Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Pharmacol ; 218: 115859, 2023 12.
Article in English | MEDLINE | ID: mdl-37863326

ABSTRACT

Cutaneous melanoma is one of the most prevalent tumors, and it is still a huge challenge in the current clinical treatment. Isoliquiritigenin (ISL), which is isolated from Glycyrrhiza uralensis Fisch., has been reported for its anti-tumor effect. However, the underlying mechanism and targets of ISL are still not be revealed clearly. In this study, differentiallyexpressedproteins were identified bylabel-free quantitative mass spectrometry. Two isoforms of the histone variant H2A.Z, including H2A.Z.1 and H2A.Z.2, were significantly down regulated after administration of ISL in melanoma. H2A.Z.1 was highly expressed in melanoma and correlated with poor prognosis of melanoma. The expression of H2A.Z was inhibited by ISL in a concentration-dependent manner. Overexpression of H2A.Z.1 in melanoma cell lines partly restored the repressed cell proliferation and cell cycle by ISL. Moreover, E2F1 was identified as one downstream target of H2A.Z.1, which was also highly expressed in melanoma and correlated with poor prognosis of melanoma. Furthermore, in vivo assays validated the inhibitory role of ISL in melanoma proliferation and the expression of H2A.Z.1 and E2F1.Aboveall,it is indicated that ISL inhibit melanoma proliferation via targeting H2A.Z.1-E2F1 pathway. These findings explain the anti-tumor mechanism of ISL and provide potential therapeutic targets for melanoma.


Subject(s)
Chalcones , Melanoma , Skin Neoplasms , Humans , Melanoma/metabolism , Histones , Skin Neoplasms/drug therapy , Cell Line, Tumor , Chalcones/pharmacology , Chalcones/therapeutic use , E2F1 Transcription Factor , Melanoma, Cutaneous Malignant
2.
Biochem Biophys Res Commun ; 611: 118-125, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35525100

ABSTRACT

Radiation resistance reduces patient survival and is an important challenge in treating lung adenocarcinoma (LUAD). Previous studies have shown that histone H2A variants can affect the radiosensitivity of tumors; however, the main role of histone H2A variants in LUAD remains unclear. Using the TCGA database, we found that histone H2A variant H2A.Z.1 is positively associated with the progression and poor prognosis of LUAD. Colony formation, scratch wound-healing, and transwell assays as well as Western blot were performed to assess the role of H2A.Z.1 in vitro. Results suggested that H2A.Z.1 promoted cell migration and invasion, epithelial-mesenchymal transition, stemness, and radiation resistance in LUAD cells. Targeting H2A.Z.1 in combination with radiation therapy could be a potential therapeutic approach for radiation resistant LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/radiotherapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy
3.
Cells ; 9(5)2020 05 08.
Article in English | MEDLINE | ID: mdl-32397240

ABSTRACT

The replication independent (RI) histone H2A.Z is one of the more extensively studied variant members of the core histone H2A family, which consists of many replication dependent (RD) members. The protein has been shown to be indispensable for survival, and involved in multiple roles from DNA damage to chromosome segregation, replication, and transcription. However, its functional involvement in gene expression is controversial. Moreover, the variant in several groups of metazoan organisms consists of two main isoforms (H2A.Z-1 and H2A.Z-2) that differ in a few (3-6) amino acids. They comprise the main topic of this review, starting from the events that led to their identification, what is currently known about them, followed by further experimental, structural, and functional insight into their roles. Despite their structural differences, a direct correlation to their functional variability remains enigmatic. As all of this is being elucidated, it appears that a strong functional involvement of isoform variability may be connected to development.


Subject(s)
Histones/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cell Cycle , Chickens , Chromatin/metabolism , DNA Methylation , Histones/chemistry , Humans , Liver/metabolism , Male , Mice , Nucleosomes/metabolism , Osmolar Concentration , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Spermatogenesis
4.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28856239

ABSTRACT

The histone variant H2A.Z is an essential and conserved regulator of eukaryotic gene transcription. However, the exact role of this histone in the transcriptional process remains perplexing. In vertebrates, H2A.Z has two hypervariants, H2A.Z.1 and H2A.Z.2, that have almost identical sequences except for three amino acid residues. Due to such similarity, functional specificity of these hypervariants in neurobiological processes, if any, remain largely unknown. In this study with dissociated rat cortical neurons, we asked if H2A.Z hypervariants have distinct functions in regulating basal and activity-induced gene transcription. Hypervariant-specific RNAi and microarray analyses revealed that H2A.Z.1 and H2A.Z.2 regulate basal expression of largely nonoverlapping gene sets, including genes that code for several synaptic proteins. In response to neuronal activity, rapid transcription of our model gene Arc is impaired by depletion of H2A.Z.2, but not H2A.Z.1. This impairment is partially rescued by codepletion of the H2A.Z chaperone, ANP32E. In contrast, under a different context (after 48 h of tetrodotoxin, TTX), rapid transcription of Arc is impaired by depletion of either hypervariant. Such context-dependent roles of H2A.Z hypervariants, as revealed by our multiplexed gene expression assays, are also evident with several other immediate early genes, where regulatory roles of these hypervariants vary from gene to gene under different conditions. Together, our data suggest that H2A.Z hypervariants have context-specific roles that complement each other to mediate activity-induced neuronal gene transcription.


Subject(s)
Cytoskeletal Proteins/metabolism , Histones/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Epigenesis, Genetic , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/genetics , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Rats, Sprague-Dawley , Transcription, Genetic/physiology
5.
Oncotarget ; 7(10): 11412-23, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26863632

ABSTRACT

H2A.Z is a highly conserved H2A variant, and two distinct H2A.Z isoforms, H2A.Z.1 and H2A.Z.2, have been identified as products of two non-allelic genes, H2AFZ and H2AFV. H2A.Z has been reported to be overexpressed in breast, prostate and bladder cancers, but most studies did not clearly distinguish between isoforms. One recent study reported a unique role for the H2A.Z isoform H2A.Z.2 as a driver of malignant melanoma. Here we first report that H2A.Z.1 plays a pivotal role in the liver tumorigenesis by selectively regulating key molecules in cell cycle and epithelial-mesenchymal transition (EMT). H2AFZ expression was significantly overexpressed in a large cohort of hepatocellular carcinoma (HCC) patients, and high expression of H2AFZ was significantly associated with their poor prognosis. H2A.Z.1 overexpression was demonstrated in a subset of human HCC and cell lines. H2A.Z.1 knockdown suppressed HCC cell growth by transcriptional deregulation of cell cycle proteins and caused apoptotic cell death of HCC cells. We also observed that H2A.Z.1 knockdown reduced the metastatic potential of HCC cells by selectively modulating epithelial-mesenchymal transition regulatory proteins such as E-cadherin and fibronectin. In addition, H2A.Z.1 knockdown reduced the in vivo tumor growth rate in a mouse xenograft model. In conclusion, our findings suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis and that it plays established role in accelerating cell cycle transition and EMT during hepatocarcinogenesis. This makes H2A.Z.1 a promising target in liver cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Histones/genetics , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Transfection
6.
Biochem Biophys Rep ; 4: 28-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-29124184

ABSTRACT

Solution structures of nucleosomes containing a human histone variant, H2A.Z.1, were measured by small-angle X-ray and neutron scatterings (SAXS and SANS). SAXS revealed that the outer shape, reflecting the DNA shape, of the H2A.Z.1 nucleosome is almost the same as that of the canonical H2A nucleosome. In contrast, SANS employing a contrast variation technique revealed that the histone octamer of the H2A.Z.1 nucleosome is smaller than that of the canonical nucleosome. The DNA within the H2A.Z.1 nucleosome was more susceptible to micrococcal nuclease than that within the canonical nucleosome. These results suggested that the DNA is loosely wrapped around the histone core in the H2A.Z.1 nucleosome.

7.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 12): 2431-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24311584

ABSTRACT

The histone H2A.Z variant is widely conserved among eukaryotes. Two isoforms, H2A.Z.1 and H2A.Z.2, have been identified in vertebrates and may have distinct functions in cell growth and gene expression. However, no structural differences between H2A.Z.1 and H2A.Z.2 have been reported. In the present study, the crystal structures of nucleosomes containing human H2A.Z.1 and H2A.Z.2 were determined. The structures of the L1 loop regions were found to clearly differ between H2A.Z.1 and H2A.Z.2, although their amino-acid sequences in this region are identical. This structural polymorphism may have been induced by a substitution that evolutionally occurred at the position of amino acid 38 and by the flexible nature of the L1 loops of H2A.Z.1 and H2A.Z.2. It was also found that in living cells nucleosomal H2A.Z.1 exchanges more rapidly than H2A.Z.2. A mutational analysis revealed that the amino-acid difference at position 38 is at least partially responsible for the distinctive dynamics of H2A.Z.1 and H2A.Z.2. These findings provide important new information for understanding the differences in the regulation and functions of H2A.Z.1 and H2A.Z.2 in cells.


Subject(s)
Histones/chemistry , Amino Acid Sequence , Crystallography, X-Ray , DNA Mutational Analysis , Fluorescence Recovery After Photobleaching , HeLa Cells , Histones/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nucleosomes/chemistry , Nucleosomes/genetics , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL