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
      • KCI등재

        Toward the Clinical Application of Therapeutic Angiogenesis Against Pediatric Ischemic Retinopathy

        조동현,김정훈 한국지질동맥경화학회 2020 지질·동맥경화학회지 Vol.9 No.2

        Therapeutic angiogenesis refers to strategies of inducing angiogenesis to treat diseases involving ischemic conditions. Historically, most attempts and achievements have been related to coronary and peripheral artery diseases. In this review, we propose the clinical application of therapeutic angiogenesis for the treatment of pediatric ischemic retinopathy, including retinopathy of prematurity, familial exudative retinopathy, and NDP-related retinopathy. These diseases are all characterized by the reduction of physiological angiogenesis and the following induction of pathological angiogenesis. Therapeutic angiogenesis, which supplements insufficient physiological angiogenesis, may be a therapeutic approach for ischemic conditions. Various molecules and modalities can be utilized to apply therapeutic angiogenesis for the treatment of ischemic retinopathy, as in coronary and peripheral artery diseases. Experiences with cardiovascular diseases provide a useful reference for the further clinical application of therapeutic angiogenesis in pediatric ischemic retinopathy. Recombinant proteins and gene therapy are powerful tools to deliver angiogenic factors to retinal tissues directly. Furthermore, endothelial progenitor or bone marrow-derived cells can be injected into the vitreous cavity of the eye for therapeutic angiogenesis. Intraocular injections are highly promising for the delivery of therapeutics for therapeutic angiogenesis. We expect that therapeutic angiogenesis will be a breakthrough in the treatment of pediatric ischemic retinopathy.

      • Microfluidic platform for angiogenesis and vascular fusion

        연주헌(Ju Hun Yeon),호청평(Qing Ping Hu),전누리(Noo Li Jeon) 대한기계학회 2010 대한기계학회 춘추학술대회 Vol.2010 No.11

        Angiogenesis and vascular fusion is a complex cellular process which has an essential role in tumor growth, tumor metastasis and other pathological processes. Various in vivo and in vitro models have been developed for studying angiogenesis and anastomosis. In vivo models, the observation of angiogenesis is technically difficult and observed effects are also difficult to quantify. For this reason, the compound screening for angiogenesis on in vivo models was limited and the development of angiogenesis research was restrained. Recently, several in vitro models have been attempted to recreate the complex sequence of events of angiogenesis with varying degrees of success. However, the formation of new blood vessels and blood vessel fusion were not attempted to study in vitro models because it was not proved the appropriate model for angiogenesis and vascular fusion. In this study, we developed a novel microfluidic platform using the appropriate micro-architecture and 3D gel for mimicking angiogenesis. This device demonstrates its capability to precise control the cellular environment, the direction of vessel growth and the distribution of new vessels. Microfluidic platform for angiogenesis and vascular fusion will improve the study about specific vascular fusion and will be beneficial to quantitative angiogenesis assay in further research.

      • KCI등재

        수종의 한약재에서 신생혈관형성 활성 검색 및 기전 연구

        허정은,백용현,이재동,최도영,박동석 대한침구의학회 2007 대한침구의학회지 Vol.24 No.5

        Objectives : Angiogenesis consists of the proliferation, migration, and differentiation of endothelial cells, and angiogenic factors and matrix protein interactions modulate this process. The aim of this study was to determine whether herbs medicine(KHBJs) could induce angiogenic activity in human umbilical vein endothelial cells(HUVECs). Methods : The angiogenic activity of KHBJs were evaluated by proliferation using BrdU assay, chemotactic migration assay, tube formation assay, and measurement of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor(VEGF) in HUVECs. Also, In order to identify enhance angiogenic activity by activity guided fractionation, the angiogenic activity of fractions of KHBJs such as KHBJB or KHBJR were evaluated in vitro and in vivo Matrigel plug angiogenesis asaay. Results : About 9 KHBJs significantly increased HUVECs proliferation in a dose-dependent manner. In addition, 9 herbs medicine(KHBJs) increased migration and tube-like formation in HUVECs. Interest- ingly the expression of bFGF and VEGF, an angiogenesis-inducing growth factor, were dose-dependently increased by KHBJs. However, angiogenic activity of fractionated KHBJs(KHBJB or KHBJR) not enhanced more than KHBJs in HUVECs and Matrigel plug in vivo angiogenesis assay. Conclusions : 9 KHBJs significantly induces angiogenesis in in vitro and in vivo. These results suggest that 9 KHBJs potent angiogenic agents and promising drug for the induction of neovascularization. Objectives : Angiogenesis consists of the proliferation, migration, and differentiation of endothelial cells, and angiogenic factors and matrix protein interactions modulate this process. The aim of this study was to determine whether herbs medicine(KHBJs) could induce angiogenic activity in human umbilical vein endothelial cells(HUVECs). Methods : The angiogenic activity of KHBJs were evaluated by proliferation using BrdU assay, chemotactic migration assay, tube formation assay, and measurement of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor(VEGF) in HUVECs. Also, In order to identify enhance angiogenic activity by activity guided fractionation, the angiogenic activity of fractions of KHBJs such as KHBJB or KHBJR were evaluated in vitro and in vivo Matrigel plug angiogenesis asaay. Results : About 9 KHBJs significantly increased HUVECs proliferation in a dose-dependent manner. In addition, 9 herbs medicine(KHBJs) increased migration and tube-like formation in HUVECs. Interest- ingly the expression of bFGF and VEGF, an angiogenesis-inducing growth factor, were dose-dependently increased by KHBJs. However, angiogenic activity of fractionated KHBJs(KHBJB or KHBJR) not enhanced more than KHBJs in HUVECs and Matrigel plug in vivo angiogenesis assay. Conclusions : 9 KHBJs significantly induces angiogenesis in in vitro and in vivo. These results suggest that 9 KHBJs potent angiogenic agents and promising drug for the induction of neovascularization.

      • SCOPUSKCI등재

        Original Article : Effects of the knockdown of hypoxia inducible factor-1α expression by adenovirus-mediated shRNA on angiogenesis and tumor growth in hepatocellular carcinoma cell Lines

        ( Sung Hoon Choi ),( Hye Won Shin ),( Jun Yong Park ),( Ji Young Yoo ),( Do Young Kim ),( Weon Sang Ro ),( Chae Ok Yun ),( Kwang Hyub Han ) 대한간학회 2010 Clinical and Molecular Hepatology(대한간학회지) Vol.16 No.3

        Background/Aims: Hypoxia-inducible factor-1α (HIF-1α) is a central transcriptional factor involved in the cellular responses related to various aspects of cancer biology, including proliferation, survival, and angiogenesis, and the metabolism of the extracellular matrix in hypoxia. This study evaluated whether adenovirus-mediated small hairpin RNA (shRNA) against HIF-1α (shHIF-1α) inhibits cell proliferation and angiogenesis in hepatocellular carcinoma (HCC) cell lines. Methods: Knockdown of HIF-1α expression was constructed by adenovirus-mediated RNA interference tools, and HCC cell lines infected with shHIF-1α coding virus were cultured under a hypoxia condition (1% O2) for 24 hours. Following infection, the expression levels of HIF-1α, angiogenesis factors, and matrix metalloproteinase (MMP) were examined using Western blotting. Cell proliferation and angiogenesis were measured by a cell proliferation assay (MTT assay) and an angiogenesis-related assay (invasion and tube-formation assay), respectively. Results: Adenovirus mediated inhibition of HIF-1α induced suppression of tumor growth in HCC cell lines. It also down-regulated the expression of angiogenesis factor and MMP proteins. Angiogenesis as well as mobility of vascular cells to tumor was suppressed by adenovirus-mediated shHIF-1α-infected groups in human umbilical vein endothelial cells (HUVECs). Conclusions: These data suggest that adenovirus-mediated inhibition of HIF-1α inhibits the invasion, tube formation, and cell growth in HUVECs and HCC cells. (Korean J Hepatol 2010;16:280-287)

