RISS 학술연구정보서비스

검색
다국어 입력

http://chineseinput.net/에서 pinyin(병음)방식으로 중국어를 변환할 수 있습니다.

변환된 중국어를 복사하여 사용하시면 됩니다.

예시)
  • 中文 을 입력하시려면 zhongwen을 입력하시고 space를누르시면됩니다.
  • 北京 을 입력하시려면 beijing을 입력하시고 space를 누르시면 됩니다.
닫기
    인기검색어 순위 펼치기

    RISS 인기검색어

      검색결과 좁혀 보기

      선택해제
      • 좁혀본 항목 보기순서

        • 원문유무
        • 음성지원유무
        • 원문제공처
          펼치기
        • 등재정보
          펼치기
        • 학술지명
          펼치기
        • 주제분류
          펼치기
        • 발행연도
          펼치기
        • 작성언어

      오늘 본 자료

      • 오늘 본 자료가 없습니다.
      더보기
      • 무료
      • 기관 내 무료
      • 유료
      • KCI등재

        ox-LDL regulates proliferation and apoptosis in VSMCs by controlling the miR-183-5p/FOXO1

        Fan Mingqiang,Huang Yinglong,Li Kunsheng,Yang Xiangxiang,Bai Jing,Si Qiaoke,Peng Zhengfei,Jia Chunwen,Zhang Qiangnu,Tao Ding 한국유전학회 2022 Genes & Genomics Vol.44 No.6

        Background: microRNA-mRNA axes that are involved in oxidized low-density lipoprotein (ox-LDL)-induced vascular smooth muscle cells (VSMCs) proliferation/apoptosis imbalance need to be further investigated. Objective: To investigate the functional role of miR-183-5p/FOXO1 in VSMCs and its interaction with ox-LDL. Methods: RNA sequencing was used to detect transcriptome changes of VSMCs treated with ox-LDL. miR-183-5p and FOXO1 expression levels in VSMCs after ox-LDL treatment were assessed using qRT-PCR and western blotting. The regulatory effect of miR-183-5p on FOXO1 has been tried to prove using a dual-luciferase reporter assay. The functions of miR-183-5p, and FOXO1 were analyzed by CCK-8 assay and flow cytometry assay. The tissue samples or serum samples of high fat-feeding mice and carotid atherosclerosis patients were collected, and the levels of miR-183-5p/FOXO1 were analyzed. Results: RNA sequencing data showed 81 miRNAs including miR-183-5p was significantly changed after ox-LDL treatment in VSMCs. FOXO1, a miR-183-5p's potential target, was also down-regulated in ox-LDL treated cells. qRT-PCR and western blot found that expression of FOXO1 mRNA and protein significantly reduced in VSMCs treated with ox-LDL, accompanied by overexpression of miR-183-5p. miR-183-5p inhibited FOXO1 mRNA by binding to its 3' UTR. Interference miR-183-5p/FOXO1 could change proliferation/apoptosis imbalance in VSMCs under ox-LDL stimulation. Higher levels of miR-183-5p but reduced FOXO1 can be found in the thoracic aorta tissues of high fat-feeding mice. In serum samples from individuals with carotid atherosclerosis, Higher levels of miR-183-5p were observed. the miR-183-5p level was positively related to the level of serum ox-LDL in patients. Conclusions: Aberrant expression of miR-183-5p/FOXO1 pathway mediated ox-LDL-induced proliferation/apoptosis imbalance in VSMCs. The miR-183-5p/FOXO1 axis can potentially be utilized as the target in the treatment of patients with atherosclerosis.

      • SIRT1-Mediated FoxO1 Deacetylation Is Essential for Multidrug Resistance-Associated Protein 2 Expression in Tamoxifen-Resistant Breast Cancer Cells

        Choi, Hoo-Kyun,Cho, Kyoung Bin,Phuong, Nguyen Thi Thuy,Han, Chang Yeob,Han, Hyo-Kyung,Hien, Tran Thi,Choi, Hong Seok,Kang, Keon Wook American Chemical Society 2013 Molecular pharmaceutics Vol.10 No.7

        <P>Our previous studies have shown that multidrug resistance protein 2 (MRP2) is overexpressed in tamoxifen-resistant MCF-7 breast cancer cells (TAMR-MCF-7 cells) and forkhead box-containing protein, O subfamily1 (FoxO1), functions as a key regulator of multidrug resistance 1 (MDR1) gene transcription. This study aimed to investigate the role of FoxO1 in regulating MRP2 gene expression in TAMR-MCF-7 cells. The proximal promoter region of the human MRP2 gene contains four putative FoxO binding sites, and MRP2 gene transcription was stimulated by FoxO1 overexpression in MCF-7 cells. Subcellular fractionation and immunoblot analyses revealed that basal MRP2 expression and nuclear levels of FoxO1 were enhanced in TAMR-MCF-7 cells compared to MCF-7 cells and the enhanced MRP2 gene transcription was suppressed by FoxO1 siRNA. Because nuclear localization of FoxO1 is regulated by SIRT1 deacetylase, we were further interested in whether SIRT1 is involved in MRP2 expression. Overexpression of SIRT1 with FoxO1 potentiated the gene transcriptional activity of MRP2, and the basal activity and expression of SIRT1 was increased in TAMR-MCF-7 cells. In addition, SIRT1 inhibition reduced both the nuclear FoxO1 levels and MRP2 expression and enhanced cytotoxic effects of paclitaxel and doxorubicin in TAMR-MCF-7 cells. These results suggest that FoxO1 activation via SIRT1-mediated deacetylation is closely related with up-regulation of MRP2 in TAMR-MCF-7 cells.</P><P><B>Graphic Abstract</B> <IMG SRC='http://pubs.acs.org/appl/literatum/publisher/achs/journals/content/mpohbp/2013/mpohbp.2013.10.issue-7/mp400287p/production/images/medium/mp-2013-00287p_0008.gif'></P>

      • KCI등재

        The FOXO1 Gene-Obesity Interaction Increases the Risk of Type 2 Diabetes Mellitus in a Chinese Han Population

        Lilin Gong,Rong Li,Wei Ren,Zengchan Wang,Zhihong Wang,Maosheng Yang,Suhua Zhang 대한의학회 2017 Journal of Korean medical science Vol.32 No.2

        Here, we aimed to study the effect of the forkhead box O1-insulin receptor substrate 2 (FOXO1-IRS2) gene interaction and the FOXO1 and IRS2 genes-environment interaction for the risk of type 2 diabetes mellitus (T2DM) in a Chinese Han population. We genotyped 7 polymorphism sites of FOXO1 gene and IRS2 gene in 780 unrelated Chinese Han people (474 cases of T2DM, 306 cases of healthy control). The risk of T2DM in individuals with AA genotype for rs7986407 and CC genotype for rs4581585 in FOXO1 gene was 2.092 and 2.57 times higher than that with GG genotype (odds ratio [OR] = 2.092; 95% confidence interval [CI] = 1.178–3.731; P = 0.011) and TT genotype (OR = 2.571; 95% CI = 1.404–4.695; P = 0.002), respectively. The risk of T2DM in individuals with GG genotype for Gly1057Asp in IRS2 gene was 1.42 times higher than that with AA genotype (OR = 1.422; 95% CI = 1.037–1.949; P = 0.029). The other 4 single nucleotide polymorphisms (SNPs) had no significant association with T2DM (P > 0.05). Multifactor dimensionality reduction (MDR) analysis showed that the interaction between SNPs rs7986407 and rs4325426 in FOXO1 gene and waist was the best model confirmed by interaction analysis, closely associating with T2DM. There was an increased risk for T2DM in the case of non-obesity with genotype combined AA/CC, AA/AC or AG/AA for rs7986407 and rs4325426, and obesity with genotype AA for rs7986407 or AA for rs4325426 (OR = 3.976; 95% CI = 1.156–13.675; P value from sign test [Psign] = 0.025; P value from permutation test [Pperm] = 0.000–0.001). Together, this study indicates an association of FOXO1 and IRS2 gene polymorphisms with T2DM in Chinese Han population, supporting FOXO1-obesity interaction as a key factor for the risk of T2DM.

      • KCI등재

        Forkhead box O-class 1 and Forkhead box G1 as Prognostic Markers for Bladder Cancer

        Kim, Tae-Hwan,Jo, Sung-Whan,Lee, Young Suk,Kim, Yong-June,Lee, Sang-Cheol,Kim, Wun-Jae,Yun, Seok Joong The Korean Academy of Medical Sciences 2009 JOURNAL OF KOREAN MEDICAL SCIENCE Vol.24 No.3

        <P>Forkhead box O-class 1 (<I>FOXO1</I>) is a key regulator of glucose homeostasis, cell-cycle progression, and apoptosis. Its functions are modulated by forkhead box G1 (<I>FOXG1</I>), which acts as a transcriptional repressor with oncogenic potential. Real-time PCR and immunohistochemical staining were performed in 174 primary bladder cancer specimens and 21 normal bladder mucosae to evaluate these genes. <I>FOXO1</I> and <I>FOXG1</I> mRNA expression in cancer tissues were higher than in normal mucosae (each <I>P</I><0.001). <I>FOXO1</I> mRNA levels were significantly higher in samples of non-progressed patients (<I>P</I><0.001), but <I>FOXG1</I> were enhanced in those of progressed patients (<I>P</I>=0.019). On univariate analysis, <I>FOXO1</I> mRNA expression was significantly associated with grade, stage, recurrence, progression and survival (each <I>P</I><0.05). On multivariate analysis, increased <I>FOXO1</I> mRNA expression was associated with both reduced disease progression (odds ratio [OR], 0.367; 95% confidence interval [CI], 0.163-0.826, <I>P</I>=0.015) and enhanced disease-free survival (OR, 3.262; 95% CI, 1.361-7.820, <I>P</I>=0.008). At a median follow-up of 33 months (range 2 to 156), the patients with a high <I>FOXO1</I> mRNA expression had a significantly prolonged survival (<I>P</I>=0.001). Immunohistochemical findings of <I>FOXO1</I> were generally concordant with mRNA expression levels. In conclusion, <I>FOXO1</I> may be a promising marker for predicting progression in human bladder cancers.</P>

      • SCISCIESCOPUS

        Loss of FOXO1 promotes gastric tumour growth and metastasis through upregulation of human epidermal growth factor receptor 2/neu expression

        Ko, Young San,Cho, Sung Jin,Park, Jinju,Kim, Younghoon,Choi, Yong Joon,Pyo, Jung-Soo,Jang, Bo Gun,Park, Jong-Wan,Kim, Woo Ho,Lee, Byung Lan Nature Publishing Group 2015 The British journal of cancer Vol. No.

        <P><B>Background:</B></P><P>The biological significance of FOXO1, a member of the forkhead box O transcription factor family, in gastric cancer (GC) remains unclear. The present study provides direct evidence of the role of FOXO1 in tumour growth and metastasis of GC in relation to human epidermal growth factor receptor 2 (HER2).</P><P><B>Methods:</B></P><P>The expressions of FOXO1 and HER2 were modulated in GC cell lines (SNU-638, MKN45, SNU-216 and NCI-N87) by stable transfections. The effects of transfection on GC phenotypes were evaluated <I>in vitro</I> and in animal models. In addition, the relationship between FOXO1 and HER2 was analysed using GC clinical specimens, cell lines and xenografts.</P><P><B>Results:</B></P><P>FOXO1 silencing in GC cells increased colony formation and mesenchymal transition <I>in vitro</I>, as well as tumour growth and metastasis in nude mice, whereas HER2 silencing induced the opposite results.. Furthermore, an inverse relationship between FOXO1 and HER2 was found in clinical specimens of GC, GC cells and GC xenograft tumours. Although a negative crosstalk between these two molecules was shown, double knockdown of both FOXO1 and HER2 in GC cells revealed that HER2 silencing reversed the FOXO1 shRNA-induced migration and invasion even without the FOXO1 restoration.</P><P><B>Conclusions:</B></P><P>Our results indicate that loss of FOXO1 promotes GC growth and metastasis by upregulating HER2 expression and that the HER2 expression is more critical to the induction of GC cell metastasis. The present study provides evidence that the FOXO1/HER2 pathway may regulate GC progression in a subgroup of GC patients.</P>

      • KCI등재

        Forkhead Transcription Factor FOXO1 Inhibits Angiogenesis in Gastric Cancer in Relation to SIRT1

        김수연,고영산,박진주,최이슬,박종완,김영훈,표정수,유영복,이재선,이병란 대한암학회 2016 Cancer Research and Treatment Vol.48 No.1

        Purpose We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expres- sion in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. Materials and Methods Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. Results In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1α (HIF-1α) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1α activation and vascular endothelial growth factor (VEGF) expression. In addi- tion, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. Conclusion Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic condi- tions via inactivation of the HIF-1α–VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.

      • KCI등재

        고강도 지구성 운동이 골격근내 p53, p-FoxO1 및 MuRF1 단백질의 발현에 미치는 영향

        강윤석 ( Youn Seok Kang ),김정하 ( Jeong Ha Kim ),유성경 ( Seong Kyeong Yu ),박대령 ( Dae Ryoung Park ),김재철 ( Jae Cheol Kim ) 한국운동생리학회(구 한국운동과학회) 2011 운동과학 Vol.20 No.4

        강윤석, 김정하, 유성경, 박대령, 김재철. 고강도 지구성 운동이 골격근내 p53, p-FoxO1 및 MuRF1 단백질의 발현에 미치는 영향. 운동과학. 제20권 제4호. 379-388. 2011. 고강도 지구력 운동(26 m/min, 10°, 15 min, 30 min, 45 min, 60 min, 90 min)을 하는 동안 쥐의 골격근내 p53, p-FoxO1 및 MuRF1 단백질의 발현을 규명하는데 연구의 목적이 있으며, 골격근내 단백질 발현은 western blotting 방법을 통해 분석하였다. p53 단백질의 발현의 경우 적색 및 백색 비복근 모두 운동 45분에 현저하게 증가하였다. p-FoxO1 단백질 발현의 경우적색 비복근에서는 운동 30분~45분의 증가한 반면에 백색 비복근에서는 운동 15분과 60분에서 높게 증가하는 것으로 나타났다. MuRF1 단백질 발현의 경우 운동 15분과 45분에서 증가한 반면에 백색 비복근에서는 운동 15분에서 높게 증가하는 것으로 나타났다. 이러한 결과는 고강도 지구력 운동을 하는 동안 p53, p-FoxO1 및 MuRF1 단백질이 근세포 손상에 대한 항상성을 유지하는데 중요하게 작용하고 있는 것으로 사료된다. Kang, Y. S., Kim, J. H., Yu, S. K., Park, D. R., Kim, J. C. The expression of p53, pFoxO-1 and MuRF1 protein in skeletal muscle during intensity endurance training. Exercise Science. 20(4): 379-388, 2011. The aim of this study was to investigate the expression of p53, p-FOXO-1, and MuRF-1 protein in the skeletal muscle of Sprague-Dawley rats during intensity endurance exercise (26 m/min, 10°, 15 min, 30 min, 45 min, 60 min, 90 min). The expression of p53, p-FoxO1, and MuRF1 protein in skeletal muscle by western blotting. The expression of p53, p-FoxO1, and MuRF1 protein significantly were increased at 15min to 30 min during intensity endurance exercise and then gradually returned to bofore exercise level. These finding suggest that p53, p-FoxO1, and MuRF1 protein plays a role in the maintennance of homeostasis in skeletal muscle during intensity endurance exercise.

      • KCI등재

        Forkhead box O-class 1 and Forkhead box G1 as Prognostic Markers for Bladder Cancer

        김태환,조성환,이영숙,김용준,이상철,김원재,윤석중 대한의학회 2009 Journal of Korean medical science Vol.24 No.3

        Forkhead box O-class 1 (FOXO1) is a key regulator of glucose homeostasis, cellcycle progression, and apoptosis. Its functions are modulated by forkhead box G1 (FOXG1), which acts as a transcriptional repressor with oncogenic potential. Realtime PCR and immunohistochemical staining were performed in 174 primary bladder cancer specimens and 21 normal bladder mucosae to evaluate these genes. FOXO1 and FOXG1 mRNA expression in cancer tissues were higher than in normal mucosae (each P<0.001). FOXO1 mRNA levels were significantly higher in samples of non-progressed patients (P<0.001), but FOXG1 were enhanced in those of progressed patients (P=0.019). On univariate analysis, FOXO1 mRNA expression was significantly associated with grade, stage, recurrence, progression and survival (each P<0.05). On multivariate analysis, increased FOXO1 mRNA expression was associated with both reduced disease progression (odds ratio [OR], 0.367; 95% confidence interval [CI], 0.163-0.826, P=0.015) and enhanced disease-free survival (OR, 3.262; 95% CI, 1.361-7.820, P=0.008). At a median follow-up of 33 months (range 2 to 156), the patients with a high FOXO1 mRNA expression had a significantly prolonged survival (P=0.001). Immunohistochemical findings of FOXO1 were generally concordant with mRNA expression levels. In conclusion, FOXO1 may be a promising marker for predicting progression in human bladder cancers.

      • Zbtb7c is a critical gluconeogenic transcription factor that induces glucose-6-phosphatase and phosphoenylpyruvate carboxykinase 1 genes expression during mice fasting

        Choi, Won-Il,Yoon, Jae-Hyeon,Song, Ji-Yang,Jeon, Bu-Nam,Park, Joo-Man,Koh, Dong-In,Ahn, Yong-ho,Kim, Kyung-Sup,Lee, In-Kyu,Hur, Man-Wook Elsevier 2019 Biochimica et biophysica acta. Gene regulatory mec Vol.1862 No.6

        <P><B>Abstract</B></P> <P>Gluconeogenesis is essential for blood glucose homeostasis during fasting and is regulated by various enzymes, which are encoded by gluconeogenic genes. Those genes are controlled by various transcription factors. Zinc finger and BTB domain–containing 7c (Zbtb7c, also called Kr-pok) is a BTB-POZ family transcription factor with proto-oncogenic activity. Previous findings have indicated that Zbtb7c is involved in the regulation of fatty acid biosynthesis, suggesting an involvement also in primary metabolism. We found here that fasting induced <I>Zbtb7c</I> expression in the mouse liver and in primary liver hepatocytes. We also observed that Zbtb7c-knockout mice have decreased blood glucose levels, so we investigated whether Zbtb7c plays a role in gluconeogenesis. Indeed, differential gene expression analysis of Zbtb7c-knockout versus wild type mouse livers showed downregulated transcription of gluconeogenic genes encoding the glucose 6-phosphatase catalytic subunit (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1), while Zbtb7c expression upregulated these two genes, under fasting conditions. Mechanistically, we found that when complexed with histone deacetylase 3 (Hdac3), Zbtb7c binds insulin response elements (IREs) within the <I>G6pc</I> and <I>Pck1</I> promoters. Moreover, complexed Zbtb7c deacetylated forkhead box O1 (Foxo1), thereby increasing Foxo1 binding to the <I>G6pc</I> and <I>Pck1</I> IREs, resulting in their transcriptional activation. These results demonstrate Zbtb7c to be a crucial metabolic regulator of blood glucose homeostasis, during mammalian fasting.</P> <P><B>Highlights</B></P> <P> <UL> <LI> Zbtb7c is induced and plays a critical role in gluconeogenesis by activating G6pc and Pck1 gene transcription during mice fasting. </LI> <LI> Zbtb7c complexed with Hdac3, and deacetylated Foxo1, which increases DNA binding of Foxo1. </LI> <LI> Zbtb7c is critical in increasing Foxo1 binding to the IREs. </LI> </UL> </P>

      • KCI등재

        Ginsenoside Rc modulates Akt/FoxO1 pathways and suppresses oxidative stress

        김대현,박찬흠,박대의,최연재,박민희,정기웅,김소라,이준식,정해영 대한약학회 2014 Archives of Pharmacal Research Vol.37 No.6

        Ginsenoside Rc (Rc), a protopanaxadiol typeginsenoside, is the active component mainly responsiblefor the therapeutic and pharmacologic properties of ginseng,which are derived from its suppression of superoxideinducedfree radicals. Forkhead box O (FoxO1) regulatesvarious genes involved in cellular metabolism related tocell death and response to oxidative stress, and Rc is knownto prevent FoxO1 phosphorylation by activation of PI3K/Akt and subsequent inhibition of AMP-activated proteinkinase (AMPK) in cells exposed to tert-butylhydroperoxide(t-BHP). In the current study, we attempted the mechanismof increased catalase expression by Rc through inhibitionof FoxO1 activation resulting from t-BHP-induced productionof reactive species (RS). We found that overexpressionof catalase induced by Rc resulted in suppressionof RS production in kidney human embryo kidney 293Tcells (HEK293T) cells, and that oxidative stress inducedactivation of PI3K/Akt and inhibition of the AMPK pathwayand FoxO1 phosphorylation, leading to down-regulationof catalase, a FoxO1-targeting gene. In addition,treatment of HEK293T cells with Rc resulted in cAMPresponseelement-binding protein (CREB)-binding protein (CBP) regulated FoxO1 acetylation. Our results suggestthat Rc modulates FoxO1 phosphorylation through activationof PI3K/Akt and inhibition of AMPK and FoxO1acetylation through interaction with CBP and SIRT1, andthat this leads to upregulation of catalase under conditionsof oxidative stress.

      연관 검색어 추천

      이 검색어로 많이 본 자료

      활용도 높은 자료

      해외이동버튼