Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
J Dent Res ; 103(1): 51-61, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37950483

RESUMO

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.


Assuntos
Proteínas do Esmalte Dentário , Proteína HMGN1 , Proteína HMGN2 , Animais , Camundongos , Ameloblastos/metabolismo , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Proteína HMGN1/genética , Proteína HMGN1/metabolismo , Epigênese Genética , Diferenciação Celular/genética , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Fatores de Transcrição/metabolismo , Proteínas do Esmalte Dentário/genética , Proteínas do Esmalte Dentário/metabolismo , Cromatina/metabolismo , Amelogenina/metabolismo
2.
Sci Rep ; 12(1): 18423, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36319719

RESUMO

Testicular germ cell tumor (TGCT) is a rare cancer but the most common tumor among adolescent and young adult males. Patients with advanced TGCT often exhibit a worse prognosis due to the acquisition of therapeutic resistance. Cisplatin-based chemotherapy is a standard treatment for advanced TGCTs initially sensitive to cisplatin, as exemplified by embryonal carcinoma. The acquisition of cisplatin resistance, however, could be a fatal obstacle for TGCT management. To identify cisplatin resistance-related genes, we performed transcriptome analysis for cisplatin-resistant TGCT cells compared to parental cells. In two types of cisplatin-resistant TGCT cell models that we established from patient-derived TGCT cells, and from the NEC8 cell line, we found that mRNA levels of the high-mobility-group nucleosome-binding gene HMGN5 and meiosis-related gene TEX11 were remarkably upregulated compared to those in the corresponding parental cells. We showed that either HMGN5 or TEX11 knockdown substantially reduced the viability of cisplatin-resistant TGCT cells in the presence of cisplatin. Notably, TEX11 silencing in cisplatin-resistant TGCT cells increased the level of cleaved PARP1 protein, and the percentage of double-strand break marker γH2AX-positive cells. We further demonstrated the therapeutic efficiency of TEX11-specific siRNA on in vivo xenograft models derived from cisplatin-resistant patient-derived TGCT cells. Taken together, the present study provides a potential insight into a mechanism of cisplatin resistance via TEX11-dependent pathways that inhibit apoptosis and DNA damage. We expect that our findings can be applied to the improvement of cisplatin-based chemotherapy for TGCT, particularly for TEX11-overexpressing tumor.


Assuntos
Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Adolescente , Humanos , Masculino , Linhagem Celular Tumoral , Cisplatino/farmacologia , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Proteínas HMGN , Neoplasias Testiculares/genética , Testículo/metabolismo , Transativadores/genética
3.
Nat Commun ; 13(1): 7303, 2022 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-36435799

RESUMO

White adipose tissue browning is a key metabolic process controlled by epigenetic factors that facilitate changes in gene expression leading to altered cell identity. We find that male mice lacking the nucleosome binding proteins HMGN1 and HMGN2 (DKO mice), show decreased body weight and inguinal WAT mass, but elevated food intake, WAT browning and energy expenditure. DKO white preadipocytes show reduced chromatin accessibility and lower FRA2 and JUN binding at Pparγ and Pparα promoters. White preadipocytes and mouse embryonic fibroblasts from DKO mice show enhanced rate of differentiation into brown-like adipocytes. Differentiating DKO adipocytes show reduced H3K27ac levels at white adipocyte-specific enhancers but elevated H3K27ac levels at brown adipocyte-specific enhancers, suggesting a faster rate of change in cell identity, from white to brown-like adipocytes. Thus, HMGN proteins function as epigenetic factors that stabilize white adipocyte cell identity, thereby modulating the rate of white adipose tissue browning and affecting energy metabolism in mice.


Assuntos
Tecido Adiposo Marrom , Nucleossomos , Masculino , Animais , Camundongos , Nucleossomos/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas HMGN/metabolismo , Epigênese Genética , Fibroblastos/metabolismo , Tecido Adiposo Branco/metabolismo , Adipócitos Marrons/metabolismo , Metabolismo Energético/genética
4.
Mol Cancer Res ; 20(12): 1724-1738, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36066963

RESUMO

Cancer progression is highly dependent on the ability of cancer cell tumor formation, in which epigenetic modulation plays an essential role. However, the epigenetic factors promoting breast tumor formation are less known. Screened from three-dimensional (3D)-sphere tumor formation model, HMGN5 that regulates chromatin structures became the candidate therapeutic target in breast cancer, though its role is obscure. HMGN5 is highly expressed in 3D-spheres of breast cancer cells and clinical tumors, also an unfavorable prognostic marker in patients. Furthermore, HMGN5 controls tumor formation and metastasis of breast cancer cells in vitro and in vivo. Mechanistically, HMGN5 is governed by active STAT3 transcriptionally and further escorts STAT3 to shape the oncogenic chromatin landscape and transcriptional program. More importantly, interference of HMGN5 by nanovehicle-packaged siRNA effectively inhibits tumor growth in breast cancer cell-derived xenograft mice model. IMPLICATIONS: Our findings reveal a novel feed-forward circuit between HMGN5 and STAT3 in promoting breast cancer tumorigenesis and suggest HMGN5 as a novel epigenetic therapeutic target in STAT3-hyperactive breast cancer.


Assuntos
Neoplasias da Mama , Proteínas HMGN , Humanos , Camundongos , Animais , Feminino , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Cromatina/genética , Linhagem Celular Tumoral , Proliferação de Células , Apoptose/genética , Transativadores/metabolismo , Neoplasias da Mama/genética , Fator de Transcrição STAT3/genética , Carcinogênese/genética
5.
Carcinogenesis ; 43(9): 874-884, 2022 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-35792800

RESUMO

High-mobility group nucleosome-binding domain 4 (HMGN4) exerts biological functions by regulating gene transcription through binding with nucleosome. As a new epigenetic regulator discovered in 2001, its biological functions have not been clarified. HMGN4 belongs to HMGNs family, in which HMGN1, 2 and 5 have been reported to play roles in oncogenesis of various cancers. However, it is reported that HMGN4 was associated with thyroid and liver cancer. In this study, we discovered for the first time that HMGN4 was highly expressed in human triple-negative breast cancer (TNBC), based on the analysis of the TCGA database. Moreover, we found that HMGN4 controlled the proliferation of human TNBC cells both in vitro and in vivo. Mechanistically, the positive correlation occurred between HMGN4 and STAT3 downstream genes while HMGN4 played an indispensable role in constitutively active STAT3 (STAT3C) induced colony formation. Interestingly, we reported that STAT3 regulated HMGN4 transcription as its transcriptional factor by chromatin immunoprecipitation and HMGN4 promoter-luc assays. That is to say, there is a feed-forward signaling circuit between HMGN4 and STAT3, which might control TNBC cell growth. Finally, we proved that the interference of HMGN4 by nanovehicle-packaged siRNA may be a potentially effective approach in TNBC treatment. In summary, our findings not only identified a novel regulator in TNBC cell proliferation but also revealed the mechanism by which HMGN4 acted as a downstream gene of STAT3 to participate in the STAT3 pathway, which indicated that HMGN4 was likely to be a potential novel target for anti-TNBC therapy.


Assuntos
Proteínas HMGN , Fator de Transcrição STAT3 , Neoplasias de Mama Triplo Negativas , Humanos , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Nucleossomos , RNA Interferente Pequeno , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas HMGN/genética
6.
Epigenetics Chromatin ; 15(1): 23, 2022 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-35761366

RESUMO

BACKGROUND: Nucleosomal binding proteins, HMGN, is a family of chromatin architectural proteins that are expressed in all vertebrate nuclei. Although previous studies have discovered that HMGN proteins have important roles in gene regulation and chromatin accessibility, whether and how HMGN proteins affect higher order chromatin status remains unknown. RESULTS: We examined the roles that HMGN1 and HMGN2 proteins play in higher order chromatin structures in three different cell types. We interrogated data generated in situ, using several techniques, including Hi-C, Promoter Capture Hi-C, ChIP-seq, and ChIP-MS. Our results show that HMGN proteins occupy the A compartment in the 3D nucleus space. In particular, HMGN proteins occupy genomic regions involved in cell-type-specific long-range promoter-enhancer interactions. Interestingly, depletion of HMGN proteins in the three different cell types does not cause structural changes in higher order chromatin, i.e., in topologically associated domains (TADs) and in A/B compartment scores. Using ChIP-seq combined with mass spectrometry, we discovered protein partners that are directly associated with or neighbors of HMGNs on nucleosomes. CONCLUSIONS: We determined how HMGN chromatin architectural proteins are positioned within a 3D nucleus space, including the identification of their binding partners in mononucleosomes. Our research indicates that HMGN proteins localize to active chromatin compartments but do not have major effects on 3D higher order chromatin structure and that their binding to chromatin is not dependent on specific protein partners.


Assuntos
Cromatina , Proteínas HMGN , Epigênese Genética , Proteínas HMGN/química , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Nucleossomos , Ligação Proteica
7.
FASEB J ; 36(7): e22345, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35635715

RESUMO

High mobility group nucleosome-binding protein 3 (HMGN3), a member of the HMGN family, modulates the structure of chromatin and regulates transcription through transcription factors. HMGN3 has been implicated in the development of various cancers; however, the underlying mechanisms remain unclear. We herein demonstrated that the high expression of HMGN3 correlated with the metastasis of liver fluke infection-induced cholangiocarcinoma (CCA) in patients in northeastern Thailand. The knockdown of HMGN3 in CCA cells significantly impaired the oncogenic properties of colony formation, migration, and invasion. HMGN3 inhibited the expression of and blocked the intracellular polarities of epithelial regulator genes, such as the CDH1/E-cadherin and TJAP1 genes in CCA cells. A chromatin immunoprecipitation sequencing analysis revealed that HMGN3 required the transcription factor SNAI2 to bind to and repress the expression of epithelial regulator genes, at least in part, due to histone deacetylases (HDACs), the pharmacological inhibition of which reactivated these epithelial regulators in CCA, leading to impairing the cell migration capacity. Therefore, the overexpression of HMGN3 represses the transcription of and blocks the polarities of epithelial regulators in CCA cells in a manner that is dependent on the SNAI2 gene and HDACs.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Regulação da Expressão Gênica , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Humanos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Cell Rep ; 38(13): 110570, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35354046

RESUMO

The appearance of trophectoderm (TE) is a hallmark event in preimplantation development during murine embryogenesis. However, little is known about the mechanisms underlying TE specification. We find that the depletion of Rif1 breaks down the barrier to the transition from embryonic stem cells (ESCs) to trophoblast stem cells (TSCs). Rif1-null-induced TSCs show typical TE properties and the potential to differentiate into terminal trophoblast lineages. Global transcriptome analysis reveal that Rif1 deletion activates 2-cell embryo (2C)-related genes and induces a totipotent-like state. Chimeric assays further confirm that Rif1-null ESCs contribute to the functional placenta in addition to the fetus on embryonic day 12.5. Furthermore, we show overexpression of Hmgn3, one of the key upregulated gene in Rif1-null ESCs, facilitates the induction of TSCs. Therefore, we report two key genes regulating the conversion of TSCs and provide insights for investigating TE specification.


Assuntos
Células-Tronco Embrionárias , Trofoblastos , Animais , Feminino , Perfilação da Expressão Gênica , Proteínas HMGN , Camundongos , Placenta , Gravidez , Proteínas de Ligação a Telômeros/genética
9.
Commun Biol ; 5(1): 159, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35197580

RESUMO

Nucleosomes containing acetylated H3K27 are a major epigenetic mark of active chromatin and identify cell-type specific chromatin regulatory regions which serve as binding sites for transcription factors. Here we show that the ubiquitous nucleosome binding proteins HMGN1 and HMGN2 bind preferentially to H3K27ac nucleosomes at cell-type specific chromatin regulatory regions. HMGNs bind directly to the acetylated nucleosome; the H3K27ac residue and linker DNA facilitate the preferential binding of HMGNs to the modified nucleosomes. Loss of HMGNs increases the levels of H3K27me3 and the histone H1 occupancy at enhancers and promoters and alters the interaction of transcription factors with chromatin. These experiments indicate that the H3K27ac epigenetic mark enhances the interaction of architectural protein with chromatin regulatory sites and identify determinants that facilitate the localization of HMGN proteins at regulatory sites to modulate cell-type specific gene expression.


Assuntos
Proteínas HMGN , Nucleossomos , Cromatina/genética , Proteínas HMGN/química , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Nucleossomos/genética , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Genes (Basel) ; 12(12)2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34946949

RESUMO

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.


Assuntos
Cognição/fisiologia , Síndrome de Down/genética , Proteínas HMGN/genética , Encéfalo/metabolismo , Mapeamento Encefálico/métodos , Bases de Dados Genéticas , Síndrome de Down/metabolismo , Epigênese Genética/genética , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Proteínas HMGN/metabolismo , Proteína HMGN1/genética , Proteína HMGN2/genética , Hipocampo/metabolismo , Humanos , Nucleossomos/genética , Córtex Pré-Frontal/metabolismo , Transativadores/genética , Fatores de Transcrição/genética , Transcriptoma/genética
11.
Nucleic Acids Res ; 49(15): 8642-8664, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34352099

RESUMO

Architectural DNA-binding proteins (ADBPs) are abundant constituents of eukaryotic or bacterial chromosomes that bind DNA promiscuously and function in diverse DNA reactions. They generate large conformational changes in DNA upon binding yet can slide along DNA when searching for functional binding sites. Here we investigate the mechanism by which ADBPs diffuse on DNA by single-molecule analyses of mutant proteins rationally chosen to distinguish between rotation-coupled diffusion and DNA surface sliding after transient unbinding from the groove(s). The properties of yeast Nhp6A mutant proteins, combined with molecular dynamics simulations, suggest Nhp6A switches between two binding modes: a static state, in which the HMGB domain is bound within the minor groove with the DNA highly bent, and a mobile state, where the protein is traveling along the DNA surface by means of its flexible N-terminal basic arm. The behaviors of Fis mutants, a bacterial nucleoid-associated helix-turn-helix dimer, are best explained by mobile proteins unbinding from the major groove and diffusing along the DNA surface. Nhp6A, Fis, and bacterial HU are all near exclusively associated with the chromosome, as packaged within the bacterial nucleoid, and can be modeled by three diffusion modes where HU exhibits the fastest and Fis the slowest diffusion.


Assuntos
Proteínas de Ligação a DNA/genética , DNA/genética , Proteínas HMGN/genética , Proteínas Mutantes/genética , Proteínas de Saccharomyces cerevisiae/genética , Cromossomos Bacterianos/genética , Proteínas Mitocondriais/genética , Simulação de Dinâmica Molecular , Ligação Proteica/genética , Saccharomyces cerevisiae/genética , Imagem Individual de Molécula
12.
Sci Rep ; 11(1): 14165, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34239016

RESUMO

The genome editing protein Cas9 faces engineering challenges in improving off-target DNA cleavage and low editing efficiency. In this study, we aimed to engineer Cas9 to be able to slide along DNA, which might facilitate genome editing and reduce off-target cleavage. We used two approaches to achieve this: reducing the sliding friction along DNA by removing the interactions of Cas9 residues with DNA and facilitating sliding by introducing the sliding-promoting tail of Nhp6A. Seven engineered mutants of Cas9 were prepared, and their performance was tested using single-molecule fluorescence microscopy. Comparison of the mutations enabled the identification of key residues of Cas9 to enhance the sliding along DNA in the presence and absence of single guide RNA (sgRNA). The attachment of the tail to Cas9 mutants enhanced sliding along DNA, particularly in the presence of sgRNA. Together, using the proposed approaches, the sliding ability of Cas9 was improved up to eightfold in the presence of sgRNA. A sliding model of Cas9 and its engineering action are discussed herein.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , DNA/metabolismo , Edição de Genes , Engenharia Genética , Proteína 9 Associada à CRISPR/genética , Proteínas HMGN/metabolismo , Modelos Biológicos , Mutação/genética , RNA Guia de Cinetoplastídeos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
13.
Genome Biol ; 22(1): 203, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253240

RESUMO

BACKGROUND: The origin of sex chromosomes requires the establishment of recombination suppression between the proto-sex chromosomes. In many fish species, the sex chromosome pair is homomorphic with a recent origin, providing species for studying how and why recombination suppression evolved in the initial stages of sex chromosome differentiation, but this requires accurate sequence assembly of the X and Y (or Z and W) chromosomes, which may be difficult if they are recently diverged. RESULTS: Here we produce a haplotype-resolved genome assembly of zig-zag eel (Mastacembelus armatus), an aquaculture fish, at the chromosomal scale. The diploid assembly is nearly gap-free, and in most chromosomes, we resolve the centromeric and subtelomeric heterochromatic sequences. In particular, the Y chromosome, including its highly repetitive short arm, has zero gaps. Using resequencing data, we identify a ~7 Mb fully sex-linked region (SLR), spanning the sex chromosome centromere and almost entirely embedded in the pericentromeric heterochromatin. The SLRs on the X and Y chromosomes are almost identical in sequence and gene content, but both are repetitive and heterochromatic, consistent with zero or low recombination. We further identify an HMG-domain containing gene HMGN6 in the SLR as a candidate sex-determining gene that is expressed at the onset of testis development. CONCLUSIONS: Our study supports the idea that preexisting regions of low recombination, such as pericentromeric regions, can give rise to SLR in the absence of structural variations between the proto-sex chromosomes.


Assuntos
Enguias/genética , Genoma , Proteínas HMGN/genética , Processos de Determinação Sexual , Telômero , Cromossomo Y/química , Animais , Centrômero , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Expressão Gênica , Proteínas HMGN/metabolismo , Heterocromatina/química , Cariótipo , Masculino , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Cromossomo X
14.
Nat Commun ; 12(1): 3534, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112801

RESUMO

Metabolic diseases are associated with an increased risk of severe COVID-19 and conversely, new-onset hyperglycemia and complications of preexisting diabetes have been observed in COVID-19 patients. Here, we performed a comprehensive analysis of pancreatic autopsy tissue from COVID-19 patients using immunofluorescence, immunohistochemistry, RNA scope and electron microscopy and detected SARS-CoV-2 viral infiltration of beta-cells in all patients. Using SARS-CoV-2 pseudoviruses, we confirmed that isolated human islet cells are permissive to infection. In eleven COVID-19 patients, we examined the expression of ACE2, TMPRSS and other receptors and factors, such as DPP4, HMBG1 and NRP1, that might facilitate virus entry. Whereas 70% of the COVID-19 patients expressed ACE2 in the vasculature, only 30% displayed ACE2-expression in beta-cells. Even in the absence of manifest new-onset diabetes, necroptotic cell death, immune cell infiltration and SARS-CoV-2 viral infection of pancreatic beta-cells may contribute to varying degrees of metabolic dysregulation in patients with COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/patologia , Células Secretoras de Insulina/virologia , Receptores de Coronavírus/metabolismo , SARS-CoV-2/isolamento & purificação , Serina Endopeptidases/metabolismo , Adulto , Idoso , Autopsia , Complicações do Diabetes/patologia , Complicações do Diabetes/virologia , Diabetes Mellitus/patologia , Dipeptidil Peptidase 4/metabolismo , Feminino , Proteínas HMGN/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Pessoa de Meia-Idade , Neuropilina-1/metabolismo , Especificidade de Órgãos/fisiologia
15.
Eur Rev Med Pharmacol Sci ; 25(3): 1330-1338, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33629303

RESUMO

OBJECTIVE: To detect the expression of high-mobility group nucleosome-binding domain 5 (HMGN5) in colorectal cancer tissues, to explore the function of HMGN5 on the proliferation and metastasis of colorectal cancer cells, and to further study the molecular mechanism of HMGN5 in the malignant progression of colorectal cancer (CRC). PATIENTS AND METHODS: The cancer tissues and para-carcinoma tissues were harvested from 40 patients with CRC. The expression of HMGN5 was detected via quantitative real-time polymerase chain reaction (qRT-PCR), and the relation between HMGN5 and clinical indexes of CRC patients was further analyzed. The CRC HT29 and HCT116 cell lines with high expression levels of HMGN5 were selected, and the HMGN5 knockdown model was established. The functions of HMGN5 on CRC cells were stated by cell counting kit-8 (CCK-8) assay and transwell migration assay. Then, the association between HMGN5 and fibroblast growth factor 12 (FGF12) was further explored via Dual-Luciferase reporter assay and reverse assay. RESULTS: The qRT-PCR showed that HMGN5 expression was significantly rising in cancer tissues compared to the control group. The incidence rate of lymph node metastasis and distant metastasis was higher in higher expression HMGN5 group than the lower expression HMGN5 group. The results of cell function experiments revealed that silence of HMGN5 could suppress the proliferation and migration of HT29 and HCT116. In addition, it was found using qRT-PCR that knockdown of HMGN5 could significantly down-regulate the expressions of FGF12, FGFR, PI3K and AKT in HT29 and HCT116 cells. The targeted binding relation between HMGN5 and FGF12 was also indicated by the dual-luciferase reporter assay. The consequence of qRT-PCR manifested that FGF12 expression markedly rose in CRC tissues, which had a positive correlation with HMGN5. Moreover, reverse assay indicated that the inhibitory effect of HMGN5 knockdown on the malignant progression of CRC could be reversed by recombinant FGF12, indicating once again that there is a mutual regulatory effect between HMGN5 and FGF12. CONCLUSIONS: HMGN5 can increase the proliferative and migrative capacity of CRC cells via targeted binding to FGF12. In addition, clinical data analyses demonstrate that HMGN5 is intimately related to the incidence rate of lymph node metastasis and distant metastasis in patients with CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas HMGN/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transativadores/metabolismo , Linhagem Celular , Movimento Celular , Neoplasias Colorretais/patologia , Feminino , Fatores de Crescimento de Fibroblastos/genética , Proteínas HMGN/genética , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Fatores de Crescimento de Fibroblastos/genética , Transativadores/genética
16.
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
17.
Nucleic Acids Res ; 48(19): 10820-10831, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-32997109

RESUMO

DNA binding proteins rapidly locate their specific DNA targets through a combination of 3D and 1D diffusion mechanisms, with the 1D search involving bidirectional sliding along DNA. However, even in nucleosome-free regions, chromosomes are highly decorated with associated proteins that may block sliding. Here we investigate the ability of the abundant chromatin-associated HMGB protein Nhp6A from Saccharomyces cerevisiae to travel along DNA in the presence of other architectural DNA binding proteins using single-molecule fluorescence microscopy. We observed that 1D diffusion by Nhp6A molecules is retarded by increasing densities of the bacterial proteins Fis and HU and by Nhp6A, indicating these structurally diverse proteins impede Nhp6A mobility on DNA. However, the average travel distances were larger than the average distances between neighboring proteins, implying Nhp6A is able to bypass each of these obstacles. Together with molecular dynamics simulations, our analyses suggest two binding modes: mobile molecules that can bypass barriers as they seek out DNA targets, and near stationary molecules that are associated with neighboring proteins or preferred DNA structures. The ability of mobile Nhp6A molecules to bypass different obstacles on DNA suggests they do not block 1D searches by other DNA binding proteins.


Assuntos
DNA/química , Proteínas HMGN/química , Proteínas de Saccharomyces cerevisiae/química , DNA/metabolismo , Proteínas HMGN/metabolismo , Simulação de Dinâmica Molecular , Movimento (Física) , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo , Imagem Individual de Molécula
18.
Biomed Res Int ; 2020: 8610271, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32596388

RESUMO

HMGN5 regulates biological function and molecular transcription via combining with a nucleosome. There has been growing evidence that aberrant expression of HMGN5 is associated with malignant neoplasm development and progression. In the present study, we found that the expression of HMGN5 is significantly higher in high-grade glioblastoma tissues than in low-grade samples. To clarify the function of HMGN5 in glioblastoma, we knocked down HMGN5 in U87 and U251 glioblastoma cells via siRNA. The results demonstrated that HMGN5 was involved in the regulation of proliferation and apoptosis, migration, and invasion of glioblastoma cells. These outcomes also indicated that silencing HMGN5 possibly suppressed the expression of p-AKT and p-ERK1/2. Taken together, our research reveals that HMGN5 might be an efficient target for glioblastoma-targeted therapy.


Assuntos
Glioblastoma , Proteínas HMGN , Sistema de Sinalização das MAP Quinases/genética , Transativadores , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Inativação Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Interferente Pequeno/genética , Transativadores/genética , Transativadores/metabolismo
19.
Aging (Albany NY) ; 12(8): 7282-7298, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32315283

RESUMO

Bladder cancer (BC) is one of the most common cancers worldwide, with a high rate of recurrence and poor outcomes. High-mobility group nucleosome-binding domain 5 (HMGN5) is overexpressed in many cancers and could cause carcinogenesis in BC. By protein-protein-interaction (PPI) analysis, we found that heat shock protein 27 (Hsp27), also a crucial functional factor in BC carcinogenesis, is significantly related to HMGN5. Hsp27 is required for IL-6-mediated EMT via STAT3/Twist signaling in prostate cancer. Here, we hypothesize that HMGN5 may interact with Hsp27 to affect IL-6-induced EMT and invasion in BC via STAT3 signaling. In the present study, we found that HMGN5 and Hsp27 are highly expressed in BC tissues and positively correlated with each other. HMGN5 interacts with Hsp27 in vitro, to modulate the cell invasion and EMT in BC. Moreover, HMGN5 could modulate IL-6-Hsp27-induced EMT and invasion in BC cells by regulating STAT3 phosphorylation and STAT3 targeting of the Twist promoter. HMGN5 interacts with Hsp27 to promote tumor growth in a human BC xenograft model in nude mice. In summary, HMGN5 interacts with Hsp27 to promote IL-6-induced EMT, therefore promoting invasion in BC and contributing to the progression of BC.


Assuntos
Carcinoma de Células de Transição/genética , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Proteínas HMGN/genética , Proteínas de Choque Térmico HSP27/genética , Interleucina-6/genética , Neoplasias da Bexiga Urinária/genética , Animais , Carcinoma de Células de Transição/metabolismo , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Proteínas HMGN/biossíntese , Proteínas de Choque Térmico HSP27/biossíntese , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Nus , Neoplasias Experimentais , Transdução de Sinais , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
20.
Int J Mol Sci ; 21(2)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936777

RESUMO

Chromatin plays a key role in regulating gene expression programs necessary for the orderly progress of development and for preventing changes in cell identity that can lead to disease. The high mobility group N (HMGN) is a family of nucleosome binding proteins that preferentially binds to chromatin regulatory sites including enhancers and promoters. HMGN proteins are ubiquitously expressed in all vertebrate cells potentially affecting chromatin function and epigenetic regulation in multiple cell types. Here, we review studies aimed at elucidating the biological function of HMGN proteins, focusing on their possible role in vertebrate development and the etiology of disease. The data indicate that changes in HMGN levels lead to cell type-specific phenotypes, suggesting that HMGN optimize epigenetic processes necessary for maintaining cell identity and for proper execution of specific cellular functions. This manuscript contains tables that can be used as a comprehensive resource for all the English written manuscripts describing research aimed at elucidating the biological function of the HMGN protein family.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/fisiologia , Animais , Cromatina , Doença , Proteínas HMGN , Proteínas de Grupo de Alta Mobilidade/classificação , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Camundongos , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...