Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Biol Chem ; 298(9): 102295, 2022 09.
Article in English | MEDLINE | ID: mdl-35872015

ABSTRACT

The chromatin-associated high mobility group protein N2 (HMGN2) cofactor regulates transcription factor activity through both chromatin and protein interactions. Hmgn2 expression is known to be developmentally regulated, but the post-transcriptional mechanisms that regulate Hmgn2 expression and its precise roles in tooth development remain unclear. Here, we demonstrate that HMGN2 inhibits the activity of multiple transcription factors as a general mechanism to regulate early development. Bimolecular fluorescence complementation, pull-down, and coimmunoprecipitation assays show that HMGN2 interacts with the transcription factor Lef-1 through its HMG-box domain as well as with other early development transcription factors, Dlx2, FoxJ1, and Pitx2. Furthermore, EMSAs demonstrate that HMGN2 binding to Lef-1 inhibits its DNA-binding activity. We found that Pitx2 and Hmgn2 associate with H4K5ac and H3K4me2 chromatin marks in the proximal Dlx2 promoter, demonstrating Hmgn2 association with open chromatin. In addition, we demonstrate that microRNAs (miRs) mir-23a and miR-23b directly target Hmgn2, promoting transcriptional activation at several gene promoters, including the amelogenin promoter. In vivo, we found that decreased Hmgn2 expression correlates with increased miR-23 expression in craniofacial tissues as the murine embryo develops. Finally, we show that ablation of Hmgn2 in mice results in increased amelogenin expression because of increased Pitx2, Dlx2, Lef-1, and FoxJ1 transcriptional activity. Taken together, our results demonstrate both post-transcriptional regulation of Hmgn2 by miR-23a/b and post-translational regulation of gene expression by Hmgn2-transcription factor interactions. We conclude that HMGN2 regulates tooth development through its interaction with multiple transcription factors.


Subject(s)
Amelogenesis , Gene Expression Regulation , HMGN2 Protein , Homeodomain Proteins , Lymphoid Enhancer-Binding Factor 1 , Transcription Factors , Transcription, Genetic , Amelogenesis/genetics , Amelogenin/genetics , Animals , Chromatin/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Homeodomain Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors/metabolism , Homeobox Protein PITX2
2.
J Biol Chem ; 295(2): 468-480, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31699896

ABSTRACT

The surface area of the human cerebral cortex undergoes dramatic expansion during late fetal development, leading to cortical folding, an evolutionary feature not present in rodents. Microcephaly is a neurodevelopmental disorder defined by an abnormally small brain, and many gene mutations have been found to be associated with primary microcephaly. However, mouse models generated by ablating primary microcephaly-associated genes often fail to recapitulate the severe loss of cortical surface area observed in individuals with this pathology. Here, we show that a mouse model with deficient expression of high-mobility group nucleosomal binding domain 2 (HMGN2) manifests microcephaly with reduced cortical surface area and almost normal radial corticogenesis, with a pattern of incomplete penetrance. We revealed that altered cleavage plane and mitotic delay of ventricular radial glia may explain the rising ratio of intermediate progenitor cells to radial glia and the displacement of neural progenitor cells in microcephalic mutant mice. These led to decreased self-renewal of the radial glia and reduction in lateral expansion. Furthermore, we found that HMGN2 protected corticogenesis by maintaining global chromatin accessibility mainly at promoter regions, thereby ensuring the correct regulation of the transcriptome. Our findings underscore the importance of the regulation of chromatin structure in cortical development and highlight a mouse model with critical insights into the etiology of microcephaly.


Subject(s)
Cerebral Cortex/embryology , Chromatin Assembly and Disassembly , HMGN2 Protein/metabolism , Microcephaly/metabolism , Animals , Cerebral Cortex/metabolism , Female , Gene Deletion , Gene Expression Regulation, Developmental , HMGN2 Protein/analysis , HMGN2 Protein/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcephaly/genetics
3.
Future Oncol ; 17(5): 541-548, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33467898

ABSTRACT

Aim: The objective of this work was to investigate the prognostic role of the HMGN family in acute myeloid leukemia (AML). Methods: A total of 155 AML patients with HMGN1-5 expression data from the Cancer Genome Atlas database were enrolled in this study. Results: In the chemotherapy-only group, patients with high HMGN2 expression had significantly longer event-free survival (EFS) and overall survival (OS) than those with low expression (all p < 0.05), whereas high HMGN5 expressers had shorter EFS and OS than the low expressers (all p < 0.05). Multivariate analysis identified that high HMGN2 expression was an independent favorable prognostic factor for patients who only received chemotherapy (all p < 0.05). HMGN family expression had no impact on EFS and OS in AML patients receiving allogeneic hematopoietic stem cell transplantation. Conclusion: High HMGN2/5 expression is a potential prognostic indicator for AML.


Subject(s)
Biomarkers, Tumor/genetics , HMGN Proteins/genetics , HMGN2 Protein/genetics , Leukemia, Myeloid, Acute/mortality , Trans-Activators/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cell Transplantation/statistics & numerical data , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Prognosis , Progression-Free Survival , Young Adult
4.
Microbiol Immunol ; 64(12): 825-834, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33034909

ABSTRACT

It has been reported that high mobility group nucleosomal binding domain 2 (HMGN2) is a nucleus-related protein that regulates gene transcription and plays a critical role in bacterial clearance. An elevated level of HMGN2 reduced integrin α5/ß1 expression of human pulmonary epithelial A549 cells was demonstrated during Klebsiella pneumoniae infection, thus weakening bacterial adhesion and invasion. However, the mechanism by which HMGN2 regulates integrin expression remains unclear. This study found that a transcription factor-nuclear factor I (NFI), which serves as the potential target of HMGN2 regulated integrin expression. The results showed that HMGN2 was able to promote NFIA and NFIB expression by increasing H3K27 acetylation of NFIA/B promoter regions. The integrin α5/ß1 expression was significantly enhanced by knockdown of NFIA/B via a siRNA approach. Meanwhile, NFIA/B silence could also compromise the inhibition effect of HMGN2 on the integrin α5/ß1 expression. Mechanistically, it was demonstrated that HMGN2 facilitated the recruitment of NFI on the promoter regions of integrin α5/ß1 according to the chromatin immunoprecipitation assay. In addition, it was further demonstrated that the knockdown of NFIA/B induced more adhesion of Klebsiella pneumoniae on pulmonary epithelial A549 cells, which could be reversed by the application of an integrin inhibitor RGD. The results revealed a regulatory role of HMGN2 on the transcription level of integrin α5/ß1, indicating a potential treatment strategy against Klebsiella pneumoniae-induced infectious lung diseases.


Subject(s)
Bacterial Adhesion/physiology , Epithelial Cells/microbiology , HMGN2 Protein/metabolism , Integrin alpha5beta1/metabolism , Klebsiella pneumoniae/metabolism , NFI Transcription Factors/metabolism , A549 Cells , Gene Expression Regulation , HMGN2 Protein/genetics , Humans , Integrin alpha5/genetics , Integrin alpha5/metabolism , Integrin alpha5beta1/genetics , Integrin beta1/genetics , Integrin beta1/metabolism , Klebsiella Infections/metabolism , Klebsiella pneumoniae/genetics , Lung , RNA, Small Interfering/metabolism , Transcriptome
5.
J Cell Mol Med ; 23(12): 7985-7998, 2019 12.
Article in English | MEDLINE | ID: mdl-31596045

ABSTRACT

Non-tuberculous mycobacteria (NTM), also known as an environmental and atypical mycobacteria, can cause the chronic pulmonary infectious diseases. Macrophages have been suggested as the main host cell to initiate the innate immune responses to NTM infection. However, the molecular mechanism to regulate the antimicrobial immune responses to NTM is still largely unknown. Current study showed that the NTM clinical groups, Mycobacterium abscessus and Mycobacterium smegmatis, significantly induced the M1 macrophage polarization with the characteristic production of nitric oxide (NO) and marker gene expression of iNOS, IFNγ, TNF-α, IL1-ß and IL-6. Interestingly, a non-histone nuclear protein, HMGN2 (high-mobility group N2), was found to be spontaneously induced during NTM-activated M1 macrophage polarization. Functional studies revealed that HMGN2 deficiency in NTM-infected macrophage promotes the expression of M1 markers and the production of NO via the enhanced activation of NF-κB and MAPK signalling. Further studies exhibited that HMGN2 knock-down also enhanced IFNγ-induced M1 macrophage polarization. Finally, we observed that silencing HMGN2 affected the survival of NTM in macrophage, which might largely relevant to enhanced macrophage polarization into M1 phenotype under the NTM infection. Collectively, current studies thus suggested a novel function of HMGN2 in regulating the anti-non-tuberculous mycobacteria innate immunity of macrophage.


Subject(s)
HMGN2 Protein/metabolism , Macrophage Activation/genetics , Macrophages/metabolism , Mycobacterium Infections/immunology , Nontuberculous Mycobacteria/growth & development , Animals , Cell Survival/genetics , Gene Knockdown Techniques , Gene Silencing , HMGN2 Protein/genetics , Humans , Immunity, Innate , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System/genetics , Mice , Mycobacterium abscessus/immunology , Mycobacterium abscessus/isolation & purification , Mycobacterium smegmatis/immunology , Mycobacterium smegmatis/isolation & purification , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , RNA Interference , Tumor Necrosis Factor-alpha/metabolism
6.
Nucleic Acids Res ; 45(6): 3031-3045, 2017 04 07.
Article in English | MEDLINE | ID: mdl-27923998

ABSTRACT

An interplay between the nucleosome binding proteins H1 and HMGN is known to affect chromatin dynamics, but the biological significance of this interplay is still not clear. We find that during embryonic stem cell differentiation loss of HMGNs leads to down regulation of genes involved in neural differentiation, and that the transcription factor OLIG2 is a central node in the affected pathway. Loss of HMGNs affects the expression of OLIG2 as well as that of OLIG1, two transcription factors that are crucial for oligodendrocyte lineage specification and nerve myelination. Loss of HMGNs increases the chromatin binding of histone H1, thereby recruiting the histone methyltransferase EZH2 and elevating H3K27me3 levels, thus conferring a repressive epigenetic signature at Olig1&2 sites. Embryonic stem cells lacking HMGNs show reduced ability to differentiate towards the oligodendrocyte lineage, and mice lacking HMGNs show reduced oligodendrocyte count and decreased spinal cord myelination, and display related neurological phenotypes. Thus, the presence of HMGN proteins is required for proper expression of neural differentiation genes during embryonic stem cell differentiation. Specifically, we demonstrate that the dynamic interplay between HMGNs and H1 in chromatin epigenetically regulates the expression of OLIG1&2, thereby affecting oligodendrocyte development and myelination, and mouse behavior.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Epigenesis, Genetic , HMGN Proteins/physiology , Histones/metabolism , Nerve Tissue Proteins/genetics , Oligodendroglia/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , HMGN1 Protein/genetics , HMGN1 Protein/physiology , HMGN2 Protein/genetics , HMGN2 Protein/physiology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2
7.
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
8.
Genome Res ; 25(9): 1295-308, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26156321

ABSTRACT

DNase I hypersensitive sites (DHSs) are a hallmark of chromatin regions containing regulatory DNA such as enhancers and promoters; however, the factors affecting the establishment and maintenance of these sites are not fully understood. We now show that HMGN1 and HMGN2, nucleosome-binding proteins that are ubiquitously expressed in vertebrate cells, maintain the DHS landscape of mouse embryonic fibroblasts (MEFs) synergistically. Loss of one of these HMGN variants led to a compensatory increase of binding of the remaining variant. Genome-wide mapping of the DHSs in Hmgn1(-/-), Hmgn2(-/-), and Hmgn1(-/-)n2(-/-) MEFs reveals that loss of both, but not a single HMGN variant, leads to significant remodeling of the DHS landscape, especially at enhancer regions marked by H3K4me1 and H3K27ac. Loss of HMGN variants affects the induced expression of stress-responsive genes in MEFs, the transcription profiles of several mouse tissues, and leads to altered phenotypes that are not seen in mice lacking only one variant. We conclude that the compensatory binding of HMGN variants to chromatin maintains the DHS landscape, and the transcription fidelity and is necessary to retain wild-type phenotypes. Our study provides insight into mechanisms that maintain regulatory sites in chromatin and into functional compensation among nucleosome binding architectural proteins.


Subject(s)
Binding Sites , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic , HMGN Proteins/metabolism , Animals , Cell Line , Chromatin/metabolism , Cluster Analysis , Gene Expression Profiling , Gene Knockout Techniques , HMGN Proteins/genetics , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Humans , Mice , Mice, Knockout , Nucleosomes/metabolism , Phenotype , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Stress, Physiological/genetics
9.
Cell Physiol Biochem ; 44(5): 1681-1695, 2017.
Article in English | MEDLINE | ID: mdl-29216626

ABSTRACT

BACKGROUND/AIMS: Hmgn2 is involved in regulating embryonic development, but its physiological function during embryo implantation and decidualization remains unknown. METHODS: In situ hybridization, real-time PCR, RNA interference, gene overexpression and MTS assay were used to examine the expression of Hmgn2 in mouse uterus during the pre-implantation period and explore its function and regulatory mechanisms in epithelial adhesion junction and stromal cell proliferation and differentiation. RESULTS: Hmgn2 was primarily accumulated in uterine luminal epithelia on day 4 of pregnancy and subluminal stromal cells around the implanting blastocyst at implantation sites on day 5. Similar results were observed during delayed implantation and activation. Meanwhile, Hmgn2 expression was visualized in the decidua. In uterine epithelial cells, silencing of Hmgn2 by specific siRNA reduced the expression of adhesion molecules Cdh1, Cdh2 and Ctnnb1 and enhanced the expression of Muc1, whereas constitutive activation of Hmgn2 exhibited the opposite effects, suggesting a role for Hmgn2 in attachment reaction during embryo implantation. Estrogen stimulated the expression of Hmgn2 in uterine epithelia, but the stimulation was abrogated by ER antagonist ICI 182,780. Further analysis evidenced that attenuation of Hmgn2 might eliminate the regulation of estrogen on the expression of Cdh1, Cdh2 and Ctnnb1. In uterine stromal cells, progesterone induced the accumulation of Hmgn2 which advanced the expression of Prl8a2 and Prl3c1, two well-known differentiation markers for decidualization, but did not affect the proliferation of stromal cells. Knockdown of Hmgn2 blocked the progesterone-induced differentiation of uterine stromal cells. Moreover, Hmgn2 might serve as an intermediate to mediate the regulation of progesterone on Hand2. CONCLUSION: Hmgn2 may play an important role during embryo implantation and decidualization.


Subject(s)
Decidua/metabolism , Embryo Implantation , HMGN2 Protein/metabolism , Animals , Cadherins/metabolism , Cdh1 Proteins/metabolism , Cell Differentiation/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/pharmacology , Female , Fulvestrant , Gene Expression Regulation, Developmental/drug effects , HMGN2 Protein/antagonists & inhibitors , HMGN2 Protein/genetics , Mice , Mucin-1/metabolism , Pregnancy , Progesterone/pharmacology , Prolactin/metabolism , RNA Interference , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Uterus/metabolism , beta Catenin/metabolism
10.
Mol Cell ; 34(5): 620-6, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524541

ABSTRACT

The high-mobility group N (HMGN) proteins are abundant nonhistone chromosomal proteins that bind specifically to nucleosomes at two high-affinity sites. Here we report that purified recombinant human HMGN1 (HMG14) and HMGN2 (HMG17) potently repress ATP-dependent chromatin remodeling by four different molecular motor proteins. In contrast, mutant HMGN proteins with double Ser-to-Glu mutations in their nucleosome-binding domains are unable to inhibit chromatin remodeling. The HMGN-mediated repression of chromatin remodeling is reversible and dynamic. With the ACF chromatin remodeling factor, HMGN2 does not directly inhibit the ATPase activity but rather appears to reduce the affinity of the factor to chromatin. These findings suggest that HMGN proteins serve as a counterbalance to the action of the many ATP-dependent chromatin remodeling activities in the nucleus.


Subject(s)
Chromatin Assembly and Disassembly/physiology , HMGN1 Protein/physiology , HMGN2 Protein/physiology , Nucleosomes/metabolism , Recombinant Proteins/metabolism , Adenosine Triphosphate/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Humans , Molecular Motor Proteins/physiology , Mutation , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
11.
Hum Mol Genet ; 23(1): 194-208, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23975681

ABSTRACT

Patients with Axenfeld-Rieger Syndrome (ARS) present various dental abnormalities, including hypodontia, and enamel hypoplasia. ARS is genetically associated with mutations in the PITX2 gene, which encodes one of the earliest transcription factors to initiate tooth development. Thus, Pitx2 has long been considered as an upstream regulator of the transcriptional hierarchy in early tooth development. However, because Pitx2 is also a major regulator of later stages of tooth development, especially during amelogenesis, it is unclear how mutant forms cause ARS dental anomalies. In this report, we outline the transcriptional mechanism that is defective in ARS. We demonstrate that during normal tooth development Pitx2 activates Amelogenin (Amel) expression, whose product is required for enamel formation, and that this regulation is perturbed by missense PITX2 mutations found in ARS patients. We further show that Pitx2-mediated Amel activation is controlled by chromatin-associated factor Hmgn2, and that Hmgn2 prevents Pitx2 from efficiently binding to and activating the Amel promoter. Consistent with a physiological significance to this interaction, we show that K14-Hmgn2 transgenic mice display a severe loss of Amel expression on the labial side of the lower incisors, as well as enamel hypoplasia-consistent with the human ARS phenotype. Collectively, these findings define transcriptional mechanisms involved in normal tooth development and shed light on the molecular underpinnings of the enamel defect observed in ARS patients who carry PITX2 mutations. Moreover, our findings validate the etiology of the enamel defect in a novel mouse model of ARS.


Subject(s)
Amelogenin/metabolism , Anterior Eye Segment/abnormalities , Eye Abnormalities/pathology , HMGN2 Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Incisor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amelogenin/genetics , Animals , Anterior Eye Segment/pathology , Cell Line , Dental Enamel/metabolism , Dental Enamel/pathology , Disease Models, Animal , Embryo, Mammalian , Eye Abnormalities/genetics , Eye Diseases, Hereditary , Gene Expression Regulation , HMGN2 Protein/genetics , Humans , Incisor/pathology , Mice , Mice, Knockout , Mutation, Missense , Promoter Regions, Genetic , Homeobox Protein PITX2
12.
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
13.
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
14.
J Dent Res ; 103(1): 51-61, 2024 01.
Article in English | MEDLINE | ID: mdl-37950483

ABSTRACT

Dental enamel formation is coordinated by ameloblast differentiation, production of enamel matrix proteins, and crystal growth. The factors regulating ameloblast differentiation are not fully understood. Here we show that the high mobility group N (HMGN) nucleosomal binding proteins modulate the rate of ameloblast differentiation and enamel formation. We found that HMGN1 and HMGN2 proteins are downregulated during mouse ameloblast differentiation. Genetically altered mice lacking HMGN1 and HMGN2 proteins show faster ameloblast differentiation and a higher rate of enamel deposition in mice molars and incisors. In vitro differentiation of induced pluripotent stem cells to dental epithelium cells showed that HMGN proteins modulate the expression and chromatin accessibility of ameloblast-specific genes and affect the binding of transcription factors epiprofin and PITX2 to ameloblast-specific genes. Our results suggest that HMGN proteins regulate ameloblast differentiation and enamel mineralization by modulating lineage-specific chromatin accessibility and transcription factor binding to ameloblast regulatory sites.


Subject(s)
Dental Enamel Proteins , HMGN1 Protein , HMGN2 Protein , Animals , Mice , Ameloblasts/metabolism , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , Epigenesis, Genetic , Cell Differentiation/genetics , HMGN Proteins/genetics , HMGN Proteins/metabolism , Transcription Factors/metabolism , Dental Enamel Proteins/genetics , Dental Enamel Proteins/metabolism , Chromatin/metabolism , Amelogenin/metabolism
15.
Cell Biol Int ; 36(2): 195-202, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21988615

ABSTRACT

Hmgn2 (high mobility group nucleosomal 2), a ubiquitous nucleosome-binding protein that unfolds chromatin fibres and enhances DNA replication, reportedly regulates differentiation of epithelial and mesenchymal cells. To investigate how Hmgn2 regulates HC (haemopoietic cell) differentiation, we quantified Hmgn2 expression in HCs of mouse FL (fetal liver) during erythroid differentiation. Hmgn2 expression levels were >10-fold higher in immature erythroid progenitors than in mature erythroid cells, suggesting that Hmgn2 antagonizes erythroid differentiation. To address this issue, Hmgn2 were transfected into both Friend erythroleukaemia cells and FL HCs. There was a 3.3-fold decrease in relatively mature c-Kit(+)/CD71(+) erythroid cells, a 2.9-fold increase in immature c-Kit(+)/CD71(-) erythroid cells in transfected Friend cells, a 1.1-fold decrease in relatively mature CD71(+)/Ter119(+) erythroid cells, and a 1.7-fold increase in relatively immature c-Kit(+)/CD71(+) erythroid cells in FL HCs accompanied by down-regulation of genes encoding the erythroid transcription factors, Gata1 and Klf1. Two days after Hmgn2 transfection of Friend erythroleukaemia cells, the number of S-phase cells increased, whereas the number of cells in G(1) decreased, while that of mitotic cells remained unchanged. We conclude that ectopic expression of Hmgn2 antagonizes mouse erythroid differentiation in vitro, which may be due to enhancement of DNA replication and/or blocking entry of mitosis at S-phase.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells/cytology , HMGN2 Protein/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Blood Group Antigens/genetics , Blood Group Antigens/metabolism , Cell Line, Tumor , Cells, Cultured , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , HMGN2 Protein/genetics , Kruppel-Like Transcription Factors/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mitosis , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , S Phase
16.
J Mol Histol ; 53(4): 741-751, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35861945

ABSTRACT

High Mobility Group Chromosomal Protein N2 (HMGN2) can recognize tumor cells and enhance the anti-tumor effect of immune cells. This study aimed to establish a lentiviral vector of recombinant HMGN2 gene, establish recombinant T cells (HMGN2-T cells), and observe their anti-tumor effects. Total RNA was isolated from peripheral blood mononuclear cells. HMGN2, cluster of differentiation (CD) 8 A, CD28, CD137, and CD3ζ genes were amplified and connected. Jurkat cells were transfected with the recombinant lentivirus vector. The viability, apoptosis, and cell cycle of HMGN2-T cells were detected using Cell Counting Kit-8 assay and flow cytometry. The co-culture was performed by adding HMGN2-T cells to tumor cells with different effect-to-target (E:T) ratios. The cytotoxic activity was measured by lactate dehydrogenase (LDH) releasing assay. The sequences of HMGN2, CD8A, CD28, CD137, and CD3ζ gene plasmids were confirmed using gene sequencing. After the lentiviral transfection for 72 h, green fluorescence cells (HMGN2-T cells) could be seen. Cell viability and apoptosis were increased in HMGN2-T cells. The cytokine levels of interleukin 2 (IL-2) and tumor necrosis factor α (TNF-α) increased in cell supernatants of HMGN2-T cells. The percentage of G0/G1 phase cells was lower, the rate of S phase cells was higher in HMGN2-T cells than control cells. The co-culture of HMGN2-T cells and tumor cells could promote the cytokines' release. The LDH level was increased with the elevation of E:T ratios. In conclusion, the HMGN2-T cells were well-established and have the effect of secreting cytokines and killing tumor cells.


Subject(s)
HMGN2 Protein , CD28 Antigens/genetics , Cytokines , HMGN2 Protein/genetics , HMGN2 Protein/metabolism , Humans , Jurkat Cells , Leukocytes, Mononuclear/metabolism
17.
Acta Biochim Biophys Sin (Shanghai) ; 43(9): 680-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21778192

ABSTRACT

Since bacterial invasion into host cells is an important step in the infection process, using the agents to interfere with bacterial internalization is an attractive approach to block the infection process. In this work, we describe a new, previously unrecognized role of the human cationic host defense peptide HMGN2 during Klebsiella pneumoniae infections. Our results revealed that the internalization of K. pneumoniae strain 03183 into cultured bladder epithelial cells (T24) was significantly reduced at HMGN2 concentrations that were unable to produce any bacteriostatic or bactericidal effect. Using microarrays and follow-up studies, we demonstrated that HMGN2 affected the internalization of K. pneumoniae strain 03183 by inhibiting the attachment of bacteria, and then decreasing bacteria-induced ERK1/2 activation and actin polymerization, which might contribute to bacterial internalization into T24 cells. This disruption of bacterial internalization implied that HMGN2 could provide protection against K. pneumoniae infections.


Subject(s)
Endocytosis/drug effects , Epithelial Cells/microbiology , HMGN2 Protein/pharmacology , Klebsiella pneumoniae/physiology , Actins/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Blotting, Western , Butadienes/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Flavonoids/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Bacterial , HMGN2 Protein/genetics , Host-Pathogen Interactions/drug effects , Humans , Klebsiella pneumoniae/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Recombinant Proteins/pharmacology , Time Factors , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/microbiology , Urinary Bladder Neoplasms/pathology
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(4): 461-5, 2011 Jul.
Article in Zh | MEDLINE | ID: mdl-21866626

ABSTRACT

OBJECTIVE: To construct a prokaryotic expression recombinant for the expression of HMGN2 and to evaluate its antiviral activity against human hepatitis virus. METHODS: The extracellular region cDNA of HMGN2 was isolated and amplified by RT-PCR, and introduced to the prokaryotic expression vector pGEX-4T-1. HMGN2 protein was expressed under IPTG induction and purified by GST protein purification system, then identified by SDS-PAGE and Western blot. The cytotoxicity of fusion HMGN2 to HBV-transfected HepG2. 2.15 cell was evaluated with MTT assay. Different concentration of fusion HMGN2 was applied on the HepG2. 2.15 cell and the cell culture supernatants were harvested after 3 and 6 days treatment. The HBsAg and HBeAg in the supernatants were detected by ELISA and the HBV DNA was detected by RT-PCR. RESULTS: In the range of tested 1-100 microg/mL of HMGN2, no cytotoxicity to HepG2. 2.15 cells was detected by MTT assay. When incubated with HMGN2 at 15 microg/mL for 72 h or 144 h, there was a significant reduction in HBeAg and HBsAg expression as well as the HBV DNA copies. CONCLUSION: pGEX-4T-1/HMGN2 vector was success constructed, and the recombinant HMGN2 protein could inhibit HBV expression and replication in vitro remarkably.


Subject(s)
Antiviral Agents/pharmacology , Escherichia coli/metabolism , HMGN2 Protein/biosynthesis , Hepatitis B virus/drug effects , Recombinant Proteins/pharmacology , Escherichia coli/genetics , Genetic Vectors/genetics , HMGN2 Protein/genetics , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
19.
Chirurgia (Bucur) ; 106(6): 775-80, 2011.
Article in Ro | MEDLINE | ID: mdl-22308916

ABSTRACT

STUDY AIM: Immunohistochemical screening of hMLH1 and hMSH2 gene mutations in patients diagnosed with colorectal cancers, suspected of having microsatellite instability, as diagnosed between January 2002 and December 2009 in the Surgery Department of the CF Clinical Hospital Cluj-Napoca (prospective non-randomised study). METHODS: Inclusion criteria were adenocarcinoma pathology finding and also minimum one of the revised Bethesda criteria for genetic testing of microsatellite instability in colorectal cancers. 110 eligible patients were divided in 2 study groups according to the number of Bethesda criteria met (group A - 1 criteria; group B - 2 or more criteria). Both groups were statistically compared considering the clinical and pathological parameters specific to the Lynch syndrome. We performed immunohistochemical staining to determine the expression of hMLH1 and hMSH2 genes in the tumors of all the patients. RESULTS: We found the differences in age, colorectal family history and right colon tumor site between the two groups to be statistically significant. Immunohistochemical stainings showed lack of hMLH1 gene expresion in 9 patients and of hMSH2 gene in 4 patients respectively. CONCLUSIONS: Immunohistochemical staining can identify patients who need to be genetically tested for mutations of the DNA mismatch repair genes, in order to establish the correct diagnostic of Lynch syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Genetic Testing , HMGN2 Protein/genetics , Microsatellite Instability , Mutation , Nuclear Proteins/genetics , Algorithms , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , MutL Protein Homolog 1 , Prospective Studies
20.
Genes (Basel) ; 12(12)2021 12 17.
Article in English | MEDLINE | ID: mdl-34946949

ABSTRACT

DNA methylation and histone posttranslational modifications are epigenetics processes that contribute to neurophenotype of Down Syndrome (DS). Previous reports present strong evidence that nonhistone high-mobility-group N proteins (HMGN) are epigenetic regulators. They play important functions in various process to maintain homeostasis in the brain. We aimed to analyze the differential expression of five human HMGN genes in some brain structures and age ranks from DS postmortem brain samples. Methodology: We performed a computational analysis of the expression of human HMGN from the data of a DNA microarray experiment (GEO database ID GSE59630). Using the transformed log2 data, we analyzed the differential expression of five HMGN genes in several brain areas associated with cognition in patients with DS. Moreover, using information from different genome databases, we explored the co-expression and protein interactions of HMNGs with the histones of nucleosome core particle and linker H1 histone. Results: We registered that HMGN1 and HMGN5 were significantly overexpressed in the hippocampus and areas of prefrontal cortex including DFC, OFC, and VFC of DS patients. Age-rank comparisons between euploid control and DS individuals showed that HMGN2 and HMGN4 were overexpressed in the DS brain at 16 to 22 gestation weeks. From the BioGRID database, we registered high interaction scores of HMGN2 and HMGN4 with Hist1H1A and Hist1H3A. Conclusions: Overall, our results give strong evidence to propose that DS would be an epigenetics-based aneuploidy. Remodeling brain chromatin by HMGN1 and HMGN5 would be an essential pathway in the modification of brain homeostasis in DS.


Subject(s)
Cognition/physiology , Down Syndrome/genetics , HMGN Proteins/genetics , Brain/metabolism , Brain Mapping/methods , Databases, Genetic , Down Syndrome/metabolism , Epigenesis, Genetic/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , HMGN Proteins/metabolism , HMGN1 Protein/genetics , HMGN2 Protein/genetics , Hippocampus/metabolism , Humans , Nucleosomes/genetics , Prefrontal Cortex/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL