Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
Nucleic Acids Res ; 45(17): 9917-9930, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973435

ABSTRACT

The structure of the nucleosome, the basic building block of the chromatin fiber, plays a key role in epigenetic regulatory processes that affect DNA-dependent processes in the context of chromatin. Members of the HMGN family of proteins bind specifically to nucleosomes and affect chromatin structure and function, including transcription and DNA repair. To better understand the mechanisms by which HMGN 1 and 2 alter chromatin, we analyzed their effect on the organization of histone tails and linker histone H1 in nucleosomes. We find that HMGNs counteract linker histone (H1)-dependent stabilization of higher order 'tertiary' chromatin structures but do not alter the intrinsic ability of nucleosome arrays to undergo salt-induced compaction and self-association. Surprisingly, HMGNs do not displace H1s from nucleosomes; rather these proteins bind nucleosomes simultaneously with H1s without disturbing specific contacts between the H1 globular domain and nucleosomal DNA. However, HMGNs do alter the nucleosome-dependent condensation of the linker histone C-terminal domain, which is critical for stabilizing higher-order chromatin structures. Moreover, HMGNs affect the interactions of the core histone tail domains with nucleosomal DNA, redirecting the tails to more interior positions within the nucleosome. Our studies provide new insights into the molecular mechanisms whereby HMGNs affect chromatin structure.


Subject(s)
DNA/chemistry , HMGN1 Protein/chemistry , HMGN2 Protein/chemistry , Histones/chemistry , Nucleosomes/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chickens , DNA/genetics , DNA/metabolism , Gene Expression , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Histones/genetics , Histones/metabolism , Humans , Nucleic Acid Conformation , Nucleosomes/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Domains , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xenopus laevis
2.
Annu Rev Biochem ; 83: 291-315, 2014.
Article in English | MEDLINE | ID: mdl-24905784

ABSTRACT

Large macromolecular assemblies, so-called molecular machines, are critical to ensuring proper cellular function. Understanding how proper function is achieved at the atomic level is crucial to advancing multiple avenues of biomedical research. Biophysical studies often include X-ray diffraction and cryo-electron microscopy, providing detailed structural descriptions of these machines. However, their inherent flexibility has complicated an understanding of the relation between structure and function. Solution NMR spectroscopy is well suited to the study of such dynamic complexes, and continued developments have increased size boundaries; insights into function have been obtained for complexes with masses as large as 1 MDa. We highlight methyl-TROSY (transverse relaxation optimized spectroscopy) NMR, which enables the study of such large systems, and include examples of applications to several cellular machines. We show how this emerging technique contributes to an understanding of cellular function and the role of molecular plasticity in regulating an array of biochemical activities.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Allosteric Site , Animals , Bacterial Proteins/chemistry , Catalytic Domain , Exosomes , HMGN2 Protein/chemistry , Heat-Shock Proteins/chemistry , Humans , Hydrogen-Ion Concentration , Macromolecular Substances/chemistry , Nucleosomes/chemistry , Potassium Channels/chemistry , Proteasome Endopeptidase Complex/chemistry , Protein Conformation , Proteins/chemistry
3.
J Biol Chem ; 289(29): 20000-11, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24872413

ABSTRACT

High mobility group nucleosomal binding domain 2 (HMGN2) is a small and unique non-histone protein that has many functions in a variety of cellular processes, including regulation of chromatin structure, transcription, and DNA repair. In addition, it may have other roles in antimicrobial activity, cell homing, and regulating cytokine release. Although the biochemical properties of HMGN2 protein are regulated by acetylation and phosphorylation, it is not yet known whether HMGN2 activity can also be regulated by SUMOylation. In this study, we demonstrated for the first time that HMGN2 is modified by covalent attachment of small ubiquitin-related modifier 1 (SUMO1) by pro-inflammatory signal and identified the major SUMOylated lysine residues that localize to the HMGN2 nucleosome-binding domain at Lys-17 and Lys-35. SENP1 can deSUMOylate SUMOylated HMGN2, and PIAS1 is the E3 ligase responsible for SUMOylation of HMGN2. Finally, using SUMO1-conjugated HMGN2 purified from a basal SUMOylation system in Escherichia coli, we demonstrated that SUMOylated HMGN2 has decreased the binding affinity to nucleosome core particles in comparison to unSUMOylated HMGN2. These observations potentially provide new perspectives for understanding the functions of HMGN2 in inflammatory reaction.


Subject(s)
HMGN2 Protein/metabolism , Nucleosomes/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , Cell Line , Cysteine Endopeptidases , Endopeptidases/genetics , Endopeptidases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , HEK293 Cells , HMGN2 Protein/chemistry , HMGN2 Protein/genetics , HeLa Cells , Humans , Lysine/chemistry , Molecular Sequence Data , Protein Binding , Protein Inhibitors of Activated STAT/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sequence Homology, Amino Acid , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Ubiquitin-Protein Ligases/genetics
4.
Eur J Nucl Med Mol Imaging ; 39(12): 1886-97, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872310

ABSTRACT

PURPOSE: Targeted therapy with α-particle emitting radionuclides is a promising new option in cancer therapy. Stable conjugates of the vascular tumour-homing peptide F3 with the α-emitter (213)Bi specifically target tumour cells. The aim of our study was to determine efficacy of combined (213)Bi-diethylenetriaminepentaacetic acid (DTPA)-F3 and paclitaxel treatment compared to treatment with either (213)Bi-DTPA-F3 or paclitaxel both in vitro and in vivo. METHODS: Cytotoxicity of treatment with (213)Bi-DTPA-F3 and paclitaxel, alone or in combination, was assayed towards OVCAR-3 cells using the alamarBlue assay, the clonogenic assay and flow cytometric analyses of the mode of cell death and cell cycle arrest. Therapeutic efficacy of the different treatment options was assayed after repeated treatment of mice bearing intraperitoneal OVCAR-3 xenograft tumours. Therapy monitoring was performed by bioluminescence imaging and histopathologic analysis. RESULTS: Treatment of OVCAR-3 cells in vitro with combined (213)Bi-DTPA-F3 and paclitaxel resulted in enhanced cytotoxicity, induction of apoptosis and G2/M phase arrest compared to treatment with either (213)Bi-DTPA-F3 or paclitaxel. Accordingly, i.p. xenograft OVCAR-3 tumours showed the best response following repeated (six times) combined therapy with (213)Bi-DTPA-F3 (1.85 MBq) and paclitaxel (120 µg) as demonstrated by bioluminescence imaging and histopathologic investigation of tumour spread on the mesentery of the small and large intestine. Moreover, mean survival of xenograft mice that received combined therapy with (213)Bi-DTPA-F3 and paclitaxel was significantly superior to mice treated with either (213)Bi-DTPA-F3 or paclitaxel alone. CONCLUSION: Combined treatment with (213)Bi-DTPA-F3 and paclitaxel significantly increased mean survival of mice with peritoneal carcinomatosis of ovarian origin, thus favouring future therapeutic application.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma/therapy , Chemoradiotherapy , Organometallic Compounds/therapeutic use , Paclitaxel/therapeutic use , Peritoneal Neoplasms/therapy , Radiopharmaceuticals/therapeutic use , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints/drug effects , HEK293 Cells , HMGN2 Protein/chemistry , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Organometallic Compounds/pharmacology , Paclitaxel/pharmacology , Protein Structure, Tertiary , Radiopharmaceuticals/pharmacology , Treatment Outcome
5.
Proc Natl Acad Sci U S A ; 108(30): 12283-8, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21730181

ABSTRACT

Chromatin structure and function are regulated by numerous proteins through specific binding to nucleosomes. The structural basis of many of these interactions is unknown, as in the case of the high mobility group nucleosomal (HMGN) protein family that regulates various chromatin functions, including transcription. Here, we report the architecture of the HMGN2-nucleosome complex determined by a combination of methyl-transverse relaxation optimized nuclear magnetic resonance spectroscopy (methyl-TROSY) and mutational analysis. We found that HMGN2 binds to both the acidic patch in the H2A-H2B dimer and to nucleosomal DNA near the entry/exit point, "stapling" the histone core and the DNA. These results provide insight into how HMGNs regulate chromatin structure through interfering with the binding of linker histone H1 to the nucleosome as well as a structural basis of how phosphorylation induces dissociation of HMGNs from chromatin during mitosis. Importantly, our approach is generally applicable to the study of nucleosome-binding interactions in chromatin.


Subject(s)
HMGN2 Protein/chemistry , Nucleosomes/chemistry , Amino Acid Sequence , Binding Sites , DNA/chemistry , DNA/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , In Vitro Techniques , Kinetics , Methylation , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Nucleosomes/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
7.
Mol Biol Cell ; 19(5): 1816-24, 2008 May.
Article in English | MEDLINE | ID: mdl-18287527

ABSTRACT

Throughout the cell cycle, the histones remain associated with DNA, but the repertoire of proteins associated with the chromatin fiber continuously changes. The chromatin interaction of HMGNs, a family of nucleosome binding proteins that modulates the structure and activity of chromatin, during the cell cycle is controversial. Immunofluorescence studies demonstrated that HMGNs are not associated with chromatin, whereas live cell imaging indicated that they are present in mitotic chromosomes. To resolve this controversy, we examined the organization of wild-type and mutated HMGN1 and HMGN2 proteins in the cell nucleus by using immunofluorescence studies, live cell imaging, gel mobility shift assays, and bimolecular fluorescence complementation (BiFC). We find that during interphase, HMGNs bind specifically to nucleosomes and form homodimeric complexes that yield distinct BiFC signals. In metaphase, the nucleosomal binding domain of the protein is inactivated, and the proteins associate with chromatin with low affinity as monomers, and they do not form specific complexes. Our studies demonstrate that the mode of binding of HMGNs to chromatin is cell cycle dependent.


Subject(s)
Cell Cycle , Chromatin/metabolism , HMGN1 Protein/metabolism , HMGN2 Protein/metabolism , Animals , Cell Survival , Chromosomes, Human/metabolism , Fluorescence , Genetic Complementation Test , HMGN1 Protein/chemistry , HMGN2 Protein/chemistry , HeLa Cells , Humans , Interphase , Mice , Mitosis , Models, Biological , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Rats
9.
J Biol Chem ; 281(15): 10182-7, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16484217

ABSTRACT

We have demonstrated that levels of specific modification in histone H3 are modulated by members of the nucleosome-binding high mobility group N (HMGN) protein family in a variant-specific manner. HMGN1 (but not HMGN2) inhibits the phosphorylation of both H3S10 and H3S28, whereas HMGN2 enhances H3K14 acetylation more robustly than HMGN1. Two HMGN domains are necessary for modulating chromatin modifications, a non-modification-specific domain necessary for chromatin binding and a modification-specific domain localized in the C terminus of the HMGNs. Thus, chromatin-binding structural proteins such as HMGNs affect the levels of specific chromatin modifications and therefore may play a role in epigenetic regulation.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation , Acetylation , Animals , Cell Nucleus/metabolism , Chickens , Chromatin/metabolism , Chromosomes/metabolism , Epigenesis, Genetic , Erythrocytes/metabolism , Escherichia coli/metabolism , HMGN1 Protein/chemistry , HMGN2 Protein/chemistry , High Mobility Group Proteins/chemistry , Histone Acetyltransferases/chemistry , Histones/chemistry , Mutation , Nucleosomes/chemistry , Nucleosomes/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Time Factors
10.
J Leukoc Biol ; 78(5): 1136-41, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16204630

ABSTRACT

Leukocytes are a central cellular element of innate-immune defense in mammals. In addition to the generation of toxic oxygen radicals and nitric oxide, leukocytes express and secrete a broad array of antimicrobial proteins and peptides. In the study, an antimicrobial polypeptide was isolated and purified from human peripheral blood mononuclear leukocytes in the presence of interleukin (IL)-2. Microsequencing provided that its N-terminal amino sequence was PKRKAEGDAK, which was identical to high mobility group nucleosomal-binding domain 2 (HMGN2). Mass spectrometric value and Western blot also indicated its individual character of HMGN2. The antimicrobial assays showed that the Escherichia coli-based production of HMGN2 had a potent antimicrobial activity against E. coli ML-35p, Pseudomonas aeruginosa ATCC 27853, and to some extent, against Candida albicans ATCC 10231. The HMGN2 alpha-helical domain had the same antimicrobial activity as HMGN2. The immunocytochemistry staining, enzyme-linked immunosorbent assay, and Western blot revealed that HMGN2 was present in the cytoplasm of mononuclear leukocytes and released to the extracellular environment when stimulated with IL-2. These results suggest that HMGN2 would be a novel antimicrobial effector molecule of human mononuclear leukocyte.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , HMGN2 Protein/chemistry , HMGN2 Protein/pharmacology , Leukocytes, Mononuclear/chemistry , Anti-Infective Agents/isolation & purification , Candida albicans/drug effects , Escherichia coli/drug effects , HMGN2 Protein/isolation & purification , Humans , Interleukin-2/pharmacology , Leukocytes, Mononuclear/drug effects , Microbial Sensitivity Tests , Peptides/chemistry , Peptides/isolation & purification , Peptides/pharmacology , Protein Conformation , Protein Structure, Secondary , Pseudomonas aeruginosa/drug effects , Reference Values
11.
Proc Natl Acad Sci U S A ; 99(11): 7444-9, 2002 May 28.
Article in English | MEDLINE | ID: mdl-12032302

ABSTRACT

We used a screening procedure to identify protein domains from phage-displayed cDNA libraries that bind both to bone marrow endothelial progenitor cells and tumor vasculature. Screening phage for binding of progenitor cell-enriched bone marrow cells in vitro, and for homing to HL-60 human leukemia cell xenograft tumors in vivo, yielded a cDNA fragment that encodes an N-terminal fragment of human high mobility group protein 2 (HMGN2, formerly HMG-17). Upon i.v. injection, phage displaying this HMGN2 fragment homed to HL-60 and MDA-MB-435 tumors. Testing of subfragments localized the full binding activity to a 31-aa peptide (F3) in the HMGN2 sequence. Fluorescein-labeled F3 peptide bound to and was internalized by HL-60 cells and human MDA-MB-435 breast cancer cells, appearing initially in the cytoplasm and then in the nuclei of these cells. Fluorescent F3 accumulated in HL-60 and MDA-MB-435 tumors after an i.v. injection, appearing in the nuclei of tumor endothelial cells and tumor cells. Thus, F3 can carry a payload (phage, fluorescein) to a tumor and into the cell nuclei in the tumor. This peptide may be suitable for targeting cytotoxic drugs and gene therapy vectors into tumors.


Subject(s)
Cell Nucleus/metabolism , Endothelium, Vascular/metabolism , HMGN2 Protein/metabolism , Peptide Fragments/metabolism , Animals , Bone Marrow Cells/physiology , Breast Neoplasms , Female , Gene Library , HL-60 Cells , HMGN2 Protein/chemistry , Hematopoietic Stem Cells/physiology , Humans , Mice , Protein Isoforms/metabolism , RNA, Messenger/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...