Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(18): 10469-10486, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36155803

ABSTRACT

Human CtIP maintains genomic integrity primarily by promoting 5' DNA end resection, an initial step of the homologous recombination (HR). A few mechanisms have been suggested as to how CtIP recruitment to damage sites is controlled, but it is likely that we do not yet have full understanding of the process. Here, we provide evidence that CtIP recruitment and functioning are controlled by the SIAH2 E3 ubiquitin ligase. We found that SIAH2 interacts and ubiquitinates CtIP at its N-terminal lysine residues. Mutating the key CtIP lysine residues impaired CtIP recruitment to DSBs and stalled replication forks, DSB end resection, overall HR repair capacity of cells, and recovery of stalled replication forks, suggesting that the SIAH2-induced ubiquitination is important for relocating CtIP to sites of damage. Depleting SIAH2 consistently phenocopied these results. Overall, our work suggests that SIAH2 is a new regulator of CtIP and HR repair, and emphasizes that SIAH2-mediated recruitment of the CtIP is an important step for CtIP's function during HR repair.


Subject(s)
DNA Repair , DNA Replication , Endodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Endodeoxyribonucleases/genetics , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Cells ; 11(18)2022 09 08.
Article in English | MEDLINE | ID: mdl-36139389

ABSTRACT

CtBP-interacting protein (CtIP) plays a critical role in controlling the homologous recombination-mediated DNA double-stranded break (DSB) repair pathway through DNA end resection, and recent studies suggest that it also plays a role in mitosis. However, the mechanism by which CtIP contributes to mitosis regulation remains elusive. Here, we show that depletion of CtIP leads to a delay in anaphase progression resulting in misaligned chromosomes, an aberrant number of centrosomes, and defects in chromosome segregation. Additionally, we demonstrate that CtIP binds and colocalizes with Targeting protein for Xklp2 (TPX2) during mitosis to regulate the recruitment of TPX2 to the spindle poles. Furthermore, depletion of CtIP resulted in both a lower concentration of Aurora A, its downstream target, and very low microtubule intensity at the spindle poles, suggesting an important role for the CtIP-TPX2-Auroa A complex in microtubule dynamics at the centrosomal spindles. Our findings reveal a novel function of CtIP in regulating spindle dynamics through interactions with TPX2 and indicate that CtIP is involved in the proper execution of the mitotic program, where deregulation may lead to chromosomal instability.


Subject(s)
Nuclear Proteins , Spindle Apparatus , DNA/metabolism , Microtubules/metabolism , Mitosis , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism
3.
Cell Death Dis ; 13(6): 528, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35668076

ABSTRACT

The cell signaling factors EGFR, EphA2, and Ephexin1 are associated with lung and colorectal cancer and play an important role in tumorigenesis. Although the respective functional roles of EGFR and EphA2 are well known, interactions between these proteins and a functional role for the complex is not understood. Here, we showed that Ephexin1, EphA2, and EGFR are each expressed at higher levels in lung and colorectal cancer patient tissues, and binding of EGFR to EphA2 was associated with both increased tumor grade and metastatic cases in both cancer types. Treatment with Epidermal Growth Factor (EGF) induced binding of the RR domain of EGFR to the kinase domain of EphA2, and this binding was promoted by Ephexin1. Additionally, the AKT-mediated phosphorylation of EphA2 (at Ser897) promoted interactions with EGFR, pointing to the importance of this pathway. Two mutations in EGFR, L858R and T790M, that are frequently observed in lung cancer patients, promoted binding to EphA2, and this binding was dependent on Ephexin1. Our results indicate that the formation of a complex between EGFR, EphA2, and Ephexin1 plays an important role in lung and colorectal cancers, and that inhibition of this complex may be an effective target for cancer therapy.


Subject(s)
Colorectal Neoplasms , Lung Neoplasms , Receptor, EphA2 , Carcinogenesis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Receptor, EphA2/genetics , Receptor, EphA2/metabolism
4.
BMC Cancer ; 22(1): 552, 2022 May 16.
Article in English | MEDLINE | ID: mdl-35578316

ABSTRACT

BACKGROUND: Scavenger receptor class A member 3 (SCARA3) is decreased in prostate cancer and myeloma. However, functions of SCARA3 in various cancers remain unclear. In this study, we tried to evaluate the functional study of SCARA3 in lung cancer. METHODS: The expression level of SCARA3 in the TCGA-database, lung cancer tissue microarray and lung cancer cells and the prognosis of lung cancer patients were measured. Lung cancer tissue microarray was analyzed pathologically using immunohistochemistry, and quantitative analysis of SCARA3 in normal lung cells and lung cancer cells was analyzed using western blot analysis. Survival curves for lung cancer patients were prepared with the Kaplan-Meier method. Migration and invasion of SCARA3 overexpressed lung cancer cells were determined using a Transwell chamber system. Proliferation of lung cancer cells was determined based on cell viability assay using cell culture in vitro and a tumorigenicity model of BALB/C nude mouse in vivo. RESULTS: The expression of SCARA3 was abnormally reduced in TCGA-database, lung tissue microarray, and various lung cancer cells. However, overexpression of SCARA3 reduced the proliferation of lung cancer. The ability of SCARA3 to inhibit cancer cell proliferation was maintained even in vivo using a mouse xenograft model. In addition, overexpression of SCARA3 reduced migration and invasion ability of lung cancer cells and induced decreases of EMT markers such as ß-catenin, vimentin, and MMP9. We aimed to prove the role of SCARA3 in the treatment of Lung cancer, and shown that the expression level of SCARA3 is important in cancer treatment using cisplatin. The enhancement of the effect of cisplatin according to SCARA3 overexpression is via the AKT and JNK pathways. CONCLUSIONS: This study confirmed an abnormal decrease in SCARA3 in lung cancer. Overexpression of SCARA3 potently inhibited tumors in lung cancer and induced apoptosis by increasing sensitivity of lung cancer to cisplatin. These results suggest that SCARA3 is a major biomarker of lung cancer and that the induction of SCARA3 overexpression can indicate an effective treatment.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins c-akt , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/metabolism , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/metabolism , Scavenger Receptors, Class A , Signal Transduction
5.
Mol Cell Oncol ; 9(1): 2054263, 2022.
Article in English | MEDLINE | ID: mdl-35372672

ABSTRACT

RAD51 loading onto chromatin is a key step during the homologous recombination (HR) repair. We recently reported a new mode of RAD51 regulation, which is mediated by TOPORS E3 SUMO ligase and RAD51 SUMOylation. ATM/ATR-induced phosphorylation of TOPORS is necessary for this event, revealing a new role of these master DNA damage response kinases in HR repair.

6.
Cell Death Dis ; 13(4): 309, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35387978

ABSTRACT

The Hsp70-binding protein 1 (HspBP1) belongs to a family of co-chaperones that regulate Hsp70 activity and whose biological significance is not well understood. In the present study, we show that when HspBP1 is either knocked down or overexpressed in BRCA1-proficient breast cancer cells, there were profound changes in tumorigenesis, including anchorage-independent cell growth in vitro and in tumor formation in xenograft models. However, HspBP1 did not affect tumorigenic properties in BRCA1-deficient breast cancer cells. The mechanisms underlying HspBP1-induced tumor suppression were found to include interactions with BRCA1 and promotion of BRCA1-mediated homologous recombination DNA repair, suggesting that HspBP1 contributes to the suppression of breast cancer by regulating BRCA1 function and thereby maintaining genomic stability. Interestingly, independent of BRCA1 status, HspBP1 facilitates cell survival in response to ionizing radiation (IR) by interfering with the association of Hsp70 and apoptotic protease-activating factor-1. These findings suggest that decreased HspBP1 expression, a common occurrence in high-grade and metastatic breast cancers, leads to genomic instability and enables resistance to IR treatment.


Subject(s)
Breast Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Repair , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Recombinational DNA Repair
7.
Nucleic Acids Res ; 50(3): 1501-1516, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35061896

ABSTRACT

Homologous recombination (HR) is critical for error-free repair of DNA double-strand breaks. Chromatin loading of RAD51, a key protein that mediates the recombination, is a crucial step in the execution of the HR repair. Here, we present evidence that SUMOylation of RAD51 is crucial for the RAD51 recruitment to chromatin and HR repair. We found that topoisomerase 1-binding arginine/serine-rich protein (TOPORS) induces the SUMOylation of RAD51 at lysine residues 57 and 70 in response to DNA damaging agents. The SUMOylation was facilitated by an ATM-induced phosphorylation of TOPORS at threonine 515 upon DNA damage. Knockdown of TOPORS or expression of SUMOylation-deficient RAD51 mutants caused reduction in supporting normal RAD51 functions during the HR repair, suggesting the physiological importance of the modification. We found that the SUMOylation-deficient RAD51 reduces the association with its crucial binding partner BRCA2, explaining its deficiency in supporting the HR repair. These findings altogether demonstrate a crucial role for TOPORS-mediated RAD51 SUMOylation in promoting HR repair and genomic maintenance.


Subject(s)
Rad51 Recombinase , Recombinational DNA Repair , Chromatin , DNA/metabolism , DNA Damage , DNA Repair/genetics , Homologous Recombination , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Sumoylation
8.
Nucleic Acids Res ; 50(3): 1465-1483, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35037047

ABSTRACT

p53-binding protein 1 (53BP1) regulates the DNA double-strand break (DSB) repair pathway and maintains genomic integrity. Here we found that 53BP1 functions as a molecular scaffold for the nucleoside diphosphate kinase-mediated phosphorylation of ATP-citrate lyase (ACLY) which enhances the ACLY activity. This functional association is critical for promoting global histone acetylation and subsequent transcriptome-wide alterations in gene expression. Specifically, expression of a replication-dependent histone biogenesis factor, stem-loop binding protein (SLBP), is dependent upon 53BP1-ACLY-controlled acetylation at the SLBP promoter. This chain of regulation events carried out by 53BP1, ACLY, and SLBP is crucial for both quantitative and qualitative histone biogenesis as well as for the preservation of genomic integrity. Collectively, our findings reveal a previously unknown role for 53BP1 in coordinating replication-dependent histone biogenesis and highlight a DNA repair-independent function in the maintenance of genomic stability through a regulatory network that includes ACLY and SLBP.


Subject(s)
ATP Citrate (pro-S)-Lyase , Histones , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Acetylation , DNA Breaks, Double-Stranded , DNA Repair , Histones/genetics , Histones/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
9.
Cell Death Dis ; 12(11): 1013, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711817

ABSTRACT

ABSTRCT: Ephexin1 was reported to be highly upregulated by oncogenic Ras, but the functional consequences of this remain poorly understood. Here, we show that Ephexin1 is highly expressed in colorectal cancer (CRC) and lung cancer (LC) patient tissues. Knockdown of Ephexin1 markedly inhibited the cell growth of CRC and LC cells with oncogenic Ras mutations. Ephexin1 contributes to the positive regulation of Ras-mediated downstream target genes and promotes Ras-induced skin tumorigenesis. Mechanically, Akt phosphorylates Ephexin1 at Ser16 and Ser18 (pSer16/18) and pSer16/18 Ephexin1 then interacts with oncogenic K-Ras to promote downstream MAPK signaling, facilitating tumorigenesis. Furthermore, pSer16/18 Ephexin1 is associated with both an increased tumor grade and metastatic cases of CRC and LC, and those that highly express pSer16/18 exhibit poor overall survival rates. These data indicate that Ephexin1 plays a critical role in the Ras-mediated CRC and LC and pSer16/18 Ephexin1 might be an effective therapeutic target for CRC and LC.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Guanine Nucleotide Exchange Factors/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncogenes , Proto-Oncogene Proteins c-akt/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , MAP Kinase Signaling System , Male , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Prognosis , Protein Binding , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
10.
Oncotarget ; 11(21): 2024-2025, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32523656

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.10275.].

11.
Nat Commun ; 9(1): 2284, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29875365

ABSTRACT

This Article contains errors in Fig. 3, Fig. 4 and Fig. 7, for which we apologize. In Fig. 3, panel 'b', the 0.5 hour time point after Ku55933 treatment images were inadvertently replaced with duplicates of the 3 hour time point after Ku55933 treatment images in Fig. 3b. Additionally, in panel 'b', the 0.5 hour time point after Nu7026 treatment images were inadvertently replaced with duplicates of the 180 min time point after siMDC1 treatment images in Fig. 3d. In Fig. 4, panel 'g', RNF168 foci in U2OS cell images were inadvertently replaced with duplicates of RNF168 foci in HeLa cell images in Fig. 4f. In Fig. 7, panel 'b', the DAPI images 0.5 hours after IR under siID3 treatment were inadvertently replaced with DAPI images of a different field of view from the same experiment. Additionally, in panel 'i', the shID3 mock-treated GFP-ID3 cells image was inadvertently replace with duplications of the shID3 mock-treated GFP-ID3 cells image in Fig. 7g.

12.
Nat Commun ; 8(1): 903, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026069

ABSTRACT

MDC1 plays a critical role in the DNA damage response (DDR) by interacting directly with several factors including γ-H2AX. However, the mechanism by which MDC1 is recruited to damaged sites remains elusive. Here, we show that MDC1 interacts with a helix-loop-helix (HLH)-containing protein called inhibitor of DNA-binding 3 (ID3). In response to double-strand breaks (DSBs) in the genome, ATM phosphorylates ID3 at serine 65 within the HLH motif, and this modification allows a direct interaction with MDC1. Moreover, depletion of ID3 results in impaired formation of ionizing radiation (IR)-induced MDC1 foci, suppression of γ-H2AX-bound MDC1, impaired DSB repair, cellular hypersensitivity to IR, and genomic instability. Disruption of the MDC1-ID3 interaction prevents accumulation of MDC1 at sites of DSBs and suppresses DSB repair. Thus, our study uncovers an ID3-dependent mechanism of recruitment of MDC1 to DNA damage sites and suggests that the ID3-MDC1 interaction is crucial for DDR.MDC1 is a key component of the DNA damage response and interacts with several factors such as γ-H2AX. Here the authors show that MDC1 interacts with ID3, facilitating MDC1 recruitment to sites of damage and repair of breaks.


Subject(s)
DNA Damage , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cattle , Cell Cycle Proteins , DNA Breaks, Double-Stranded , Genomic Instability , HEK293 Cells , HeLa Cells , Helix-Loop-Helix Motifs , Histones/metabolism , Humans , Inhibitor of Differentiation Proteins , Mice , Neoplasm Proteins , Radiation, Ionizing , Rats
13.
Oncotarget ; 7(29): 45976-45994, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27351285

ABSTRACT

Inflammation is a potent inducer of tumorigenesis. Increased DNA damage or loss of genome integrity is thought to be one of the mechanisms linking inflammation and cancer development. It has been suggested that NF-κB-induced microRNA-146 (miR146a) may be a mediator of the inflammatory response. Based on our initial observation that miR146a overexpression strongly increases DNA damage, we investigated its potential role as a modulator of DNA repair. Here, we demonstrate that FANCM, a component in the Fanconi Anemia pathway, is a novel target of miR146a. miR146a suppressed FANCM expression by directly binding to the 3' untranslated region of the gene. miR146a-induced downregulation of FANCM was associated with inhibition of FANCD2 monoubiquitination, reduced DNA homologous recombination repair and checkpoint response, failed recovery from replication stress, and increased cellular sensitivity to cisplatin. These phenotypes were recapitulated when miR146a expression was induced by overexpressing the NF-κB subunit p65/RelA or Helicobacter pylori infection in a human gastric cell line; the phenotypes were effectively reversed with an anti-miR146a antagomir. These results suggest that undesired inflammation events caused by a pathogen or over-induction of miR146a can impair genome integrity via suppression of FANCM.


Subject(s)
DNA Helicases/biosynthesis , Gene Expression Regulation/genetics , MicroRNAs/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Damage/physiology , DNA Helicases/genetics , DNA Repair/physiology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology
14.
Free Radic Biol Med ; 93: 12-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26496208

ABSTRACT

Human 8-oxoguanine DNA glycosylase 1 (OGG1) is the major DNA repair enzyme that plays a key role in excision of oxidative damaged DNA bases such as 8-oxoguainine (8-oxoG). Recent studies suggest another function of OGG1, namely that it may be involved in the endotoxin- or oxidative stress-induced inflammatory response. In this study, we investigated the role of OGG1 in the inflammatory response. OGG1 expression is increased in the organs of endotoxin-induced or myelin oligodendrocyte glycoprotein (MOG)-immunized mice and immune cells, resulting in induction of the expression of pro-inflammatory mediators at the transcriptional levels. Biochemical studies showed that signal transducer and activator of transcription 1 (STAT1) plays a key role in endotoxin-induced OGG1 expression and inflammatory response. STAT1 regulates the transcriptional activity of OGG1 through recruiting and binding to the gamma-interferon activation site (GAS) motif of the OGG1 promoter region, and chromatin remodeling by acetylation and dimethylation of lysine-14 and -4 residues of histone H3. In addition, OGG1 acts as a STAT1 coactivator and has transcriptional activity in the presence of endotoxin. The data presented here identifies a novel mechanism, and may provide new therapeutic strategies for the treatment of endotoxin-mediated inflammatory diseases.


