Ginsenoside-Rb1-Mediated Anti-angiogenesis via Regulating PEDF and miR-33a through the Activation of PPAR-γ Pathway

Angiogenesis is the formation of new blood vessels from the existing vasculature, which is involved in multiple biological processes, including atherosclerosis, ischemic heart disease, and cancer. Ginsenoside-Rb1 (Rb1), the most abundant ginsenoside isolated form Panax ginseng, has been identified as a promising anti-angiogenic agent via the up-regulation of PEDF. However, the underlying molecular mechanisms still unknown. In the present study, human umbilical vein endothelial cells (HUVECs) were selected to perform in vitro assays. Rb1 (0–20 nM) treatment induced pigment epithelial-derived factor (PEDF) protein expression in concentration and time-dependent manners. Interestingly, it was also demonstrated that the exposure of Rb1 (10 nM) could increase PEDF protein expression without any alteration on mRNA level, suggesting the involvement of posttranscriptional regulation. Furthermore, bioinformatics predictions indicated the regulation of miR-33a on PEDF mRNA 3′-UTR, which was further confirmed by luciferase reporter gene assay and real-time PCR. Over-expression of pre-miR-33a was found to regress partly Rb1-mediated PEDF increment and anti-angiogenic effect in HUVECs. Additionally, Rb1-reduced miR-33a and increased PEDF expression was prevented by pre-incubation with peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist (GW9662) or transfection with PPAR-γ siRNA in HUVECs. Taken together, our findings demonstrated that Rb1 exerted anti-angiogenic effects through PPAR-γ signaling pathway via modulating miR-33a and PEDF expressions. Thus, Rb1 may have the potential of being developed as an anti-angiogenic agent, however, further appropriate studies are warranted to evaluate the effect in vivo.

[1]  Daniel S. Ory,et al.  miR-33 links SREBP-2 induction to repression of sterol transporters , 2010, Proceedings of the National Academy of Sciences.

[2]  T. Imaizumi,et al.  Pigment epithelium-derived factor (PEDF) administration inhibits occlusive thrombus formation in rats: a possible participation of reduced intraplatelet PEDF in thrombosis of acute coronary syndromes. , 2008, Atherosclerosis.

[3]  M. Norenberg,et al.  Role of Matricellular Proteins in Disorders of the Central Nervous System , 2016, Neurochemical Research.

[4]  K. Moore,et al.  MiR-33 Contributes to the Regulation of Cholesterol Homeostasis , 2010, Science.

[5]  W. Benedict,et al.  Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. , 1999, Science.

[6]  Shiladitya Sengupta,et al.  Modulating Angiogenesis: The Yin and the Yang in Ginseng , 2004, Circulation.

[7]  S. Surapureddi,et al.  Pigment epithelium-derived factor is an angiogenesis and lipid regulator that activates peroxisome proliferator-activated receptor alpha. , 2008, Advances in experimental medicine and biology.

[8]  L. W. Cheung,et al.  Ginsenoside Rb1 inhibits tube‐like structure formation of endothelial cells by regulating pigment epithelium‐derived factor through the oestrogen β receptor , 2007, British journal of pharmacology.

[9]  Yong Cheng,et al.  Anti-amnestic and anti-aging effects of ginsenoside Rg1 and Rb1 and its mechanism of action , 2005, Acta Pharmacologica Sinica.

[10]  V. Koppaka,et al.  anti* , 2002, Annals of Internal Medicine.

[11]  Yuen-Kit Cheng,et al.  Pharmacogenomics and the Yin/Yang actions of ginseng: anti-tumor, angiomodulating and steroid-like activities of ginsenosides , 2007, Chinese medicine.

[12]  E. Edelman,et al.  miRNAs in atherosclerotic plaque initiation, progression, and rupture. , 2015, Trends in molecular medicine.

[13]  C. Yuan,et al.  Ginseng pharmacology: multiple constituents and multiple actions. , 1999, Biochemical pharmacology.

[14]  L. Castellano,et al.  Noncoding RNAs and the control of hormonal signaling via nuclear receptor regulation. , 2014, Journal of molecular endocrinology.

[15]  R. Wong,et al.  Ginsenoside-Rb1 Promotes Adipogenesis Through Regulation of PPARγ and MicroRNA-27b , 2012, Hormone and Metabolic Research.

[16]  N. Bouck,et al.  Prevention of ischemia-induced retinopathy by the natural ocular antiangiogenic agent pigment epithelium-derived factor , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[17]  Longhou Fang,et al.  Targeted cholesterol efflux , 2013, Cell cycle.

[18]  A. Griffioen,et al.  Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? , 2017, Angiogenesis.

[19]  R. Wong,et al.  Role of microRNA-214 in ginsenoside-Rg1-induced angiogenesis. , 2009, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[20]  Katsuaki Sato,et al.  Inhibition of tumor angiogenesis and metastasis by a saponin of Panax ginseng, ginsenoside-Rb2. , 1994, Biological & pharmaceutical bulletin.

[21]  D. Bartel MicroRNAs Genomics, Biogenesis, Mechanism, and Function , 2004, Cell.

[22]  Huoming Zhang,et al.  Role of G3BP1 in glucocorticoid receptor-mediated microRNA-15b and microRNA-23a biogenesis in endothelial cells , 2017, Cellular and Molecular Life Sciences.

[23]  A. Tall,et al.  ATP-Binding Cassette Transporters and HDL Suppress Hematopoietic Stem Cell Proliferation , 2010, Science.

[24]  R. Wong,et al.  Ginsenoside Rb₁ induces type I collagen expression through peroxisome proliferator-activated receptor-delta. , 2012, Biochemical pharmacology.

[25]  S. Ko,et al.  Effects of ginsenosides Rg3 and Rh2 on the proliferation of prostate cancer cells , 2004, Archives of pharmacal research.

[26]  Jong-Hoon Kim,et al.  A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases , 2014, Journal of ginseng research.

[27]  Chao Liu,et al.  Control of Angiogenesis by AIBP-mediated Cholesterol Efflux , 2013, Nature.

[28]  L. Chan,et al.  MicroRNA-15b contributes to ginsenoside-Rg1-induced angiogenesis through increased expression of VEGFR-2. , 2013, Biochemical pharmacology.

[29]  W. Filipowicz,et al.  The widespread regulation of microRNA biogenesis, function and decay , 2010, Nature Reviews Genetics.

[30]  S. Yamagishi,et al.  Anti-atherothrombogenic properties of PEDF. , 2010, Current molecular medicine.

[31]  K. Huber,et al.  Pigment epithelium-derived factor (PEDF) as a therapeutic target in cardiovascular disease , 2009, Expert opinion on therapeutic targets.

[32]  J. Tombran-Tink,et al.  PEDF in angiogenic eye diseases. , 2010, Current molecular medicine.

[33]  S. Woods,et al.  Antiobesity and Antihyperglycemic Effects of Ginsenoside Rb1 in Rats , 2010, Diabetes.

[34]  P. Y.-K. Yue,et al.  Ginsenoside-Rb 1 Promotes Adipogenesis Through , 2022 .

[35]  I. Gérin,et al.  Expression of miR-33 from an SREBP2 Intron Inhibits Cholesterol Export and Fatty Acid Oxidation* , 2010, The Journal of Biological Chemistry.

[36]  M. Wen Getting miRNA Therapeutics into the Target Cells for Neurodegenerative Diseases: A Mini-Review , 2016, Front. Mol. Neurosci..

[37]  Microvascular Plasticity: Angiogenesis in Health and Disease – Preface , 2016, Microcirculation.

[38]  Huixia Lu,et al.  PEDF improves atherosclerotic plaque stability by inhibiting macrophage inflammation response. , 2017, International journal of cardiology.

[39]  Y. Tsao,et al.  PEDF induces p53-mediated apoptosis through PPAR gamma signaling in human umbilical vein endothelial cells. , 2007, Cardiovascular research.

[40]  I. Chowers,et al.  Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. , 2013, Cell metabolism.

[41]  S. Yamagishi,et al.  Pigment epithelium-derived factor (PEDF) and cardiometabolic disorders. , 2014, Current pharmaceutical design.

[42]  Chunxiang Zhang MicroRNAs in Vascular Biology and Vascular Disease , 2010, Journal of cardiovascular translational research.

[43]  V. Notario,et al.  The effects of PEDF on cancer biology: mechanisms of action and therapeutic potential , 2013, Nature Reviews Cancer.

[44]  N. Bouck PEDF: anti-angiogenic guardian of ocular function. , 2002, Trends in molecular medicine.

[45]  I. Cho Effects of Panax ginseng in Neurodegenerative Diseases , 2012, Journal of ginseng research.