Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38587186

ABSTRACT

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.

2.
Int J Cancer ; 151(2): 275-286, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35239184

ABSTRACT

Radiotherapy can induce various adverse effects including fibrosis in cancer patients. Radiation-induced aberrant expression of profibrotic genes has been associated with dysregulated epigenetic mechanisms. Pan-BET (bromodomain and extraterminal domain) inhibitors, such as JQ1 and I-BET151, have been reported to attenuate the profibrotic response after irradiation. Despite their profound preclinical efficacy, the clinical utility of pan-inhibitors is limited due to observed cytotoxicicities. Recently, inhibitors were developed that selectively target the first (BD1) and second (BD2) bromodomain of the BET proteins (iBET-BD1 [GSK778] and iBET-BD2 [GSK046]). Here, their potential to attenuate radiation-induced fibroblast activation with low-toxicity was investigated. Our results indicated that cell proliferation and cell cycle progression in fibroblasts from BJ cells and six donors were reduced when treated with I-BET151 and iBET-BD1, but not with iBET-BD2. After irradiation, induction of DGKA and profibrotic markers, especially COL1A1 and ACTA2, was attenuated with all BET inhibitors. H3K27ac enrichment was similar at the DGKA enhancer region after I-BET151 treatment and irradiation, but was reduced at the COL1A1 transcription start site and the ACTA2 enhancer site. iBET-BD2 did not change H3K27ac levels in these regions. BRD4 occupancy at these regions was not altered by any of the compounds. Cell migration activity was measured as a characteristic independent of extracellular matrix production and was unchanged in fibroblasts after irradiation and BET inhibitor-treatment. In conclusion, iBET-BD2 efficiently suppressed radiation-induced expression of DGKA and profibrotic markers without showing cytotoxicity. Thus BD2-selective targeting is a promising new therapeutic avenue for further investigations to prevent or attenuate radiotherapy-induced fibrosis.


Subject(s)
Antineoplastic Agents , Nuclear Proteins , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Nuclear Proteins/metabolism , Protein Domains , Transcription Factors/metabolism
3.
Cell Death Dis ; 13(2): 131, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136016

ABSTRACT

Worldwide, nasopharyngeal carcinoma (NPC) is a rare head and neck cancer; however, it is a common malignancy in southern China. Radiotherapy is the most important treatment strategy for NPC. However, although radiotherapy is a strong tool to kill cancer cells, paradoxically it also promotes aggressive phenotypes. Therefore, we mimicked the treatment process in NPC cells in vitro. Upon exposure to radiation, a subpopulation of NPC cells gradually developed resistance to radiation and displayed cancer stem-cell characteristics. Radiation-induced stemness largely depends on the accumulation of the antiapoptotic myeloid cell leukemia 1 (MCL-1) protein. Upregulated MCL-1 levels were caused by increased stability and more importantly, enhanced protein synthesis. We showed that repeated ionizing radiation resulted in persistently enhanced reactive oxygen species (ROS) production at a higher basal level, further promoting protein kinase B (AKT) signaling activation. Intracellular ROS and AKT activation form a positive feedback loop in the process of MCL-1 protein synthesis, which in turn induces stemness and radioresistance. AKT/MCL-1 axis inhibition attenuated radiation-induced resistance, providing a potential target to reverse radiation therapy-induced radioresistance.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein , Nasopharyngeal Neoplasms , Proto-Oncogene Proteins c-akt , Cell Line, Tumor , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/radiotherapy , Radiation Tolerance/genetics , Reactive Oxygen Species
4.
Nucleic Acids Res ; 49(20): 11666-11689, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718742

ABSTRACT

The inhibitor of DNA-binding 3 (ID3) is a transcriptional regulator that limits interaction of basic helix-loop-helix transcription factors with their target DNA sequences. We previously reported that ID3 loss is associated with mutational signatures linked to DNA repair defects. Here we demonstrate that ID3 exhibits a dual role to promote DNA double-strand break (DSB) repair, particularly homologous recombination (HR). ID3 interacts with the MRN complex and RECQL helicase to activate DSB repair and it facilitates RAD51 loading and downstream steps of HR. In addition, ID3 promotes the expression of HR genes in response to ionizing radiation by regulating both chromatin accessibility and activity of the transcription factor E2F1. Consistently, analyses of TCGA cancer patient data demonstrate that low ID3 expression is associated with impaired HR. The loss of ID3 leads to sensitivity of tumor cells to PARP inhibition, offering new therapeutic opportunities in ID3-deficient tumors.


Subject(s)
Homologous Recombination , Inhibitor of Differentiation Proteins/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , Drug Resistance, Neoplasm , E2F1 Transcription Factor/metabolism , HEK293 Cells , Humans , Inhibitor of Differentiation Proteins/chemistry , Male , Neoplasm Proteins/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism
5.
Cancers (Basel) ; 13(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070078

ABSTRACT

Radiotherapy, a common component in cancer treatment, can induce adverse effects including fibrosis in co-irradiated tissues. We previously showed that differential DNA methylation at an enhancer of diacylglycerol kinase alpha (DGKA) in normal dermal fibroblasts is associated with radiation-induced fibrosis. After irradiation, the transcription factor EGR1 is induced and binds to the hypomethylated enhancer, leading to increased DGKA and pro-fibrotic marker expression. We now modulated this DGKA induction by targeted epigenomic and genomic editing of the DGKA enhancer and administering epigenetic drugs. Targeted DNA demethylation of the DGKA enhancer in HEK293T cells resulted in enrichment of enhancer-related histone activation marks and radiation-induced DGKA expression. Mutations of the EGR1-binding motifs decreased radiation-induced DGKA expression in BJ fibroblasts and caused dysregulation of multiple fibrosis-related pathways. EZH2 inhibitors (GSK126, EPZ6438) did not change radiation-induced DGKA increase. Bromodomain inhibitors (CBP30, JQ1) suppressed radiation-induced DGKA and pro-fibrotic marker expression. Similar drug effects were observed in donor-derived fibroblasts with low DNA methylation. Overall, epigenomic manipulation of DGKA expression may offer novel options for a personalized treatment to prevent or attenuate radiotherapy-induced fibrosis.

6.
Front Oncol ; 10: 737, 2020.
Article in English | MEDLINE | ID: mdl-32477950

ABSTRACT

Radiotherapy is an efficient tool in cancer treatment, but it brings along the risk of side effects such as fibrosis in the irradiated healthy tissue thus limiting tumor control and impairing quality of life of cancer survivors. Knowledge on radiation-related fibrosis risk and therapeutic options is still limited and requires further research. Recent studies demonstrated that epigenetic regulation of diacylglycerol kinase alpha (DGKA) is associated with radiation-induced fibrosis. However, the specific mechanisms are still unknown. In this review, we scrutinized the role of DGKA in the radiation response and in further cellular functions to show the potential of DGKA as a predictive marker or a novel target in fibrosis treatment. DGKA was reported to participate in immune response, lipid signaling, exosome production, and migration as well as cell proliferation, all processes which are suggested to be critical steps in fibrogenesis. Most of these functions are based on the conversion of diacylglycerol (DAG) to phosphatidic acid (PA) at plasma membranes, but DGKA might have also other, yet not well-known functions in the nucleus. Current evidence summarized here underlines that DGKA activation may play a central role in fibrosis formation post-irradiation and shows a potential of direct DGKA inhibitors or epigenetic modulators to attenuate pro-fibrotic reactions, thus providing novel therapeutic choices.

7.
Oncogene ; 38(3): 317-331, 2019 01.
Article in English | MEDLINE | ID: mdl-30097606

ABSTRACT

Aged hepatocyte-specific-Mcl-1 knockout (MKO-hep) mice are prone to develop liver tumors mimicking human hepatocellular carcinoma (HCC). Here we reported that a protein named UDP-N-acetylglucosamine pyrophosphorylase-1-like-1 (Uap1l1) is upregulated in the liver of young MKO-hep mice without any macroscopically detectable tumor nodules and is prominently expressed in the hepatic tumors developed in the aged MKO-hep mice. Intriguingly, human UAP1L1 is also significantly upregulated in a distinct subset of HCC tissues and patients with upregulated expression of UAP1L1 appeared to have poor prognosis. Overexpression of UAP1L1 significantly promoted, whereas UAP1L1 knockdown markedly reduced the proliferation of human hepatoma cells both in vitro and in vivo. UAP1L1 shows ~59% sequence identity to UDP-N-acetylglucosamine pyrophosphorylase-1 (UAP1), which is directly involved in the synthesis of the sugar donor (UDP-GlcNac) for N-acetylglucosamine modification (O-GlcNAcylation) of proteins. However, unlike UAP1, UAP1L1 harbors very limited UDP-GlcNAc synthesis activity. Moreover, although both UAP1 and UAP1L1 are required for O-GlcNAc transferase (OGT)-mediated protein O-GlcNAcylation, they appear to function distinctly from each other. UAP1L1 directly interacts with OGT, but does not seem to be an OGT substrate. In addition, UAP1L1 alone is not sufficient to activate OGT activity in vitro, suggesting that UAP1L1 may function together with other proteins to modulate OGT activity in vivo. Lastly, UAP1L1 knockdown attenuated c-MYC O-GlcNAcylation and protein stability, and overexpression of c-MYC significantly rescued the proliferation defect of UAP1L1 knockdown HepG2 cells, suggesting that c-MYC is one downstream target of UAP1L1 that contributes to UAP1L1-mediated cell proliferation, at least in HepG2 cells.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/enzymology , Neoplasm Proteins/physiology , Nucleotidyltransferases/metabolism , Protein Processing, Post-Translational/physiology , Animals , Carcinoma, Hepatocellular/pathology , Cell Division , Enzyme Activation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycosylation , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Mice , N-Acetylglucosaminyltransferases/metabolism , Nucleotidyltransferases/genetics , Prognosis , Protein Stability , Proto-Oncogene Proteins c-myc/metabolism , Transcriptome , Up-Regulation
8.
Sci Rep ; 7(1): 4613, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28676638

ABSTRACT

JAK2 activation is crucial for cytokine receptor signal transduction and leukemogenesis. However, the underlying processes that lead to full activation of JAK2 are unclear. Here, we report a positive role for ubiquitination of JAK2 during GM-CSF-induced activation. Upon GM-CSF stimulation, JAK2 ubiquitination is significantly enhanced through K63-linked poly-ubiquitination. Studies employing both knockout and overexpression of Cbl, an E3 ubiquitin ligase, led to the conclusion that Cbl specifically promotes JAK2 ubiquitination, and this was further confirmed in vitro using a Cbl ubiquitination assay. Moreover, following GM-CSF stimulation, the levels of phospho-JAK2 and -STAT5 and a STAT5 luciferase reporter assay were all reduced in Cbl knockout cells and this effect could be rescued by Cbl expression. Mechanistically, Cbl can interact with, and ubiquitinate JAK2 FERM and kinase domains via the Cbl TKB domain. Using lysine-to-arginine site-directed mutagenesis, K970 in the kinase domain of JAK2 was identified as the ubiquitination site important for promoting full JAK2 activation by Cbl via K63-conjugated poly-ubiquitination. Our study suggests that GM-CSF-induced JAK2 activation is enhanced by Cbl-mediated ubiquitination of JAK2. Targeting ubiquitination of JAK2 might offer a novel therapeutic strategy against JAK2-mediated disorders.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Janus Kinase 2/metabolism , Lysine/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Mutagenesis, Site-Directed , Phosphorylation/drug effects , Protein Binding , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
9.
Article in English | MEDLINE | ID: mdl-24023582

ABSTRACT

To evaluate the effectiveness of long-round needle therapy for pain relief in patients with knee osteoarthritis, 192 patients were included in a multicenter, randomized, controlled trial. 97 patients were randomized to the long-round needle therapy group (EG), and 95 patients were randomized to the control group (CG). In EG, the long-round needle therapy was performed once every 7 days for 3 therapy sessions. Ibuprofen sustained-release capsules were administered orally in CG, 1 pill each time, twice daily for 3 weeks. Curative effect was measured after the therapy and was evaluated at a 3-month follow-up interview. In EG, the treatment resulted in a basic cure for 79 patients, was effective for 15 patients, and was ineffective for 1 patient. In CG, the treatment resulted in a basic cure for 30 patients, was effective for 38 patients, and was ineffective for 21 patients. In the follow-up examination in EG, 75 patients were basically cured, and the treatment was effective for 11 patients and ineffective for 9. In CG, 22 were basically cured, 31 found the treatment effective, and 36 found the treatment ineffective. The curative effects in EG after both the treatment and the 3-mouth followup were significantly more superior than that in CG (P < 0.01) which should be adopted more widely.

10.
Biosci Biotechnol Biochem ; 77(8): 1682-8, 2013.
Article in English | MEDLINE | ID: mdl-23924730

ABSTRACT

Oxidative stress due to the over-production of reactive oxygen species (ROS) is associated with human skin aging. This study was designed to identify the bioactive phenolics in detoxified Rhus verniciflua Stokes (DRVS) that may protect human skin against oxidative stress. Under oxidative stress caused by H2O2, the 40% (v/v) aqueous methanol extract of DRVS protected human keratinocytes in a dose-dependent manner. The expression of matrix metalloproteinase-1 (MMP-1) was also inhibited by the DRVS extract in human dermal fibroblasts-neonatal cells exposed to ultraviolet A. The major bioactive phenolics of DRVS were tentatively identified by LC/Q-TOF-ESI-MS/MS, and included gallic acid, 2-(ethoxymethoxy)-3-hydroxyphenol, fustin, a fustin isomer, tetragalloyl glucose, pentagalloyl glucose, fisetin, sulfuretin, a sulfuretin isomer, and butein. The results suggest that a DRVS extract may be effective in slowing skin aging through its antioxidative properties and by down-regulating MMP-1 expression. Further studies are needed to examine whether this effect would be mediated by the phenolics identified in this study.


Subject(s)
Fibroblasts/drug effects , Keratinocytes/drug effects , Phenols/isolation & purification , Plant Extracts/pharmacology , Toxicodendron/chemistry , Cell Line , Fibroblasts/radiation effects , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/toxicity , Keratinocytes/radiation effects , Matrix Metalloproteinase 1/biosynthesis , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Phenols/chemistry , Phenols/pharmacology , Plant Extracts/chemistry , Reactive Oxygen Species , Tandem Mass Spectrometry , Ultraviolet Rays
11.
Cell Cycle ; 11(19): 3611-26, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22935703

ABSTRACT

It is well known that ligand binding to the high-affinity GM-CSF receptor (GMR) activates JAK2. However, how and where this event occurs in a cellular environment remains unclear. Here, we demonstrate that clathrin- but not lipid raft-mediated endocytosis is crucial for GMR signaling. Knockdown expression of clathrin heavy chain or intersectin 2 (ITSN2) attenuated GMR-mediated activation of JAK2, whereas inhibiting clathrin-coated pits or plagues to bud off the membrane by the dominant-negative mutant of dynamin enhanced such event. Moreover, unlike the wild-type receptor, an ITSN2-non-binding mutant of GMR defective in targeting to clathrin-coated pits or plagues [collectively referred to as clathrin-coated structures (CCSs) here] failed to activate JAK2 at such locations. Additional experiments demonstrate that ligand treatment not only enhanced JAK2/GMR association at CCSs, but also induced a conformational change of JAK2 which is required for JAK2 to be activated by CCS-localized CK2. Interestingly, ligand-independent activation of the oncogenic mutant of JAK2 (JAK2V617F) also requires the targeting of this mutant to CCSs. But JAK2V617F seems to be constitutively in an open conformation for CK2 activation. Together, this study reveals a novel functional role of CCSs in GMR signaling and the oncogenesis of JAK2V617F.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Janus Kinase 2/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Endocytosis , Enzyme Activation , HeLa Cells , Humans , Ligands , Mice , Models, Biological , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Conformation , Protein Transport , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Signal Transduction
12.
FEBS J ; 277(5): 1202-18, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121949

ABSTRACT

Snail is a multifunctional transcriptional factor that has been described as a repressor in many different contexts. It is also proposed as an activator in a few cases relevant to tumor progression and cell-cycle arrest. This study investigated the detailed mechanisms by which Snail upregulates gene expression of the CDK inhibitor p15(INK4b) in HepG2 induced by the tumor promoter tetradecanoyl phorbol acetate (TPA). Using deletion mapping, the TPA-responsive element on the p15(INK4b) promoter was located between 77 and 228 bp upstream of the transcriptional initiation site, within which the putative binding regions of early growth response gene 1 (EGR-1) and stimulatory protein 1 (SP-1) were found. Gene expression of EGR-1, Snail and SP-1 can be induced by TPA within 0.5-6 h. In addition, basal levels of SP-1, but not of the other two transcriptional factors, were observed. Blockade of TPA-induced gene expression of Snail, EGR-1 or SP-1 suppressed activation of the p15-pro228 reporter plasmid harboring the TPA-responsive element. More detailed deletion mapping and site-directed mutagenesis further concluded that the overlapping EGR-1/SP-1-binding site was required for TPA-induced p15-pro228 activation. In an EMSA, a DNA-protein complex was elevated by TPA, which can be blocked by antibodies against EGR-1, SP-1 or Snail at 6 h. Immunoprecipitation/western blotting demonstrated that TPA could trigger the association of EGR-1 with Snail or SP-1. Furthermore, a double chromatin immunoprecipitation assay verified that EGR-1 could form a complex with Snail or SP-1 on the TPA-responsive element after treatment with TPA for 2-6 h. Finally, we demonstrated a novel Snail-target region which could be bound by Snail and was also required for TPA-induced p15-pro228 activation. In conclusion, Snail associates with EGR-1 and SP-1 to mediate TPA-induced transcriptional upregulation of p15(INK4b) in HepG2.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/metabolism , Early Growth Response Protein 1/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation , Cyclin-Dependent Kinase Inhibitor p15/genetics , Early Growth Response Protein 1/genetics , Hep G2 Cells , Humans , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/metabolism , Snail Family Transcription Factors , Sp1 Transcription Factor/genetics , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...