Subject(s)
DNA Glycosylases/biosynthesis , Inflammation/genetics , STAT1 Transcription Factor/genetics , Transcriptional Activation/genetics , Animals , Chromatin Assembly and Disassembly/genetics , DNA Damage/genetics , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA Repair/genetics , Endotoxins/toxicity , Gene Expression Regulation , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/metabolism , Mice , Myelin-Oligodendrocyte Glycoprotein/metabolism , Oxidative Stress/genetics , Promoter Regions, Genetic , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/metabolism
15.
Cancer Res ; 75(7): 1298-310, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25627978

ABSTRACT

MDC1 is critical component of the DNA damage response (DDR) machinery and orchestrates the ensuring assembly of the DDR protein at the DNA damage sites, and therefore loss of MDC1 results in genomic instability and tumorigenicity. However, the molecular mechanisms controlling MDC1 expression are currently unknown. Here, we show that miR-22 inhibits MDC1 translation via direct binding to its 3' untranslated region, leading to impaired DNA damage repair and genomic instability. We demonstrated that activated Akt1 and senescence hinder DDR function of MDC1 by upregulating endogenous miR-22. After overexpression of constitutively active Akt1, homologous recombination was inhibited by miR-22-mediated MDC1 repression. In addition, during replicative senescence and stress-induced premature senescence, MDC1 was downregulated by upregulating miR-22 and thereby accumulating DNA damage. Our results demonstrate a central role of miR-22 in the physiologic regulation of MDC1-dependent DDR and suggest a molecular mechanism for how aberrant Akt1 activation and senescence lead to increased genomic instability, fostering an environment that promotes tumorigenesis.


Subject(s)
DNA Repair , Genomic Instability , MicroRNAs/physiology , Nuclear Proteins/genetics , Trans-Activators/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Aged , Animals , Cell Cycle Proteins , Cellular Senescence , DNA Damage , Down-Regulation , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Mice , Middle Aged , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Trans-Activators/metabolism , Young Adult
16.
Biomol Ther (Seoul) ; 21(4): 258-63, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-24244809

ABSTRACT

We demonstrate herein that silibinin, a polyphenolic flavonoid compound isolated from milk thistle (Silybum marianum), inhibits LPS-induced activation of macrophages and production of nitric oxide (NO) in RAW 264.7 cells. Western blot analysis showed silibinin inhibits iNOS gene expression. RT-PCR showed that silibinin inhibits iNOS, TNF-α, and IL1ß. We also showed that silibinin strongly inhibits p38 MAPK phosphorylation, whereas the ERK1/2 and JNK pathways are not inhibited. The p38 MAPK inhibitor abrogated the LPS-induced nitrite production, whereas the MEK-1 inhibitor did not affect the nitrite production. A molecular modeling study proposed a binding pose for silibinin targeting the ATP binding site of p38 MAPK (1OUK). Collectively, this series of experiments indicates that silibinin inhibits macrophage activation by blocking p38 MAPK signaling.

17.
Korean J Physiol Pharmacol ; 17(4): 315-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23946691

ABSTRACT

Here, we show that radicicol, a fungal antibiotic, resulted in marked inhibition of inducible nitric oxide synthase (iNOS) transcription by the pancreatic beta cell line MIN6N8a in response to cytokine mixture (CM: TNF-α, IFN-γ, and IL-1ß). Treatment of MIN6N8a cells with radicicol inhibited CM-stimulated activation of NF-κB/Rel, which plays a critical role in iNOS transcription, in a dose-related manner. Nitrite production in the presence of PD98059, a specific inhibitor of the extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) pathway, was dramatically diminished, suggesting that the ERK1/2 pathway is involved in CM-induced iNOS expression. In contrast, SB203580, a specific inhibitor of p38, had no effect on nitrite generation. Collectively, this series of experiments indicates that radicicol inhibits iNOS gene expression by blocking ERK1/2 signaling. Due to the critical role that NO release plays in mediating destruction of pancreatic beta cells, the inhibitory effects of radicicol on iNOS expression suggest that radicicol may represent a useful anti-diabetic activity.

18.
J Clin Invest ; 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23863623

ABSTRACT

Aberrant expression of apurinic-apyrimidinic endonuclease-1 (APEX1) has been reported in numerous human solid tumors and is positively correlated with cancer progression; however, the role of APEX1 in tumor progression is poorly defined. Here, we show that APEX1 contributes to aggressive colon cancer behavior and functions as an upstream activator in the Jagged1/Notch signaling pathway. APEX1 overexpression or knockdown in human colon cancer cell lines induced profound changes in malignant properties such as cell proliferation, anchorage-independent growth, migration, invasion, and angiogenesis in vitro and in tumor formation and metastasis in mouse xenograft models. These oncogenic effects of APEX1 were mediated by the upregulation of Jagged1, a major Notch ligand. Furthermore, APEX1 expression was associated with Jagged1 in various colon cancer cell lines and in tissues from colon cancer patients. This finding identifies APEX1 as a positive regulator of Jagged1/Notch activity and suggests that it is a potential therapeutic target in colon cancers that exhibit high levels of Jagged1/Notch signaling.

19.
Biol Trace Elem Res ; 151(2): 294-300, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23203814

ABSTRACT

This study investigated the anti-obesity effects of Jeju ground water containing the vanadium components S1 (8.0 ± 0.9 µg/l) and S3 (26.0 ± 2.09 µg/l) on the differentiation of 3 T3-L1 preadipocytes and obesity in mice that were fed a high-fat diet (HFD). The 3 T3-L1 preadipocyte cells were cultured and differentiated in media consisting of Jeju ground water (S1, S3) or deionized water (DW) containing dexamethasone, isobutylmethylxanthine, and insulin. Oil Red O staining showed that lipid accumulation was attenuated in adipocyte cells treated with Jeju ground water. S3 significantly decreased peroxisome-activated receptor γ and CCAAT-enhancer-binding protein α mRNA expression levels, which play major roles in the transcriptional control of adipogenesis, compared to DW. Furthermore, mRNA expression levels of targeted genes, such as adipocyte fatty acid, lipoprotein lipase, and leptin, were decreased by S3 treatment compared with the control group. In mice with HFD-induced obesity, Jeju ground water decreased HFD-induced body weight gain and reduced total cholesterol, triglyceride, and glucose levels in the plasma compared to control mice. Taken together, Jeju ground water inhibits preadipocyte differentiation and adipogenesis in obesity animal models.


Subject(s)
Anti-Obesity Agents/pharmacology , Groundwater/chemistry , Obesity/therapy , Vanadium/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Azo Compounds/chemistry , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/chemistry , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation/drug effects , Cells, Cultured , Culture Media/chemistry , Dexamethasone/chemistry , Diet, High-Fat/adverse effects , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Insulin/chemistry , Lipid Metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR gamma/chemistry , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Republic of Korea , Transcription, Genetic , Vanadium/chemistry
20.
Biochim Biophys Acta ; 1823(12): 2099-108, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22982065

ABSTRACT

The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) plays an essential role in double-strand break repair by initially recognizing and binding to DNA breaks. Here, we show that DNA-PKcs interacts with the regulatory γ1 subunit of AMP-activated protein kinase (AMPK), a heterotrimeric enzyme that has been proposed to function as a "fuel gauge" to monitor changes in the energy status of cells and is controlled by the upstream kinases LKB1 and Ca²âº/calmodulin-dependent kinase kinase (CaMKK). In co-immunoprecipitation analyses, DNA-PKcs and AMPKγ1 interacted physically in DNA-PKcs-proficient M059K cells but not in DNA-PKcs-deficient M059J cells. Glucose deprivation-stimulated phosphorylation of AMPKα on Thr172 and of acetyl-CoA carboxylase (ACC), a downstream target of AMPK, is substantially reduced in M059J cells compared with M059K cells. The inhibition or down-regulation of DNA-PKcs by the DNA-PKcs inhibitors, wortmannin and Nu7441, or by DNA-PKcs siRNA caused a marked reduction in AMPK phosphorylation, AMPK activity, and ACC phosphorylation in response to glucose depletion in M059K, WI38, and IMR90 cells. In addition, DNA-DNA-PKcs(-/-) mouse embryonic fibroblasts (MEFs) exhibited decreased AMPK activation in response to glucose-free conditions. Furthermore, the knockdown of DNA-PKcs led to the suppression of AMPK (Thr172) phosphorylation in LKB1-deficient HeLa cells under glucose deprivation. Taken together, these findings support the positive regulation of AMPK activation by DNA-PKcs under glucose-deprived conditions in mammalian cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , DNA-Activated Protein Kinase/metabolism , Glioma/metabolism , Glucose/deficiency , AMP-Activated Protein Kinase Kinases , Animals , Blotting, Western , Cells, Cultured , DNA Repair/genetics , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Glioma/genetics , Glioma/pathology , HeLa Cells , Humans , Immunoprecipitation , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , RNA, Small Interfering/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...