Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
J Dent Res ; 103(1): 51-61, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37950483

RESUMEN

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.


Asunto(s)
Proteínas del Esmalte Dental , Proteína HMGN1 , Proteína HMGN2 , Animales , Ratones , Ameloblastos/metabolismo , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Proteína HMGN1/genética , Proteína HMGN1/metabolismo , Epigénesis Genética , Diferenciación Celular/genética , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Factores de Transcripción/metabolismo , Proteínas del Esmalte Dental/genética , Proteínas del Esmalte Dental/metabolismo , Cromatina/metabolismo , Amelogenina/metabolismo
2.
Nat Commun ; 13(1): 7303, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36435799

RESUMEN

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.


Asunto(s)
Tejido Adiposo Pardo , Nucleosomas , Masculino , Animales , Ratones , Nucleosomas/metabolismo , Tejido Adiposo Pardo/metabolismo , Proteínas HMGN/metabolismo , Epigénesis Genética , Fibroblastos/metabolismo , Tejido Adiposo Blanco/metabolismo , Adipocitos Marrones/metabolismo , Metabolismo Energético/genética
3.
Mol Cancer Res ; 20(12): 1724-1738, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36066963

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Proteínas HMGN , Humanos , Ratones , Animales , Femenino , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Cromatina/genética , Línea Celular Tumoral , Proliferación Celular , Apoptosis/genética , Transactivadores/metabolismo , Neoplasias de la Mama/genética , Factor de Transcripción STAT3/genética , Carcinogénesis/genética
4.
Epigenetics Chromatin ; 15(1): 23, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35761366

RESUMEN

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.


Asunto(s)
Cromatina , Proteínas HMGN , Epigénesis Genética , Proteínas HMGN/química , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Nucleosomas , Unión Proteica
5.
FASEB J ; 36(7): e22345, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35635715

RESUMEN

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.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Regulación de la Expresión Génica , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Humanos , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Commun Biol ; 5(1): 159, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197580

RESUMEN

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.


Asunto(s)
Proteínas HMGN , Nucleosomas , Cromatina/genética , Proteínas HMGN/química , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Nucleosomas/genética , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Genes (Basel) ; 12(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34946949

RESUMEN

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.


Asunto(s)
Cognición/fisiología , Síndrome de Down/genética , Proteínas HMGN/genética , Encéfalo/metabolismo , Mapeo Encefálico/métodos , Bases de Datos Genéticas , Síndrome de Down/metabolismo , Epigénesis Genética/genética , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Proteínas HMGN/metabolismo , Proteína HMGN1/genética , Proteína HMGN2/genética , Hipocampo/metabolismo , Humanos , Nucleosomas/genética , Corteza Prefrontal/metabolismo , Transactivadores/genética , Factores de Transcripción/genética , Transcriptoma/genética
8.
Sci Rep ; 11(1): 14165, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34239016

RESUMEN

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.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , ADN/metabolismo , Edición Génica , Ingeniería Genética , Proteína 9 Asociada a CRISPR/genética , Proteínas HMGN/metabolismo , Modelos Biológicos , Mutación/genética , ARN Guía de Kinetoplastida/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
Genome Biol ; 22(1): 203, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253240

RESUMEN

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.


Asunto(s)
Anguilas/genética , Genoma , Proteínas HMGN/genética , Procesos de Determinación del Sexo , Telómero , Cromosoma Y/química , Animales , Centrómero , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Expresión Génica , Proteínas HMGN/metabolismo , Heterocromatina/química , Cariotipo , Masculino , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Cromosoma X
10.
Nat Commun ; 12(1): 3534, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112801

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/patología , Células Secretoras de Insulina/virología , Receptores de Coronavirus/metabolismo , SARS-CoV-2/aislamiento & purificación , Serina Endopeptidasas/metabolismo , Adulto , Anciano , Autopsia , Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/virología , Diabetes Mellitus/patología , Dipeptidil Peptidasa 4/metabolismo , Femenino , Proteínas HMGN/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Neuropilina-1/metabolismo , Especificidad de Órganos/fisiología
11.
Eur Rev Med Pharmacol Sci ; 25(3): 1330-1338, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33629303

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas HMGN/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transactivadores/metabolismo , Línea Celular , Movimiento Celular , Neoplasias Colorrectales/patología , Femenino , Factores de Crecimiento de Fibroblastos/genética , Proteínas HMGN/genética , Humanos , Masculino , Persona de Mediana Edad , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transactivadores/genética
12.
Nucleic Acids Res ; 48(19): 10820-10831, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-32997109

RESUMEN

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.


Asunto(s)
ADN/química , Proteínas HMGN/química , Proteínas de Saccharomyces cerevisiae/química , ADN/metabolismo , Proteínas HMGN/metabolismo , Simulación de Dinámica Molecular , Movimiento (Física) , Unión Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo , Imagen Individual de Molécula
13.
Biomed Res Int ; 2020: 8610271, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32596388

RESUMEN

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.


Asunto(s)
Glioblastoma , Proteínas HMGN , Sistema de Señalización de MAP Quinasas/genética , Transactivadores , Animales , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Silenciador del Gen , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Interferente Pequeño/genética , Transactivadores/genética , Transactivadores/metabolismo
14.
Mol Biol (Mosk) ; 53(6): 1038-1048, 2019.
Artículo en Ruso | MEDLINE | ID: mdl-31876282

RESUMEN

Transcriptional enhancers in the cell nuclei typically interact with the target promoters in cis over long stretches of chromatin, but the mechanism of this communication remains unknown. Previously we have developed a defined in vitro system for quantitative analysis of the rate of distant enhancer-promoter communication (EPC) and have shown that the chromatin fibers maintain efficient distant EPC in cis. Here we investigate the roles of linker histone H1 and HMGN5 protein in EPC. A considerable negative effect of histone H1 on EPC depending on its C- and N-tails was shown. Protein HMGN5 that affects chromatin compaction and is associated with active chromatin counteracts EPC inhibition by H1. The data suggest that the efficiency of the interaction between the enhancer and the promoter depends on the structure and dynamics of the chromatin fiber localized between them and can be regulated by proteins associated with chromatin.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Proteínas HMGN/metabolismo , Histonas/metabolismo , Cromatina/química , Elementos de Facilitación Genéticos/genética , Regiones Promotoras Genéticas/genética
15.
DNA Cell Biol ; 38(8): 840-848, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31314587

RESUMEN

microRNAs are a class of noncoding RNAs that play important roles in cancer progression. microRNA-183-3p (miR-183-3p) is a novel microRNA that is dysregulated in many kinds of cancers. Our previous studies found high expression and oncologic role of high-mobility group nucleosome binding domain 5 (HMGN5) in prostate cancer. In this study, we found that miR-183-3p was downregulated in prostate cancer cells and primary tissues compared with normal controls. In addition, miR-183-3p expression was negatively correlated with HMGN5 expression. On the basis of bioinformatics predication and quantitative polymerase chain reaction and Western blot verification, it is demonstrated that miR-183-3p regulated HMGN5 expression. Luciferase reporter assay confirmed that miR-183-3p directly targeted the 3'-untranslated region of HMGN5. Interestingly, cell proliferation and migration inhibition and apoptosis induction were also observed in miR-183-3p transfected human prostate cancer VCap and C4-2 cells. Moreover, overexpression of HMGN5 significantly reversed the inhibitory effect on cell proliferation and migration and promoted effect on cell apoptosis by miR-183-3p. Our data suggest that dysfunction of miR-183-3p-HMGN5 axis plays an oncogenic role and can be a therapeutic target for prostate cancer.


Asunto(s)
Proteínas HMGN/genética , MicroARNs/genética , Neoplasias de la Próstata/genética , Transactivadores/genética , Regiones no Traducidas 3' , Anciano , Apoptosis/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteínas HMGN/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Neoplasias de la Próstata/patología , Transactivadores/metabolismo
16.
Nucleic Acids Res ; 47(6): 2871-2883, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30698746

RESUMEN

The yeast Nhp6A protein (yNhp6A) is a member of the eukaryotic HMGB family of chromatin factors that enhance apparent DNA flexibility. yNhp6A binds DNA nonspecifically with nM affinity, sharply bending DNA by >60°. It is not known whether the protein binds to unbent DNA and then deforms it, or if bent DNA conformations are 'captured' by protein binding. The former mechanism would be supported by discovery of conditions where unbent DNA is bound by yNhp6A. Here, we employed an array of conformational probes (FRET, fluorescence anisotropy, and circular dichroism) to reveal solution conditions in which an 18-base-pair DNA oligomer indeed remains bound to yNhp6A while unbent. In 100 mM NaCl, yNhp6A-bound DNA unbends as the temperature is raised, with no significant dissociation of the complex detected up to ∼45°C. In 200 mM NaCl, DNA unbending in the intact yNhp6A complex is again detected up to ∼35°C. Microseconds-resolved laser temperature-jump perturbation of the yNhp6a-DNA complex revealed relaxation kinetics that yielded unimolecular DNA bending/unbending rates on timescales of 500 µs-1 ms. These data provide the first direct observation of bending/unbending dynamics of DNA in complex with yNhp6A, suggesting a bind-then-bend mechanism for this protein.


Asunto(s)
ADN de Hongos/química , ADN de Hongos/metabolismo , Proteínas HMGN/química , Proteínas HMGN/metabolismo , Conformación de Ácido Nucleico , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Ensamble y Desensamble de Cromatina/genética , Transferencia Resonante de Energía de Fluorescencia , Proteínas HMGN/fisiología , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Estructura Cuaternaria de Proteína , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología
17.
Nucleic Acids Res ; 47(2): 666-678, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30445475

RESUMEN

Nucleosome disruption plays a key role in many nuclear processes including transcription, DNA repair and recombination. Here we combine atomic force microscopy (AFM) and optical tweezers (OT) experiments to show that high mobility group B (HMGB) proteins strongly disrupt nucleosomes, revealing a new mechanism for regulation of chromatin accessibility. We find that both the double box yeast Hmo1 and the single box yeast Nhp6A display strong binding preferences for nucleosomes over linker DNA, and both HMGB proteins destabilize and unwind DNA from the H2A-H2B dimers. However, unlike Nhp6A, Hmo1 also releases half of the DNA held by the (H3-H4)2 tetramer. This difference in nucleosome destabilization may explain why Nhp6A and Hmo1 function at different genomic sites. Hmo1 is enriched at highly transcribed ribosomal genes, known to be depleted of histones. In contrast, Nhp6A is found across euchromatin, pointing to a significant difference in cellular function.


Asunto(s)
Proteínas HMGN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Microscopía de Fuerza Atómica , Nucleosomas/química , Nucleosomas/ultraestructura , Pinzas Ópticas
18.
J Cell Physiol ; 234(4): 4851-4863, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30272824

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of death from cancer in the world. To comprehensively investigate the utility of microRNAs (miRNAs) and protein-encoding transcripts (messenger RNAs [mRNAs]) in HCC as potential biomarkers for early detection and diagnosis, we exhaustively mined genomic data from three available omics datasets (GEO, Oncomine, and TCGA), analyzed the overlaps among gene expression studies from 920 hepatocellular carcinoma samples and 508 healthy (or adjacent normal) liver tissue samples available from six laboratories, and identified 178 differentially expressed genes (DEGs) associated with HCC. Paired with miRNA and lncRNA data, we identified 23 core genes that were targeted by nine differentially expressed miRNAs and 21 HCC-specific lncRNAs. We further demonstrated that alterations in these 23 genes were quite frequent, with five genes altered in over 5% of the population. Patients with high levels of YWHAZ, ENAH, and HMGN4 tended to have high-grade tumors and shorter overall survival, suggesting that these genes could be promising candidate biomarkers for disease and poor prognosis in patients with HCC. Our comprehensive mRNA, miRNA, and lncRNA omics analyses from multiple independent datasets identified robust molecules that may be used as biomarkers for early HCC detection and diagnosis.


Asunto(s)
Proteínas 14-3-3/genética , Carcinoma Hepatocelular/genética , Proteínas HMGN/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteínas de Microfilamentos/genética , ARN Largo no Codificante/genética , Proteínas 14-3-3/metabolismo , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Proteínas HMGN/metabolismo , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/patología , Proteínas de Microfilamentos/metabolismo , Pronóstico , ARN Mensajero/genética
19.
PLoS Comput Biol ; 14(7): e1006362, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30052627

RESUMEN

The non-homologous end joining of a DNA double strand break is initiated by the MRE11-NBS1-RAD50 complex whose subunits are the first three proteins to arrive to the breakage site thereby making the recruitment time of MRE11, NBS1 and RAD50 essential for cell survival. In the present investigation, the nature of MRE11 and NBS1 transportation from the cytoplasm to the nucleus, hosting the damaged DNA strand, is hypothesized to be a passive diffusive process. The feasibility of such a mechanism is addressed through theoretical and computational approaches which permit establishing the characteristic recruitment time of MRE11 and NBS1 by the nucleus. A computational model of a cell is constructed from a set of biological parameters and the kinetic Monte Carlo algorithm is used to simulate the diffusing MRE11 and NBS1 particles as a random walk process. To accurately describe the experimented data, it is discovered that MRE11 and NBS1 should start diffusion from significantly different starting positions which suggests that diffusion might not be the only transport mechanism of repair protein recruitment to the DNA break.


Asunto(s)
Simulación por Computador , Reparación del ADN , Proteínas HMGN/metabolismo , Proteína Homóloga de MRE11/metabolismo , Transactivadores/metabolismo , Algoritmos , Roturas del ADN , Reparación del ADN por Unión de Extremidades , Difusión , Humanos , Modelos Teóricos , Método de Montecarlo
20.
J Biochem Mol Toxicol ; 31(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28914995

RESUMEN

Previous study has demonstrated that high mobility group nucleosome-binding domain 5 (HMGN5) is involved in tumorigenesis and the development of multidrug resistance in several human cancers. However, the role of HMGN5 in esophageal squamous cell carcinoma (ESCC) remains unclear. Here, we showed that HMGN5 was significantly upregulated in ESCC cells. Knockdown of HMGN5 significantly inhibited cell growth and induced cell apoptosis of ESCC cells. Moreover, knockdown of HMGN5 increased the sensitivity of ESCC cells towards cisplatin. By contrast, overexpression of HMGN5 showed the opposite effects. Further experiments demonstrated that HMGN5 regulated the expression of multidrug resistance 1, cyclin B1, and Bcl-2. Overall, our results reveal that HMGN5 promotes tumor progression of ESCC and is also an important regulator of chemoresistance. Our study suggests that inhibition of HMGN5 may be a potential strategy for improving effectiveness of ESCC treatment.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Cisplatino/farmacología , Neoplasias Esofágicas/metabolismo , Proteínas HMGN/genética , Transactivadores/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HMGN/metabolismo , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA