• 제목/요약/키워드: VEGFR2 phosphorylation

검색결과 9건 처리시간 0.024초

복분자 온수추출물의 VEGF-유도성 혈관신생 억제효과 (Hot-water Extract of Rubus Coreanus Miquel Suppresses VEGF-induced Angiogenesis)

  • 김억천;김혜진;김택중
    • 생명과학회지
    • /
    • 제24권12호
    • /
    • pp.1345-1355
    • /
    • 2014
  • 약초 추출물을 이용한 혈관신생 억제는 많은 고형종양을 치료하기 위한 효과적인 방안으로 인식되어 왔다. 현재까지 가장 효과적으로 종양을 억제하는 방법은 VEGF-유도성 혈관형성 경로를 목표로 하는 것이다. 본 연구에서는 처음으로 복분자 온수추출물의 혈관형성 억제효과를 in vitro와 ex vivo 실험을 통해서 확인하였다. 복분자 온수추출물은 VEGF-유도성 혈관신생을 억제할 뿐만 아니라 ERK와 p38의 인산화, MMP의 활성화를 억제하였다. 또한, 복분자 온수추출물은 VEGF에 의해서 유도된 VEGFR2 인산화를 억제하였다. 이 결과들은 복분자 온수추출물이 VEGFR2의 인산화를 저해함으로써 혈관신생을 억제하고 이것은 혈관신생과 관련된 질병을 치료하는데 좋은 소재가 될 수 있을 것으로 사료된다.

구강 편평상피세포암에서 상피성장인자 수용체와 혈관내피성장인자 수용체 타이로신 활성화효소의 동시 억제 (CONCOMITANT INHIBITION OF EPIDERMAL GROWTH FACTOR AND VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR TYROSINE KINASES IN ORAL SQUAMOUS CELL CARCINOMA)

  • 박영욱;이상신
    • Maxillofacial Plastic and Reconstructive Surgery
    • /
    • 제28권3호
    • /
    • pp.193-201
    • /
    • 2006
  • Squamous cell carcinoma(SCC) of head and neck(SCCHN) is the sixth most common human malignant tumor. However, despite advances in prevention and treatment of SCC, the five-year survival rates for patients remain still low. To improve the outcome for patients with SCCHN, novel treatment strategies are needed. Overexpression of the epidermal growth factor(EGF) and activation of its receptor(EGFR) are associated with progressive growth of SCCHN. Vascular endothelial growth factor(VEGF) signaling molecules are related with neoangiogenesis and vascular metastasis of SCC. In this study, we determined the therapeutic effect of AEE788(Novartis Pharma AG, Basel, Switzerland), which is a dual inhibitor of EGFR/ErbB2 and VEGFR tyrosine kinases, on human oral SCC. At first, we screened the expression of EGFR, c-ErbB2(HER-2) and VEGFR-2 in a series of human oral SCC cell lines. And then we evaluated the effects of AEE788 on the phosphorylation of EGFR and VEGFR-2 in a oral SCC cell line expressing EGFR/HER-2 and VEGFR-2. We also evaluated the effects of AEE788 alone, or with paclitaxel(Taxol) on the oral SCC cell growth and apoptosis. As a result, all oral SCC cells expressed EGFR and VEGFR-2. Treatment of oral SCC cells with AEE788 led to dose-dependent inhibition of EGFR and VEGFR-2 phosphorylation, growth inhibition, and induction of apoptosis. Moreover, AEE788 sensitizes the cells to paclitaxel-mediated toxicity and apoptosis. These data mean EGFR and VEGFR-2 can be reliable targets for molecular therapy of oral SCC, and therefore warrant clinical use of EGFR/VEGFR inhibition in the treatment of patients with recurrent or metastatic oral SCC.

Protein Tyrosine Phosphatase, Receptor Type B (PTPRB) Inhibits Brown Adipocyte Differentiation through Regulation of VEGFR2 Phosphorylation

  • Kim, Ji Soo;Kim, Won Kon;Oh, Kyoung-Jin;Lee, Eun-Woo;Han, Baek Soo;Lee, Sang Chul;Bae, Kwang-Hee
    • Journal of Microbiology and Biotechnology
    • /
    • 제29권4호
    • /
    • pp.645-650
    • /
    • 2019
  • Brown adipocytes have an important role in the regulation of energy balance through uncoupling protein-1 (UCP-1)-mediated nonshivering thermogenesis. Although brown adipocytes have been highlighted as a new therapeutic target for the treatment of metabolic diseases, such as obesity and type II diabetes in adult humans, the molecular mechanism underlying brown adipogenesis is not fully understood. We recently found that protein tyrosine phosphatase receptor type B (PTPRB) expression dramatically decreased during brown adipogenic differentiation. In this study, we investigated the functional roles of PTPRB and its regulatory mechanism during brown adipocyte differentiation. Ectopic expression of PTPRB led to a reduced brown adipocyte differentiation by suppressing the tyrosine phosphorylation of VEGFR2, whereas a catalytic inactive PTPRB mutant showed no effects on differentiation and phosphorylation. Consistently, the expression of brown adipocyte-related genes, such as UCP-1, $PGC-1{\alpha}$, PRDM16, $PPAR-{\gamma}$, and CIDEA, were significantly inhibited by PTPRB overexpression. Overall, these results suggest that PTPRB functions as a negative regulator of brown adipocyte differentiation through its phosphatase activity-dependent mechanism and may be used as a target protein for the regulation of obesity and type II diabetes.

Novel Anti-Angiogenic and Anti-Tumour Activities of the N-Terminal Domain of NOEY2 via Binding to VEGFR-2 in Ovarian Cancer

  • Rho, Seung Bae;Lee, Keun Woo;Lee, Seung-Hoon;Byun, Hyun Jung;Kim, Boh-Ram;Lee, Chang Hoon
    • Biomolecules & Therapeutics
    • /
    • 제29권5호
    • /
    • pp.506-518
    • /
    • 2021
  • The imprinted tumour suppressor NOEY2 is downregulated in various cancer types, including ovarian cancers. Recent data suggest that NOEY2 plays an essential role in regulating the cell cycle, angiogenesis and autophagy in tumorigenesis. However, its detailed molecular function and mechanisms in ovarian tumours remain unclear. In this report, we initially demonstrated the inhibitory effect of NOEY2 on tumour growth by utilising a xenograft tumour model. NOEY2 attenuated the cell growth approximately fourfold and significantly reduced tumour vascularity. NOEY2 inhibited the phosphorylation of the signalling components downstream of phosphatidylinositol-3'-kinase (PI3K), including phosphoinositide-dependent protein kinase 1 (PDK-1), tuberous sclerosis complex 2 (TSC-2) and p70 ribosomal protein S6 kinase (p70S6K), during ovarian tumour progression via direct binding to vascular endothelial growth factor receptor-2 (VEGFR-2). Particularly, the N-terminal domain of NOEY2 (NOEY2-N) had a potent anti-angiogenic activity and dramatically downregulated VEGF and hypoxia-inducible factor-1α (HIF-1α), key regulators of angiogenesis. Since no X-ray or nuclear magnetic resonance structures is available for NOEY2, we constructed the three-dimensional structure of this protein via molecular modelling methods, such as homology modelling and molecular dynamic simulations. Thereby, Lys15 and Arg16 appeared as key residues in the N-terminal domain. We also found that NOEY2-N acts as a potent inhibitor of tumorigenesis and angiogenesis. These findings provide convincing evidence that NOEY2-N regulates endothelial cell function and angiogenesis by interrupting the VEGFR-2/PDK-1/GSK-3β signal transduction and thus strongly suggest that NOEY2-N might serve as a novel anti-tumour and anti-angiogenic agent against many diseases, including ovarian cancer.

혈관내피세포에서 꽃송이버섯(Sparassis crispa) 소수성 추출물의 항혈관신생 활성 (Antiangiogenic activity of non-aqueous fraction from Sparassis crispa extract in human umbilical vein endothelial cells)

  • 한장미;공소연;송재경;강예재;정혜진
    • 한국식품과학회지
    • /
    • 제51권2호
    • /
    • pp.141-146
    • /
    • 2019
  • 본 연구에서는 꽃송이버섯 소수성 추출물(SCF4)의 항혈관신생활성을 혈관내피세포인 HUVECs을 사용하여 확인하였다. 그 결과, SCF4는 세포 독성을 나타내지 않는 $5-25{\mu}g/mL$의 농도에서 VEGF에 의해 유도된 혈관내피세포 증식을 유의적으로 감소시켰을 뿐만 아니라, 혈관내피세포 침윤성과 관 형성 능력을 농도의 존적으로 감소시켜 in vitro 혈관신생을 효과적으로 저해함을 확인하였다. 또한, SCF4는 독성을 나타내지 않고 CAM의 혈관신생을 저해함으로써, in vivo 혈관신생을 효과적으로 저해함을 확인하였다. 마지막으로 SCF4가 혈관신생을 유도하는 주요 신호전달 경로인 VEGFR2, AKT 및 ERK1/2의 전체 단백질 발현 수준의 변화없이 인산화를 저해함을 확인하였다. 따라서, 본 연구는 꽃송이버섯 소수성 추출물이 혈관신생 저해활성을 나타내고 이러한 현상은 VEGFR2 신호전달경로의 억제를 통해 진행됨을 입증하여, 혈관신생 관련 질환 예방 및 치료를 위한 천연물 소재로서의 적용 가능성을 새롭게 제시하였다.

Arg-Leu-Tyr-Glu Suppresses Retinal Endothelial Permeability and Choroidal Neovascularization by Inhibiting the VEGF Receptor 2 Signaling Pathway

  • Park, Wonjin;Baek, Yi-Yong;Kim, Joohwan;Jo, Dong Hyun;Choi, Seunghwan;Kim, Jin Hyoung;Kim, Taesam;Kim, Suji;Park, Minsik;Kim, Ji Yoon;Won, Moo-Ho;Ha, Kwon-Soo;Kim, Jeong Hun;Kwon, Young-Guen;Kim, Young-Myeong
    • Biomolecules & Therapeutics
    • /
    • 제27권5호
    • /
    • pp.474-483
    • /
    • 2019
  • Vascular endothelial growth factor (VEGF) plays a pivotal role in pathologic ocular neovascularization and vascular leakage via activation of VEGF receptor 2 (VEGFR2). This study was undertaken to evaluate the therapeutic mechanisms and effects of the tetrapeptide Arg-Leu-Tyr-Glu (RLYE), a VEGFR2 inhibitor, in the development of vascular permeability and choroidal neovascularization (CNV). In cultured human retinal microvascular endothelial cells (HRMECs), treatment with RLYE blocked VEGF-A-induced phosphorylation of VEGFR2, Akt, ERK, and endothelial nitric oxide synthase (eNOS), leading to suppression of VEGF-A-mediated hyper-production of NO. Treatment with RLYE also inhibited VEGF-A-stimulated angiogenic processes (migration, proliferation, and tube formation) and the hyperpermeability of HRMECs, in addition to attenuating VEGF-A-induced angiogenesis and vascular permeability in mice. The anti-vascular permeability activity of RLYE was correlated with enhanced stability and positioning of the junction proteins VE-cadherin, ${\beta}$-catenin, claudin-5, and ZO-1, critical components of the cortical actin ring structure and retinal endothelial barrier, at the boundary between HRMECs stimulated with VEGF-A. Furthermore, intravitreally injected RLYE bound to retinal microvascular endothelium and inhibited laser-induced CNV in mice. These findings suggest that RLYE has potential as a therapeutic drug for the treatment of CNV by preventing VEGFR2-mediated vascular leakage and angiogenesis.

Inhibitory effect of ginsenglactone A from Panax ginseng on the tube formation of human umbilical vein endothelial cells and migration of human ovarian cancer cells

  • Dahae Lee;Ranhee Kim;So-Ri Son;Ji-Young Kim;Sungyoul Choi;Ki Sung Kang;Dae Sik Jang
    • Journal of Ginseng Research
    • /
    • 제47권2호
    • /
    • pp.246-254
    • /
    • 2023
  • Background: Here, we aimed to assess the inhibitory effect of a new compound from Panax ginseng on the migration of human ovarian cancer cells and tube formation of human umbilical vein endothelial cells (HUVECs). Methods: A new compound, ginsenglactone A (1), was isolated from ginseng roots, together with seven known compounds (2-8). Spectroscopic data were used to elucidate the chemical structure of 1. The tubular structure formation in HUVECs was assessed by Mayer's hematoxylin staining. The migration of A2780 cells was evaluated using the scratch wound healing assay. Results: HUVECs treated with 1 had the statistically significant decrease in tubular structure formation compared to the HUVECs treated with compounds 2-8. This effect was enhanced by co-treatment with inhibitors for phosphatidylinositol 3-kinase (PI3K) (LY294002) and extracellular signal-regulated kinase (ERK) (U0126). Treatment with 1 decreased the expression of phosphorylation of ERK, PI3K, vascular endothelial growth factor receptor2 (VEGFR2), Akt, and mammalian target of rapamycin (mTOR). In addition, the ability of A2780 cells to cover the scratched area were also decreased. This effect was enhanced by co-treatment with U0126. Lastly, treatment with 1 decreased the phosphorylation of ERK, matrix metalloproteinase-9 (MMP-9), and MMP-2. Conclusion: These results suggest that ginsenglactone A is a potential inhibitor of HUVEC tubular structure formation and A2780 cellular migration, which may be helpful for understanding its anticancer mechanism.

The Histone Methyltransferase Inhibitor BIX01294 Inhibits HIF-1α Stability and Angiogenesis

  • Oh, Su Young;Seok, Ji Yoon;Choi, Young Sun;Lee, Sung Hee;Bae, Jong-Sup;Lee, You Mie
    • Molecules and Cells
    • /
    • 제38권6호
    • /
    • pp.528-534
    • /
    • 2015
  • Hypoxia-inducible factor (HIF) is a key regulator of tumor growth and angiogenesis. Recent studies have shown that, BIX01294, a G9a histone methyltransferase (HMT)-specific inhibitor, induces apoptosis and inhibits the proliferation, migration, and invasion of cancer cells. However, not many studies have investigated whether inhibition of G9a HMT can modulate HIF-$1{\alpha}$ stability and angiogenesis. Here, we show that BIX01294 dose-dependently decreases levels of HIF-$1{\alpha}$ in HepG2 human hepatocellular carcinoma cells. The half-life of HIF-$1{\alpha}$, expression of proline hydroxylase 2 (PHD2), hydroxylated HIF-$1{\alpha}$ and von Hippel-Lindau protein (pVHL) under hypoxic conditions were decreased by BIX01294. The mRNA expression and secretion of vascular endothelial growth factor (VEGF) were also significantly reduced by BIX01294 under hypoxic conditions in HepG2 cells. BIX01294 remarkably decreased angiogenic activity induced by VEGF in vitro, ex vivo, and in vivo, as demonstrated by assays using human umbilical vein endothelial cells (HUVECs), mouse aortic rings, and chick chorioallantoic membranes (CAMs), respectively. Furthermore, BIX01294 suppressed VEGF-induced matrix metalloproteinase 2 (MMP2) activity and inhibited VEGF-induced phosphorylation of VEGF receptor 2 (VEGFR-2), focal adhesion kinase (FAK), and paxillin in HUVECs. In addition, BIX01294 inhibited VEGF-induced formation of actin cytoskeletal stress fibers. In conclusion, we demonstrated that BIX01294 inhibits HIF-$1{\alpha}$ stability and VEGF-induced angiogenesis through the VEGFR-2 signaling pathway and actin cytoskeletal remodeling, indicating a promising approach for developing novel therapeutics to stop tumor progression.

과루인 에탄올 추출물의 혈관신생 억제효과 (Inhibitory Effect of the Ethanolic Seed Extract of Trichosanthes kirilowii on Angiogenesis in Human Umbilical Vein Endothelial Cells)

  • 박신형;박현지
    • 동의생리병리학회지
    • /
    • 제36권5호
    • /
    • pp.175-180
    • /
    • 2022
  • The seeds of Trichosanthes kirilowii (STK) used in traditional Oriental medicine for the treatment of dry cough and constipation have diverse pharmacological activities, including hypolipidemic, antioxidant, immunosuppressive, and anticancer effects. However, the effect of STK on angiogenesis has not been studied yet. In this study, we investigated whether the ethanolic extract of STK (ESTK) can regulate the migration and tube formation of human umbilical vein endothelial cells (HUVECs) and explored the underlying mechanism. Results of transwell assay showed that ESTK treatment dose-dependently suppressed the migration of HUVECs. The conditioned medium collected from H1299 human lung cancer cells was used as a chemoattractant. Our observation suggests that ESTK would inhibit the recruitment of endothelial cells into tumors. In addition, ESTK treatment significantly reduced the tube formation of HUVECs. As a molecular mechanism, we found that vascular endothelial growth factor (VEGF)-induced phosphorylation of VEGF receptor 2 (VEGFR2) was completely blocked by ESTK treatment. The expression of angiogenic factors, including VEGFA, fibroblast growth factor 2 (FGF2), angiopoietin, placental growth factor (PGF), platelet derived growth factor (PDGF), angiogenin, and tumor necrosis factor (TNF)-α, was commonly decreased by ESTK treatment in H1299 cells, indicating that ESTK would reduce the production of angiogenic factors from cancer cells. Taken together, our results clearly demonstrated that ESTK exhibited anti-angiogenic effects in HUVECs, which provides another possible mechanism underlying the anticancer activities of STK.