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
        • 저자
          펼치기

      오늘 본 자료

      • 오늘 본 자료가 없습니다.
      더보기
      • 무료
      • 기관 내 무료
      • 유료
      • A new multifunctional Schiff base as a fluorescence sensor for Al<sup>3+</sup> and a colorimetric sensor for CN<sup>−</sup> in aqueous media: an application to bioimaging

        Lee, Seul Ah,You, Ga Rim,Choi, Ye Won,Jo, Hyun Yong,Kim, Ah Ram,Noh, Insup,Kim, Sung-Jin,Kim, Youngmee,Kim, Cheal The Royal Society of Chemistry 2014 Dalton Transactions Vol.43 No.18

        <P>A multifunctional fluorescent and colorimetric receptor <B>1</B> ((<I>E</I>)-<I>N</I>′-((8-hydroxy-1,2,3,5,6,7-hexahydropyrido[3,2,1-<I>ij</I>]quinolin-9-yl)methylene)benzohydrazide) for the detection of both Al<SUP>3+</SUP> and CN<SUP>−</SUP> in aqueous solution has been developed. Receptor <B>1</B> exhibited an excellent selective fluorescence response toward Al<SUP>3+</SUP>. The sensitivity of the fluorescent based assay (0.193 μM) for Al<SUP>3+</SUP> is far below the limit in the World Health Organization (WHO) guidelines for drinking water (7.41 μM). In addition, receptor <B>1</B> showed an excellent detection ability in a wide pH range of 4–10 and also in living cells. Moreover, receptor <B>1</B> showed a highly selective colorimetric response to CN<SUP>−</SUP> by changing its color from colorless to yellow immediately without any interference from other anions.</P> <P>Graphic Abstract</P><P>A new multifunctional Schiff-base receptor <B>1</B> which exhibits an excellent cell-permeable fluorescence for Al<SUP>3+</SUP> and a color change for CN<SUP>−</SUP> in aqueous media has been prepared. <IMG SRC='http://pubs.rsc.org/services/images/RSCpubs.ePlatform.Service.FreeContent.ImageService.svc/ImageService/image/GA?id=c4dt00361f'> </P>

      • KCI등재후보

        Trifolium pratense induces apoptosis through caspase pathway in FaDu human hypopharynx squamous carcinoma cells

        Seul Ah Lee,Bo-Ram Park,Chun Sung Kim 대한구강생물학회 2019 International Journal of Oral Biology Vol.44 No.3

        Trifolium pratense leaves (red clover) has been used in Oriental and European folk medicine for the treatment of whooping cough, asthma, and eczema, and is now being used to treat and alleviate the symptoms, such as hot flushes, cardiovascular health effects that occur in postmenopausal women. However, relatively little scientific data is available on the physiological activity of this plant. Therefore, in this study, we investigated the anti-cancer activity of T. pratense leaves using methanol extract of T. pratense leaves (MeTP) on human FaDu hypopharyngeal squamous carcinoma cells. MeTP inhibited the viability of FaDu cells by inducing apoptosis through the cleavage of pro- caspase-3, -7, and -9 and poly (adenosine diphosphate ribose-ribose) polymerase (PARP), downregulation of Bcl-2, and upregulation of Bax, as determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, Live & dead assay, 4’6-diamidino-2-phenylindole stain, fluorescence-activated cell sorting analysis, and Western blot analysis. In addition, colony formation was slightly inhibited when FaDu cells were treated with a non-cytotoxic concentration (0.125 mg/mL) of MeTP and almost completely inhibited when cells were treated with 0.25 mg/mL MeTP. Collectively, these results indicate that MeTP induced cell apoptosis via caspase- and mitochondrial-dependent apoptotic pathways, and inhibited colony formation of cancer cells in FaDu human hypopharyngeal squamous carcinoma cells. These findings suggest MeTP should be considered for clinical development as a chemotherapeutic option in oral cancer.

      • KCI등재후보

        Methanol Extracts of Codium fragile Induces Apoptosis through G1/S Cell Cycle Arrest in FaDu Human Hypopharynx Squamous Carcinoma Cells

        Seul Ah Lee,Bo-Ram Park,Sung Min Moon,Do Kyung Kim,Chun Sung Kim 대한구강생물학회 2018 International Journal of Oral Biology Vol.43 No.2

        Codium fragile (Suringar) Hariot is an edible green seaweed that belong to the Codiaceae family and has been used in Oriental medicine for the treatment of enterobiasis, dropsy, and dysuria. Methanol extract of codium fragile has anti-oxidant, anti-inflammatory and anti-cancer properties, although the anti-cancer effect on oral cancer has not yet been reported. In this study, we investigated the anti-cancer activity and the mechanism of cell death by methanol extracts of Codium fragile (MeCF) on human FaDu hypopharyngeal squamous carcinoma cells. Our data showed that MeCF inhibits cell viability in a dose-dependent manner, and markedly induced apoptosis, as determined by the MTT assay, Live/Dead assay, and DAPI stain. In addition, MeCF induced the proteolytic cleavage of procaspase -3, -7, -9 and poly(ADP-ribose) polymerase(PARP), and upregulated or downregulated the expression of mitochondrial-apoptosis factor, Bax(pro-apoptotic factor), and Bcl-2(anti-apoptotic factor), . Futhermore, MeCF induced a cell cycle arrest at the G1/S phase through suppressing the expression of the cell cycle cascade proteins, p21, CDK4, CyclinD1, and phospho-Rb. Taken together, these results indicated that MeCF inhibits cell growth, and this inhibition is mediated by caspase- and mitochondrial-dependent apoptotic pathways through cell cycle arrest at the G1/S phase in human FaDu hypopharyngeal squamous carcinoma cells. Therefore, methanol extracts of Codium fragile can be provided as a novel chemotherapeutic drug due to its growth inhibition effects and induction of apoptosis in human oral cancer cells.

      • SCISCIESCOPUS

        Chondroprotective effects of aqueous extract of <i>Anthriscus sylvestris</i> leaves on osteoarthritis <i>in vitro</i> and <i>in vivo</i> through MAPKs and NF-κB signaling inhibition

        Lee, Seul Ah,Moon, Sung-Min,Han, Seul Hee,Hwang, Eun Ju,Park, Bo-Ram,Kim, Jae-Sung,Kim, Do Kyung,Kim, Chun Sung Elsevier 2018 BIOMEDICINE AND PHARMACOTHERAPY Vol.103 No.-

        <P><B>Abstract</B></P> <P>Osteoarthritis (OA) is a common degenerative joint disease, characterized by cartilage degradation and inflammation, in the elderly population. <I>Anthriscus sylvestris</I> has been used in Korean traditional medicine and contains many polyphenolic compounds such as cynaroside and chlorogenic acid, which are major active components responsible for its antioxidant effect. In this study, we aimed to evaluate the chondroprotective effect of an aqueous extract of <I>A. sylvestris</I> leaves (AE-ASL) on OA, both <I>in vitro</I> and <I>in vivo</I>. Rat primary chondrocytes were pretreated with AE-ASL for 1 h before interleukin-1β (20 ng/mL) stimulation. The production of nitrite, PGE<SUB>2</SUB>, aggrecan, and collagen type II were detected by Griess reagent and ELISAs. The mRNA levels of iNOS, COX-2, MMP-3, and MMP-13 were measured by RT-PCR. In addition, protein levels of iNOS, COX-2, MMP-3, MMP-13, ADAMTS-4, MAPKs, and NF-κB p65 subunit were measured by western blot analysis. Sulfated glycosaminoglycan (sGAGs) were detected by dimethylmethylene blue (DMMB) assay. During <I>in vivo</I> study, the effects of AE-ASL were evaluated for 8 weeks in a rat model of destabilization of the medial meniscus (DMM) surgery-induced OA. AE-ASL significantly inhibited expression of nitrite, iNOS, PGE<SUB>2</SUB>, COX-2, MMP-3, MMP-13, and ADAMTS-4 in IL-1β-stimulated chondrocytes. Moreover, it decreased the IL-1β-induced degradation of aggrecan, collagen type II, and proteoglycan. In addition, AE-ASL suppressed IL-1β-induced phosphorylation of MAPKs and NF-κB p65 subunit translocation to nucleus. <I>In vivo</I>, AE-ASL inhibited DMM surgery-induced cartilage destruction and proteoglycan loss. Taken together, these results suggest that AE-ASL may be a potential therapeutic agent for the alleviation of OA progression.</P> <P><B>Highlights</B></P> <P> <UL> <LI> AE-ASL inhibits production of inflammation mediator by IL-1β in rat chondrocytes. </LI> <LI> AE-ASL inhibits release of cartilage degrading enzyme by IL-1β in rat chondrocytes. </LI> <LI> Chondroprotective effect of AE-ASL mediated by MAPKs and NF-κB pathway. </LI> <LI> Oral administration of AE-ASL inhibits the cartilage destruction in DMM rat model. </LI> </UL> </P> <P><B>Graphical abstract</B></P> <P>[DISPLAY OMISSION]</P>

      • SCISCIESCOPUS

        Aqueous extract of <i>Codium fragile</i> suppressed inflammatory responses in lipopolysaccharide-stimulated RAW264.7 cells and carrageenan-induced rats

        Lee, Seul Ah,Moon, Sung-Min,Choi, Yun Hee,Han, Seul Hee,Park, Bo-Ram,Choi, Mi Suk,Kim, Jae-Sung,Kim, Yong Hwan,Kim, Do Kyung,Kim, Chun Sung Elsevier 2017 BIOMEDICINE AND PHARMACOTHERAPY Vol.93 No.-

        <P><B>Abstract</B></P> <P> <I>Codium fragile</I> (Suringar) Hariot has been used in Oriental medicine for the treatment of enterobiasis, dropsy, and dysuria and has been shown to have various biological effects. In this study, we evaluated the anti-inflammatory effects of aqueous extract of <I>C. fragile</I> (AECF) using <I>in vitro</I> and <I>in vivo</I> models. Nitric oxide (NO), prostaglandin E<SUB>2</SUB> (PGE<SUB>2</SUB>), inflammatory-related mRNAs, and proteins were determined using the Griess assay, enzyme-linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction (RT-PCR), and western blotting, respectively. Our results indicate that pretreatment of cells with AECF (50, 100 and 200μg/mL) significantly inhibited LPS-induced secretion of NO and PGE<SUB>2</SUB> in RAW264.7 cells without cytotoxicity. We also found that AECF (100 and 200μg/mL) inhibited LPS-induced inducible NO synthase (iNOS) and cyclooxygenase (COX)-2 expression in a dose-dependent manner. Additionally, pretreatment of cells with AECF (100 and 200μg/mL) inhibited LPS-induced production of inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6. It also prevented the nuclear translocation of nuclear factor (NF)-κB by suppressing the phosphorylation and degradation of inhibitor of NF-κB (IκB)-α. Furthermore, AECF (100 and 200μg/mL) inhibited the phosphorylation of the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase (ERK) 1/2, c-Jun N-terminal kinase (JNK), and p38. In addition, orally administered 50, 100, and 200mg/kg body weight of AECF dose-dependently suppressed carrageenan-induced rat paw edema thickness by 6%, 31%, and 50% respectively, after 4h. Furthermore, the anti-inflammatory effect was comparable to that observed in animals treated with the standard drug diclofenac sodium (56%) <I>in vivo</I>. Collectively, our results suggest that AECF exerts potential anti-inflammatory effects by suppressing NF-κB activation and MAPKs pathways <I>in vitro</I>, as well as inhibiting carrageenan-induced rat paw edema thickness <I>in vivo</I>. These findings indicate that AECF could be further developed as an anti-inflammatory drug.</P>

      • KCI등재후보

        Methanol extracts of Asarum sieboldii Miq. inducesapoptosis via the caspase pathway in human FaDuhypopharynx squamous carcinoma cells

        Seul Ah Lee,Bo-Ram Park,Chun Sung Kim 대한구강생물학회 2021 International Journal of Oral Biology Vol.46 No.2

        Asarum sieboldii Miq. (Aristolochiaceae) is a perennial herbaceous plant and has been used as traditional medicine for treating diseases, cold, fever, phlegm, allergies, chronic gastritis, and acute toothaches. Also, it has various biological activities, such as antiallergic, antiinflammatory, antinociceptive, and antifungal. However, the anticancer effect of A. sieboldii have been rarely reported, except anticancer effect on lung cancer cell (A549) of water extracts of A. sieboldii . This study investigated the anticancer activity of methanol extracts of A. sieboldii (MeAS) and the underlying mechanism in human FaDu hypopharyngeal squamous carcinoma cells. MeAS inhibited FaDu cells grown dose-dependently without affecting normal cells (L929), as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyl tetrazolium bromide and live and dead assay. In addition, concentration of MeAS without cytotoxicity (0.05 and 0.1 mg/mL) inhibited migration and colony formation. Moreover, MeAS treatment significantly induced apoptosis through the proteolytic cleavage of caspase-3, -7, -9, poly (ADP-ribose) polymerase, and downregulation of Bcl-2 and upregulation of Bax in FaDu cells, as determined by fluorescence-activated cell sorting analysis, 4`6-diamidino- 2-phenylindole stain, and western blotting. Altogether, these results suggest that MeAS exhibits strong anticancer effects by suppressing the growth of oral cancer cells and the migration and colony formation via caspase- and mitochondrial-dependent apoptotic pathways in human FaDu hypopharyngeal squamous carcinoma cells. Therefore, MeAS can serve as a natural chemotherapeutic for human oral cancer.

      • KCI등재

        Induction of apopotosis by methanol extracts of Ficus carica L. in FaDu human hypopharynx squamous carcinoma cells

        Seul Ah Lee,Bo-Ram Park,Chun Sung Kim 대한구강생물학회 2020 International Journal of Oral Biology Vol.45 No.3

        Ficus carica L. (fig ) is one of the first cultivated crops and is as old as humans. This plant has been extensively used as a traditional medicine for treating diseases, such as cough, indigestion, nutritional anemia, and tuberculosis. However, the physiological activity of fig leaves on oral cancer is as yet unknown. In this study, we investigated the anticancer effect of methanol extracts of Ficus carica (MeFC) and the mechanism of cell death in human FaDu hypopharyngeal squamous carcinoma cells. MeFC decreased the viability of oral cancer (FaDu) cells but did not affect the viability of normal (L929) cells, as determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay and Live and Dead assay. In addition, MeFC induced apoptosis through the proteolytic cleavage of procaspase-3, -9, poly (ADP-ribose) polymerase (PARP), downregulation of Bcl-2, and upregulation of Bax, as determined by 4′,6-diamidino-2-phenylindole dihydrochloride staining and western blot analysis. Moreover, a concentration of MeFC without cytotoxicity (0.25 mg/mL) significantly suppressed colony formation, a hallmark of cancer development, and completely inhibited the colony formation at 1 mg/mL. Collectively, these results suggest that MeFC exhibits a potent anticancer effect by suppressing the growth of oral cancer cells and colony formation via caspase- and mitochondrial-dependent apoptotic pathways in FaDu human hypopharyngeal squamous carcinoma cells. Therefore, the methanol extract of Ficus carcica leaves provide a natural chemotherapeutic drug for human oral cancer.

      • KCI등재후보

        Apoptosis induced by water extracts of Nypa fruticans wurmb via a mitochondria-dependent pathway in human FaDu hypopharyngeal squamous carcinoma cells

        Seul Ah Lee,Mi Suk Choi,Bo-Ram Park,Jin-Soo Kim,Chun Sung Kim 대한구강생물학회 2021 International Journal of Oral Biology Vol.46 No.4

        Nypa fruticans Wurmb (NFW) contains a large amount of phenolic acid and flavonoids, and is popular as a superfood in Myanmar. NFW has various biological activities, such as anti-inflammatory, anti-oxidant, and neuroprotective properties; however, the anti-cancer effect of NFW have not been reported. In this study, we investigated the anticancer activity of water extracts of NFW (WeNFW) and the underlying mechanism in human FaDu hypopharyngeal squamous carcinoma cells. The WeNFW inhibited FaDu cell growth in a dose-dependent manner without affecting normal cells (L929), as determined by an MTT assay and Live and Dead assay. In addition, the concentrations of WeNFW without cytotoxicity (0.025, 0.05, and 0.1 mg/mL) inhibited wound healing and colony formation. Furthermore, WeNFW significantly induced apoptosis through the proteolytic cleavage of caspase-3 and -9, poly (ADP-ribose) polymerase, and downregulation of Bcl-2 and upregulation of Bax in FaDu cells, as determined by DAPI staining, FACS analysis, and western blot analysis. Taken together, these results suggest that WeNFW exhibits potent anti-cancer effects by suppressing the growth of oral cancer cells, wound healing and colony formation activity. Via mitrochondrial-dependent apoptotic pathways in human FaDu hypopharyngeal squamous carcinoma cells. Therefore, WeNFW can provide a natural chemotherapeutic drug for oral cancer in humans.

      연관 검색어 추천

      이 검색어로 많이 본 자료

      활용도 높은 자료

      해외이동버튼