of this protocol, please refer to Pathak et al. (2022), Sureka et al. (2018), and Pathak et al. (2013).


Subject(s)
Drosophila melanogaster , Nuclear Matrix , Animals , Cell Nucleus/genetics , Chromatin/genetics , Drosophila/genetics , Drosophila melanogaster/genetics , Nuclear Matrix/genetics
5.
Mol Cell ; 81(17): 3509-3525.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34320406

ABSTRACT

Nuclear chromosomes transcribe far more RNA than required to encode protein. Here we investigate whether non-coding RNA broadly contributes to cytological-scale chromosome territory architecture. We develop a procedure that depletes soluble proteins, chromatin, and most nuclear RNA from the nucleus but does not delocalize XIST, a known architectural RNA, from an insoluble chromosome "scaffold." RNA-seq analysis reveals that most RNA in the nuclear scaffold is repeat-rich, non-coding, and derived predominantly from introns of nascent transcripts. Insoluble, repeat-rich (C0T-1) RNA co-distributes with known scaffold proteins including scaffold attachment factor A (SAF-A), and distribution of these components inversely correlates with chromatin compaction in normal and experimentally manipulated nuclei. We further show that RNA is required for SAF-A to interact with chromatin and for enrichment of structurally embedded "scaffold attachment regions" prevalent in euchromatin. Collectively, the results indicate that long nascent transcripts contribute a dynamic structural role that promotes the open architecture of active chromosome territories.


Subject(s)
Chromatin/metabolism , Nuclear Matrix/metabolism , RNA, Untranslated/metabolism , Animals , Cell Line , Cell Nucleus/physiology , Chromatin/genetics , Chromosomes/genetics , Chromosomes/metabolism , Euchromatin/metabolism , Humans , Mice , Nuclear Matrix/genetics , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Transcription, Genetic/genetics
6.
Mol Cancer Res ; 19(7): 1196-1207, 2021 07.
Article in English | MEDLINE | ID: mdl-33771882

ABSTRACT

Nuclear envelope proteins play an important role in regulating nuclear size and structure in cancer. Altered expression of nuclear lamins are found in many cancers and its expression is correlated with better clinical outcomes. The nucleus is the largest organelle in the cell with a diameter between 10 and 20 µm. Nuclear size significantly impacts cell migration. Nuclear structural changes are predicted to impact cancer metastasis by regulating cancer cell migration. Here we show emerin regulates nuclear structure in invasive breast cancer cells to impact cancer metastasis. Invasive breast cancer cells had 40% to 50% less emerin than control cells, which resulted in decreased nuclear size. Overexpression of GFP-emerin in invasive breast cancer cells rescued nuclear size and inhibited migration through 3.0 and 8.0 µm pores. Mutational analysis showed emerin binding to nucleoskeletal proteins was important for its regulation of nuclear structure, migration, and invasion. Importantly, emerin expression inhibited lung metastasis by 91% in orthotopic mouse models of breast cancer. Emerin nucleoskeleton-binding mutants failed to inhibit metastasis. These results support a model whereby emerin binding to the nucleoskeleton regulates nuclear structure to impact metastasis. In this model, emerin plays a central role in metastatic transformation, because decreased emerin expression during transformation causes the nuclear structural defects required for increased cell migration, intravasation, and extravasation. IMPLICATIONS: Modulating emerin expression and function represents new targets for therapeutic interventions of metastasis, because increased emerin expression rescued cancer metastasis.


Subject(s)
Breast Neoplasms/genetics , Cell Movement/genetics , Cell Nucleus/genetics , Membrane Proteins/genetics , Nuclear Matrix/genetics , Nuclear Proteins/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice, Nude , Microscopy, Confocal/methods , Neoplasm Metastasis , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism , Protein Binding , Transplantation, Heterologous
7.
J Biochem ; 169(3): 259-264, 2021 Apr 18.
Article in English | MEDLINE | ID: mdl-32745171

ABSTRACT

In the last decades, it has become increasingly clear how the modulation of spatial organization of chromatin over time and through the cell cycle is closely connected to gene function regulation. Different physicochemical stimuli contribute to the realization of specific transcriptional programs and finally to a specific cellular phenotype. In this review, we aim to describe the current knowledge about the dynamics regulating the movements and the interactions of molecules within the nucleus and their impact on gene functions. In particular, taking into account that these forces exert their effect in a nuclear environment characterized by a high concentration of molecules, we will discuss the role of proteins and structures that regulate these movements and transduce physicochemical signals acting on the cell to the nucleus.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation , Animals , Cell Compartmentation , Chromatin/metabolism , Humans , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype
8.
J Biochem ; 169(3): 243-257, 2021 Apr 18.
Article in English | MEDLINE | ID: mdl-33351909

ABSTRACT

Actin is an essential regulator of cellular functions. In the eukaryotic cell nucleus, actin regulates chromatin as a bona fide component of chromatin remodelling complexes, it associates with nuclear RNA polymerases to regulate transcription and is involved in co-transcriptional assembly of nascent RNAs into ribonucleoprotein complexes. Actin dynamics are, therefore, emerging as a major regulatory factor affecting diverse cellular processes. Importantly, the involvement of actin dynamics in nuclear functions is redefining the concept of nucleoskeleton from a rigid scaffold to a dynamic entity that is likely linked to the three-dimensional organization of the nuclear genome. In this review, we discuss how nuclear actin, by regulating chromatin structure through phase separation may contribute to the architecture of the nuclear genome during cell differentiation and facilitate the expression of specific gene programs. We focus specifically on mitochondrial genes and how their dysregulation in the absence of actin raises important questions about the role of cytoskeletal proteins in regulating chromatin structure. The discovery of a novel pool of mitochondrial actin that serves as 'mitoskeleton' to facilitate organization of mtDNA supports a general role for actin in genome architecture and a possible function of distinct actin pools in the communication between nucleus and mitochondria.


Subject(s)
Actins/metabolism , Chromatin/metabolism , Nuclear Matrix/metabolism , Actins/genetics , Animals , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation , Genes, Mitochondrial , Genome , Humans , Nuclear Matrix/genetics , Transcription, Genetic
9.
Development ; 147(23)2020 12 13.
Article in English | MEDLINE | ID: mdl-33168584

ABSTRACT

DNA endoreplication has been implicated as a cell strategy for cell growth and in tissue injury. Here, we demonstrate that barrier-to-autointegration factor (BAF) represses endoreplication in Drosophila myofibers. We show that BAF localization at the nuclear envelope is eliminated in flies with mutations of the linker of nucleoskeleton and cytoskeleton (LINC) complex in which the LEM-domain protein Otefin is excluded, or after disruption of the nucleus-sarcomere connections. Furthermore, BAF localization at the nuclear envelope requires the activity of the BAF kinase VRK1/Ball, and, consistently, non-phosphorylatable BAF-GFP is excluded from the nuclear envelope. Importantly, removal of BAF from the nuclear envelope correlates with increased DNA content in the myonuclei. E2F1, a key regulator of endoreplication, overlaps BAF localization at the myonuclear envelope, and BAF removal from the nuclear envelope results in increased E2F1 levels in the nucleoplasm and subsequent elevated DNA content. We suggest that LINC-dependent and phosphosensitive attachment of BAF to the nuclear envelope, through its binding to Otefin, tethers E2F1 to the nuclear envelope thus inhibiting its accumulation in the nucleoplasm.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Endoreduplication/genetics , Membrane Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Cytoskeleton/genetics , DNA Replication/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental/genetics , Mutation/genetics , Myofibrils/genetics , Nuclear Envelope/genetics , Nuclear Matrix/genetics , Protamine Kinase/genetics
10.
J Mol Biol ; 432(23): 6028-6041, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33058875

ABSTRACT

Linker of nucleoskeleton and cytoskeleton (LINC) complexes are molecular tethers that span the nuclear envelope (NE) and physically connect the nucleus to the cytoskeleton. They transmit mechanical force across the NE in processes such as nuclear anchorage, nuclear migration, and homologous chromosome pairing during meiosis. LINC complexes are composed of KASH proteins traversing the outer nuclear membrane, and SUN proteins crossing the inner nuclear membrane. Humans have several SUN- and KASH-containing proteins, yet what governs their proper engagement is poorly understood. To investigate this question, we solved high resolution crystal structures of human SUN2 in complex with the KASH-peptides of Nesprin3, Nesprin4, and KASH5. In comparison to the published structures of SUN2-KASH1/2 we observe alternative binding modes for these KASH peptides. While the core interactions between SUN and the C-terminal residues of the KASH peptide are similar in all five complexes, the extended KASH-peptide adopts at least two different conformations. The much-improved resolution allows for a more detailed analysis of other elements critical for KASH interaction, including the KASH-lid and the cation loop, and a possible self-locked state for unbound SUN. In summary, we observe distinct differences between the examined SUN-KASH complexes. These differences may have an important role in regulating the SUN-KASH network.


Subject(s)
Cell Cycle Proteins/ultrastructure , Membrane Proteins/ultrastructure , Microfilament Proteins/ultrastructure , Multiprotein Complexes/ultrastructure , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Nucleolus/genetics , Cell Nucleolus/ultrastructure , Chromosome Pairing/genetics , Crystallography, X-Ray , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Meiosis/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nuclear Envelope/genetics , Nuclear Envelope/ultrastructure , Nuclear Matrix/genetics , Nuclear Matrix/ultrastructure , Peptides/chemistry , Peptides/genetics , Protein Conformation
11.
J Cell Biol ; 219(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32790861

ABSTRACT

LINC complexes are transmembrane protein assemblies that physically connect the nucleoskeleton and cytoskeleton through the nuclear envelope. Dysfunctions of LINC complexes are associated with pathologies such as cancer and muscular disorders. The mechanical roles of LINC complexes are poorly understood. To address this, we used genetically encoded FRET biosensors of molecular tension in a nesprin protein of the LINC complex of fibroblastic and epithelial cells in culture. We exposed cells to mechanical, genetic, and pharmacological perturbations, mimicking a range of physiological and pathological situations. We show that nesprin experiences tension generated by the cytoskeleton and acts as a mechanical sensor of cell packing. Moreover, nesprin discriminates between inductions of partial and complete epithelial-mesenchymal transitions. We identify the implicated mechanisms, which involve α-catenin capture at the nuclear envelope by nesprin upon its relaxation, thereby regulating ß-catenin transcription. Our data thus implicate LINC complex proteins as mechanotransducers that fine-tune ß-catenin signaling in a manner dependent on the epithelial-mesenchymal transition program.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Mechanotransduction, Cellular/genetics , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , beta Catenin/genetics , Animals , Biosensing Techniques , Dogs , Fluorescence Resonance Energy Transfer , Humans , Madin Darby Canine Kidney Cells , Mice , Microtubules/genetics , NIH 3T3 Cells , Nuclear Envelope/genetics , Nuclear Matrix/genetics
12.
Int J Mol Sci ; 20(21)2019 Oct 26.
Article in English | MEDLINE | ID: mdl-31717803

ABSTRACT

The cross-talk between stem cells and their microenvironment has been shown to have a direct impact on stem cells' decisions about proliferation, growth, migration, and differentiation. It is well known that stem cells, tissues, organs, and whole organisms change their internal architecture and composition in response to external physical stimuli, thanks to cells' ability to sense mechanical signals and elicit selected biological functions. Likewise, stem cells play an active role in governing the composition and the architecture of their microenvironment. Is now being documented that, thanks to this dynamic relationship, stemness identity and stem cell functions are maintained. In this work, we review the current knowledge in mechanobiology on stem cells. We start with the description of theoretical basis of mechanobiology, continue with the effects of mechanical cues on stem cells, development, pathology, and regenerative medicine, and emphasize the contribution in the field of the development of ex-vivo mechanobiology modelling and computational tools, which allow for evaluating the role of forces on stem cell biology.


Subject(s)
Cell Differentiation/physiology , Mechanotransduction, Cellular/physiology , Stem Cells/cytology , Animals , Biocompatible Materials , Biomechanical Phenomena , Computational Biology , Cytoskeleton/metabolism , Extracellular Matrix/physiology , Humans , Integrins/genetics , Integrins/metabolism , Nuclear Matrix/genetics , Nuclear Matrix/physiology , Regenerative Medicine , Stem Cell Niche , Stem Cells/metabolism
13.
Aging Cell ; 18(6): e13047, 2019 12.
Article in English | MEDLINE | ID: mdl-31576648

ABSTRACT

Cellular mechanisms that act in concert to maintain protein homeostasis (proteostasis) are vital for organismal functionality and survival. Nevertheless, subsets of aggregation-prone proteins form toxic aggregates (proteotoxicity) that in some cases, underlie the development of neurodegenerative diseases. Proteotoxic aggregates are often deposited in the vicinity of the nucleus, a process that is cytoskeleton-dependent. Accordingly, cytoskeletal dysfunction contributes to pathological hallmarks of various neurodegenerative diseases. Here, we asked whether the linker of nucleoskeleton and cytoskeleton (LINC) complex, which bridges these filaments across the nuclear envelope, is needed for the maintenance of proteostasis. Employing model nematodes, we discovered that knocking down LINC components impairs the ability of the worm to cope with proteotoxicity. Knocking down anc-1, which encodes a key component of the LINC complex, modulates the expression of transcription factors and E3 ubiquitin ligases, thereby affecting the rates of protein ubiquitination and impairing proteasome-mediated protein degradation. Our results establish a link between the LINC complex, protein degradation, and neurodegeneration-associated proteotoxicity.


Subject(s)
Caenorhabditis elegans/genetics , Cytoskeleton/genetics , Gene Expression Regulation , Nuclear Matrix/genetics , Proteasome Endopeptidase Complex/genetics , Proteostasis/genetics , Animals , Caenorhabditis elegans/metabolism , Cytoskeleton/metabolism , Gene Expression Profiling , Nuclear Matrix/metabolism , Proteasome Endopeptidase Complex/metabolism , Sequence Analysis, RNA
14.
Curr Genet ; 65(6): 1281-1285, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31147736

ABSTRACT

Correlation between nuclear and cell size, the nucleocytoplasmic ratio, is a cellular phenomenon that has been reported throughout eukaryotes for more than a century but the mechanisms that achieve it are not well understood. Here, we review work that has shed light on the cellular processes involved in nuclear size control. These studies have implicated nucleocytoplasmic transport, LINC complexes, RNA processing, regulation of nuclear envelope expansion and partitioning of importin α in nuclear size control, moving us closer to a mechanistic understanding of this phenomenon.


Subject(s)
Cell Nucleus Size/genetics , Cell Nucleus/genetics , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Cell Nucleus/physiology , Cell Nucleus Size/physiology , Cytoplasm/genetics , Cytoplasm/metabolism , Humans , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Matrix/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizosaccharomyces/genetics
15.
J Cell Biol ; 218(5): 1478-1490, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30862629

ABSTRACT

Bipolar spindle formation in yeast requires insertion of centrosomes (known as spindle pole bodies [SPBs]) into fenestrated regions of the nuclear envelope (NE). Using structured illumination microscopy and bimolecular fluorescence complementation, we map protein distribution at SPB fenestrae and interrogate protein-protein interactions with high spatial resolution. We find that the Sad1-UNC-84 (SUN) protein Mps3 forms a ring-like structure around the SPB, similar to toroids seen for components of the SPB insertion network (SPIN). Mps3 and the SPIN component Mps2 (a Klarsicht-ANC-1-Syne-1 domain [KASH]-like protein) form a novel noncanonical linker of nucleoskeleton and cytoskeleton (LINC) complex that is connected in both luminal and extraluminal domains at the site of SPB insertion. The LINC complex also controls the distribution of a soluble SPIN component Bbp1. Taken together, our work shows that Mps3 is a fifth SPIN component and suggests both direct and indirect roles for the LINC complex in NE remodeling.


Subject(s)
Centrosome/metabolism , Cytoskeleton/metabolism , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Spindle Pole Bodies/metabolism , Cell Cycle , Nuclear Matrix/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
16.
J Biosci ; 43(4): 621-633, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30207309

ABSTRACT

Nuclear Matrix (NuMat) is the structural and functional framework of the nucleus. It has been shown that attachment of chromatin to NuMat brings significant regulation of the transcriptional activity of particular genes; however, key components of NuMat involved in this process remain elusive. We have identified Lid (Little imaginal discs) as one of the components of NuMat. It belongs to the TrxG group of proteins involved in activation of important developmental genes. However, unlike other activator proteins of TrxG, Lid is a Jumonji protein involved in H3K4me3 demethylation. Here, we report the association of Lid and its various domains with NuMat which implicates its structural role in chromatin organization and epigenetic basis of cellular memory. We have found that both N and C terminal regions of this protein are capable of associating with NuMat. We have further mapped the association of individual domains and found that, PHD, ARID and JmjC domains can associate with NuMat individually. Moreover, deletion of N-terminal PHD finger does not alter Lid's NuMat association implying that although it is sufficient, yet, it is not necessary for Lid's structural role in NuMat. Based on our findings, we hypothesize that C terminal region of Lid which contains PHD fingers might be responsible for its NuMat association via protein-DNA interactions. However, for the N terminal region harboring both a PHD and an ARID finger, Lid anchors to the NuMat via both protein-protein and protein-DNA interactions. The association of JmjC domain with NuMat is the first report of the association of a demethylase domain with NuMat suggesting that Lid, a demethylase, being part of NuMat might be involved in regulating the chromatin dynamics via its NuMat association.


Subject(s)
Drosophila Proteins/genetics , Embryonic Development/genetics , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Nuclear Matrix/genetics , Animals , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Histones/genetics , PHD Zinc Fingers/genetics , Protein Domains/genetics
17.
Hum Mol Genet ; 27(12): 2154-2170, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29868845

ABSTRACT

Heterozygosity for the TOR1A-Δgag mutation causes semi-penetrant childhood-onset dystonia (OMIM #128100). More recently, homozygous TOR1A mutations were shown to cause severe neurological dysfunction in infants. However, there is little known about the recessive cases, including whether existing reports define the full spectrum of recessive TOR1A disease. Here we describe abnormal brain morphogenesis in ∼30% of Tor1a-/- mouse embryos while, in contrast, this is not found in Tor1aΔgag/Δgag mice. The abnormal Tor1a-/- brains contain excess neural tissue, as well as proliferative zone cytoarchitectural defects related to radial glial cell polarity and cytoskeletal organization. In cultured cells torsinA effects the linker of nucleoskeleton and cytoskeleton (LINC) complex that couples the nucleus and cytoskeleton. Here we identify that torsinA loss elevates LINC complex levels in the proliferative zone, and that genetic reduction of LINC complexes prevents abnormal brain morphogenesis in Tor1a-/- embryos. These data show that Tor1a affects radial glial cells via a LINC complex mediated mechanism. They also predict human TOR1A disease will include incompletely penetrant defects in embryonic brain morphogenesis in cases where mutations ablate TOR1A function.


Subject(s)
Dystonia/genetics , Molecular Chaperones/genetics , Morphogenesis/genetics , Neurogenesis/genetics , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Dystonia/physiopathology , Heterozygote , Homozygote , Humans , Mice , Mice, Knockout , Neurons , Nuclear Matrix/genetics
18.
Genome Res ; 28(2): 192-202, 2018 02.
Article in English | MEDLINE | ID: mdl-29273625

ABSTRACT

Eukaryotic chromosomes are folded into higher-order conformations to coordinate genome functions. In addition to long-range chromatin loops, recent chromosome conformation capture (3C)-based studies have indicated higher levels of chromatin structures including compartments and topologically associating domains (TADs), which may serve as units of genome organization and functions. However, the molecular machinery underlying these hierarchically three-dimensional (3D) chromatin architectures remains poorly understood. Via high-throughput assays, including in situ Hi-C, DamID, ChIP-seq, and RNA-seq, we investigated roles of the Heterogeneous Nuclear Ribonucleoprotein U (HNRNPU), a nuclear matrix (NM)-associated protein, in 3D genome organization. Upon the depletion of HNRNPU in mouse hepatocytes, the coverage of lamina-associated domains (LADs) in the genome increases from 53.1% to 68.6%, and a global condensation of chromatin was observed. Furthermore, disruption of HNRNPU leads to compartment switching on 7.5% of the genome, decreases TAD boundary strengths at borders between A (active) and B (inactive) compartments, and reduces chromatin loop intensities. Long-range chromatin interactions between and within compartments or TADs are also significantly remodeled upon HNRNPU depletion. Intriguingly, HNRNPU mainly associates with active chromatin, and 80% of HNRNPU peaks coincide with the binding of CTCF or RAD21. Collectively, we demonstrated that HNRNPU functions as a major factor maintaining 3D chromatin architecture, suggesting important roles of NM-associated proteins in genome organization.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromosomes/genetics , Genome/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Animals , Chromatin/genetics , Hepatocytes/metabolism , Mice , Nuclear Matrix/genetics
19.
Biochem Biophys Res Commun ; 506(2): 378-386, 2018 11 25.
Article in English | MEDLINE | ID: mdl-29203242

ABSTRACT

In eukaryotic cells gene regulation is dependent on global genome organization. This is achieved, in response to favorable environmental conditions, through spatial redistribution of chromatin and changes in global epigenetic levels. This eventually drives movement of gene-rich chromatin loops and formation of DNA loops, consolidating neighborhoods of gene expression and silencing. One of the challenges for future work is to examine how these neighborhoods are formed and whether they host genes involved in the same cellular functions for sustained expression or silencing over time. In the present review, we summarize evidence that actin and actin-associated proteins regulate gene activity. Furthermore we discuss how these specific nuclear tasks in which actin is engaged are important to organize and consolidate the mammalian genome, ensuring gene activation and repression of gene programs important to establish cellular identity. We propose that these mechanisms are essential to control cellular development and differentiation.


Subject(s)
Actins/metabolism , Chromatin/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genome , Actins/genetics , Alveolata , Animals , Chromatin/chemistry , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Histones/genetics , Histones/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , Nuclear Matrix/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plants , Transcription, Genetic
20.
Sci Rep ; 7(1): 6475, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743923

ABSTRACT

In higher eukaryotic nuclei, DNA is periodically anchored to an extraction-resistant protein structure, via matrix attachment regions. We describe a refined and accessible method to non-subjectively, rapidly and reproducibly measure both size and stability of the intervening chromatin loops, and use it to demonstrate that malignant transformation compromises the DNA-nuclear matrix interface.


Subject(s)
DNA/chemistry , High-Throughput Screening Assays/methods , Nuclear Matrix/chemistry , Antigens, Polyomavirus Transforming/chemistry , Cell Line , Cell Line, Tumor , Chromatin/chemistry , DNA/metabolism , DNA, Superhelical/chemistry , DNA, Superhelical/metabolism , Humans , Image Processing, Computer-Assisted , Nuclear Matrix/genetics , Nuclear Matrix/metabolism , Oncogenes
SELECTION OF CITATIONS
SEARCH DETAIL