Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Mol Histol ; 53(4): 741-751, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35861945

RESUMO

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.


Assuntos
Proteína HMGN2 , Antígenos CD28/genética , Citocinas , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Humanos , Células Jurkat , Leucócitos Mononucleares/metabolismo
2.
Endocrinology ; 162(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33589921

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteína HMGN2/metabolismo , Desacetilase 6 de Histona/metabolismo , Prolactina/metabolismo , Fator de Transcrição STAT5/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Feminino , Regulação Neoplásica da Expressão Gênica , Proteína HMGN2/genética , Desacetilase 6 de Histona/genética , Humanos , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Elementos de Resposta , Fator de Transcrição STAT5/genética , Proteínas Supressoras de Tumor/genética
3.
Future Oncol ; 17(5): 541-548, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33467898

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Proteínas HMGN/genética , Proteína HMGN2/genética , Leucemia Mieloide Aguda/mortalidade , Transativadores/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Feminino , Seguimentos , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas/estatística & dados numéricos , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Prognóstico , Intervalo Livre de Progressão , Adulto Jovem
4.
Microbiol Immunol ; 64(12): 825-834, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33034909

RESUMO

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.


Assuntos
Aderência Bacteriana/fisiologia , Células Epiteliais/microbiologia , Proteína HMGN2/metabolismo , Integrina alfa5beta1/metabolismo , Klebsiella pneumoniae/metabolismo , Fatores de Transcrição NFI/metabolismo , Células A549 , Regulação da Expressão Gênica , Proteína HMGN2/genética , Humanos , Integrina alfa5/genética , Integrina alfa5/metabolismo , Integrina alfa5beta1/genética , Integrina beta1/genética , Integrina beta1/metabolismo , Infecções por Klebsiella/metabolismo , Klebsiella pneumoniae/genética , Pulmão , RNA Interferente Pequeno/metabolismo , Transcriptoma
5.
Epigenetics Chromatin ; 12(1): 73, 2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31831052

RESUMO

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.


Assuntos
Cromatina/metabolismo , Proteína HMGN2/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Autorrenovação Celular , Proteína HMGN1/genética , Proteína HMGN1/metabolismo , Proteína HMGN2/genética , Histonas/metabolismo , Camundongos , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Processamento de Proteína Pós-Traducional
6.
J Cell Mol Med ; 23(12): 7985-7998, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31596045

RESUMO

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.


Assuntos
Proteína HMGN2/metabolismo , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Infecções por Mycobacterium/imunologia , Micobactérias não Tuberculosas/crescimento & desenvolvimento , Animais , Sobrevivência Celular/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Proteína HMGN2/genética , Humanos , Imunidade Inata , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Mycobacterium abscessus/imunologia , Mycobacterium abscessus/isolamento & purificação , Mycobacterium smegmatis/imunologia , Mycobacterium smegmatis/isolamento & purificação , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Interferência de RNA , Fator de Necrose Tumoral alfa/metabolismo
7.
Acta Biochim Pol ; 65(1): 93-100, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29549670

RESUMO

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.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteína HMGN2/fisiologia , Proteínas de Junções Íntimas/metabolismo , Urotélio/microbiologia , Células Epiteliais/metabolismo , Proteína HMGN2/genética , Humanos , Regulação para Cima , Bexiga Urinária/citologia , Bexiga Urinária/patologia , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/patogenicidade , Urotélio/citologia , Urotélio/fisiologia
8.
Cell Physiol Biochem ; 44(5): 1681-1695, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29216626

RESUMO

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.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Proteína HMGN2/metabolismo , Animais , Caderinas/metabolismo , Proteínas Cdh1/metabolismo , Diferenciação Celular/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fulvestranto , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína HMGN2/antagonistas & inibidores , Proteína HMGN2/genética , Camundongos , Mucina-1/metabolismo , Gravidez , Progesterona/farmacologia , Prolactina/metabolismo , Interferência de RNA , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Útero/metabolismo , beta Catenina/metabolismo
9.
J Zhejiang Univ Sci B ; 18(5): 410-420, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28471113

RESUMO

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.


Assuntos
Antibacterianos/administração & dosagem , Permeabilidade da Membrana Celular/efeitos dos fármacos , Escherichia coli K12/efeitos dos fármacos , Escherichia coli K12/fisiologia , Proteína HMGN2/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Proteína HMGN2/genética , Humanos , Proteínas Recombinantes/administração & dosagem
10.
Int J Mol Med ; 38(3): 737-46, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27460641

RESUMO

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.


Assuntos
Células Epiteliais/microbiologia , Proteína HMGN2/genética , Integrina alfa5beta1/genética , Klebsiella pneumoniae/fisiologia , Interferência de RNA , Células A549 , Actinas/metabolismo , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Endocitose/fisiologia , Células Epiteliais/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Proteína HMGN2/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Microscopia de Fluorescência , Fosforilação , Polimerização , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Sistema Respiratório/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Oncol Rep ; 33(3): 1300-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25530340

RESUMO

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.


Assuntos
Neoplasias Ósseas/genética , Proteína HMGN2/genética , Osteossarcoma/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Ciclina D1/biossíntese , Ciclina E/biossíntese , Progressão da Doença , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/genética , Técnicas de Inativação de Genes , Células HEK293 , Proteína HMGN2/biossíntese , Humanos , Antígeno Ki-67/biossíntese , Lentivirus , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteínas Oncogênicas/biossíntese , Antígeno Nuclear de Célula em Proliferação/biossíntese , Transplante Heterólogo
12.
J Biol Chem ; 289(29): 20000-11, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24872413

RESUMO

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.


Assuntos
Proteína HMGN2/metabolismo , Nucleossomos/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação/genética , Linhagem Celular , Cisteína Endopeptidases , Endopeptidases/genética , Endopeptidases/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Proteína HMGN2/química , Proteína HMGN2/genética , Células HeLa , Humanos , Lisina/química , Dados de Sequência Molecular , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação , Ubiquitina-Proteína Ligases/genética
13.
PLoS One ; 8(2): e56587, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437179

RESUMO

EMT (epithelial-mesenchymal transition) is crucial for cancer cells to acquire invasive phenotypes. In A549 lung adenocarcinoma cells, TGF-ß elicited EMT in Smad-dependent manner and TNF-α accelerated this process, as confirmed by cell morphology, expression of EMT markers, capacity of gelatin lysis and cell invasion. TNF-α stimulated the phosphorylation of Smad2 linker region, and this effect was attenuated by inhibiting MEK or JNK pathway. Comprehensive expression analysis unraveled genes differentially regulated by TGF-ß and TNF-α, such as cytokines, chemokines, growth factors and ECM (extracellular matrices), suggesting the drastic change in autocrine/paracrine signals as well as cell-to-ECM interactions. Integrated analysis of microRNA signature enabled us to identify a subset of genes, potentially regulated by microRNAs. Among them, we confirmed TGF-ß-mediated induction of miR-23a in lung epithelial cell lines, target genes of which were further identified by gene expression profiling. Combined with in silico approaches, we determined HMGN2 as a downstream target of miR-23a. These findings provide a line of evidence that the effects of TGF-ß and TNF-α were partially mediated by microRNAs, and shed light on the complexity of molecular events elicited by TGF-ß and TNF-α.


Assuntos
Adenocarcinoma/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Simulação por Computador , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética
14.
Cell Biol Int ; 36(2): 195-202, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21988615

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Células Eritroides/citologia , Proteína HMGN2/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Fator de Transcrição GATA1/metabolismo , Perfilação da Expressão Gênica , Proteína HMGN2/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Fase S
15.
Acta Biochim Biophys Sin (Shanghai) ; 43(9): 680-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21778192

RESUMO

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.