      • KCI등재

        Effects of Astragalus membranaceus on Angiogenesis

        Seo Dong-Min,Choi Do-Young,Lee Jae-Dong 대한침구의학회 2007 대한침구의학회지 Vol.24 No.2

        Objectives : The aim of this study is to identify and characterize whether Astragalus membranaceus (AM) could induce angiogenesis in vitro and in vivo. Therefore, this study may allow better understanding of the relationship between angiogenesis and wound healing, and ischemia control by AM in oriental medicines and be the basement of developing herbal acupuncture for treatment of wound healing, ischemia and bone fracture. Methods : This study investigated the effects on angeogenesis of AM using human umbilical vein endothelial cells (HUVECs) and Matrigel angiogenesis model. Results : AM significantly increased HUVECs proliferation in a dose-dependent manner. In addition, AM increased migration and tube-like formation in HUVECs. The expression of basic fibroblast growth factor (bFGF), an angiogenesis-stimulating growth factor, was dose-dependently increased by AM. The angiogenic activity of AM was confirmed using an in vivo Matrigel angiogenesis model, showing promotion of blood vessel formation. 목적 : 황기가 혈관 신생 작용이 있는지에 관하여 관찰한다. 황기는 상처의 치유나 허혈성 질환에 효과를 나타내는 것으로 알려져 있다. 이러한 효과가 황기의 혈관 신생 작용과의 관련성을 이해하며 향후 임상에 쓰일 수 있는 황기 약침액 개발을 위한 기초 자료를 목표로 한다. 방법 : 황기의 혈관 신생 작용의 관찰을 위하여 human umbilical vein endothelial cells (HUVECs)와 Matrigel angiogenesis model 을 이용하여 연구하였다. 결과 : 황기는 용량에 따라서 HUVECs의 증식을 나타내었다. 또한 혈관 내피 세포의 이동과 관형 형성을 보였다. 혈관 신생 물질인 basic fibroblast growth factor (bFGF) 가 황기에 의해 증가하였다. Matrigel angiogenesis model에서 황기는 조직학적으로 혈관 형성을 촉진하였으며, 헤모글로빈의 증가를 나타내었다.

      • KCI등재

        Exosomes Derived from Human Amniotic Mesenchymal Stem Cells Facilitate Diabetic Wound Healing by Angiogenesis and Enrich Multiple lncRNAs

        Fu Shangfeng,Zhang Hongyan,Li Xiancai,Zhang Qiling,Guo Chunyan,Qiu Keqing,Feng Junyun,Liu Xiaoxiao,Liu Dewu 한국조직공학과 재생의학회 2023 조직공학과 재생의학 Vol.20 No.2

        BACKGROUND: Diabetic wound healing remains a major challenge due to the impaired functionality of angiogenesis by persistent hyperglycemia. Mesenchymal stem cell exosomes are appropriate candidates for regulating the formation of angiogenesis in tissue repair and regeneration. Here, we explored the effects of exosomes derived from human amniotic mesenchymal stem cell (hAMSC-Exos) on the biological activities of human umbilical vein endothelial cells (HUVECs) treated with high glucose and on diabetic wound healing and investigate lncRNAs related to angiogenesis in hAMSC-Exos. METHODS: hAMSCs and hAMSC-Exos were isolated and identified by flow cytometry or western blot. A series of functional assays such as cell counting kit-8, scratching, transwell and tube formation assays were performed to evaluate the potential effect of hAMSC-Exos on high glucose-treated HUVECs. The effect of hAMSC-Exos on diabetic wound healing were tested by measuring wound closure rates and immunohistochemical staining of CD31. Subsequently, the lncRNAs profiles in hAMSC-Exos and hAMSCs were examined to screen the lncRNAs related to angiogenesis. RESULTS: The isolated hAMSC-Exos had a size range of 30–150 nm and were positive for CD9, CD63 and CD81. The hAMSC-Exos facilitate the functional properties of high glucose-treated HUVECs including the proliferation, migration and the angiogenic activities as well as wound closure and angiogenesis in diabetic wound. hAMSC-Exos were enriched lncRNAs that related to angiogenesis, including PANTR1, H19, OIP5-AS1 and NR2F1-AS1. CONCLUSION: Our findings demonstrated hAMSC-Exos facilitate diabetic wound healing by angiogenesis and contain several exosomal lncRNAs related to angiogenesis, which may represent a promising strategy for diabetic wound healing.

      • KCI등재

        RalA-binding Protein 1 is an Important Regulator of Tumor Angiogenesis

        Seunghyung Lee(이승형) 한국생명과학회 2014 생명과학회지 Vol.24 No.5

        본 논문은 RLIP76 단백질이 암, 종양 혈관 신생 및 그 치료에 미치는 중요성을 보고함에 있다. 암의 연구에 있어서, 종양의 혈관 신생을 억제시키는 인자와 영향을 끼치는 인자를 밝혀내는 것은 암의 억제와 치료를 위한 분자 생물학적 기전에 중대한 영향을 미친다. 최근 연구에서, RLIP76 단백질이 혈관 신생에 영향을 끼치는 역할을 발견하였다. RLIP76 제거 마우스의 종양은 일반 종양과 비교하여 혈관의 크기가 작으며, 가늘고, 그 혈관의 수가 적고 길이가 짧은 것으로 보고되고 있다. 게다가, Matrigel plugs을 이용한 혈관 신생 실험에서, RLIP76이 제거된 마우스에서는 혈관 생성이 억제 되었으나, 일반 마우스에서는 혈관이 생성되었다. 또한, 혈관세포를 이용한 in vitro 실험에 있어서, proliferation, migration 및 cord formation 모두가 RLIP76에 의해서 조절되었다. 일반적으로 RLIP76은 대부분의 인간 조직과 종양에서 발현되며, 약의 저항 기전 연구에 이용되고 있기도 한다. 또한, 이 RLIP76은 small GTPase R-Ras와 상호작용을 통하여 세포 spreading 및 migration에 관여하고 있다. 이러한 결과는 RLIP76와 암 연구의 중요성을 보고하고 있으며, 혈관 세포의 기능의 기전 및 종양의 혈관 신생을 위한 RLIP76단백질의 중요성을 알리고 있고, RLIP76의 추가적인 연구를 통하여 종양의 혈관 신생의 기전을 밝히는 것이 필요함을 제안하는 바이다. Tumor angiogenesis is important in tumorigenesis and therapeutic interventions in cancer. To know inhibitor and effector of tumor angiogenesis in cancer, the specific gene of tumor and angiogenesis may develop the mechanisms of cancer suppression and therapy. Recently, we described the role of RalA-binding protein 1 (RLIP76) in tumor angiogenesis. Tumor vascular volumes were diminished, and vessels were fewer in number, shorter, and narrower in RLIP76 knockout mice than in wild-type mice. Moreover, angiogenesis in basement membrane matrix plugs was blunted in the knockout mice in the absence of tumor cells, with endothelial cells isolated from the lungs of these animals exhibiting defects in migration, proliferation, and cord formation in vitro. RLIP76 is expressed in most human tissues and is overexpressed in many tumor types. In addition, the protein regulates tumorigenesis and angiogenesis in vivo and in vitro. As the export of chemotherapy agents is a prominent cellular function of RLIP76, it is a major factor in mechanisms of drug resistance. Moreover, RLIP76 acts as a selective effector of the small GTPase, R-Ras, and regulates R-Ras signaling, leading to cell spreading and migration. Furthermore, in skin carcinogenesis, RLIP76 knockout mice are resistant, with tumors that form showing diminished angiogenesis. Thus, RLIP76 is required for efficient endothelial cell function and angiogenesis in solid tumors.

      • KCI등재

        Caffeine-induced endothelial cell death and the inhibition of angiogenesis

        Hua Li,Sheng-Yu Jin,Hyun-Joon Son,Je Hoon Seo,Goo-Bo Jeong 대한해부학회 2013 Anatomy & Cell Biology Vol.46 No.1

        Numerous studies have shown that adenosine or adenosine agonists can stimulate angiogenesis. However, the effect of caffeine (a known adenosine receptor antagonist) on angiogenesis has not been previously studied. Accordingly, this study was undertaken to examine the effect of caffeine on angiogenesis and to clarify the mechanism involved. Chick chorioallantoic membrane assays were used to investigate the effect of caffeine on angiogenesis and proliferation assays using human umbilical vein endothelial cells (HUVECs), were used to study its effects on specific aspects of angiogenesis. The expressions of caspase-3 and Bcl-2 were examined by western blotting, immunofluorescence staining was used to identify HUVEC morphological changes, and fluorescence activated cell sorting (FACS) and DAPI staining were used to detect HUVEC apoptosis. Caffeine was found to inhibit blood vessel formation dose-dependently and to inhibit the proliferation of HUVECs time- and dosedependently. FACS analysis and DAPI staining showed that inhibitory effect of caffeine on HUVEC proliferation was the result of apoptosis and the up-regulation of thrombospondin-1 (TSP-1). Furthermore, TSP-1 levels were down-regulated by NECA but were unaffected by CGS21680, indicating that caffeine regulated TSP-1 expression via adenosine A2B receptor. In addition, caffeine up-regulated caspase-3 and down-regulated Bcl-2 at the protein level. These results suggest that the inhibitory effect of caffeine on angiogenesis is associated, at least in part, with its induction of endothelial cell apoptosis, probably mediated by a caspase-3 dependent mechanism.

      • KCI등재후보

        혈관신생에 대한 분자영상

        이경한 대한의사협회 2009 대한의사협회지 Vol.52 No.2

        Angiogenesis, the process whereby new capillaries are formed by outgrowth from existing microvessels, is required for tumor growth and metastasis, as well as for healing of ischemic injuries. Because angiogenesis is a promising target for molecular therapies, there is a real need to develop molecular imaging methods to monitor angiogenesis activity. Direct imaging of angiogenesis can help define the pathophysiology of angiogenic processes in vivo, and foster personized medicine by identifying patients likely to respond to angiogenesis-targeted drugs and accurately monitor the therapeutic efficacy. Promising imaging targets include integrins, vascular endothelial growth factor (VEGF) receptors, and matrix metalloproteinases. While MRI and optical imaging modalities are also workable, radiolabeled RGD (arginine-glycine-aspartate) probes that target αvβ3 integrins overexpressed on activated endothelia are the most extensively investigated and successful angiogenesis imaging technique to date. This technique has repeatedly been validated in preclinical models of cancers and ischemic diseases, and clinical studies are presently ongoing to elucidate the value of RGD positron image tomography (PET) imaging in human patients. Herein, we review the current status of angiogenesis imaging research with special emphasis on integrin-targeted techniques.

      • The PPARδ ligand L‐165041 inhibits vegf‐induced angiogenesis, but the antiangiogenic effect is not related to PPARδ

        Park, Jin‐,Hee,Lee, Kuy‐,Sook,Lim, Hyun‐,Joung,Kim, Hanna,Kwak, Hyun‐,Jeong,Park, Hyun‐,Young Wiley Subscription Services, Inc., A Wiley Company 2012 Journal of cellular biochemistry Vol.113 No.6

        <P><B>Abstract</B></P><P>Peroxisome proliferator‐activated receptor (PPAR)δ is known to be expressed ubiquitously and involved in lipid and glucose metabolism. Recent studies have demonstrated that PPARδ is expressed in endothelial cells (ECs) and plays a potential role in endothelial survival and proliferation. Although PPARα and PPARγ are well recognized to play anti‐inflammatory, antiproliferative, and antiangiogenic roles in ECs, the general effect of PPARδ on angiogenesis in ECs remains unclear. Thus, we investigated the effect of the PPARδ ligand L‐165041 on vascular EC proliferation and angiogenesis in vitro as well as in vivo. Our data show that L‐165041 inhibited VEGF‐induced cell proliferation and migration in human umbilical vein ECs (HUVECs). L‐165041 also inhibited angiogenesis in the Matrigel plug assay and aortic ring assay. Flow cytometric analysis indicated that L‐165041 reduced the number of ECs in the S phase and the expression levels of cell cycle regulatory proteins such as cyclin A, cyclin E, CDK2, and CDK4; phosphorylation of the retinoblastoma protein was suppressed by pretreatment with L‐165041. We confirmed whether these antiangiogenic effects of L‐165041 were PPARδ‐dependent using GW501516 and PPARδ siRNA. GW501516 treatment did not inhibit VEGF‐induced angiogenesis, and transfection of PPARδ siRNA did not reverse this antiangiogenic effect of L‐165041, suggesting that the antiangiogenic effect of L‐165041 on ECs is PPARδ‐independent. Together, these data indicate that the PPARδ ligand L‐165041 inhibits VEGF‐stimulated angiogenesis by suppressing the cell cycle progression independently of PPARδ. This study highlights the therapeutic potential of L‐165041 in the treatment of many disorders related to pathological angiogenesis. J. Cell. Biochem. 113: 1947–1954, 2012. © 2012 Wiley Periodicals, Inc.</P>

      연관 검색어 추천

      이 검색어로 많이 본 자료

      활용도 높은 자료

      해외이동버튼