Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Kidney Int ; 104(1): 163-180, 2023 07.
Article in English | MEDLINE | ID: mdl-37088425

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by autoreactive B cells and dysregulation of many other types of immune cells including myeloid cells. Lupus nephritis (LN) is a common target organ manifestations of SLE. Tonicity-responsive enhancer-binding protein (TonEBP, also known as nuclear factor of activated T-cells 5 (NFAT5)), was initially identified as a central regulator of cellular responses to hypertonic stress and is a pleiotropic stress protein involved in a variety of immunometabolic diseases. To explore the role of TonEBP, we examined kidney biopsy samples from patients with LN. Kidney TonEBP expression was found to be elevated in these patients compared to control patients - in both kidney cells and infiltrating immune cells. Kidney TonEBP mRNA was elevated in LN and correlated with mRNAs encoding inflammatory cytokines and the degree of proteinuria. In a pristane-induced SLE model in mice, myeloid TonEBP deficiency blocked the development of SLE and LN. In macrophages, engagement of various toll-like receptors (TLRs) that respond to damage-associated molecular patterns induced TonEBP expression via stimulation of its promoter. Intracellular signaling downstream of the TLRs was dependent on TonEBP. Therefore, TonEBP can act as a transcriptional cofactor for NF-κB, and activated mTOR-IRF3/7 via protein-protein interactions. Additionally, TonEBP-deficient macrophages displayed elevated efferocytosis and animals with myeloid deficiency of TonEBP showed reduced Th1 and Th17 differentiation, consistent with macrophages defective in TLR signaling. Thus, our data show that myeloid TonEBP may be an attractive therapeutic target for SLE and LN.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Mice , Kidney , Signal Transduction , Macrophages , NFATC Transcription Factors
2.
Exp Mol Med ; 53(10): 1602-1611, 2021 10.
Article in English | MEDLINE | ID: mdl-34697388

ABSTRACT

Transcription-replication conflicts lead to DNA damage and genomic instability, which are closely related to human diseases. A major source of these conflicts is the formation of R-loops, which consist of an RNA-DNA hybrid and a displaced single-stranded DNA. Although these structures have been studied, many aspects of R-loop biology and R-loop-mediated genome instability remain unclear. Here, we demonstrate that thyroid hormone receptor-associated protein 3 (Thrap3) plays a critical role in regulating R-loop resolution. In cancer cells, Thrap3 interacts with DEAD-box helicase 5 (DDX5) and localizes to R-loops. Arginine-mediated methylation of DDX5 is required for its interaction with Thrap3, and the Thrap3-DDX5 axis induces the recruitment of 5'-3' exoribonuclease 2 (XRN2) into R-loops. Loss of Thrap3 increases R-loop accumulation and DNA damage. These findings suggest that Thrap3 mediates resistance to cell death by preventing R-loop accumulation in cancer cells.


Subject(s)
R-Loop Structures , Transcription Factors , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , RNA , Transcription Factors/genetics
3.
DNA Repair (Amst) ; 104: 103132, 2021 08.
Article in English | MEDLINE | ID: mdl-34049076

ABSTRACT

Lack of coordination between the DNA replication and transcription machineries can increase the frequency of transcription-replication conflicts, leading ultimately to DNA damage and genomic instability. A major source of these conflicts is the formation of R-loops, which consist of a transcriptionally generated RNA-DNA hybrid and the displaced single-stranded DNA. R-loops play important physiological roles and have been implicated in human diseases. Although these structures have been extensively studied, many aspects of R-loop biology and R-loop-mediated genome instability remain unclear. We found that in cancer cells, tonicity-responsive enhancer-binding protein (TonEBP, also called NFAT5) interacted with PARP1 and localized to R-loops in response to DNA-damaging agent camptothecin (CPT), which is associated with R-loop formation. PARP1-mediated PARylation was required for recruitment of TonEBP to the sites of R-loop-associated DNA damage. Loss of TonEBP increased levels of R-loop accumulation and DNA damage, and promoted cell death in response to CPT. These findings suggest that TonEBP mediates resistance to CPT-induced cell death by preventing R-loop accumulation in cancer cells.


Subject(s)
DNA Damage , DNA Replication , Genomic Instability , Poly (ADP-Ribose) Polymerase-1/metabolism , R-Loop Structures , Transcription Factors/metabolism , Transcription, Genetic , Camptothecin/toxicity , Cell Line , DNA/metabolism , DNA, Single-Stranded/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Poly ADP Ribosylation
4.
Nucleic Acids Res ; 49(1): 269-284, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33313823

ABSTRACT

R-loops are three-stranded, RNA-DNA hybrid, nucleic acid structures produced due to inappropriate processing of newly transcribed RNA or transcription-replication collision (TRC). Although R-loops are important for many cellular processes, their accumulation causes genomic instability and malignant diseases, so these structures are tightly regulated. It was recently reported that R-loop accumulation is resolved by methyltransferase-like 3 (METTL3)-mediated m6A RNA methylation under physiological conditions. However, it remains unclear how R-loops in the genome are recognized and induce resolution signals. Here, we demonstrate that tonicity-responsive enhancer binding protein (TonEBP) recognizes R-loops generated by DNA damaging agents such as ultraviolet (UV) or camptothecin (CPT). Single-molecule imaging and biochemical assays reveal that TonEBP preferentially binds a R-loop via both 3D collision and 1D diffusion along DNA in vitro. In addition, we find that TonEBP recruits METTL3 to R-loops through the Rel homology domain (RHD) for m6A RNA methylation. We also show that TonEBP recruits RNaseH1 to R-loops through a METTL3 interaction. Consistent with this, TonEBP or METTL3 depletion increases R-loops and reduces cell survival in the presence of UV or CPT. Collectively, our results reveal an R-loop resolution pathway by TonEBP and m6A RNA methylation by METTL3 and provide new insights into R-loop resolution processes.


Subject(s)
Adenosine/analogs & derivatives , DNA Replication/genetics , Methyltransferases/physiology , R-Loop Structures/genetics , Transcription Factors/physiology , Adenosine/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA Adducts/metabolism , DNA Damage , Diffusion , HEK293 Cells , Humans , Methylation , Protein Binding , Protein Interaction Mapping , R-Loop Structures/radiation effects , Ribonuclease H/physiology , Ultraviolet Rays
5.
Cells ; 9(9)2020 08 20.
Article in English | MEDLINE | ID: mdl-32825390

ABSTRACT

The endoplasmic reticulum (ER) stress response and autophagy are important cellular responses that determine cell fate and whose dysregulation is implicated in the perturbation of homeostasis and diseases. Tonicity-responsive enhancer-binding protein (TonEBP, also called NFAT5) is a pleiotropic stress protein that mediates both protective and pathological cellular responses. Here, we examined the role of TonEBP in ß-cell survival under ER stress. We found that TonEBP increases ß-cell survival under ER stress by enhancing autophagy. The level of TonEBP protein increased under ER stress due to a reduction in its degradation via the ubiquitin-proteasome pathway. In response to ER stress, TonEBP increased autophagosome formations and suppressed the accumulation of protein aggregates and ß-cell death. The Rel-homology domain of TonEBP interacted with FIP200, which is essential for the initiation of autophagy, and was required for autophagy and cell survival upon exposure to ER stress. Mice in which TonEBP was specifically deleted in pancreatic endocrine progenitor cells exhibited defective glucose homeostasis and a loss of islet mass. Taken together, these findings demonstrate that TonEBP protects against ER stress-induced ß-cell death by enhancing autophagy.


Subject(s)
Endoplasmic Reticulum Stress/physiology , NFATC Transcription Factors/metabolism , Autophagy , Cell Survival , Humans
6.
EBioMedicine ; 58: 102926, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32739873

ABSTRACT

BACKGROUND: High recurrence and chemoresistance drive the high mortality in hepatocellular carcinoma (HCC). Although cancer stem cells are considered to be the source of recurrent and chemoresistant tumors, they remain poorly defined in HCC. Tonicity-responsive enhancer binding protein (TonEBP) is elevated in almost all HCC tumors and associated with recurrence and death. We aimed to identify function of TonEBP in stemness and chemoresistance of liver cancer. METHODS: Tumors obtained from 280 HCC patients were analyzed by immunohistochemical analyses. Stemness and chemoresistance of liver CSCs (LCSCs) were investigated using cell culture. Tumor-initiating activity was measured by implanting LCSCs into BALB/c nude mice. FINDINGS: Expression of TonEBP is higher in LCSCs in HCC cell lines and correlated with markers of LCSCs whose expression is significantly associated with poor prognosis of HCC patients. TonEBP mediates ATM-mediated activation of NF-κB, which stimulates the promoter of a key stem cell transcription factor SOX2. As expected, TonEBP is required for the tumorigenesis and self-renewal of LSCSs. Cisplatin induces the recruitment of the ERCC1/XPF dimer to the chromatin in a TonEBP-dependent manner leading to DNA repair and cisplatin resistance. The cisplatin-induced inflammation in LSCSs is also dependent on the TonEBP-ERCC1/XPF complex, and leads to enhanced stemness via the ATM-NF-κB-SOX2 pathway. In HCC patients, tumor expression of ERCC1/XPF predicts recurrence and death in a TonEBP-dependent manner. INTERPRETATION: TonEBP promotes stemness and cisplatin resistance of HCC via ATM-NF-κB. TonEBP is a key regulator of LCSCs and a promising therapeutic target for HCC and its recurrence.


Subject(s)
Carcinoma, Hepatocellular/pathology , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Endonucleases/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Transcription Factors/genetics , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplastic Stem Cells/metabolism , Prognosis , Up-Regulation , Xenograft Model Antitumor Assays
7.
Cell Death Dis ; 11(6): 421, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499518

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells that link the innate and adaptive immune responses; as such they play pivotal roles in initiation and progression of rheumatoid arthritis (RA). Here, we report that the tonicity-responsive enhancer-binding protein (TonEBP or NFAT5), a Rel family protein involved in the pathogenesis of autoimmune disease and inflammation, is required for maturation and function of DCs. Myeloid cell-specific TonEBP deletion reduces disease severity in a murine model of collagen-induced arthritis; it also inhibits maturation of DCs and differentiation of pathogenic Th1 and Th17 cells in vivo. Upon stimulation by TLR4, TonEBP promotes surface expression of major histocompatibility complex class II and co-stimulatory molecules via p38 mitogen-activated protein kinase. This is followed by DC-mediated differentiation of pro-inflammatory Th1 and Th17 cells. Taken together, these findings provide mechanistic basis for the pathogenic role of TonEBP in RA and possibly other autoimmune diseases.


Subject(s)
Dendritic Cells/metabolism , Inflammation/immunology , NFATC Transcription Factors/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Differentiation/immunology , Cell Proliferation , Disease Models, Animal , Lipopolysaccharides , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Models, Biological , Myeloid Cells/metabolism , NFATC Transcription Factors/deficiency , Severity of Illness Index , T-Lymphocytes/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Cells ; 8(10)2019 10 20.
Article in English | MEDLINE | ID: mdl-31635160

ABSTRACT

TonEBP (tonicity-responsive enhancer binding protein) is a transcriptional regulator whose expression is elevated in response to various forms of stress including hyperglycemia, inflammation, and hypoxia. Here we investigated the role of TonEBP in acute kidney injury (AKI) using a line of TonEBP haplo-deficient mice subjected to bilateral renal ischemia followed by reperfusion (I/R). In the TonEBP haplo-deficient animals, induction of TonEBP, oxidative stress, inflammation, cell death, and functional injury in the kidney in response to I/R were all reduced. Analyses of renal transcriptome revealed that genes in several cellular pathways including peroxisome and mitochondrial inner membrane were suppressed in response to I/R, and the suppression was relieved in the TonEBP deficiency. Production of reactive oxygen species (ROS) and the cellular injury was reproduced in a renal epithelial cell line in response to hypoxia, ATP depletion, or hydrogen peroxide. The knockdown of TonEBP reduced ROS production and cellular injury in correlation with increased expression of the suppressed genes. The cellular injury was also blocked by inhibitors of necrosis. These results demonstrate that ischemic insult suppresses many genes involved in cellular metabolism leading to local oxidative stress by way of TonEBP induction. Thus, TonEBP is a promising target to prevent AKI.


Subject(s)
Acute Kidney Injury/metabolism , NFATC Transcription Factors/metabolism , Acute Kidney Injury/genetics , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Peroxisomes/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
9.
iScience ; 19: 177-190, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31376680

ABSTRACT

Polyubiquitination of proliferating cell nuclear antigen (PCNA) regulates the error-free template-switching mechanism for the bypass of DNA lesions during DNA replication. PCNA polyubiquitination is critical for the maintenance of genomic integrity; however, the underlying mechanism is poorly understood. Here, we demonstrate that tonicity-responsive enhancer-binding protein (TonEBP) regulates PCNA polyubiquitination in response to DNA damage. TonEBP was recruited to DNA damage sites with bulky adducts and sequentially recruited E3 ubiquitin ligase SHPRH, followed by deubiquitinase USP1, to DNA damage sites, in correlation with the dynamics of PCNA polyubiquitination. Similarly, TonEBP was found to be required for replication fork protection in response to DNA damage. The Rel-homology domain of TonEBP, which encircles DNA, was essential for the interaction with SHPRH and USP1, PCNA polyubiquitination, and cell survival after DNA damage. The present findings suggest that TonEBP is an upstream regulator of PCNA polyubiquitination and of the DNA damage bypass pathway.

10.
Nat Commun ; 10(1): 3536, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31387996

ABSTRACT

Tonicity-responsive enhancer binding protein (TonEBP or NFAT5) is a regulator of cellular adaptation to hypertonicity, macrophage activation and T-cell development. Here we report that TonEBP is an epigenetic regulator of thermogenesis and obesity. In mouse subcutaneous adipocytes, TonEBP expression increases > 50-fold in response to high-fat diet (HFD) feeding. Mice with TonEBP haplo-deficiency or adipocyte-specific TonEBP deficiency are resistant to HFD-induced obesity and metabolic defects (hyperglycemia, hyperlipidemia, and hyperinsulinemia). They also display increased oxygen consumption, resistance to hypothermia, and beiging of subcutaneous fat tissues. TonEBP suppresses the promoter of ß3-adrenoreceptor gene, a critical regulator of lipolysis and thermogenesis, in ex vivo and cultured adipocytes. This involves recruitment of DNMT1 DNA methylase and methylation of the promoter. In human subcutaneous adipocytes TonEBP expression displays a correlation with body mass index but an inverse correlation with ß3-adrenoreceptor expression. Thus, TonEBP is an attractive therapeutic target for obesity, insulin resistance, and hyperlipidemia.


Subject(s)
Epigenesis, Genetic , Insulin Resistance/genetics , Obesity/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/metabolism , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/metabolism , Animals , Body Mass Index , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/etiology , Primary Cell Culture , Receptors, Adrenergic, beta-3/metabolism , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Thermogenesis/genetics , Transcription Factors/genetics
11.
Front Immunol ; 10: 850, 2019.
Article in English | MEDLINE | ID: mdl-31057560

ABSTRACT

TonEBP is a key transcriptional activator in macrophages with an M1 phenotype. High expression of TonEBP is associated with many inflammatory diseases. Heme oxygenase-1 (HO-1), a stress-inducible protein, is induced by various oxidative and inflammatory signals, and its expression is regarded as an adaptive cellular response to inflammation and oxidative injury. Here, we show that TonEBP suppresses expression of HO-1 by blocking Nrf2 binding to the HO-1 promoter, thereby inducing polarization of macrophages to the M1 phenotype. Inhibition of HO-1 expression or activity significantly reduced the inhibitory responses on M1 phenotype and stimulatory effects on M2 phenotype by TonEBP knockdown. Additional experiments showed that HO-1 plays a role in the paracrine anti-inflammatory effects of TonEBP knockdown in macrophages. Identification of HO-1 as a downstream effector of TonEBP provides new possibilities for improved therapeutic approaches to inflammatory diseases.


Subject(s)
Heme Oxygenase-1/genetics , Membrane Proteins/genetics , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Animals , Humans , Inflammation/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype
12.
Gut ; 68(2): 347-358, 2019 02.
Article in English | MEDLINE | ID: mdl-29420225

ABSTRACT

OBJECTIVES: Hepatocellular carcinoma (HCC) is a common cancer with high rate of recurrence and mortality. Diverse aetiological agents and wide heterogeneity in individual tumours impede effective and personalised treatment. Tonicity-responsive enhancer-binding protein (TonEBP) is a transcriptional cofactor for the expression of proinflammatory genes. Although inflammation is intimately associated with the pathogenesis of HCC, the role of TonEBP is unknown. We aimed to identify function of TonEBP in HCC. DESIGN: Tumours with surrounding hepatic tissues were obtained from 296 patients with HCC who received completion resection. TonEBP expression was analysed by quantitative reverse transcription-quantitative real-time PCR (RT-PCR) and immunohfistochemical analyses of tissue microarrays. Mice with TonEBP haplodeficiency, and hepatocyte-specific and myeloid-specific TonEBP deletion were used along with HCC and hepatocyte cell lines. RESULTS: TonEBP expression is higher in tumours than in adjacent non-tumour tissues in 92.6% of patients with HCC regardless of aetiology associated. The TonEBP expression in tumours and adjacent non-tumour tissues predicts recurrence, metastasis and death in multivariate analyses. TonEBP drives the expression of cyclo-oxygenase-2 (COX-2) by stimulating the promoter. In mouse models of HCC, three common sites of TonEBP action in response to diverse aetiological agents leading to tumourigenesis and tumour growth were found: cell injury and inflammation, induction by oxidative stress and stimulation of the COX-2 promoter. CONCLUSIONS: TonEBP is a key component of the common pathway in tumourigenesis and tumour progression of HCC in response to diverse aetiological insults. TonEBP is involved in multiple steps along the pathway, rendering it an attractive therapeutic target as well as a prognostic biomarker.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Transcription Factors/metabolism , Animals , Cyclooxygenase 2/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Oxidative Stress , Predictive Value of Tests , Republic of Korea , Survival Rate
13.
Sci Rep ; 6: 25726, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27160066

ABSTRACT

TonEBP is a key transcriptional activator of M1 phenotype in macrophage, and its high expression is associated with many inflammatory diseases. During the progression of the inflammatory responses, the M1 to M2 phenotypic switch enables the dual role of macrophages in controlling the initiation and resolution of inflammation. Here we report that in human and mouse M1 macrophages TonEBP suppresses IL-10 expression and M2 phenotype. TonEBP knockdown promoted the transcription of the IL-10 gene by enhancing chromatin accessibility and Sp1 recruitment to its promoter. The enhanced expression of M2 genes by TonEBP knockdown was abrogated by antagonism of IL-10 by either neutralizing antibodies or siRNA-mediated silencing. In addition, pharmacological suppression of TonEBP leads to similar upregulation of IL-10 and M2 genes. Thus, TonEBP suppresses M2 phenotype via downregulation of the IL-10 in M1 macrophages.


Subject(s)
Immunomodulation , Interleukin-10/metabolism , NFATC Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Chromatin/metabolism , Humans , Immunomodulation/drug effects , Interleukin-10/genetics , Interleukin-4/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Models, Biological , NFATC Transcription Factors/genetics , Naphthoquinones/pharmacology , Phenotype , Promoter Regions, Genetic/genetics , RAW 264.7 Cells , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Tissue Donors , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...