Assuntos
Endocitose/efeitos dos fármacos , Células Epiteliais/microbiologia , Proteína HMGN2/farmacologia , Klebsiella pneumoniae/fisiologia , Actinas/metabolismo , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Western Blotting , Butadienos/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Flavonoides/farmacologia , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Proteína HMGN2/genética , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Klebsiella pneumoniae/genética , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Recombinantes/farmacologia , Fatores de Tempo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/microbiologia , Neoplasias da Bexiga Urinária/patologia
16.
FEBS J ; 278(12): 2152-66, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21518253

RESUMO

Human ß-defensin-2 (HBD-2) is an antimicrobial peptide produced by the epithelial cells that plays an important role in innate and adaptive immunity. Here we report that high mobility group protein N2 (HMGN2), a member of the high mobility group superfamily that affects chromatin function, modulates the expression of HBD-2 in A549 cells treated by lipopolysaccharide. Mechanistically, HMGN2 prolongs the retention time and enhances the accumulation of nuclear factor κB p65 in the nucleus, and promotes the acetylation of p65 through increasing histone acetyltransferase activity and enhancing p65-Ser536 phosphorylation. Additionally, chromatin immunoprecipitation reveals that HMGN2 and p65 synergistically promote their specific binding to HBD-2 promoter, thereby affecting the downstream transcription. Taken together, these results suggest that HMGN2 acts as a positive modulator of nuclear factor κB signalling to promote lipopolysaccharide-induced ß-defensin expression.


Assuntos
Proteína HMGN2/metabolismo , beta-Defensinas/genética , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Proteína HMGN2/antagonistas & inibidores , Proteína HMGN2/genética , Humanos , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Transdução de Sinais , Fator de Transcrição RelA/metabolismo
17.
Chirurgia (Bucur) ; 106(6): 775-80, 2011.
Artigo em Romano | MEDLINE | ID: mdl-22308916

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Testes Genéticos , Proteína HMGN2/genética , Instabilidade de Microssatélites , Mutação , Proteínas Nucleares/genética , Algoritmos , Neoplasias Colorretais Hereditárias sem Polipose/genética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Estudos Prospectivos
18.
Mol Cell ; 34(5): 620-6, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19524541

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Proteína HMGN1/fisiologia , Proteína HMGN2/fisiologia , Nucleossomos/metabolismo , Proteínas Recombinantes/metabolismo , Trifosfato de Adenosina/metabolismo , Cromatina/metabolismo , DNA Helicases/metabolismo , Proteína HMGN1/genética , Proteína HMGN1/metabolismo , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Humanos , Proteínas Motores Moleculares/fisiologia , Mutação , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo
19.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 22(4): 773-7, 2005 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-16156270

RESUMO

Total RNA was extracted from human LAK cell, and a cDNA encoding mature peptide HMG-17 and its alpha helix domain was amplified by RT-PCR. The recombinant prokaryotic expression vector pGEX-1lambdaT-HMG-17 and pGEX-1lambdaT HMG-17alpha helix was constructed. Using affinity chromatography, thrombin cleaving and AU-PAGE elution, we obtained the purified HMG-17. Analyses of MIC, MEC and MBC indicated that HMG-17 and HMG-17alpha had strong antibacterial activity. MIC of the alpha-helic domain was almost the same as that of HMG17, suggesting that the alpha-helic structure would be essential for the antibacterial activity of HMG-17.


Assuntos
Antibacterianos/biossíntese , Escherichia coli/metabolismo , Proteína HMGN2/biossíntese , Proteína HMGN2/farmacologia , Antibacterianos/farmacologia , Escherichia coli/genética , Proteína HMGN2/genética , Humanos , Células Matadoras Ativadas por Linfocina/química , Peptídeos/genética , Peptídeos/farmacologia , Células Procarióticas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
20.
Protein Pept Lett ; 11(6): 515-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15579118

RESUMO

HMG-17 proteins are nucleosomal proteins, implicated in control of chromatin structure and transcription, in a way that has not yet been fully understood. In this report, quantification of HMG-17 in porcine tissues was performed, by ELISA, using previously produced and characterized specific rabbit anti-HMG-17 antibodies. Our results showed high levels of HMG-17 compared to the DNA and H1 content of the tissues (on the contrary to previous reports), and more specifically that there were 1.7 molecules of HMG-17 per molecule of histone H1.


Assuntos
Cromossomos/metabolismo , Proteína HMGN2/análise , Técnicas Imunoenzimáticas/métodos , Animais , Ensaio de Imunoadsorção Enzimática/métodos , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Suínos/genética , Suínos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA