Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
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
2.
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
3.
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
4.
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
5.
Endocrinology ; 162(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33589921

ABSTRACT

The hormone prolactin has been implicated in breast cancer pathogenesis and regulates chromatin engagement by the transcription factor, STAT5A. STAT5A is known to inducibly bind promoters and cis-regulatory elements genome-wide, though the mechanisms by which it exerts specificity and regulation of target gene expression remain enigmatic. We previously identified HDAC6 and HMGN2 as cofactors that facilitate prolactin-induced, STAT5A-mediated gene expression. Here, multicondition STAT5A, HDAC6, and HMGN2 chromatin immunoprecipitation and sequencing with parallel condition RNA-seq are utilized to reveal the cis-regulatory landscape and cofactor dynamics underlying prolactin-stimulated gene expression in breast cancer. We find that prolactin-regulated genes are significantly enriched for cis-regulatory elements bound by HDAC6 and HMGN2, and that inducible STAT5A binding at enhancers, rather than promoters, conveys specificity for prolactin-regulated genes. The selective HDAC6 inhibitor, ACY-241, blocks prolactin-induced STAT5A chromatin engagement at cis-regulatory elements as well as a significant proportion of prolactin-stimulated gene expression. We identify functional pathways known to contribute to the development and/or progression of breast cancer that are activated by prolactin and inhibited by ACY-241. Additionally, we find that the DNA sequences underlying shared STAT5A and HDAC6 binding sites at enhancers are differentially enriched for estrogen response elements (ESR1 and ESR2 motifs) relative to enhancers bound by STAT5A alone. Gene set enrichment analysis identifies significant overlap of ERα-regulated genes with genes regulated by prolactin, particularly prolactin-regulated genes with promoters or enhancers co-occupied by both STAT5A and HDAC6. Lastly, the therapeutic efficacy of ACY-241 is demonstrated in in vitro and in vivo breast cancer models, where we identify synergistic ACY-241 drug combinations and observe differential sensitivity of ER+ models relative to ER- models.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , HMGN2 Protein/metabolism , Histone Deacetylase 6/metabolism , Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Neoplastic , HMGN2 Protein/genetics , Histone Deacetylase 6/genetics , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Response Elements , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics
6.
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
7.
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
8.
Sci Rep ; 10(1): 4332, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152397

ABSTRACT

Transcription-coupled repair (TCR) removes DNA lesions from the transcribed strand of active genes. Stalling of RNA polymerase II (RNAPII) at DNA lesions initiates TCR through the recruitment of the CSB and CSA proteins. The full repertoire of proteins required for human TCR - particularly in a chromatin context - remains to be determined. Studies in mice have revealed that the nucleosome-binding protein HMGN1 is required to enhance the repair of UV-induced lesions in transcribed genes. However, whether HMGN1 is required for human TCR remains unaddressed. Here, we show that knockout or knockdown of HMGN1, either alone or in combination with HMGN2, does not render human cells sensitive to UV light or Illudin S-induced transcription-blocking DNA lesions. Moreover, transcription restart after UV irradiation was not impaired in HMGN-deficient cells. In contrast, TCR-deficient cells were highly sensitive to DNA damage and failed to restart transcription. Furthermore, GFP-tagged HMGN1 was not recruited to sites of UV-induced DNA damage under conditions where GFP-CSB readily accumulated. In line with this, HMGN1 did not associate with the TCR complex, nor did TCR proteins require HMGN1 to associate with DNA damage-stalled RNAPII. Together, our findings suggest that HMGN1 and HMGN2 are not required for human TCR.


Subject(s)
DNA Repair , HMGN1 Protein/genetics , HMGN2 Protein/genetics , Transcription, Genetic , Cell Line , DNA Damage/genetics , DNA Damage/radiation effects , Gene Knockout Techniques , HMGN1 Protein/metabolism , HMGN2 Protein/metabolism , Humans , Radiation Tolerance , Telomerase/genetics , Telomerase/metabolism , Transcription, Genetic/radiation effects , Ultraviolet Rays
9.
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
10.
Epigenetics Chromatin ; 12(1): 73, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831052

ABSTRACT

BACKGROUND: Members of the HMGN protein family modulate chromatin structure and influence epigenetic modifications. HMGN1 and HMGN2 are highly expressed during early development and in the neural stem/progenitor cells of the developing and adult brain. Here, we investigate whether HMGN proteins contribute to the chromatin plasticity and epigenetic regulation that is essential for maintaining pluripotency in stem cells. RESULTS: We show that loss of Hmgn1 or Hmgn2 in pluripotent embryonal carcinoma cells leads to increased levels of spontaneous neuronal differentiation. This is accompanied by the loss of pluripotency markers Nanog and Ssea1, and increased expression of the pro-neural transcription factors Neurog1 and Ascl1. Neural stem cells derived from these Hmgn-knockout lines also show increased spontaneous neuronal differentiation and Neurog1 expression. The loss of HMGN2 leads to a global reduction in H3K9 acetylation, and disrupts the profile of H3K4me3, H3K9ac, H3K27ac and H3K122ac at the Nanog and Oct4 loci. At endodermal/mesodermal genes, Hmgn2-knockout cells show a switch from a bivalent to a repressive chromatin configuration. However, at neuronal lineage genes whose expression is increased, no epigenetic changes are observed and their bivalent states are retained following the loss of HMGN2. CONCLUSIONS: We conclude that HMGN1 and HMGN2 maintain the identity of pluripotent embryonal carcinoma cells by optimising the pluripotency transcription factor network and protecting the cells from precocious differentiation. Our evidence suggests that HMGN2 regulates active and bivalent genes by promoting an epigenetic landscape of active histone modifications at promoters and enhancers.


Subject(s)
Chromatin/metabolism , HMGN2 Protein/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Self Renewal , HMGN1 Protein/genetics , HMGN1 Protein/metabolism , HMGN2 Protein/genetics , Histones/metabolism , Mice , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Processing, Post-Translational
11.
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
12.
Nat Commun ; 9(1): 5240, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30532006

ABSTRACT

The dynamic nature of the chromatin epigenetic landscape plays a key role in the establishment and maintenance of cell identity, yet the factors that affect the dynamics of the epigenome are not fully known. Here we find that the ubiquitous nucleosome binding proteins HMGN1 and HMGN2 preferentially colocalize with epigenetic marks of active chromatin, and with cell-type specific enhancers. Loss of HMGNs enhances the rate of OSKM induced reprogramming of mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells (iPSCs), and the ASCL1 induced conversion of fibroblast into neurons. During transcription factor induced reprogramming to pluripotency, loss of HMGNs accelerates the erasure of the MEF-specific epigenetic landscape and the establishment of an iPSCs-specific chromatin landscape, without affecting the pluripotency potential and the differentiation potential of the reprogrammed cells. Thus, HMGN proteins modulate the plasticity of the chromatin epigenetic landscape thereby stabilizing, rather than determining cell identity.


Subject(s)
Cell Membrane/metabolism , Fibroblasts/metabolism , HMGN1 Protein/metabolism , HMGN2 Protein/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Embryo, Mammalian/cytology , Epigenesis, Genetic , Fibroblasts/cytology , HEK293 Cells , HMGN1 Protein/genetics , HMGN2 Protein/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice, Knockout , Mice, Nude , Protein Binding
13.
Acta Biochim Pol ; 65(1): 93-100, 2018.
Article in English | MEDLINE | ID: mdl-29549670

ABSTRACT

The urinary tract is vulnerable to frequent challenges from environmental microflora. Uropathogenic Escherichia coli (UPEC) makes a major contribution to urinary tract infection (UTI). Previous studies have characterized positive roles of non-histone nuclear protein HMGN2 in lung epithelial innate immune response. In the study presented here, we found HMGN2 expression was up-regulated in UPEC J96-infected urothelium. Surprisingly, over-expression of HMGN2 promoted disruption of BECs 5637 cells' intercellular junctions by down-regulating tight junction (TJs) components' expression and physical structure under J96 infection. Further investigation showed that BECs 5637 monolayer, in which HMGN2 was over-expressed, had significantly increased permeability to J96. Our study systemically explored the regulatory roles of HMGN2 in BECs barrier function during UPEC infection and suggested different modulations of intracellular and paracellular routes through which UPEC invades the bladder epithelium.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , HMGN2 Protein/physiology , Tight Junction Proteins/metabolism , Urothelium/microbiology , Epithelial Cells/metabolism , HMGN2 Protein/genetics , Humans , Up-Regulation , Urinary Bladder/cytology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/pathogenicity , Urothelium/cytology , Urothelium/physiology
14.
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
15.
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
16.
J Zhejiang Univ Sci B ; 18(5): 410-420, 2017 May.
Article in English | MEDLINE | ID: mdl-28471113

ABSTRACT

OBJECTIVE: To investigate the antibacterial mechanism of high-mobility group nucleosomal-binding domain 2 (HMGN2) on Escherichia coli K12, focusing on the antibacterial and antibiofilm formation effects. Its chemotactic activity on human neutrophils was also investigated. METHODS: Human tissue-derived HMGN2 (tHMGN2) was extracted from fresh uterus fiber cystadenoma and purified by HP1100 reversed-phase high-performance liquid chromatography (RP-HPLC). Recombinant human HMGN2 (rHMGN2) was generated in E. coli DE3 carrying PET-32a-c(+)-HMGN2. Antibacterial activity of HMGN2 was determined using an agarose diffusion assay and minimum inhibitory concentration (MIC) of HMGN2 was determined by the microdilution broth method. Bacterial membrane permeability assay and DNA binding assay were performed. The antibiofilm effect of HMGN2 was investigated using a crystal violet assay and electron microscopy scanning. The activating effect and chemotactic activity of HMGN2 on neutrophils were determined using a nitroblue tetrazolium (NBT) reduction assay and Transwell chamber cell migration assay, respectively. RESULTS: HMGN2 showed a relatively high potency against Gram-negative bacteria E. coli and the MIC of HMGN2 was 16.25 µg/ml. Elevated bacterial membrane permeability was observed in HMGN2-treated E. coli K12. HMGN2 could also bind the bacterial plasmid and genomic DNA in a dose-dependent manner. The antibiofilm effect of HMGN2 on E. coli K12 was confirmed by crystal violet staining and scanning electron microscopy. However, the activating effects and chemotactic effects of HMGN2 on human neutrophils were not observed. CONCLUSIONS: As an antimicrobial peptide (AMP), HMGN2 possessed a good capacity for antibacterial and antibiofilm activities on E. coli K12. This capacity might be associated with disruption of the bacterial membrane and combination of DNA, which might affect the growth and viability of E. coli.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cell Membrane Permeability/drug effects , Escherichia coli K12/drug effects , Escherichia coli K12/physiology , HMGN2 Protein/administration & dosage , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , HMGN2 Protein/genetics , Humans , Recombinant Proteins/administration & dosage
17.
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
18.
Int J Mol Med ; 38(3): 737-46, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27460641

ABSTRACT

Integrin receptors, a large family of adhesion receptors, are involved in the attachment of Klebsiella pneumoniae to respiratory epithelial cells, and subsequently cause the internalization of K. pneumoniae by host cells. Although a number of molecules have been reported to regulate the expression and activity of integrin receptors in respiratory epithelial cells, the specific underlying molecular mechanisms remain largely unknown. High mobility group nucleosomal binding domain 2 (HMGN2), a non-histone nuclear protein, is present in eukaryotic cells as a ubiquitous nuclear protein. Our previous studies have demonstrated that HMGN2 affects chromatin function and modulates the expression of antibacterial peptide in A549 cells exposed to lipopolysaccharide, which indicates the critical role of HMGN2 in innate immune responses. In addition, our cDNA microarray analysis suggested that HMGN2 knockdown induced the enhanced expression of α5ß1 integrin in A549 cells. Therefore, we hypothesized that intercellular HMGN2 may mediate the internalization of K. pneumoniae by altering the expression of α5ß1 integrin. Using the A549 cell line, we demonstrated that HMGN2 knockdown induced the increased expression of α5ß1 integrin on cell membranes, which resulted in a significant increase in K. pneumoniae internalization. Further results revealed that HMGN2 silencing induced the expression of talin and the activation of α5ß1 integrin, which led to actin polymerization following the phosphorylation of FAK and Src. This study suggests a possible therapeutic application for bacterial internalization by targeting HMGN2 in order to treat K. pneumoniae infection.


Subject(s)
Epithelial Cells/microbiology , HMGN2 Protein/genetics , Integrin alpha5beta1/genetics , Klebsiella pneumoniae/physiology , RNA Interference , A549 Cells , Actins/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Endocytosis/physiology , Epithelial Cells/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HMGN2 Protein/metabolism , Humans , Integrin alpha5beta1/metabolism , Microscopy, Fluorescence , Phosphorylation , Polymerization , Proto-Oncogene Proteins pp60(c-src)/metabolism , Respiratory System/cytology , Reverse Transcriptase Polymerase Chain Reaction
19.
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
20.
Oncol Rep ; 33(3): 1300-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25530340

ABSTRACT

High mobility group N (HMGNs) are members of the high mobility group protein family, and are involved in the development and progression of several tumors. HMGN1 and HMGN5 were previously shown to be associated with the bioactivities of osteosarcoma. However, the effects and molecular mechanisms of HMGN2 on osteosarcoma progression remain to be determined. In order to characterize the endogenous expression of HMGN2 in osteosarcoma cell lines, RT-PCR and western blot analysis were performed. Recombinant HMGN2 lentivirus was used to infect the osteosarcoma cell lines with relatively low HMGN2 expression to determine the functional relevance of HMGN2 overexpression in osteosarcoma cell growth and migration in vitro and in vivo, and to investigate the expression levels of Ki-67, PCNA, cyclin D1 and cyclin E. The results showed that osteosarcoma cell proliferation and migration were significantly reduced by HMGN2, as indicated by cell count and wound-healing assays. Cell apoptosis was markedly induced and HMGN2 increased the sensitivity to chemotherapy. When HMGN2 expression was enhanced, the expression of cyclin D1 and PCNA was downregulated in osteosarcoma cells. In addition, the tumor volumes in SaO2 and U2-OS subcutaneous nude mouse models treated with HMGN2 lentivirus were significantly decreased as compared to those of the GFP group. These results suggested that the enhanced expression of HMGN2 in osteosarcoma cells by HMGN2 lentivirus, exerts inhibitory effects on growth and migration of osteosarcoma cells.


Subject(s)
Bone Neoplasms/genetics , HMGN2 Protein/genetics , Osteosarcoma/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Knockout Techniques , HEK293 Cells , HMGN2 Protein/biosynthesis , Humans , Ki-67 Antigen/biosynthesis , Lentivirus , Mice , Mice, Nude , Neoplasm Transplantation , Oncogene Proteins/biosynthesis , Proliferating Cell Nuclear Antigen/biosynthesis , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...