PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 33793882-12 2021 Moreover, we found that Rg3 could bind to NLRP3 suggesting the anti-inflammatory effects of Rg3 by molecular docking study. ginsenoside Rg3 24-27 NLR family, pyrin domain containing 3 Mus musculus 42-47 10204677-4 1999 On the other hand, ginsenoside-Rg3 (1-100 microM) reduced not only the acetylcholine-, the gamma-aminobutyric acid- and the neurotensin-induced secretions but also, at a higher concentration (100 microM), the angiotensin II-, the bradykinin- and the histamine-induced secretions from the bovine chromaffin cells. ginsenoside Rg3 19-34 neurotensin Bos taurus 124-135 10204677-4 1999 On the other hand, ginsenoside-Rg3 (1-100 microM) reduced not only the acetylcholine-, the gamma-aminobutyric acid- and the neurotensin-induced secretions but also, at a higher concentration (100 microM), the angiotensin II-, the bradykinin- and the histamine-induced secretions from the bovine chromaffin cells. ginsenoside Rg3 19-34 kininogen 1 Bos taurus 230-240 33793882-12 2021 Moreover, we found that Rg3 could bind to NLRP3 suggesting the anti-inflammatory effects of Rg3 by molecular docking study. ginsenoside Rg3 92-95 NLR family, pyrin domain containing 3 Mus musculus 42-47 34799220-0 2022 Ginsenoside Rg3 inhibits angiogenesis in gastric precancerous lesions through downregulation of Glut1 and Glut4. ginsenoside Rg3 0-15 solute carrier family 2 member 1 Rattus norvegicus 96-101 34799220-0 2022 Ginsenoside Rg3 inhibits angiogenesis in gastric precancerous lesions through downregulation of Glut1 and Glut4. ginsenoside Rg3 0-15 solute carrier family 2 member 4 Rattus norvegicus 106-111 34799220-11 2022 Consistent with in vitro results, GRg3 administration significantly reduced the protein expression levels of GLUT1 and GLUT4 in both AGS and HGC-27 human gastric cancer cells in vitro. ginsenoside Rg3 34-38 solute carrier family 2 member 1 Homo sapiens 109-114 34799220-11 2022 Consistent with in vitro results, GRg3 administration significantly reduced the protein expression levels of GLUT1 and GLUT4 in both AGS and HGC-27 human gastric cancer cells in vitro. ginsenoside Rg3 34-38 solute carrier family 2 member 4 Homo sapiens 119-124 34943075-0 2021 Ginsenoside Rg3 Attenuates TNF-alpha-Induced Damage in Chondrocytes through Regulating SIRT1-Mediated Anti-Apoptotic and Anti-Inflammatory Mechanisms. ginsenoside Rg3 0-15 tumor necrosis factor Homo sapiens 27-36 34930287-0 2021 Ginsenoside Rg3 alleviates septic liver injury by regulating the lncRNA TUG1/miR-200c-3p/SIRT1 axis. ginsenoside Rg3 0-15 taurine upregulated gene 1 Mus musculus 72-76 34930287-0 2021 Ginsenoside Rg3 alleviates septic liver injury by regulating the lncRNA TUG1/miR-200c-3p/SIRT1 axis. ginsenoside Rg3 0-15 microRNA 200c Mus musculus 77-85 34930287-0 2021 Ginsenoside Rg3 alleviates septic liver injury by regulating the lncRNA TUG1/miR-200c-3p/SIRT1 axis. ginsenoside Rg3 0-15 sirtuin 1 Mus musculus 89-94 34930287-10 2021 RESULTS: Rg3 upregulated TUG1 expression in liver tissues of CLP mice and LPS-induced hepatocytes. ginsenoside Rg3 9-12 taurine upregulated gene 1 Mus musculus 25-29 34930287-10 2021 RESULTS: Rg3 upregulated TUG1 expression in liver tissues of CLP mice and LPS-induced hepatocytes. ginsenoside Rg3 9-12 hyaluronan and proteoglycan link protein 1 Mus musculus 61-64 34930287-11 2021 Rg3 could activate autophagy to improve mitochondrial dysfunction in LPS-treated hepatocytes, which was partially reversed by TUG1 depletion or miR-200a-3p overexpression. ginsenoside Rg3 0-3 taurine upregulated gene 1 Mus musculus 126-130 34930287-14 2021 CONCLUSION: Our study revealed that Rg3 increased TUG1 expression and reduced miR-200a-3p expression to stimulate the SIRT1/AMPK pathway, thereby enhancing autophagy to improve sepsis-induced liver injury and mitochondrial dysfunction. ginsenoside Rg3 36-39 taurine upregulated gene 1 Mus musculus 50-54 34930287-14 2021 CONCLUSION: Our study revealed that Rg3 increased TUG1 expression and reduced miR-200a-3p expression to stimulate the SIRT1/AMPK pathway, thereby enhancing autophagy to improve sepsis-induced liver injury and mitochondrial dysfunction. ginsenoside Rg3 36-39 sirtuin 1 Mus musculus 118-123 34930332-0 2021 Ginsenoside Rg3 inhibits osteosarcoma progression by reducing circ_0003074 expression in a miR-516b-5p/KPNA4-dependent manner. ginsenoside Rg3 0-15 karyopherin (importin) alpha 4 Mus musculus 103-108 34930332-13 2021 Decreasing miR-516b-5p expression also promoted Rg3-treated OS cell malignancy through KPNA4, which was identified as a target mRNA of miR-516b-5p. ginsenoside Rg3 48-51 karyopherin (importin) alpha 4 Mus musculus 87-92 34930332-16 2021 CONCLUSION: Rg3 inhibited OS progression through circ_0003074/miR-516b-5p/KPNA4 axis, showing the potential of Rg3 as a therapeutic agent for OS. ginsenoside Rg3 12-15 karyopherin (importin) alpha 4 Mus musculus 74-79 34930332-16 2021 CONCLUSION: Rg3 inhibited OS progression through circ_0003074/miR-516b-5p/KPNA4 axis, showing the potential of Rg3 as a therapeutic agent for OS. ginsenoside Rg3 111-114 karyopherin (importin) alpha 4 Mus musculus 74-79 34916608-0 2021 Aminoacylase-1 plays a key role in myocardial fibrosis and the therapeutic effects of 20(S)-ginsenoside Rg3 in mouse heart failure. ginsenoside Rg3 86-107 aminoacylase 1 Mus musculus 0-14 34916608-10 2021 We showed that a high dose of Rg3 (30 mg kg-1 d-1) significantly decreased the content of N-acetylglutamine, increased the expression of ACY1, and inhibited TGF-beta1/Smad3 pathway in CAL mice; Rg3 (25 muM) exerted similar effects in Ang II-treated MCFs. ginsenoside Rg3 30-33 aminoacylase 1 Mus musculus 140-144 34916608-10 2021 We showed that a high dose of Rg3 (30 mg kg-1 d-1) significantly decreased the content of N-acetylglutamine, increased the expression of ACY1, and inhibited TGF-beta1/Smad3 pathway in CAL mice; Rg3 (25 muM) exerted similar effects in Ang II-treated MCFs. ginsenoside Rg3 30-33 transforming growth factor, beta 1 Mus musculus 160-169 34916608-10 2021 We showed that a high dose of Rg3 (30 mg kg-1 d-1) significantly decreased the content of N-acetylglutamine, increased the expression of ACY1, and inhibited TGF-beta1/Smad3 pathway in CAL mice; Rg3 (25 muM) exerted similar effects in Ang II-treated MCFs. ginsenoside Rg3 30-33 SMAD family member 3 Mus musculus 170-175 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 39-42 transforming growth factor, beta 1 Mus musculus 70-79 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 39-42 SMAD family member 3 Mus musculus 80-85 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 39-42 aminoacylase 1 Mus musculus 138-142 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 39-42 aminoacylase 1 Mus musculus 276-280 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 208-211 aminoacylase 1 Mus musculus 152-156 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 229-232 transforming growth factor, beta 1 Mus musculus 70-79 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 229-232 SMAD family member 3 Mus musculus 80-85 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 229-232 aminoacylase 1 Mus musculus 138-142 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 229-232 aminoacylase 1 Mus musculus 152-156 34916608-12 2021 In Ang II-treated MCFs, the effects of Rg3 on collagen deposition and TGF-beta1/Smad3 pathway were slightly enhanced by overexpression of ACY1, whereas ACY1 siRNA partially weakened the beneficial effects of Rg3, suggesting that Rg3 might suppress myocardial fibrosis through ACY1. ginsenoside Rg3 229-232 aminoacylase 1 Mus musculus 276-280 34916608-14 2021 Rg3 attenuates myocardial fibrosis to ameliorate HF through increasing ACY1 expression and inhibiting TGF-beta1/Smad3 pathway, which provides some references for further development of anti-fibrotic drugs for HF. ginsenoside Rg3 0-3 aminoacylase 1 Mus musculus 71-75 34916608-14 2021 Rg3 attenuates myocardial fibrosis to ameliorate HF through increasing ACY1 expression and inhibiting TGF-beta1/Smad3 pathway, which provides some references for further development of anti-fibrotic drugs for HF. ginsenoside Rg3 0-3 transforming growth factor, beta 1 Mus musculus 102-111 34916608-14 2021 Rg3 attenuates myocardial fibrosis to ameliorate HF through increasing ACY1 expression and inhibiting TGF-beta1/Smad3 pathway, which provides some references for further development of anti-fibrotic drugs for HF. ginsenoside Rg3 0-3 SMAD family member 3 Mus musculus 112-117 34943075-0 2021 Ginsenoside Rg3 Attenuates TNF-alpha-Induced Damage in Chondrocytes through Regulating SIRT1-Mediated Anti-Apoptotic and Anti-Inflammatory Mechanisms. ginsenoside Rg3 0-15 sirtuin 1 Homo sapiens 87-92 34943075-8 2021 We showed Rg3 reversed the TNF-alpha-inhibited SIRT1 expression. ginsenoside Rg3 10-13 sirtuin 1 Homo sapiens 47-52 34943075-9 2021 Moreover, the activation of the SIRT1/PGC-1alpha/SIRT3 pathway by Rg3 suppressed the TNF-alpha-induced acetylation of CypD, resulting in less mitochondrial dysfunction and accumulation of reactive oxygen species (ROS). ginsenoside Rg3 66-69 sirtuin 1 Homo sapiens 32-37 34943075-9 2021 Moreover, the activation of the SIRT1/PGC-1alpha/SIRT3 pathway by Rg3 suppressed the TNF-alpha-induced acetylation of CypD, resulting in less mitochondrial dysfunction and accumulation of reactive oxygen species (ROS). ginsenoside Rg3 66-69 PPARG coactivator 1 alpha Homo sapiens 38-48 34943075-9 2021 Moreover, the activation of the SIRT1/PGC-1alpha/SIRT3 pathway by Rg3 suppressed the TNF-alpha-induced acetylation of CypD, resulting in less mitochondrial dysfunction and accumulation of reactive oxygen species (ROS). ginsenoside Rg3 66-69 sirtuin 3 Homo sapiens 49-54 34943075-9 2021 Moreover, the activation of the SIRT1/PGC-1alpha/SIRT3 pathway by Rg3 suppressed the TNF-alpha-induced acetylation of CypD, resulting in less mitochondrial dysfunction and accumulation of reactive oxygen species (ROS). ginsenoside Rg3 66-69 tumor necrosis factor Homo sapiens 85-94 34943075-9 2021 Moreover, the activation of the SIRT1/PGC-1alpha/SIRT3 pathway by Rg3 suppressed the TNF-alpha-induced acetylation of CypD, resulting in less mitochondrial dysfunction and accumulation of reactive oxygen species (ROS). ginsenoside Rg3 66-69 peptidylprolyl isomerase F Homo sapiens 118-122 34943075-11 2021 Furthermore, the Rg3-reverted SIRT1/PGC-1alpha/SIRT3 activation mediated the repression of p38 MAPK, which downregulated the NF-kappaB translocation in the TNF-alpha-treated cells. ginsenoside Rg3 17-20 sirtuin 1 Homo sapiens 30-35 34943075-11 2021 Furthermore, the Rg3-reverted SIRT1/PGC-1alpha/SIRT3 activation mediated the repression of p38 MAPK, which downregulated the NF-kappaB translocation in the TNF-alpha-treated cells. ginsenoside Rg3 17-20 PPARG coactivator 1 alpha Homo sapiens 36-46 34943075-11 2021 Furthermore, the Rg3-reverted SIRT1/PGC-1alpha/SIRT3 activation mediated the repression of p38 MAPK, which downregulated the NF-kappaB translocation in the TNF-alpha-treated cells. ginsenoside Rg3 17-20 sirtuin 3 Homo sapiens 47-52 34943075-11 2021 Furthermore, the Rg3-reverted SIRT1/PGC-1alpha/SIRT3 activation mediated the repression of p38 MAPK, which downregulated the NF-kappaB translocation in the TNF-alpha-treated cells. ginsenoside Rg3 17-20 nuclear factor kappa B subunit 1 Homo sapiens 125-134 34943075-11 2021 Furthermore, the Rg3-reverted SIRT1/PGC-1alpha/SIRT3 activation mediated the repression of p38 MAPK, which downregulated the NF-kappaB translocation in the TNF-alpha-treated cells. ginsenoside Rg3 17-20 tumor necrosis factor Homo sapiens 156-165 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 C-X-C motif chemokine ligand 8 Homo sapiens 77-90 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 C-X-C motif chemokine ligand 8 Homo sapiens 92-96 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 matrix metallopeptidase 9 Homo sapiens 102-127 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 matrix metallopeptidase 9 Homo sapiens 129-134 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 sirtuin 1 Homo sapiens 156-161 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 PPARG coactivator 1 alpha Homo sapiens 162-172 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 sirtuin 3 Homo sapiens 173-178 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 nuclear factor kappa B subunit 1 Homo sapiens 188-197 34943075-12 2021 Our results revealed that administration of Rg3 diminished the production of interleukin 8 (IL-8) and matrix metallopeptidase 9 (MMP-9) in chondrocytes via SIRT1/PGC-1alpha/SIRT3/p38 MAPK/NF-kappaB signaling in response to TNF-alpha stimulation. ginsenoside Rg3 44-47 tumor necrosis factor Homo sapiens 223-232 34130098-0 2021 Lactoferrin-ginsenoside Rg3 complex ingredients: Study of interaction mechanism and preparation of oil-in-water emulsion. ginsenoside Rg3 12-27 lactotransferrin Bos taurus 0-11 34130594-2 2021 Therefore, the objective of our study was to explore the impact of RG3 on cell migration and invasion by regulating the lncRNA HOX antisense intergenic (HOTAIR) expression involving PI3K/AKT signaling pathway. ginsenoside Rg3 67-70 HOX transcript antisense RNA Homo sapiens 153-159 34130594-2 2021 Therefore, the objective of our study was to explore the impact of RG3 on cell migration and invasion by regulating the lncRNA HOX antisense intergenic (HOTAIR) expression involving PI3K/AKT signaling pathway. ginsenoside Rg3 67-70 AKT serine/threonine kinase 1 Homo sapiens 187-190 34130594-9 2021 Ginsenoside-Rg3 reduced the level of lncRNA HOTAIR. ginsenoside Rg3 0-15 HOX transcript antisense RNA Homo sapiens 44-50 34130594-10 2021 Overexpressed lncRNA HOTAIR offset ginsenoside-Rg3 inhibited proliferation, migration and invasion of HCC cells. ginsenoside Rg3 35-50 HOX transcript antisense RNA Homo sapiens 21-27 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 AKT serine/threonine kinase 1 Homo sapiens 59-62 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 matrix metallopeptidase 2 Homo sapiens 72-98 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 matrix metallopeptidase 2 Homo sapiens 100-104 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 matrix metallopeptidase 9 Homo sapiens 110-136 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 matrix metallopeptidase 9 Homo sapiens 138-142 34130594-11 2021 Furthermore, ginsenoside-Rg3 decreased the expression of p-AKT, p-PI3K, matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9), which was reversed after the treatment of HOTAIR. ginsenoside Rg3 13-28 HOX transcript antisense RNA Homo sapiens 187-193 34130594-13 2021 Ginsenoside-Rg3 could reduce the expression of lncRNA HOTAIR, resulting in the inhibited cell proliferation, migration and invasion. ginsenoside Rg3 0-15 HOX transcript antisense RNA Homo sapiens 54-60 34130594-14 2021 Furthermore, ginsenoside-Rg3 inhibited cell proliferation and invasion ability through the PI3k/AKT pathway. ginsenoside Rg3 13-28 AKT serine/threonine kinase 1 Homo sapiens 96-99 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 72-93 lactotransferrin Bos taurus 51-62 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 72-93 lactotransferrin Bos taurus 64-66 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 72-93 lactotransferrin Bos taurus 128-130 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 95-98 lactotransferrin Bos taurus 51-62 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 95-98 lactotransferrin Bos taurus 64-66 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 121-124 lactotransferrin Bos taurus 51-62 34130098-1 2021 Revealing the interaction mechanism between bovine lactoferrin (LF) and 20(S)-ginsenoside Rg3 (Rg3), thereby introducing Rg3 to LF and even into stable emulsions will contribute significantly to food valorization and food industry. ginsenoside Rg3 121-124 lactotransferrin Bos taurus 64-66 34130098-2 2021 Adding Rg3 to LF caused slight absorbance increment and static fluorescence quench of LF, implying the successful combination. ginsenoside Rg3 7-10 lactotransferrin Bos taurus 14-16 34130098-2 2021 Adding Rg3 to LF caused slight absorbance increment and static fluorescence quench of LF, implying the successful combination. ginsenoside Rg3 7-10 lactotransferrin Bos taurus 86-88 34130098-3 2021 Synchronous fluorescence, three-dimensional fluorescence and circular dichroism spectroscopy indicated the conformation changing of LF after binding with Rg3. ginsenoside Rg3 154-157 lactotransferrin Bos taurus 132-134 34694773-0 2021 Ginsenoside Rg3 Alleviates Aluminum Chloride-Induced Bone Impairment in Rats by Activating the TGF-beta1/Smad Signaling Pathway. ginsenoside Rg3 0-15 transforming growth factor, beta 1 Rattus norvegicus 95-104 34694773-3 2021 Previously, we demonstrated that Rg3 can reverse aluminum chloride (AlCl3)-induced oxidative stress and metabolic disorder of bones; however, whether the TGF-beta1/Smad signaling pathway is involved in it remains unclear. ginsenoside Rg3 33-36 transforming growth factor, beta 1 Rattus norvegicus 154-163 34694773-4 2021 First, we found that Rg3 attenuated Al-induced bone impairment in vivo and in vitro by relieving structural damage to the femur, increasing MC3T3-E1 cell activity, differentiation, mineralization, inhibition of cell apoptosis, and upregulating the extracellular matrix (ECM) synthesis and the expression of TGF-beta1/Smad signaling pathway key factors. ginsenoside Rg3 21-24 transforming growth factor, beta 1 Mus musculus 307-316 34694773-5 2021 Subsequently, in the signal pathway intervention experiment, the protective effect of Rg3 on bone impairment induced by Al was weakened; these results indicate that activating the TGF-beta1/Smad signaling pathway is one of the mechanisms of Rg3-attenuated Al-induced bone impairment. ginsenoside Rg3 86-89 transforming growth factor, beta 1 Mus musculus 180-189 34694773-5 2021 Subsequently, in the signal pathway intervention experiment, the protective effect of Rg3 on bone impairment induced by Al was weakened; these results indicate that activating the TGF-beta1/Smad signaling pathway is one of the mechanisms of Rg3-attenuated Al-induced bone impairment. ginsenoside Rg3 241-244 transforming growth factor, beta 1 Mus musculus 180-189 34423507-0 2021 Ginsenoside Rg3 attenuates cisplatin-induced kidney injury through inhibition of apoptosis and autophagy-inhibited NLRP3. ginsenoside Rg3 0-15 NLR family pyrin domain containing 3 Homo sapiens 115-120 34428586-0 2021 Rg3 promotes the SUMOylation of SERCA2a and corrects cardiac dysfunction in heart failure. ginsenoside Rg3 0-3 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 Mus musculus 32-39 34425474-10 2021 Besides, combining Rg3 benefited DCR, ORR and QOL, and alleviated nausea and vomiting, hyperbilirubinemia, leukopenia, myelosuppression, thrombocytopenia and alpha-fetoprotein, while combining GS alleviated nausea and vomiting, ache and hyperbilirubinemia, combining Rh2 alleviated thrombocytopenia, and combining CK alleviated nausea and vomiting, pyrexia, ache and leukopenia, respectively. ginsenoside Rg3 19-22 alpha fetoprotein Homo sapiens 158-175 34764720-10 2021 Inflammatory BEAS-2B cells treated with ginsenoside Rg3 reduced the eotaxin and pro-inflammatory cytokine expressions, and monocyte adherence to BEAS-2B cells was significantly reduced as a result of decreased ICAM-1 expression. ginsenoside Rg3 40-55 C-C motif chemokine ligand 11 Homo sapiens 68-75 34764720-10 2021 Inflammatory BEAS-2B cells treated with ginsenoside Rg3 reduced the eotaxin and pro-inflammatory cytokine expressions, and monocyte adherence to BEAS-2B cells was significantly reduced as a result of decreased ICAM-1 expression. ginsenoside Rg3 40-55 intercellular adhesion molecule 1 Homo sapiens 210-216 34659631-4 2021 Some studies have showed that ginsenoside Rg3 decelerated hERG K(+) channel tail current deactivation. ginsenoside Rg3 30-45 ETS transcription factor ERG Homo sapiens 58-62 34659631-5 2021 Therefore, in this study, we aim to confirm whether ginsenoside Rg3 targeting hERG K(+) channel could be used to reverse the vandetanib-induced QT interval prolongation. ginsenoside Rg3 52-67 ETS transcription factor ERG Homo sapiens 78-82 34659631-13 2021 Furthermore, ginsenoside Rg3 alleviated vandetanib-induced hERG current inhibition and accelerated the process of the channel activation. ginsenoside Rg3 13-28 ETS transcription factor ERG Homo sapiens 59-63 34659631-14 2021 Ginsenoside Rg3 may be a promising cardioprotective agent against vandetanib-induced QT interval prolongation through targeting hERG channel. ginsenoside Rg3 0-15 ETS transcription factor ERG Homo sapiens 128-132 34428586-5 2021 Among mice undergoing transverse aortic constriction, animals that received Rg3 showed improvements in cardiac function and Ca2+ homeostasis, accompanied by increases in the SUMOylation level and SERCA2a activity. ginsenoside Rg3 76-79 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 Mus musculus 196-203 34428586-7 2021 Gene knockout of SUMO1 in mice inhibited the cardioprotective effect of Rg3, and SUMO1 knockout mice that received Rg3 did not exhibit improved Ca2+ homeostasis in cardiomyocytes. ginsenoside Rg3 72-75 small ubiquitin-like modifier 1 Mus musculus 17-22 34428586-8 2021 Additionally, mutation of the SUMOylation sites of SERCA2a blocked the positive effect of Rg3 on the ISO-induced abnormal Ca2+ cycle in HL-1 cells, and was accompanied by an abnormal endoplasmic reticulum stress response and generation of ROS. ginsenoside Rg3 90-93 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 Mus musculus 51-58 34428586-10 2021 SUMO1 is an important factor that mediates the protective effect of Rg3. ginsenoside Rg3 68-71 small ubiquitin-like modifier 1 Mus musculus 0-5 34153662-14 2021 CONCLUSION: Rg3 prevents Ang II-induced myocardial hypertrophy via inactivating NLRP3 inflammasome and oxidative stress by modulating the SIRT1/NF-kappaB pathway. ginsenoside Rg3 12-15 angiotensinogen Rattus norvegicus 25-31 34589493-0 2021 Ginsenoside Rg3 Promotes Cell Growth Through Activation of mTORC1. ginsenoside Rg3 0-15 CREB regulated transcription coactivator 1 Mus musculus 59-65 34589493-2 2021 Previous studies have shown that Rg3 treatment downregulates the activity of rapamycin complex 1 (mTORC1) activity and inhibits the growth of cancer cells. ginsenoside Rg3 33-36 CREB regulated transcription coactivator 1 Mus musculus 98-104 34589493-9 2021 We showed that at lower concentrations, Rg3 activates mTORC1 independent of AKT and AMP-activated protein kinase (AMPK). ginsenoside Rg3 40-43 CREB regulated transcription coactivator 1 Mus musculus 54-60 34589493-12 2021 These findings demonstrate a pro-growth activity of Rg3 that acts through mTORC1 and mitochondrial biogenesis and suggest a dose-dependent effect of Rg3 on tumor cell growth. ginsenoside Rg3 52-55 CREB regulated transcription coactivator 1 Mus musculus 74-80 34153662-0 2021 Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-kappaB pathway. ginsenoside Rg3 0-15 angiotensinogen Rattus norvegicus 27-41 34153662-14 2021 CONCLUSION: Rg3 prevents Ang II-induced myocardial hypertrophy via inactivating NLRP3 inflammasome and oxidative stress by modulating the SIRT1/NF-kappaB pathway. ginsenoside Rg3 12-15 NLR family, pyrin domain containing 3 Rattus norvegicus 80-85 34153662-0 2021 Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-kappaB pathway. ginsenoside Rg3 0-15 NLR family, pyrin domain containing 3 Rattus norvegicus 92-97 34153662-14 2021 CONCLUSION: Rg3 prevents Ang II-induced myocardial hypertrophy via inactivating NLRP3 inflammasome and oxidative stress by modulating the SIRT1/NF-kappaB pathway. ginsenoside Rg3 12-15 sirtuin 1 Rattus norvegicus 138-143 34153662-0 2021 Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-kappaB pathway. ginsenoside Rg3 0-15 sirtuin 1 Rattus norvegicus 147-152 34803431-8 2021 Rg3 down-regulated the MDM2 expression level increased by PMACI stimulation. ginsenoside Rg3 0-3 MDM2 proto-oncogene Homo sapiens 23-27 34153662-8 2021 Furthermore, pharmacological intervention on sirtuin-1 (SIRT1) was performed to clarify the function of SIRT1 in Rg3-mediated effects. ginsenoside Rg3 113-116 sirtuin 1 Rattus norvegicus 104-109 34153662-9 2021 RESULTS: Rg3 dose-dependently attenuated the Ang II-induced myocardial hypertrophy and fibrosis. ginsenoside Rg3 9-12 angiotensinogen Homo sapiens 45-51 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 NLR family, pyrin domain containing 3 Rattus norvegicus 36-41 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 PYD and CARD domain containing Rattus norvegicus 42-45 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 caspase 1 Rattus norvegicus 46-54 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 heme oxygenase 1 Rattus norvegicus 99-103 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 NFE2 like bZIP transcription factor 2 Rattus norvegicus 109-113 34153662-10 2021 What"s more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. ginsenoside Rg3 13-16 angiotensinogen Homo sapiens 146-152 34153662-11 2021 Mechanistically, Rg3 attenuated NF-kappaB activation and promoted SIRT1 expression. ginsenoside Rg3 17-20 sirtuin 1 Rattus norvegicus 66-71 34153662-12 2021 Inhibiting SIRT1 (by AGK2) mostly reversed Rg3-mediated effects against Ang II-induced myocardial hypertrophy and fibrosis. ginsenoside Rg3 43-46 sirtuin 1 Rattus norvegicus 11-16 34443430-6 2021 Treatment with FA and Rg3 significantly attenuated DAergic neurodegeneration induced by 6-OHDA in BZ555 strain, improved basal slowing rate, and prolonged lifespan in the 6-OHDA-induced wild-type strain with downregulation of the apoptosis mediators, egl-1 and ced-3, and upregulation of sod-3 and cat-2. ginsenoside Rg3 22-25 Programmed cell death activator egl-1 Caenorhabditis elegans 251-256 34443430-6 2021 Treatment with FA and Rg3 significantly attenuated DAergic neurodegeneration induced by 6-OHDA in BZ555 strain, improved basal slowing rate, and prolonged lifespan in the 6-OHDA-induced wild-type strain with downregulation of the apoptosis mediators, egl-1 and ced-3, and upregulation of sod-3 and cat-2. ginsenoside Rg3 22-25 Cell death protein 3 subunit p17 Caenorhabditis elegans 261-266 34443430-6 2021 Treatment with FA and Rg3 significantly attenuated DAergic neurodegeneration induced by 6-OHDA in BZ555 strain, improved basal slowing rate, and prolonged lifespan in the 6-OHDA-induced wild-type strain with downregulation of the apoptosis mediators, egl-1 and ced-3, and upregulation of sod-3 and cat-2. ginsenoside Rg3 22-25 Superoxide dismutase [Mn] 2, mitochondrial Caenorhabditis elegans 288-293 34443430-6 2021 Treatment with FA and Rg3 significantly attenuated DAergic neurodegeneration induced by 6-OHDA in BZ555 strain, improved basal slowing rate, and prolonged lifespan in the 6-OHDA-induced wild-type strain with downregulation of the apoptosis mediators, egl-1 and ced-3, and upregulation of sod-3 and cat-2. ginsenoside Rg3 22-25 BH4_AAA_HYDROXYL_2 domain-containing protein;Tyrosine 3-hydroxylase;Tyrosine 3-monooxygenase Caenorhabditis elegans 298-303 34459133-0 2021 Activation of Ca2+ -AMPK-mediated autophagy by ginsenoside Rg3 attenuates cellular senescence in human dermal fibroblasts. ginsenoside Rg3 47-62 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 20-24 34803431-0 2021 Ginsenoside Rg3 attenuates skin disorders via down-regulation of MDM2/HIF1alpha signaling pathway. ginsenoside Rg3 0-15 MDM2 proto-oncogene Homo sapiens 65-69 34803431-0 2021 Ginsenoside Rg3 attenuates skin disorders via down-regulation of MDM2/HIF1alpha signaling pathway. ginsenoside Rg3 0-15 hypoxia inducible factor 1 subunit alpha Homo sapiens 70-79 34803431-3 2021 However, a modulatory effect of Rg3 on TSLP expression in the inflammatory responses remains poorly understood. ginsenoside Rg3 32-35 thymic stromal lymphopoietin Mus musculus 39-43 34803431-7 2021 Results: Rg3 treatment restrained the production and mRNA expression levels of TSLP and VEGF in activated HMC-1 cells. ginsenoside Rg3 9-12 thymic stromal lymphopoietin Homo sapiens 79-83 34803431-7 2021 Results: Rg3 treatment restrained the production and mRNA expression levels of TSLP and VEGF in activated HMC-1 cells. ginsenoside Rg3 9-12 vascular endothelial growth factor A Homo sapiens 88-92 34481534-5 2021 Furthermore, S-Rg3 had significant regulatory effects on PTCH1 and GLI1, key proteins in the Hh pathway, causing significant upregulation of PTCH1 levels and downregulation of GLI1 expression. ginsenoside Rg3 13-18 patched 1 Homo sapiens 57-62 34481534-5 2021 Furthermore, S-Rg3 had significant regulatory effects on PTCH1 and GLI1, key proteins in the Hh pathway, causing significant upregulation of PTCH1 levels and downregulation of GLI1 expression. ginsenoside Rg3 13-18 GLI family zinc finger 1 Homo sapiens 67-71 34481534-5 2021 Furthermore, S-Rg3 had significant regulatory effects on PTCH1 and GLI1, key proteins in the Hh pathway, causing significant upregulation of PTCH1 levels and downregulation of GLI1 expression. ginsenoside Rg3 13-18 patched 1 Homo sapiens 141-146 34481534-5 2021 Furthermore, S-Rg3 had significant regulatory effects on PTCH1 and GLI1, key proteins in the Hh pathway, causing significant upregulation of PTCH1 levels and downregulation of GLI1 expression. ginsenoside Rg3 13-18 GLI family zinc finger 1 Homo sapiens 176-180 34481534-7 2021 Molecular dynamics simulations showed that Rg3 molecules could bind stably to PTCH1 protein through hydrophobic interactions, hydrogen bonds, and pi-pi stacking forces. ginsenoside Rg3 43-46 patched 1 Homo sapiens 78-83 34803431-11 2021 Rg3 inhibited the TSLP and VEGF levels in the serum and ear tissue homogenate. ginsenoside Rg3 0-3 thymic stromal lymphopoietin Mus musculus 18-22 34803431-11 2021 Rg3 inhibited the TSLP and VEGF levels in the serum and ear tissue homogenate. ginsenoside Rg3 0-3 vascular endothelial growth factor A Mus musculus 27-31 34803431-12 2021 Moreover, the MDM2 and HIF1alpha expression levels in the ear tissue homogenate were suppressed by Rg3. ginsenoside Rg3 99-102 transformed mouse 3T3 cell double minute 2 Mus musculus 14-18 34803431-12 2021 Moreover, the MDM2 and HIF1alpha expression levels in the ear tissue homogenate were suppressed by Rg3. ginsenoside Rg3 99-102 hypoxia inducible factor 1, alpha subunit Mus musculus 23-32 34803431-13 2021 Conclusion: Taken together, the current study identifies new mechanistic evidence about MDM2/HIF1alpha pathway in the antiinflammatory effect of Rg3, providing a new effective therapeutic strategy for the treatment of skin inflammatory diseases. ginsenoside Rg3 145-148 transformed mouse 3T3 cell double minute 2 Mus musculus 88-92 34803431-13 2021 Conclusion: Taken together, the current study identifies new mechanistic evidence about MDM2/HIF1alpha pathway in the antiinflammatory effect of Rg3, providing a new effective therapeutic strategy for the treatment of skin inflammatory diseases. ginsenoside Rg3 145-148 hypoxia inducible factor 1, alpha subunit Mus musculus 93-102 34111025-7 2021 Remarkably, Rg3-CNT, but not Rg3, attenuated PD-L1 expression in TNBC cells. ginsenoside Rg3 12-15 CD274 molecule Sus scrofa 45-50 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 33-36 insulin-like growth factor I receptor Mus musculus 77-83 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 33-36 epidermal growth factor receptor Mus musculus 94-98 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 33-36 erb-b2 receptor tyrosine kinase 2 Mus musculus 100-105 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 33-36 platelet derived growth factor receptor, beta polypeptide Mus musculus 109-114 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 33-36 mechanistic target of rapamycin kinase Mus musculus 133-137 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 161-164 insulin-like growth factor I receptor Mus musculus 77-83 34208799-5 2021 Molecular docking predicted that Rg3 epimers had a better binding score with IGF-1R than with EGFR, HER-2 or PDGFR, and predicted an mTOR inhibitory function of Rg3. ginsenoside Rg3 161-164 mechanistic target of rapamycin kinase Mus musculus 133-137 34111025-8 2021 Rg3-CNT decreased the PD-L1 upregulation induced by interferon-gamma (IFN-gamma) in breast cancer cells. ginsenoside Rg3 0-3 CD274 molecule Sus scrofa 22-27 34111025-8 2021 Rg3-CNT decreased the PD-L1 upregulation induced by interferon-gamma (IFN-gamma) in breast cancer cells. ginsenoside Rg3 0-3 interferon gamma Sus scrofa 52-68 34111025-8 2021 Rg3-CNT decreased the PD-L1 upregulation induced by interferon-gamma (IFN-gamma) in breast cancer cells. ginsenoside Rg3 0-3 interferon gamma Sus scrofa 70-79 34111025-10 2021 Specifically, Rg3-CNT reduced the PD-1/PD-L1 axis in a T cell/triple-negative TNBC cell co-culture system. ginsenoside Rg3 14-17 CD274 molecule Sus scrofa 39-44 34111025-13 2021 The Rg3-CNT improved the anti-cancer effect of Rg3 toward TNBC by inhibiting the PD-1/PD-L1 axis. ginsenoside Rg3 4-7 CD274 molecule Sus scrofa 86-91 34111025-13 2021 The Rg3-CNT improved the anti-cancer effect of Rg3 toward TNBC by inhibiting the PD-1/PD-L1 axis. ginsenoside Rg3 47-50 CD274 molecule Sus scrofa 86-91 34066403-8 2021 Using molecular docking and vascular endothelial growth factor (VEGF) bioassay, we showed that Rg3 has an allosteric modulatory effect on vascular endothelial growth factor receptor 2 (VEGFR2). ginsenoside Rg3 95-98 vascular endothelial growth factor A Homo sapiens 28-62 34066403-8 2021 Using molecular docking and vascular endothelial growth factor (VEGF) bioassay, we showed that Rg3 has an allosteric modulatory effect on vascular endothelial growth factor receptor 2 (VEGFR2). ginsenoside Rg3 95-98 vascular endothelial growth factor A Homo sapiens 64-68 34066403-8 2021 Using molecular docking and vascular endothelial growth factor (VEGF) bioassay, we showed that Rg3 has an allosteric modulatory effect on vascular endothelial growth factor receptor 2 (VEGFR2). ginsenoside Rg3 95-98 kinase insert domain receptor Homo sapiens 138-183 34066403-8 2021 Using molecular docking and vascular endothelial growth factor (VEGF) bioassay, we showed that Rg3 has an allosteric modulatory effect on vascular endothelial growth factor receptor 2 (VEGFR2). ginsenoside Rg3 95-98 kinase insert domain receptor Homo sapiens 185-191 35582998-0 2022 Ginsenoside Rg3 ameliorates acute pancreatitis by activating the NRF2/HO-1-mediated ferroptosis pathway. ginsenoside Rg3 0-15 NFE2 like bZIP transcription factor 2 Rattus norvegicus 65-69 34374655-3 2021 In human microglia, Rg3 and its stereoisomers significantly restored inflammatory M1 to normal M0 state and promoted M2 activation by up-regulating acute cytokines such as interleukin-10 and Arginase 1. ginsenoside Rg3 20-23 interleukin 10 Homo sapiens 172-186 34374655-3 2021 In human microglia, Rg3 and its stereoisomers significantly restored inflammatory M1 to normal M0 state and promoted M2 activation by up-regulating acute cytokines such as interleukin-10 and Arginase 1. ginsenoside Rg3 20-23 arginase 1 Homo sapiens 191-201 34374655-4 2021 Moreover, scavenger receptor type A (SRA) was significantly elevated in the presence of ginsenoside Rg3 and 20(S)-Rg3. ginsenoside Rg3 88-103 macrophage scavenger receptor 1 Homo sapiens 10-35 34374655-4 2021 Moreover, scavenger receptor type A (SRA) was significantly elevated in the presence of ginsenoside Rg3 and 20(S)-Rg3. ginsenoside Rg3 88-103 macrophage scavenger receptor 1 Homo sapiens 37-40 34374655-6 2021 We also observed that soluble amyloid precursor protein-alpha (sAPPalpha) and ADAM10 levels were increased in APP swe-transfected Nuro-2a neuronal cells, whereas sAPPbeta was not processed, suggesting that ginsenoside Rg3 was involved in non-amyloidogenic processing. ginsenoside Rg3 206-221 ADAM metallopeptidase domain 10 Homo sapiens 78-84 34374655-7 2021 In immunocytochemistry, SRA and a disintegrin and metalloproteinase 10 (desintegrin and metalloproteinase-containing protein 10, ADAM10) were coincidently upregulated in the presence of ginsenoside Rg3 and its stereoisomers compared to those in normal control. ginsenoside Rg3 186-201 macrophage scavenger receptor 1 Homo sapiens 24-27 34374655-7 2021 In immunocytochemistry, SRA and a disintegrin and metalloproteinase 10 (desintegrin and metalloproteinase-containing protein 10, ADAM10) were coincidently upregulated in the presence of ginsenoside Rg3 and its stereoisomers compared to those in normal control. ginsenoside Rg3 186-201 ADAM metallopeptidase domain 10 Homo sapiens 129-135 34461812-0 2021 Protective Effect of 20(R)-Ginsenoside Rg3 Against Cisplatin-Induced Renal Toxicity via PI3K/AKT and NF-(Formula: see text)B Signaling Pathways Based on the Premise of Ensuring Anticancer Effect. ginsenoside Rg3 21-42 thymoma viral proto-oncogene 1 Mus musculus 93-96 34461812-8 2021 We used western blotting analysis to demonstrate that R-Rg3 restored cisplatin-induced AKI might be related to PI3K/AKT and NF-(Formula: see text)B mediated apoptosis and inflammation pathways. ginsenoside Rg3 54-59 thymoma viral proto-oncogene 1 Mus musculus 116-119 34461812-9 2021 In the meantime, we also verified that R-Rg3 could activate different sites of p53 to control renal cell apoptosis induced by cisplatin without affecting its antitumor effect. ginsenoside Rg3 39-44 transformation related protein 53, pseudogene Mus musculus 79-82 35582998-0 2022 Ginsenoside Rg3 ameliorates acute pancreatitis by activating the NRF2/HO-1-mediated ferroptosis pathway. ginsenoside Rg3 0-15 heme oxygenase 1 Rattus norvegicus 70-74 35582998-8 2022 Western blot analysis revealed that the decrease in glutathione peroxidase 4 (GPX4) and cystine/glutamate transporter (xCT) levels induced by Cn were reversed by Rg3 treatment in the AR42J cells. ginsenoside Rg3 162-165 glutathione peroxidase 4 Rattus norvegicus 52-76 35582998-8 2022 Western blot analysis revealed that the decrease in glutathione peroxidase 4 (GPX4) and cystine/glutamate transporter (xCT) levels induced by Cn were reversed by Rg3 treatment in the AR42J cells. ginsenoside Rg3 162-165 glutathione peroxidase 4 Rattus norvegicus 78-82 35582998-8 2022 Western blot analysis revealed that the decrease in glutathione peroxidase 4 (GPX4) and cystine/glutamate transporter (xCT) levels induced by Cn were reversed by Rg3 treatment in the AR42J cells. ginsenoside Rg3 162-165 solute carrier family 7 (cationic amino acid transporter, y+ system), member 11 Mus musculus 119-122 35582998-12 2022 In comparison, the NRF2/HO-1/xCT/GPX4 pathway was activated in pancreatic tissues following Rg3 administration. ginsenoside Rg3 92-95 nuclear factor, erythroid derived 2, like 2 Mus musculus 19-23 35582998-12 2022 In comparison, the NRF2/HO-1/xCT/GPX4 pathway was activated in pancreatic tissues following Rg3 administration. ginsenoside Rg3 92-95 heme oxygenase 1 Mus musculus 24-28 35582998-12 2022 In comparison, the NRF2/HO-1/xCT/GPX4 pathway was activated in pancreatic tissues following Rg3 administration. ginsenoside Rg3 92-95 solute carrier family 7 (cationic amino acid transporter, y+ system), member 11 Mus musculus 29-32 35582998-12 2022 In comparison, the NRF2/HO-1/xCT/GPX4 pathway was activated in pancreatic tissues following Rg3 administration. ginsenoside Rg3 92-95 glutathione peroxidase 4 Mus musculus 33-37 35582998-13 2022 Taken together, the present study, to the best of our knowledge, is the first to reveal a protective role for Rg3 in mice with AP by suppressing oxidative stress-related ferroptosis and the activation of the NRF2/HO-1 pathway. ginsenoside Rg3 110-113 nuclear factor, erythroid derived 2, like 2 Mus musculus 208-212 35582998-13 2022 Taken together, the present study, to the best of our knowledge, is the first to reveal a protective role for Rg3 in mice with AP by suppressing oxidative stress-related ferroptosis and the activation of the NRF2/HO-1 pathway. ginsenoside Rg3 110-113 heme oxygenase 1 Mus musculus 213-217 35509820-6 2022 Next, our results showed that Rg3 treatment activates the PINK1-Parkin signaling pathway and recruits Parkin and ubiquitin proteins to mitochondria to induce mitophagy. ginsenoside Rg3 30-33 PTEN induced kinase 1 Homo sapiens 58-63 35219164-5 2022 G-Rg3 increased NK cell cytotoxicity and simultaneously increased the expression of NK-activating receptors, NKp44, NKp46, and NKp30. ginsenoside Rg3 0-5 natural cytotoxicity triggering receptor 2 Homo sapiens 109-114 35219164-5 2022 G-Rg3 increased NK cell cytotoxicity and simultaneously increased the expression of NK-activating receptors, NKp44, NKp46, and NKp30. ginsenoside Rg3 0-5 natural cytotoxicity triggering receptor 1 Homo sapiens 116-121 35219164-5 2022 G-Rg3 increased NK cell cytotoxicity and simultaneously increased the expression of NK-activating receptors, NKp44, NKp46, and NKp30. ginsenoside Rg3 0-5 natural cytotoxicity triggering receptor 3 Homo sapiens 127-132 35219164-6 2022 Additionally, G-Rg3 increased the mRNA expression of NK cytolytic molecules, granzyme B and perforin. ginsenoside Rg3 14-19 granzyme B Homo sapiens 77-87 35219164-7 2022 The expression of CD107a, a marker of NK cell degranulation, also increased in G-Rg3-treated NK cells. ginsenoside Rg3 79-84 lysosomal associated membrane protein 1 Homo sapiens 18-24 35219164-8 2022 We therefore proceeded to identify which MAPK signaling pathway was involved in G-Rg3-mediated cytolytic activity. ginsenoside Rg3 80-85 mitogen-activated protein kinase 1 Homo sapiens 41-45 35219164-9 2022 Treatment with G-Rg3 increased the phosphorylation levels of extracellular signal-regulated kinase (ERK), whereas ERK inhibition eliminated G-Rg3-induced NK cell cytotoxicity, suggesting the involvement of the ERK pathway. ginsenoside Rg3 15-20 mitogen-activated protein kinase 1 Homo sapiens 61-98 35219164-9 2022 Treatment with G-Rg3 increased the phosphorylation levels of extracellular signal-regulated kinase (ERK), whereas ERK inhibition eliminated G-Rg3-induced NK cell cytotoxicity, suggesting the involvement of the ERK pathway. ginsenoside Rg3 15-20 mitogen-activated protein kinase 1 Homo sapiens 100-103 35219164-11 2022 The G-Rg3 isomer, 20(R)-Rg3, effectively activated NK cells via the extracellular signal-regulated kinase (ERK) signaling pathway, whereas 20(S)-Rg3 had no effect on NK cell activity. ginsenoside Rg3 4-9 mitogen-activated protein kinase 1 Homo sapiens 107-110 35600776-3 2022 Artesunate (ART) and 20(R)-ginsenoside Rg3 (Rg3) have anti-hepatoma effects and can inhibit STAT3 signaling in cancer cells. ginsenoside Rg3 21-42 signal transducer and activator of transcription 3 Mus musculus 92-97 35600776-3 2022 Artesunate (ART) and 20(R)-ginsenoside Rg3 (Rg3) have anti-hepatoma effects and can inhibit STAT3 signaling in cancer cells. ginsenoside Rg3 44-47 signal transducer and activator of transcription 3 Mus musculus 92-97 35600776-11 2022 Mechanistic studies revealed that Rg3-plus-ART inhibited activation/phosphorylation of Src and STAT3 in HepG2-SR cultures and tumors. ginsenoside Rg3 34-37 Rous sarcoma oncogene Mus musculus 87-90 35600776-11 2022 Mechanistic studies revealed that Rg3-plus-ART inhibited activation/phosphorylation of Src and STAT3 in HepG2-SR cultures and tumors. ginsenoside Rg3 34-37 signal transducer and activator of transcription 3 Mus musculus 95-100 35600776-14 2022 Conclusions: Rg3-plus-ART overcomes sorafenib resistance in experimental models, and inhibition of Src/STAT3 signaling and modulation of ROS/STAT3 signaling contribute to the underlying mechanisms. ginsenoside Rg3 13-16 Rous sarcoma oncogene Mus musculus 99-102 35600776-14 2022 Conclusions: Rg3-plus-ART overcomes sorafenib resistance in experimental models, and inhibition of Src/STAT3 signaling and modulation of ROS/STAT3 signaling contribute to the underlying mechanisms. ginsenoside Rg3 13-16 signal transducer and activator of transcription 3 Mus musculus 103-108 35600776-14 2022 Conclusions: Rg3-plus-ART overcomes sorafenib resistance in experimental models, and inhibition of Src/STAT3 signaling and modulation of ROS/STAT3 signaling contribute to the underlying mechanisms. ginsenoside Rg3 13-16 signal transducer and activator of transcription 3 Mus musculus 141-146 35509820-9 2022 Moreover, GAPDH participates in the Rg3-induced mitophagy and regulates the translocation of Parkin to mitochondria. ginsenoside Rg3 36-39 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 10-15 35509820-10 2022 Functionally, Rg3 exerts the inhibitory effect through regulating the nonglycolytic activity of GAPDH, which could be associated with the cellular oxidative stress. ginsenoside Rg3 14-17 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 96-101 35059739-5 2022 Furthermore, Rg3 efficiently prevented DEX-triggered mitochondrial dysfunction of myotubes through peroxisome proliferator-activated receptor-gamma coactivator1alpha activities and its mitochondrial biogenetic transcription factors, nuclear respiratory factor-1 and mitochondrial transcription factor A. ginsenoside Rg3 13-16 PPARG coactivator 1 alpha Homo sapiens 99-165 35059739-5 2022 Furthermore, Rg3 efficiently prevented DEX-triggered mitochondrial dysfunction of myotubes through peroxisome proliferator-activated receptor-gamma coactivator1alpha activities and its mitochondrial biogenetic transcription factors, nuclear respiratory factor-1 and mitochondrial transcription factor A. ginsenoside Rg3 13-16 nuclear respiratory factor 1 Homo sapiens 233-261 35059739-5 2022 Furthermore, Rg3 efficiently prevented DEX-triggered mitochondrial dysfunction of myotubes through peroxisome proliferator-activated receptor-gamma coactivator1alpha activities and its mitochondrial biogenetic transcription factors, nuclear respiratory factor-1 and mitochondrial transcription factor A. ginsenoside Rg3 13-16 transcription factor A, mitochondrial Homo sapiens 266-302 33509760-6 2021 RESULTS: Treatment of SKOV3 cells with 20(S)-Rg3 significantly upregulated VHL and downregulated DNMT3A expressions at both the mRNA and protein levels (P < 0.05) and upregulated DNMT3B expression only at the mRNA level, but did not cause significant changes in either the mRNA or protein level of DNMT1. ginsenoside Rg3 45-48 von Hippel-Lindau tumor suppressor Homo sapiens 75-78 35519255-4 2022 These results verify the effectiveness of using the THz TD-ATR spectroscopy to detect the action of G-Rg3 on glioma cells in vitro. ginsenoside Rg3 100-105 ATR serine/threonine kinase Homo sapiens 59-62 34038042-0 2021 Ginsenoside Rg3 suppresses ovarian cancer cell proliferation and invasion by inhibiting the expression of lncRNA H19. ginsenoside Rg3 0-15 H19 imprinted maternally expressed transcript Homo sapiens 113-116 34038042-10 2021 In addition, the expression of N-cadherin was downregulated, and the expression of E-cadherin was upregulated with Rg3 treatment. ginsenoside Rg3 115-118 cadherin 2 Homo sapiens 31-41 34038042-10 2021 In addition, the expression of N-cadherin was downregulated, and the expression of E-cadherin was upregulated with Rg3 treatment. ginsenoside Rg3 115-118 cadherin 1 Homo sapiens 83-93 34038042-12 2021 In terms of the mechanism, knockdown of H19 inhibited cell proliferation, migration and invasion, while overexpression of H19 reversed the inhibitory effect of Rg3 on the OC cells. ginsenoside Rg3 160-163 H19 imprinted maternally expressed transcript Homo sapiens 122-125 34038042-13 2021 In conclusion, ginsenoside Rg3 suppresses the proliferation, migration and invasion of OC cells by partially inhibiting the expression of lncRNA H19. ginsenoside Rg3 15-30 H19 imprinted maternally expressed transcript Homo sapiens 145-148 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 interleukin 1 alpha Mus musculus 111-134 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 interleukin 6 Mus musculus 136-140 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 tumor necrosis factor Mus musculus 146-173 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 arginase, liver Mus musculus 239-249 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 interleukin 10 Mus musculus 254-259 33643049-0 2021 Combination of Ginsenosides Rb2 and Rg3 Promotes Angiogenic Phenotype of Human Endothelial Cells via PI3K/Akt and MAPK/ERK Pathways. ginsenoside Rg3 36-39 AKT serine/threonine kinase 1 Homo sapiens 106-109 33643049-0 2021 Combination of Ginsenosides Rb2 and Rg3 Promotes Angiogenic Phenotype of Human Endothelial Cells via PI3K/Akt and MAPK/ERK Pathways. ginsenoside Rg3 36-39 mitogen-activated protein kinase 1 Homo sapiens 114-118 33643049-0 2021 Combination of Ginsenosides Rb2 and Rg3 Promotes Angiogenic Phenotype of Human Endothelial Cells via PI3K/Akt and MAPK/ERK Pathways. ginsenoside Rg3 36-39 mitogen-activated protein kinase 1 Homo sapiens 119-122 33643049-9 2021 mRNA expression of CXCL8, CYR61, FGF16 and FGFRL1 was significantly elevated by the Rb2/Rg3 treatment, and representative signaling pathways induced by these genes were found. ginsenoside Rg3 88-91 C-X-C motif chemokine ligand 8 Homo sapiens 19-24 33643049-9 2021 mRNA expression of CXCL8, CYR61, FGF16 and FGFRL1 was significantly elevated by the Rb2/Rg3 treatment, and representative signaling pathways induced by these genes were found. ginsenoside Rg3 88-91 cellular communication network factor 1 Homo sapiens 26-31 33643049-9 2021 mRNA expression of CXCL8, CYR61, FGF16 and FGFRL1 was significantly elevated by the Rb2/Rg3 treatment, and representative signaling pathways induced by these genes were found. ginsenoside Rg3 88-91 fibroblast growth factor 16 Homo sapiens 33-38 33643049-9 2021 mRNA expression of CXCL8, CYR61, FGF16 and FGFRL1 was significantly elevated by the Rb2/Rg3 treatment, and representative signaling pathways induced by these genes were found. ginsenoside Rg3 88-91 fibroblast growth factor receptor like 1 Homo sapiens 43-49 33643049-9 2021 mRNA expression of CXCL8, CYR61, FGF16 and FGFRL1 was significantly elevated by the Rb2/Rg3 treatment, and representative signaling pathways induced by these genes were found. ginsenoside Rg3 88-91 RB transcriptional corepressor like 2 Homo sapiens 84-87 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 C-X-C motif chemokine receptor 1 Homo sapiens 95-102 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 C-X-C motif chemokine ligand 8 Homo sapiens 103-108 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 C-X-C motif chemokine ligand 8 Homo sapiens 110-113 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 AKT serine/threonine kinase 1 Homo sapiens 129-132 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 mitogen-activated protein kinase 1 Homo sapiens 137-141 33643049-11 2021 The present study provides the hypothesis that SBP, via ginsenosides Rb2 and Rg3, involves the CXCR1/2 CXCL8 (IL8)-mediated PI3K/Akt and MAPK/ERK signaling pathways in achieving its proangiogenic effects. ginsenoside Rg3 77-80 mitogen-activated protein kinase 1 Homo sapiens 142-145 35111231-6 2022 Ginsenosides Rg3, Rk1 + Rg5, F2, and CK in the BRG group showed a higher C max, AUC(0-t), and AUC(0- ) and shorter T max (for CK) than those in the RG group. ginsenoside Rg3 0-16 cytidine/uridine monophosphate kinase 1 Homo sapiens 126-128 34025136-4 2021 Methods: Here, we investigated whether Rg3-KRGE may improve the symptoms and pathological features of myelin oligodendroglial glycoprotein (MOG)35-55 peptide - induced chronic EAE mice through improving the integrity of the BBB. ginsenoside Rg3 39-42 myelin oligodendrocyte glycoprotein Mus musculus 140-143 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 prostaglandin-endoperoxide synthase 2 Mus musculus 60-76 34025136-7 2021 Rg3-KRGE repressed increases of proinflammatory transcripts cyclooxygenase-2, inducible nitric oxide synthase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor-alpha, but enhanced expression levels of anti-inflammatory transcripts arginase-1 and IL-10 in the spinal cord following EAE induction. ginsenoside Rg3 0-3 nitric oxide synthase 2, inducible Mus musculus 78-109 33639908-0 2021 Ginsenoside Rg3 inhibits pulmonary fibrosis by preventing HIF-1alpha nuclear localisation. ginsenoside Rg3 0-15 hypoxia inducible factor 1 subunit alpha Homo sapiens 58-68 33639908-6 2021 In addition, biacore was used to detect the binding between ginsenoside Rg3 and HIF-1alpha. ginsenoside Rg3 60-75 hypoxia inducible factor 1 subunit alpha Homo sapiens 80-90 33639908-8 2021 In addition, molecular docking and biacore results indicated that ginsenoside Rg3 can bind HIF-1alpha. ginsenoside Rg3 66-81 hypoxia inducible factor 1 subunit alpha Homo sapiens 91-101 33639908-9 2021 Therefore, Ginsenoside Rg3 can slow down the progression of pulmonary fibrosis by inhibiting the nuclear localisation of HIF-1alpha. ginsenoside Rg3 11-26 hypoxia inducible factor 1 subunit alpha Homo sapiens 121-131 33509760-6 2021 RESULTS: Treatment of SKOV3 cells with 20(S)-Rg3 significantly upregulated VHL and downregulated DNMT3A expressions at both the mRNA and protein levels (P < 0.05) and upregulated DNMT3B expression only at the mRNA level, but did not cause significant changes in either the mRNA or protein level of DNMT1. ginsenoside Rg3 45-48 DNA methyltransferase 3 alpha Homo sapiens 97-103 33509760-6 2021 RESULTS: Treatment of SKOV3 cells with 20(S)-Rg3 significantly upregulated VHL and downregulated DNMT3A expressions at both the mRNA and protein levels (P < 0.05) and upregulated DNMT3B expression only at the mRNA level, but did not cause significant changes in either the mRNA or protein level of DNMT1. ginsenoside Rg3 45-48 DNA methyltransferase 3 beta Homo sapiens 179-185 33509760-6 2021 RESULTS: Treatment of SKOV3 cells with 20(S)-Rg3 significantly upregulated VHL and downregulated DNMT3A expressions at both the mRNA and protein levels (P < 0.05) and upregulated DNMT3B expression only at the mRNA level, but did not cause significant changes in either the mRNA or protein level of DNMT1. ginsenoside Rg3 45-48 DNA methyltransferase 1 Homo sapiens 298-303 33509760-9 2021 Immunohistochemisty showed a significantly increased VHL expression but a lowered DNMT3A expression in subcutaneous SKOV3 cell xenografts in 20 (S)-Rg3-treated nude mice. ginsenoside Rg3 148-151 von Hippel-Lindau tumor suppressor Homo sapiens 53-56 33371813-0 2021 Ginsenoside Rg3 Suppresses Epithelial-Mesenchymal Transition via Downregulating Notch-Hes1 Signaling in Colon Cancer Cells. ginsenoside Rg3 0-15 hes family bHLH transcription factor 1 Homo sapiens 86-90 33477683-0 2021 Ginsenoside Rg3 Prevents Oncogenic Long Noncoding RNA ATXN8OS from Inhibiting Tumor-Suppressive microRNA-424-5p in Breast Cancer Cells. ginsenoside Rg3 0-15 ATXN8 opposite strand lncRNA Homo sapiens 54-61 33477683-7 2021 The in silico miR-target-gene prediction identified 200 potential target genes of miR-424-5p, which were subsequently narrowed down to seven that underwent hypermethylation at their promoter by Rg3. ginsenoside Rg3 194-197 membrane associated ring-CH-type finger 8 Homo sapiens 14-17 33477683-7 2021 The in silico miR-target-gene prediction identified 200 potential target genes of miR-424-5p, which were subsequently narrowed down to seven that underwent hypermethylation at their promoter by Rg3. ginsenoside Rg3 194-197 microRNA 424 Homo sapiens 82-89 33477683-9 2021 When taken together, Rg3 downregulated ATXN8OS that inhibited the tumor-suppressive miR-424-5p, leading to the downregulation of the oncogenic target genes. ginsenoside Rg3 21-24 ATXN8 opposite strand lncRNA Homo sapiens 39-46 33477683-9 2021 When taken together, Rg3 downregulated ATXN8OS that inhibited the tumor-suppressive miR-424-5p, leading to the downregulation of the oncogenic target genes. ginsenoside Rg3 21-24 microRNA 424 Homo sapiens 84-91 33371813-5 2021 In addition, Rg3 suppressed the epithelial-mesenchymal transition (EMT) of HCT15 cells and SW48 cells evidenced by detecting EMT related markers E-cadherin, vimentin, and snail expression. ginsenoside Rg3 13-16 cadherin 1 Homo sapiens 145-155 33371813-5 2021 In addition, Rg3 suppressed the epithelial-mesenchymal transition (EMT) of HCT15 cells and SW48 cells evidenced by detecting EMT related markers E-cadherin, vimentin, and snail expression. ginsenoside Rg3 13-16 vimentin Homo sapiens 157-165 33371813-5 2021 In addition, Rg3 suppressed the epithelial-mesenchymal transition (EMT) of HCT15 cells and SW48 cells evidenced by detecting EMT related markers E-cadherin, vimentin, and snail expression. ginsenoside Rg3 13-16 snail family transcriptional repressor 1 Homo sapiens 171-176 33371813-7 2021 Meanwhile, the expression of NICD and Hes1 was obviously decreased in the presence of Rg3. ginsenoside Rg3 86-89 hes family bHLH transcription factor 1 Homo sapiens 38-42 33371813-9 2021 In particular, Rg3 significantly reversed IL-6-induced EMT promotion and blocked IL-6- induced NICD and Hes1 upregulations. ginsenoside Rg3 15-18 interleukin 6 Homo sapiens 42-46 33371813-9 2021 In particular, Rg3 significantly reversed IL-6-induced EMT promotion and blocked IL-6- induced NICD and Hes1 upregulations. ginsenoside Rg3 15-18 interleukin 6 Homo sapiens 81-85 33371813-9 2021 In particular, Rg3 significantly reversed IL-6-induced EMT promotion and blocked IL-6- induced NICD and Hes1 upregulations. ginsenoside Rg3 15-18 hes family bHLH transcription factor 1 Homo sapiens 104-108 33371813-10 2021 Overall, these findings suggested that Rg3 could inhibit colon cancer migration and metastasis via suppressing Notch-Hes1-EMT signaling. ginsenoside Rg3 39-42 hes family bHLH transcription factor 1 Homo sapiens 117-121 33193843-7 2020 In the heart tissues of the MI rats, Rg3 attenuated myocardial pathological changes and cell apoptosis caused by MI, decreased the gene expression levels of TNF-alpha, IL-1beta and IL-6, but increased the gene expression level of IL-10. ginsenoside Rg3 37-40 interleukin 6 Rattus norvegicus 181-185 33456471-1 2020 Osimertinib is a novel oral, potent, and irreversible epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) for treatment of advanced T790M mutation-positive advanced non-small cell lung cancer, which is commonly combined with ginsenoside Rg3 in clinic to enhance the efficacy and minimize adverse reactions. ginsenoside Rg3 243-258 epidermal growth factor receptor Rattus norvegicus 114-118 33193843-0 2020 Ginsenoside Rg3 alleviates inflammation in a rat model of myocardial infarction via the SIRT1/NF-kappaB pathway. ginsenoside Rg3 0-15 sirtuin 1 Rattus norvegicus 88-93 33193843-2 2020 Ginsenoside Rg3 (Rg3), an activator of sirtuin 1 (SIRT1), has been identified to elicit anti-inflammatory effects via the NF-kappaB pathway. ginsenoside Rg3 0-15 sirtuin 1 Rattus norvegicus 39-48 33193843-2 2020 Ginsenoside Rg3 (Rg3), an activator of sirtuin 1 (SIRT1), has been identified to elicit anti-inflammatory effects via the NF-kappaB pathway. ginsenoside Rg3 0-15 sirtuin 1 Rattus norvegicus 50-55 33193843-2 2020 Ginsenoside Rg3 (Rg3), an activator of sirtuin 1 (SIRT1), has been identified to elicit anti-inflammatory effects via the NF-kappaB pathway. ginsenoside Rg3 12-15 sirtuin 1 Rattus norvegicus 39-48 33193843-2 2020 Ginsenoside Rg3 (Rg3), an activator of sirtuin 1 (SIRT1), has been identified to elicit anti-inflammatory effects via the NF-kappaB pathway. ginsenoside Rg3 12-15 sirtuin 1 Rattus norvegicus 50-55 33193843-4 2020 In the present study, a MI rat model was established by coronary artery ligation and treated with Rg3 to explore whether Rg3 could inhibit inflammation in MI rats by inhibiting the SIRT1/NF-kappaB pathway. ginsenoside Rg3 121-124 sirtuin 1 Rattus norvegicus 181-186 33193843-5 2020 At 28 days post-MI, it was identified that Rg3 not only decreased the ST-segment ECG values in MI rats, but also significantly decreased serum LDH, CK-MB and cTnI levels in MI rats. ginsenoside Rg3 43-46 troponin I3, cardiac type Rattus norvegicus 158-162 33193843-6 2020 In addition, Rg3 also significantly decreased serum tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1beta and IL-6 levels and increased serum IL-10 levels in MI rats. ginsenoside Rg3 13-16 tumor necrosis factor Rattus norvegicus 52-79 33193843-6 2020 In addition, Rg3 also significantly decreased serum tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1beta and IL-6 levels and increased serum IL-10 levels in MI rats. ginsenoside Rg3 13-16 tumor necrosis factor Rattus norvegicus 81-90 33193843-6 2020 In addition, Rg3 also significantly decreased serum tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1beta and IL-6 levels and increased serum IL-10 levels in MI rats. ginsenoside Rg3 13-16 interleukin 1 alpha Rattus norvegicus 93-115 33193843-6 2020 In addition, Rg3 also significantly decreased serum tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1beta and IL-6 levels and increased serum IL-10 levels in MI rats. ginsenoside Rg3 13-16 interleukin 6 Rattus norvegicus 120-124 33193843-6 2020 In addition, Rg3 also significantly decreased serum tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1beta and IL-6 levels and increased serum IL-10 levels in MI rats. ginsenoside Rg3 13-16 interleukin 10 Rattus norvegicus 152-157 33193843-7 2020 In the heart tissues of the MI rats, Rg3 attenuated myocardial pathological changes and cell apoptosis caused by MI, decreased the gene expression levels of TNF-alpha, IL-1beta and IL-6, but increased the gene expression level of IL-10. ginsenoside Rg3 37-40 tumor necrosis factor Rattus norvegicus 157-166 33193843-7 2020 In the heart tissues of the MI rats, Rg3 attenuated myocardial pathological changes and cell apoptosis caused by MI, decreased the gene expression levels of TNF-alpha, IL-1beta and IL-6, but increased the gene expression level of IL-10. ginsenoside Rg3 37-40 interleukin 1 alpha Rattus norvegicus 168-176 33193843-7 2020 In the heart tissues of the MI rats, Rg3 attenuated myocardial pathological changes and cell apoptosis caused by MI, decreased the gene expression levels of TNF-alpha, IL-1beta and IL-6, but increased the gene expression level of IL-10. ginsenoside Rg3 37-40 interleukin 10 Rattus norvegicus 230-235 33193843-8 2020 In addition, the expression levels of the SIRT1 and transcription factor RelB proteins were significantly increased following Rg3 treatment, and the expression level of p-p65/p65 protein was significantly decreased in the heart tissues of MI rats with Rg3 treatment compared with that in heart tissues of MI rats without Rg3 treatment. ginsenoside Rg3 126-129 sirtuin 1 Rattus norvegicus 42-47 33193843-8 2020 In addition, the expression levels of the SIRT1 and transcription factor RelB proteins were significantly increased following Rg3 treatment, and the expression level of p-p65/p65 protein was significantly decreased in the heart tissues of MI rats with Rg3 treatment compared with that in heart tissues of MI rats without Rg3 treatment. ginsenoside Rg3 252-255 sirtuin 1 Rattus norvegicus 42-47 33193843-8 2020 In addition, the expression levels of the SIRT1 and transcription factor RelB proteins were significantly increased following Rg3 treatment, and the expression level of p-p65/p65 protein was significantly decreased in the heart tissues of MI rats with Rg3 treatment compared with that in heart tissues of MI rats without Rg3 treatment. ginsenoside Rg3 252-255 synaptotagmin 1 Rattus norvegicus 171-174 33193843-8 2020 In addition, the expression levels of the SIRT1 and transcription factor RelB proteins were significantly increased following Rg3 treatment, and the expression level of p-p65/p65 protein was significantly decreased in the heart tissues of MI rats with Rg3 treatment compared with that in heart tissues of MI rats without Rg3 treatment. ginsenoside Rg3 252-255 sirtuin 1 Rattus norvegicus 42-47 33193843-8 2020 In addition, the expression levels of the SIRT1 and transcription factor RelB proteins were significantly increased following Rg3 treatment, and the expression level of p-p65/p65 protein was significantly decreased in the heart tissues of MI rats with Rg3 treatment compared with that in heart tissues of MI rats without Rg3 treatment. ginsenoside Rg3 252-255 synaptotagmin 1 Rattus norvegicus 171-174 33193843-9 2020 In conclusion, Rg3 alleviates inflammation in a rat model of MI via the SIRT1/NF-kappaB pathway. ginsenoside Rg3 15-18 sirtuin 1 Rattus norvegicus 72-77 32653593-0 2020 20(S)-ginsenoside Rg3 promotes myoblast differentiation and protects against myotube atrophy via regulation of the Akt/mTOR/FoxO3 pathway. ginsenoside Rg3 0-21 Akt1 Drosophila melanogaster 115-118 32898934-0 2020 Ginsenoside Rg3 attenuates ovariectomy-induced osteoporosis via AMPK/mTOR signaling pathway. ginsenoside Rg3 0-15 mechanistic target of rapamycin kinase Homo sapiens 69-73 32898934-1 2020 Ginsenoside Rg3, a ginsenoside isolated from Panax ginseng, can regulate autophagy via AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) signaling pathway. ginsenoside Rg3 0-15 protein kinase AMP-activated non-catalytic subunit beta 1 Homo sapiens 147-151 32898934-1 2020 Ginsenoside Rg3, a ginsenoside isolated from Panax ginseng, can regulate autophagy via AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) signaling pathway. ginsenoside Rg3 0-15 mechanistic target of rapamycin kinase Homo sapiens 152-156 32898934-7 2020 10 and 20 mumol/L Rg3, which had no significant effect on cell viability and significantly affected AMPK/mTOR signaling, were chosen for further analysis. ginsenoside Rg3 18-21 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 100-104 32898934-7 2020 10 and 20 mumol/L Rg3, which had no significant effect on cell viability and significantly affected AMPK/mTOR signaling, were chosen for further analysis. ginsenoside Rg3 18-21 mechanistic target of rapamycin kinase Mus musculus 105-109 32898934-11 2020 Rg3 significantly alleviated OVX-induced BW increases, BMD declines and histological changes of femur tissues, promoted osteogenesis, autophagy, and AMPK signaling, but inhibited mTOR signaling in vivo. ginsenoside Rg3 0-3 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 149-153 32898934-11 2020 Rg3 significantly alleviated OVX-induced BW increases, BMD declines and histological changes of femur tissues, promoted osteogenesis, autophagy, and AMPK signaling, but inhibited mTOR signaling in vivo. ginsenoside Rg3 0-3 mechanistic target of rapamycin kinase Mus musculus 179-183 33091036-6 2020 Mechanistic investigations suggested that these inhibitory effects of Rg3 on MDSCs and corresponding cancer progression depend upon suppression of the STAT3-dependent pathway, tumor-derived cytokines, and the NOTCH signaling pathway. ginsenoside Rg3 70-73 signal transducer and activator of transcription 3 Mus musculus 151-156 33116644-10 2020 In addition, we found that ginsenoside Rg3 can block the interaction of NRP1 and FN1 and inhibit the progression of gastric cancer. ginsenoside Rg3 27-42 neuropilin 1 Homo sapiens 72-76 33116644-10 2020 In addition, we found that ginsenoside Rg3 can block the interaction of NRP1 and FN1 and inhibit the progression of gastric cancer. ginsenoside Rg3 27-42 fibronectin 1 Homo sapiens 81-84 32898934-12 2020 Moreover, Rg3 significantly enhanced AMPK signaling, autophagy, osteogenic differentiation, and mineralization, but suppressed mTOR signaling in vitro. ginsenoside Rg3 10-13 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 37-41 32898934-12 2020 Moreover, Rg3 significantly enhanced AMPK signaling, autophagy, osteogenic differentiation, and mineralization, but suppressed mTOR signaling in vitro. ginsenoside Rg3 10-13 mechanistic target of rapamycin kinase Mus musculus 127-131 32898934-13 2020 However, Compound C significantly reversed Rg3-induced alterations in vitro, indicating that Rg3 regulated autophagy, osteogenic differentiation, and mineralization via AMPK/mTOR signaling. ginsenoside Rg3 93-96 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 169-173 32898934-13 2020 However, Compound C significantly reversed Rg3-induced alterations in vitro, indicating that Rg3 regulated autophagy, osteogenic differentiation, and mineralization via AMPK/mTOR signaling. ginsenoside Rg3 93-96 mechanistic target of rapamycin kinase Mus musculus 174-178 32898934-14 2020 Hence, it was speculated that Rg3 might attenuate OVX-induced osteoporosis via AMPK/mTOR signaling pathway. ginsenoside Rg3 30-33 mechanistic target of rapamycin kinase Mus musculus 84-88 33192122-7 2020 Interestingly, KRGE or representative ginsenosides (Rb1, Rg1, and Rg3(s)) inhibited the activity of inflammatory enzymes cyclooxygenase-2 and iNOS, cytosolic p-IkB, and nuclear p-nuclear factor kappa-light-chain-enhancer of activated B in lipopolysaccharide-induced RAW264.7 cells, whereas they increased nuclear factor erythroid-derived 2-related factor 2 nuclear translocation. ginsenoside Rg3 66-69 prostaglandin-endoperoxide synthase 2 Mus musculus 121-137 33192122-7 2020 Interestingly, KRGE or representative ginsenosides (Rb1, Rg1, and Rg3(s)) inhibited the activity of inflammatory enzymes cyclooxygenase-2 and iNOS, cytosolic p-IkB, and nuclear p-nuclear factor kappa-light-chain-enhancer of activated B in lipopolysaccharide-induced RAW264.7 cells, whereas they increased nuclear factor erythroid-derived 2-related factor 2 nuclear translocation. ginsenoside Rg3 66-69 nitric oxide synthase 2, inducible Mus musculus 142-146 32653593-0 2020 20(S)-ginsenoside Rg3 promotes myoblast differentiation and protects against myotube atrophy via regulation of the Akt/mTOR/FoxO3 pathway. ginsenoside Rg3 0-21 Megator Drosophila melanogaster 119-123 32653593-0 2020 20(S)-ginsenoside Rg3 promotes myoblast differentiation and protects against myotube atrophy via regulation of the Akt/mTOR/FoxO3 pathway. ginsenoside Rg3 0-21 forkhead box O3 Mus musculus 124-129 32653593-4 2020 This effect was likely caused by S-Rg3 treatment-induced promotion of Akt/mTOR phosphorylation and inhibition of FoxO3 nuclear transcription. ginsenoside Rg3 33-38 Akt1 Drosophila melanogaster 70-73 32653593-4 2020 This effect was likely caused by S-Rg3 treatment-induced promotion of Akt/mTOR phosphorylation and inhibition of FoxO3 nuclear transcription. ginsenoside Rg3 33-38 Megator Drosophila melanogaster 74-78 32653593-4 2020 This effect was likely caused by S-Rg3 treatment-induced promotion of Akt/mTOR phosphorylation and inhibition of FoxO3 nuclear transcription. ginsenoside Rg3 33-38 forkhead box O3 Mus musculus 113-118 32653593-6 2020 Our study provides a mechanistic framework for understanding how S-Rg3 enhances myoblast differentiation and inhibits myotube atrophy through activation of the Akt/mTOR/FoxO3 signaling pathway, as demonstrated in vitro in C2C12 cells and in vivo in fruit flies. ginsenoside Rg3 65-70 thymoma viral proto-oncogene 1 Mus musculus 160-163 32653593-6 2020 Our study provides a mechanistic framework for understanding how S-Rg3 enhances myoblast differentiation and inhibits myotube atrophy through activation of the Akt/mTOR/FoxO3 signaling pathway, as demonstrated in vitro in C2C12 cells and in vivo in fruit flies. ginsenoside Rg3 65-70 mechanistic target of rapamycin kinase Mus musculus 164-168 32653593-6 2020 Our study provides a mechanistic framework for understanding how S-Rg3 enhances myoblast differentiation and inhibits myotube atrophy through activation of the Akt/mTOR/FoxO3 signaling pathway, as demonstrated in vitro in C2C12 cells and in vivo in fruit flies. ginsenoside Rg3 65-70 forkhead box O3 Mus musculus 169-174 32945504-8 2020 In addition, the PI3K/AKT signaling pathway in colon cancer cells was suppressed by Rg3 and 5-FU. ginsenoside Rg3 84-87 AKT serine/threonine kinase 1 Homo sapiens 22-25 32945504-0 2020 Ginsenoside Rg3 enhances the anticancer effect of 5-FU in colon cancer cells via the PI3K/AKT pathway. ginsenoside Rg3 0-15 AKT serine/threonine kinase 1 Homo sapiens 90-93 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 apoptotic peptidase activating factor 1 Homo sapiens 131-136 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 caspase 9 Homo sapiens 137-146 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 caspase 3 Homo sapiens 147-156 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 cyclin D1 Homo sapiens 259-268 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 cyclin dependent kinase 2 Homo sapiens 270-274 32945504-7 2020 We also found that combined treatment of Rg3 and 5-FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. ginsenoside Rg3 41-44 cyclin dependent kinase 4 Homo sapiens 279-283 32945504-11 2020 Collectively, the present study demonstrated that ginsenoside Rg3 enhances the anticancer effect of 5-FU in colon cancer cells via the PI3K/AKT pathway. ginsenoside Rg3 50-65 AKT serine/threonine kinase 1 Homo sapiens 140-143 32372871-2 2020 Although our previous studies have demonstrated that ginsenoside Rg3 significantly attenuated the activation of NMDA receptors (NMDARs) in hippocampal neurons, the effects of ginsenosides Rg5 and Rk1, which are derived from heat-mediated dehydration of ginsenoside Rg3, on neuronal NMDARs have not yet been elucidated. ginsenoside Rg3 253-268 kallikrein 1-related peptidase B3 Rattus norvegicus 196-199 32714406-12 2020 Molecular docking technology suggests that ginsenoside Rg3 and ginsenoside Rh2 can easily establish good docking modes and have a high affinity with EpCAM. ginsenoside Rg3 43-58 epithelial cell adhesion molecule Homo sapiens 149-154 32714406-13 2020 The 6"-hydroxyl and 6""-hydroxyl on the 3-glycosyl of ginsenoside Rg3 are prone to form hydrogen bonds (Lys151 and Lys221) with the active sites of EpCAM ligand binding domain. ginsenoside Rg3 54-69 epithelial cell adhesion molecule Homo sapiens 148-153 32714406-15 2020 The formation of hydrogen bonds plays an important role in binding of ginsenoside Rg3 and ginsenoside Rh2 to EpCAM, as well as the stability of EpCAM conformation. ginsenoside Rg3 70-85 epithelial cell adhesion molecule Homo sapiens 109-114 32884696-6 2020 PTEN and P53 protein expression levels were significantly increased, and p-PI3K and AKT protein expression levels were significantly depressed in ginsenoside Rg3-treated groups in a dose-dependent manner (p < .05). ginsenoside Rg3 146-161 phosphatase and tensin homolog Homo sapiens 0-4 32884696-6 2020 PTEN and P53 protein expression levels were significantly increased, and p-PI3K and AKT protein expression levels were significantly depressed in ginsenoside Rg3-treated groups in a dose-dependent manner (p < .05). ginsenoside Rg3 146-161 AKT serine/threonine kinase 1 Homo sapiens 84-87 32884696-7 2020 Conclusion: Ginsenoside Rg3 suppresses gastric cancer via regulation of the PTEN/p-PI3K/AKT pathway. ginsenoside Rg3 12-27 phosphatase and tensin homolog Homo sapiens 76-80 32884696-7 2020 Conclusion: Ginsenoside Rg3 suppresses gastric cancer via regulation of the PTEN/p-PI3K/AKT pathway. ginsenoside Rg3 12-27 AKT serine/threonine kinase 1 Homo sapiens 88-91 32532964-5 2020 Treatment with G-Rg3 alleviated hepatic pathological changes and reversed hepatic fibrosis in the TAA-chronic models with decreased deposition of collagen fibers, reduced expression of HSCs activation marker (alpha-SMA), and reduced secretion of profibrogenic factors (TGF-beta1). ginsenoside Rg3 15-20 transforming growth factor, beta 1 Rattus norvegicus 269-278 32532964-8 2020 G-Rg3 dose-dependently inhibited autophagy in vitro with less expression of p62 and fewer LC3a transformation into LC3b in inflammatory inducer lipopolysaccharide (LPS)-induced rat HSC-T6 cells. ginsenoside Rg3 0-5 KH RNA binding domain containing, signal transduction associated 1 Rattus norvegicus 76-79 32532964-9 2020 Furthermore, G-Rg3 enhanced the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) in vivo and in vitro. ginsenoside Rg3 13-18 phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit beta Rattus norvegicus 51-80 32532964-9 2020 Furthermore, G-Rg3 enhanced the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) in vivo and in vitro. ginsenoside Rg3 13-18 AKT serine/threonine kinase 1 Rattus norvegicus 110-113 32532964-10 2020 Besides, mTOR inhibitor Rapamycin and PI3K inhibitors LY294002 were employed in LPS-treated HSC-T6 cell cultures to verify that Rg3 partially reversed the increase in autophagy in hepatic fibrosis in vitro. ginsenoside Rg3 128-131 mechanistic target of rapamycin kinase Rattus norvegicus 9-13 31916386-1 2020 In the present study, we found that Ginsenoside Rg3 attenuated the stemness of non-small cell lung cancer (NSCLC) cells, evident by decreasing spheroid formation ability, ALDH1 activity and stemness marker expression. ginsenoside Rg3 36-51 aldehyde dehydrogenase 1 family member A1 Homo sapiens 171-176 31916386-4 2020 RNA-sequencing revealed that Hippo signaling was shown on the top of the most upregulated pathways regulated by Ginsenoside Rg3 in NSCLC cells, and YAP/TAZ expression was suppressed by Ginsenoside Rg3. ginsenoside Rg3 185-200 Yes1 associated transcriptional regulator Homo sapiens 148-151 31916386-4 2020 RNA-sequencing revealed that Hippo signaling was shown on the top of the most upregulated pathways regulated by Ginsenoside Rg3 in NSCLC cells, and YAP/TAZ expression was suppressed by Ginsenoside Rg3. ginsenoside Rg3 185-200 tafazzin, phospholipid-lysophospholipid transacylase Homo sapiens 152-155 32104955-0 2020 Ginsenoside Rg3 suppresses the growth of gemcitabine-resistant pancreatic cancer cells by upregulating lncRNA-CASC2 and activating PTEN signaling. ginsenoside Rg3 0-15 cancer susceptibility 2 Homo sapiens 110-115 32104955-0 2020 Ginsenoside Rg3 suppresses the growth of gemcitabine-resistant pancreatic cancer cells by upregulating lncRNA-CASC2 and activating PTEN signaling. ginsenoside Rg3 0-15 phosphatase and tensin homolog Homo sapiens 131-135 32104955-7 2020 The level of long noncoding RNA cancer susceptibility candidate 2 (CASC2) and PTEN expression was upregulated by the ginsenoside Rg3 treatment, and CASC2/PTEN signaling was involved in the ginsenoside Rg3-induced cell growth suppression and apoptosis in GEM-resistant pancreatic cancer cells. ginsenoside Rg3 117-132 cancer susceptibility 2 Homo sapiens 67-72 32104955-7 2020 The level of long noncoding RNA cancer susceptibility candidate 2 (CASC2) and PTEN expression was upregulated by the ginsenoside Rg3 treatment, and CASC2/PTEN signaling was involved in the ginsenoside Rg3-induced cell growth suppression and apoptosis in GEM-resistant pancreatic cancer cells. ginsenoside Rg3 117-132 phosphatase and tensin homolog Homo sapiens 78-82 32104955-7 2020 The level of long noncoding RNA cancer susceptibility candidate 2 (CASC2) and PTEN expression was upregulated by the ginsenoside Rg3 treatment, and CASC2/PTEN signaling was involved in the ginsenoside Rg3-induced cell growth suppression and apoptosis in GEM-resistant pancreatic cancer cells. ginsenoside Rg3 189-204 cancer susceptibility 2 Homo sapiens 67-72 32104955-7 2020 The level of long noncoding RNA cancer susceptibility candidate 2 (CASC2) and PTEN expression was upregulated by the ginsenoside Rg3 treatment, and CASC2/PTEN signaling was involved in the ginsenoside Rg3-induced cell growth suppression and apoptosis in GEM-resistant pancreatic cancer cells. ginsenoside Rg3 189-204 cancer susceptibility 2 Homo sapiens 148-153 32104955-7 2020 The level of long noncoding RNA cancer susceptibility candidate 2 (CASC2) and PTEN expression was upregulated by the ginsenoside Rg3 treatment, and CASC2/PTEN signaling was involved in the ginsenoside Rg3-induced cell growth suppression and apoptosis in GEM-resistant pancreatic cancer cells. ginsenoside Rg3 189-204 phosphatase and tensin homolog Homo sapiens 154-158 32372871-5 2020 However, ginsenoside Rk1 inhibited NMDARs most effectively among the five compounds (Rg3, Rg5, Rk1, Rg5/Rk1 mixture, and protopanaxadiol) tested in cultured hippocampal neurons. ginsenoside Rg3 85-88 kallikrein 1-related peptidase B3 Rattus norvegicus 21-24 32258169-10 2020 Mechanistically, 20(S)-Rg3 dramatically downregulated the expression of TGF-beta1, NF-kappaB65, and TNF-alpha in the kidney. ginsenoside Rg3 17-26 transforming growth factor, beta 1 Rattus norvegicus 72-81 32365500-7 2020 Rg3 and C-K were further identified for their interaction efficacy with PD-1/PD-L1, which supported our results demonstrating the blocking activity of these compounds against PD-1/PD-L1 binding interactions. ginsenoside Rg3 0-3 programmed cell death 1 Homo sapiens 72-76 32365500-7 2020 Rg3 and C-K were further identified for their interaction efficacy with PD-1/PD-L1, which supported our results demonstrating the blocking activity of these compounds against PD-1/PD-L1 binding interactions. ginsenoside Rg3 0-3 CD274 molecule Homo sapiens 77-82 32365500-7 2020 Rg3 and C-K were further identified for their interaction efficacy with PD-1/PD-L1, which supported our results demonstrating the blocking activity of these compounds against PD-1/PD-L1 binding interactions. ginsenoside Rg3 0-3 programmed cell death 1 Homo sapiens 175-179 32365500-7 2020 Rg3 and C-K were further identified for their interaction efficacy with PD-1/PD-L1, which supported our results demonstrating the blocking activity of these compounds against PD-1/PD-L1 binding interactions. ginsenoside Rg3 0-3 CD274 molecule Homo sapiens 180-185 32365500-8 2020 Collectively, our findings suggest that some ginsenosides, including Rg3 and C-K, inhibit PD-1/PD-L1 binding interactions. ginsenoside Rg3 69-72 programmed cell death 1 Homo sapiens 90-94 32365500-8 2020 Collectively, our findings suggest that some ginsenosides, including Rg3 and C-K, inhibit PD-1/PD-L1 binding interactions. ginsenoside Rg3 69-72 CD274 molecule Homo sapiens 95-100 32275817-0 2020 Ginsenoside Rg3 Alleviates Complete Freund"s Adjuvant-Induced Rheumatoid Arthritis in Mice by Regulating CD4+CD25+Foxp3+Treg Cells. ginsenoside Rg3 0-15 CD4 antigen Mus musculus 105-108 32275817-0 2020 Ginsenoside Rg3 Alleviates Complete Freund"s Adjuvant-Induced Rheumatoid Arthritis in Mice by Regulating CD4+CD25+Foxp3+Treg Cells. ginsenoside Rg3 0-15 interleukin 2 receptor, alpha chain Mus musculus 109-113 32275817-0 2020 Ginsenoside Rg3 Alleviates Complete Freund"s Adjuvant-Induced Rheumatoid Arthritis in Mice by Regulating CD4+CD25+Foxp3+Treg Cells. ginsenoside Rg3 0-15 forkhead box P3 Mus musculus 114-119 32390845-0 2020 Ginsenoside Rg3 Alleviates ox-LDL Induced Endothelial Dysfunction and Prevents Atherosclerosis in ApoE-/- Mice by Regulating PPARgamma/FAK Signaling Pathway. ginsenoside Rg3 0-15 apolipoprotein E Mus musculus 98-102 32390845-0 2020 Ginsenoside Rg3 Alleviates ox-LDL Induced Endothelial Dysfunction and Prevents Atherosclerosis in ApoE-/- Mice by Regulating PPARgamma/FAK Signaling Pathway. ginsenoside Rg3 0-15 peroxisome proliferator activated receptor gamma Mus musculus 125-134 32390845-0 2020 Ginsenoside Rg3 Alleviates ox-LDL Induced Endothelial Dysfunction and Prevents Atherosclerosis in ApoE-/- Mice by Regulating PPARgamma/FAK Signaling Pathway. ginsenoside Rg3 0-15 PTK2 protein tyrosine kinase 2 Mus musculus 135-138 32390845-5 2020 GW9662, the PPARgamma-specific inhibitor, can repressed the effects of Rg3 on ox-LDL-stimulated HUVECs. ginsenoside Rg3 71-74 peroxisome proliferator activated receptor gamma Homo sapiens 12-21 32390845-6 2020 For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE-/- mice fed with HFD, up-regulated PPARgamma, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. ginsenoside Rg3 19-22 apolipoprotein E Mus musculus 85-89 32390845-6 2020 For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE-/- mice fed with HFD, up-regulated PPARgamma, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. ginsenoside Rg3 19-22 peroxisome proliferator activated receptor gamma Mus musculus 125-134 32390845-6 2020 For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE-/- mice fed with HFD, up-regulated PPARgamma, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. ginsenoside Rg3 19-22 PTK2 protein tyrosine kinase 2 Mus musculus 161-164 32390845-6 2020 For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE-/- mice fed with HFD, up-regulated PPARgamma, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. ginsenoside Rg3 19-22 vascular cell adhesion molecule 1 Mus musculus 204-210 32390845-6 2020 For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE-/- mice fed with HFD, up-regulated PPARgamma, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. ginsenoside Rg3 19-22 intercellular adhesion molecule 1 Mus musculus 212-218 32390845-7 2020 We conclude that Rg3 may protect endothelial cells and inhibit atherosclerosis by up-regulating PPARgamma via repressing FAK-mediated pathways, indicating that Rg3 have good potential in preventing dyslipidemia induced atherosclerosis. ginsenoside Rg3 17-20 peroxisome proliferator activated receptor gamma Mus musculus 96-105 32390845-7 2020 We conclude that Rg3 may protect endothelial cells and inhibit atherosclerosis by up-regulating PPARgamma via repressing FAK-mediated pathways, indicating that Rg3 have good potential in preventing dyslipidemia induced atherosclerosis. ginsenoside Rg3 17-20 PTK2 protein tyrosine kinase 2 Mus musculus 121-124 32390845-7 2020 We conclude that Rg3 may protect endothelial cells and inhibit atherosclerosis by up-regulating PPARgamma via repressing FAK-mediated pathways, indicating that Rg3 have good potential in preventing dyslipidemia induced atherosclerosis. ginsenoside Rg3 160-163 peroxisome proliferator activated receptor gamma Mus musculus 96-105 32390845-7 2020 We conclude that Rg3 may protect endothelial cells and inhibit atherosclerosis by up-regulating PPARgamma via repressing FAK-mediated pathways, indicating that Rg3 have good potential in preventing dyslipidemia induced atherosclerosis. ginsenoside Rg3 160-163 PTK2 protein tyrosine kinase 2 Mus musculus 121-124 32420095-7 2020 Results: We found that Rg3 significantly reduced ISO induced myocardial injury indicated by the downregulation of serum BNP and LDH. ginsenoside Rg3 23-26 natriuretic peptide type B Mus musculus 120-123 32258169-10 2020 Mechanistically, 20(S)-Rg3 dramatically downregulated the expression of TGF-beta1, NF-kappaB65, and TNF-alpha in the kidney. ginsenoside Rg3 17-26 tumor necrosis factor Rattus norvegicus 100-109 31817227-0 2019 Fermented Korean Red Ginseng Extract Enriched in Rd and Rg3 Protects against Non-Alcoholic Fatty Liver Disease through Regulation of mTORC1. ginsenoside Rg3 56-59 CREB regulated transcription coactivator 1 Mus musculus 133-139 32076440-1 2020 Meyer (Rg3) Ameliorates Gastric Precancerous Lesions in Atp4a-/- Mice via Inhibition of Glycolysis through PI3K/AKT/miRNA-21 Pathway. ginsenoside Rg3 7-10 ATPase, H+/K+ exchanging, gastric, alpha polypeptide Mus musculus 56-61 32076440-1 2020 Meyer (Rg3) Ameliorates Gastric Precancerous Lesions in Atp4a-/- Mice via Inhibition of Glycolysis through PI3K/AKT/miRNA-21 Pathway. ginsenoside Rg3 7-10 thymoma viral proto-oncogene 1 Mus musculus 112-115 32076440-6 2020 Herein, the aim of the present study was to clarify the potential role for glycolysis pathogenesis in Rg3-treated GPL in Atp4a-/- mice. ginsenoside Rg3 102-105 ATPase, H+/K+ exchanging, gastric, alpha polypeptide Mus musculus 121-126 32076440-11 2020 Furthermore, the increased gene levels of Bcl-2 and caspase-3 were attenuated in Rg3-treated GPL mice. ginsenoside Rg3 81-84 B cell leukemia/lymphoma 2 Mus musculus 42-47 32076440-11 2020 Furthermore, the increased gene levels of Bcl-2 and caspase-3 were attenuated in Rg3-treated GPL mice. ginsenoside Rg3 81-84 caspase 3 Mus musculus 52-61 32076440-12 2020 In conclusion, the findings of this study imply that abnormal glycolysis in GPL mice was relieved by Rg3 via regulation of the expressions of PI3K, AKT, mTOR, HIF-1alpha, LDHA, HK-II, and miRNA-21. ginsenoside Rg3 101-104 thymoma viral proto-oncogene 1 Mus musculus 148-151 32076440-12 2020 In conclusion, the findings of this study imply that abnormal glycolysis in GPL mice was relieved by Rg3 via regulation of the expressions of PI3K, AKT, mTOR, HIF-1alpha, LDHA, HK-II, and miRNA-21. ginsenoside Rg3 101-104 mechanistic target of rapamycin kinase Mus musculus 153-157 32076440-12 2020 In conclusion, the findings of this study imply that abnormal glycolysis in GPL mice was relieved by Rg3 via regulation of the expressions of PI3K, AKT, mTOR, HIF-1alpha, LDHA, HK-II, and miRNA-21. ginsenoside Rg3 101-104 hypoxia inducible factor 1, alpha subunit Mus musculus 159-169 32076440-12 2020 In conclusion, the findings of this study imply that abnormal glycolysis in GPL mice was relieved by Rg3 via regulation of the expressions of PI3K, AKT, mTOR, HIF-1alpha, LDHA, HK-II, and miRNA-21. ginsenoside Rg3 101-104 lactate dehydrogenase A Mus musculus 171-175 32076440-12 2020 In conclusion, the findings of this study imply that abnormal glycolysis in GPL mice was relieved by Rg3 via regulation of the expressions of PI3K, AKT, mTOR, HIF-1alpha, LDHA, HK-II, and miRNA-21. ginsenoside Rg3 101-104 hexokinase 2 Mus musculus 177-182 31914640-0 2020 Ginsenoside Rg3 suppresses the NLRP3 inflammasome activation through inhibition of its assembly. ginsenoside Rg3 0-15 NLR family, pyrin domain containing 3 Mus musculus 31-36 31914640-4 2020 Here we show that ginsenoside Rg3 blocks IL-1beta secretion and caspase-1 activation through inhibiting LPS priming and the NLRP3 inflammasome activation in human and mouse macrophages. ginsenoside Rg3 18-33 interleukin 1 beta Homo sapiens 41-49 31914640-4 2020 Here we show that ginsenoside Rg3 blocks IL-1beta secretion and caspase-1 activation through inhibiting LPS priming and the NLRP3 inflammasome activation in human and mouse macrophages. ginsenoside Rg3 18-33 caspase 1 Homo sapiens 64-73 31914640-4 2020 Here we show that ginsenoside Rg3 blocks IL-1beta secretion and caspase-1 activation through inhibiting LPS priming and the NLRP3 inflammasome activation in human and mouse macrophages. ginsenoside Rg3 18-33 NLR family pyrin domain containing 3 Homo sapiens 124-129 31914640-5 2020 Rg3 specifically inhibits activation of NLRP3 but not the NLRC4 or AIM2 inflammasomes. ginsenoside Rg3 0-3 NLR family, pyrin domain containing 3 Mus musculus 40-45 31914640-7 2020 Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. ginsenoside Rg3 17-20 NIMA (never in mitosis gene a)-related expressed kinase 7 Mus musculus 31-35 31914640-7 2020 Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. ginsenoside Rg3 17-20 NLR family, pyrin domain containing 3 Mus musculus 36-41 31914640-7 2020 Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. ginsenoside Rg3 17-20 NLR family, pyrin domain containing 3 Mus musculus 81-86 31914640-7 2020 Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. ginsenoside Rg3 17-20 steroid sulfatase Mus musculus 87-90 31914640-7 2020 Mechanistically, Rg3 abrogates NEK7-NLRP3 interaction, and subsequently inhibits NLRP3-ASC interaction, ASC oligomerization, and speckle formation. ginsenoside Rg3 17-20 steroid sulfatase Mus musculus 104-107 31914640-8 2020 More importantly, Rg3 can reduce IL-1beta secretion induced by LPS in mice and protect mice from lethal endotoxic shock. ginsenoside Rg3 18-21 interleukin 1 beta Mus musculus 33-41 31914640-9 2020 Thus, our findings reveal an anti-inflammatory mechanism for Rg3 and suggest its potential use in NLRP3-driven diseases. ginsenoside Rg3 61-64 NLR family, pyrin domain containing 3 Mus musculus 98-103 30601070-1 2020 The purpose of this work was to prepare and characterize Angiopep-2 functionalized ginsenoside-Rg3 loaded nanoparticles (ANG-Rg3-NP) and evaluate the therapeutic effect on C6 glioma cells. ginsenoside Rg3 95-98 angiogenin Homo sapiens 121-124 31783047-7 2020 We identified FoxO3a as the therapeutic target of Rg3 using molecular docking and gene silencing. ginsenoside Rg3 50-53 forkhead box O3 Homo sapiens 14-20 31783047-9 2020 The mechanism of action was established as Rg3 targeting of FoxO3a, which inhibited the promotion of oxidative stress, inflammation, and fibrosis via downstream signaling pathways. ginsenoside Rg3 43-46 forkhead box O3 Homo sapiens 60-66 31563698-8 2020 The treatment with Rg3 significantly diminished the level of NF-kappaB p65 subunit as well as TNF-alpha induced nuclear translocation of NF-kappaB p65 subunit in ectopic endometriotic cells. ginsenoside Rg3 19-22 RELA proto-oncogene, NF-kB subunit Homo sapiens 61-74 31563698-8 2020 The treatment with Rg3 significantly diminished the level of NF-kappaB p65 subunit as well as TNF-alpha induced nuclear translocation of NF-kappaB p65 subunit in ectopic endometriotic cells. ginsenoside Rg3 19-22 tumor necrosis factor Homo sapiens 94-103 31563698-8 2020 The treatment with Rg3 significantly diminished the level of NF-kappaB p65 subunit as well as TNF-alpha induced nuclear translocation of NF-kappaB p65 subunit in ectopic endometriotic cells. ginsenoside Rg3 19-22 RELA proto-oncogene, NF-kB subunit Homo sapiens 137-150 31563698-9 2020 Moreover, Rg3 upregulated the expression of caspases3 but suppressed the expression of VEGF. ginsenoside Rg3 10-13 vascular endothelial growth factor A Homo sapiens 87-91 31629252-0 2019 Ginsenoside Rg3 regulates DNA damage in non-small cell lung cancer cells by activating VRK1/P53BP1 pathway. ginsenoside Rg3 0-15 vaccinia related kinase 1 Mus musculus 87-91 31629252-0 2019 Ginsenoside Rg3 regulates DNA damage in non-small cell lung cancer cells by activating VRK1/P53BP1 pathway. ginsenoside Rg3 0-15 transformation related protein 53 binding protein 1 Mus musculus 92-98 31629252-9 2019 Ginsenoside Rg3 increased the mRNA and protein expression of VRK1 in NSCLC cells as measured by RT-qPCR and western blot, respectively. ginsenoside Rg3 0-15 vaccinia related kinase 1 Mus musculus 61-65 31629252-10 2019 These findings suggests that ginsenoside Rg3 regulates VRK1 signaling. ginsenoside Rg3 29-44 vaccinia related kinase 1 Mus musculus 55-59 31629252-13 2019 Taken together, these results show that ginsenoside Rg3 upregulate VRK1 expression and P53BP1 foci formation in response to DNA damage thereby inhibiting the tumorigenesis and viability of cancer cells. ginsenoside Rg3 40-55 vaccinia related kinase 1 Mus musculus 67-71 31629252-13 2019 Taken together, these results show that ginsenoside Rg3 upregulate VRK1 expression and P53BP1 foci formation in response to DNA damage thereby inhibiting the tumorigenesis and viability of cancer cells. ginsenoside Rg3 40-55 transformation related protein 53 binding protein 1 Mus musculus 87-93 31658733-9 2019 20(S)-GRg3 blocked the late stage of autophagy (fusion of lysosomes and degradation of autophagic lysosomes), including a decrease in acidic vesicular organelle fluorescence, increased LC3 I-II conversion, accumulation of EGFP-LC3 fluorescence, GFP-mRFP-LC3 red-green fluorescence ratio, degradation of the substrate p62, and loss of the balance between autophagy and apoptosis, which induced apoptosis. ginsenoside Rg3 6-10 nucleoporin 62 Homo sapiens 317-320 31631976-0 2019 Erratum: 20(S)-Ginsenoside Rg3 Promotes Senescence And Apoptosis In Gallbladder Cancer Cells Via The P53 Pathway [Corrigendum]. ginsenoside Rg3 9-30 tumor protein p53 Homo sapiens 101-104 31695563-9 2019 Furthermore, Glide XP from Schrodinger was used for molecular docking to clarify the interactions between 20 (S)-ginsenoside Rg3 and EGF and NOS2. ginsenoside Rg3 109-128 epidermal growth factor Mus musculus 133-136 31695563-9 2019 Furthermore, Glide XP from Schrodinger was used for molecular docking to clarify the interactions between 20 (S)-ginsenoside Rg3 and EGF and NOS2. ginsenoside Rg3 109-128 nitric oxide synthase 2, inducible Mus musculus 141-145 31695563-10 2019 Results: 20 (S)-ginsenoside Rg3 significantly decreased the UI scores and UI ratios in all the three GU models, and it demonstrated antiulcer effects by decreasing the ET-1 and NOS2 levels and increasing the NO, superoxide dismutase, EGF, and epidermal growth factor receptor levels. ginsenoside Rg3 12-31 endothelin 1 Mus musculus 168-172 31695563-10 2019 Results: 20 (S)-ginsenoside Rg3 significantly decreased the UI scores and UI ratios in all the three GU models, and it demonstrated antiulcer effects by decreasing the ET-1 and NOS2 levels and increasing the NO, superoxide dismutase, EGF, and epidermal growth factor receptor levels. ginsenoside Rg3 12-31 nitric oxide synthase 2, inducible Mus musculus 177-181 31695563-10 2019 Results: 20 (S)-ginsenoside Rg3 significantly decreased the UI scores and UI ratios in all the three GU models, and it demonstrated antiulcer effects by decreasing the ET-1 and NOS2 levels and increasing the NO, superoxide dismutase, EGF, and epidermal growth factor receptor levels. ginsenoside Rg3 12-31 epidermal growth factor Mus musculus 234-237 31695563-10 2019 Results: 20 (S)-ginsenoside Rg3 significantly decreased the UI scores and UI ratios in all the three GU models, and it demonstrated antiulcer effects by decreasing the ET-1 and NOS2 levels and increasing the NO, superoxide dismutase, EGF, and epidermal growth factor receptor levels. ginsenoside Rg3 12-31 epidermal growth factor receptor Mus musculus 243-275 31700260-0 2019 Ginsenoside Rg3 and Korean Red Ginseng extract epigenetically regulate the tumor-related long noncoding RNAs RFX3-AS1 and STXBP5-AS1. ginsenoside Rg3 0-15 RFX3 divergent transcript Homo sapiens 109-117 31700260-0 2019 Ginsenoside Rg3 and Korean Red Ginseng extract epigenetically regulate the tumor-related long noncoding RNAs RFX3-AS1 and STXBP5-AS1. ginsenoside Rg3 0-15 prostaglandin D2 receptor Homo sapiens 122-132 31700260-2 2019 Here, we investigate two long noncoding RNAs (lncRNAs), STXBP5-AS1 and RFX3-AS1, which are hypomethylated and hypermethylated in the promoter region by Rg3 in MCF-7 cancer cells. ginsenoside Rg3 152-155 STXBP5 antisense RNA 1 Homo sapiens 56-66 31700260-2 2019 Here, we investigate two long noncoding RNAs (lncRNAs), STXBP5-AS1 and RFX3-AS1, which are hypomethylated and hypermethylated in the promoter region by Rg3 in MCF-7 cancer cells. ginsenoside Rg3 152-155 RFX3 divergent transcript Homo sapiens 71-79 31700260-7 2019 Results: STXBP5-AS1 and RFX3-AS1 exhibited anti- and pro-proliferation effects, respectively, in the cancer cells, and the effects of Rg3 and KRG extract on apoptosis and cell proliferation were weakened after deregulating the lncRNAs. ginsenoside Rg3 134-137 syntaxin binding protein 5 Homo sapiens 9-15 31700260-7 2019 Results: STXBP5-AS1 and RFX3-AS1 exhibited anti- and pro-proliferation effects, respectively, in the cancer cells, and the effects of Rg3 and KRG extract on apoptosis and cell proliferation were weakened after deregulating the lncRNAs. ginsenoside Rg3 134-137 RFX3 divergent transcript Homo sapiens 24-32 31632547-0 2019 MicroRNA-181b blocks gensenoside Rg3-mediated tumor suppression of gallbladder carcinoma by promoting autophagy flux via CREBRF/CREB3 pathway. ginsenoside Rg3 21-36 CREB3 regulatory factor Homo sapiens 121-127 31632547-0 2019 MicroRNA-181b blocks gensenoside Rg3-mediated tumor suppression of gallbladder carcinoma by promoting autophagy flux via CREBRF/CREB3 pathway. ginsenoside Rg3 21-36 cAMP responsive element binding protein 3 Homo sapiens 128-133 31632547-14 2019 However, exogenous miR-181b blunted Rg3-evoked anti-tumor effect possibly by inhibiting CREBRF/CREB pathway. ginsenoside Rg3 36-39 CREB3 regulatory factor Homo sapiens 88-94 31632547-14 2019 However, exogenous miR-181b blunted Rg3-evoked anti-tumor effect possibly by inhibiting CREBRF/CREB pathway. ginsenoside Rg3 36-39 cAMP responsive element binding protein 1 Homo sapiens 88-92 31632547-15 2019 CONCLUSION: Collectively, these data indicates that miR-181b possibly mediates the pathologic progression of GBC by CREBRF/CREB3 signaling pathways and impairs anti-tumor effects of Rg3 on GBC development, which suggests that miR-181b might be an key switch in the process of Rg3-mediated tumor cytotoxicity in the progression of GBC. ginsenoside Rg3 182-185 CREB3 regulatory factor Homo sapiens 116-122 31414228-11 2019 Immunofluorescence and western blotting studies showed that PEM/Rg3 inhibited the expressions of interleukin 1 (IL-1), interleukin 6 (IL-6), and reactive oxygen species modulator-1 (ROMO1). ginsenoside Rg3 64-67 interleukin 6 Mus musculus 119-132 31527568-0 2019 Ginsenoside Rg3 Reduces Epithelial-Mesenchymal Transition Induced by Transforming Growth Factor-beta1 by Inactivation of AKT in HMrSV5 Peritoneal Mesothelial Cells. ginsenoside Rg3 0-15 transforming growth factor beta 1 Homo sapiens 69-101 31527568-0 2019 Ginsenoside Rg3 Reduces Epithelial-Mesenchymal Transition Induced by Transforming Growth Factor-beta1 by Inactivation of AKT in HMrSV5 Peritoneal Mesothelial Cells. ginsenoside Rg3 0-15 AKT serine/threonine kinase 1 Homo sapiens 121-124 31527568-14 2019 Ginsenoside Rg3 reduced TGF-ss1-induced activation of AKT and Smurf2. ginsenoside Rg3 0-15 AKT serine/threonine kinase 1 Homo sapiens 54-57 31527568-14 2019 Ginsenoside Rg3 reduced TGF-ss1-induced activation of AKT and Smurf2. ginsenoside Rg3 0-15 SMAD specific E3 ubiquitin protein ligase 2 Homo sapiens 62-68 31527568-15 2019 SC79 reversed the effects of ginsenoside Rg3 on TGF-ss1-induced EMT in HMrSV5 cells. ginsenoside Rg3 29-44 major histocompatibility complex, class II, DR beta 1 Homo sapiens 52-55 31527568-16 2019 CONCLUSIONS Ginsenoside Rg3 inhibited EMT induced by TGF-ss1 in HMrSV5 human peritoneal mesothelial cells by inhibiting the activation of AKT. ginsenoside Rg3 12-27 AKT serine/threonine kinase 1 Homo sapiens 138-141 31414228-11 2019 Immunofluorescence and western blotting studies showed that PEM/Rg3 inhibited the expressions of interleukin 1 (IL-1), interleukin 6 (IL-6), and reactive oxygen species modulator-1 (ROMO1). ginsenoside Rg3 64-67 interleukin 6 Mus musculus 134-138 31414228-11 2019 Immunofluorescence and western blotting studies showed that PEM/Rg3 inhibited the expressions of interleukin 1 (IL-1), interleukin 6 (IL-6), and reactive oxygen species modulator-1 (ROMO1). ginsenoside Rg3 64-67 reactive oxygen species modulator 1 Mus musculus 182-187 31296984-0 2019 Ginsenoside Rg3 protects mouse leydig cells against triptolide by downregulation of miR-26a. ginsenoside Rg3 0-15 microRNA 26a-1 Mus musculus 84-91 31296984-10 2019 Moreover, the level of miR-26a was obviously downregulated by Rg3 treatment. ginsenoside Rg3 62-65 microRNA 26a-1 Mus musculus 23-30 31296984-11 2019 The protective effect of Rg3 against TP-induced toxicity in MLTC-1 cells was abolished by miR-26a upregulation. ginsenoside Rg3 25-28 microRNA 26a-1 Mus musculus 90-97 31296984-14 2019 As expected, upregulation of miR-26a could abrogate the protective effects of Rg3 against TP-induced cytotoxicity via inhibiting the expression of GSK3beta. ginsenoside Rg3 78-81 microRNA 26a-1 Mus musculus 29-36 31296984-14 2019 As expected, upregulation of miR-26a could abrogate the protective effects of Rg3 against TP-induced cytotoxicity via inhibiting the expression of GSK3beta. ginsenoside Rg3 78-81 glycogen synthase kinase 3 alpha Mus musculus 147-155 31296984-15 2019 Conclusion: These results indicated that Rg3 could protect MLTC-1 against TP by downregulation of miR-26a. ginsenoside Rg3 41-44 microRNA 26a-1 Mus musculus 98-105 30930425-0 2019 Ginsenoside Rg3 Combined with Oxaliplatin Inhibits the Proliferation and Promotes Apoptosis of Hepatocellular Carcinoma Cells via Downregulating PCNA and Cyclin D1. ginsenoside Rg3 0-15 proliferating cell nuclear antigen Homo sapiens 145-149 30930425-0 2019 Ginsenoside Rg3 Combined with Oxaliplatin Inhibits the Proliferation and Promotes Apoptosis of Hepatocellular Carcinoma Cells via Downregulating PCNA and Cyclin D1. ginsenoside Rg3 0-15 cyclin D1 Homo sapiens 154-163 30930425-6 2019 Meanwhile, ginsenoside Rg3, oxaliplatin or ginsenoside Rg3 + oxaliplatin also significantly inhibited the expressions of PCNA and cyclin D1. ginsenoside Rg3 11-26 proliferating cell nuclear antigen Homo sapiens 121-125 30930425-6 2019 Meanwhile, ginsenoside Rg3, oxaliplatin or ginsenoside Rg3 + oxaliplatin also significantly inhibited the expressions of PCNA and cyclin D1. ginsenoside Rg3 11-26 cyclin D1 Homo sapiens 130-139 30930425-6 2019 Meanwhile, ginsenoside Rg3, oxaliplatin or ginsenoside Rg3 + oxaliplatin also significantly inhibited the expressions of PCNA and cyclin D1. ginsenoside Rg3 43-58 proliferating cell nuclear antigen Homo sapiens 121-125 30930425-6 2019 Meanwhile, ginsenoside Rg3, oxaliplatin or ginsenoside Rg3 + oxaliplatin also significantly inhibited the expressions of PCNA and cyclin D1. ginsenoside Rg3 43-58 cyclin D1 Homo sapiens 130-139 25175462-11 2014 Western blotting and densitometric assay showed that the expression of Bcl-2 was decreased after the combined treatment of ginsenoside Rg3 and cisplatin, whereas the expression of cytochrome c and caspase-3 were increased, suggesting the activation of the intrinsic apoptotic pathway. ginsenoside Rg3 123-138 BCL2 apoptosis regulator Homo sapiens 71-76 29156516-0 2018 Synergistic anticancer activity of 20(S)-Ginsenoside Rg3 and Sorafenib in hepatocellular carcinoma by modulating PTEN/Akt signaling pathway. ginsenoside Rg3 35-56 phosphatase and tensin homolog Homo sapiens 113-117 29156516-0 2018 Synergistic anticancer activity of 20(S)-Ginsenoside Rg3 and Sorafenib in hepatocellular carcinoma by modulating PTEN/Akt signaling pathway. ginsenoside Rg3 35-56 AKT serine/threonine kinase 1 Homo sapiens 118-121 29156516-8 2018 The levels of PTEN, Bax and cleaved caspase-3 expression increased, while the levels of phospho-PDK1 and phospho-Akt expression decreased by the treatment of Sorafenib combined with 20(S)-Ginsenoside Rg3. ginsenoside Rg3 182-203 BCL2 associated X, apoptosis regulator Homo sapiens 20-23 29156516-8 2018 The levels of PTEN, Bax and cleaved caspase-3 expression increased, while the levels of phospho-PDK1 and phospho-Akt expression decreased by the treatment of Sorafenib combined with 20(S)-Ginsenoside Rg3. ginsenoside Rg3 182-203 pyruvate dehydrogenase kinase 1 Homo sapiens 96-100 29156516-8 2018 The levels of PTEN, Bax and cleaved caspase-3 expression increased, while the levels of phospho-PDK1 and phospho-Akt expression decreased by the treatment of Sorafenib combined with 20(S)-Ginsenoside Rg3. ginsenoside Rg3 182-203 AKT serine/threonine kinase 1 Homo sapiens 113-116 29156516-10 2018 The results demonstrated the synergistic anticancer activity of 20(S)-Ginsenoside Rg3 and Sorafenib in HCC by modulating PTEN/Akt signaling pathway. ginsenoside Rg3 64-85 phosphatase and tensin homolog Homo sapiens 121-125 29156516-10 2018 The results demonstrated the synergistic anticancer activity of 20(S)-Ginsenoside Rg3 and Sorafenib in HCC by modulating PTEN/Akt signaling pathway. ginsenoside Rg3 64-85 AKT serine/threonine kinase 1 Homo sapiens 126-129 31939450-0 2019 Inhibition of the hypoxia-induced factor-1alpha and vascular endothelial growth factor expression through ginsenoside Rg3 in human gastric cancer cells. ginsenoside Rg3 106-121 vascular endothelial growth factor A Homo sapiens 52-86 31939450-1 2019 Objective: The aim of this study is to probe in the inhibitory effects of ginsenoside Rg3 on the expression of hypoxia-induced factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) in human gastric cancer cells. ginsenoside Rg3 74-89 hypoxia inducible factor 1 subunit alpha Homo sapiens 142-152 31939450-1 2019 Objective: The aim of this study is to probe in the inhibitory effects of ginsenoside Rg3 on the expression of hypoxia-induced factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) in human gastric cancer cells. ginsenoside Rg3 74-89 vascular endothelial growth factor A Homo sapiens 158-192 31939450-1 2019 Objective: The aim of this study is to probe in the inhibitory effects of ginsenoside Rg3 on the expression of hypoxia-induced factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) in human gastric cancer cells. ginsenoside Rg3 74-89 vascular endothelial growth factor A Homo sapiens 194-198 31939450-3 2019 Results: Under hypoxia, expression of HIF-1alpha and VEGF in human gastric cancer BGC823 cells showed an increasing trend, and that was remarkably lower in experiment group than in the control group after applying Rg3, which was obvious at 12 and 24 h (P < 0.05). ginsenoside Rg3 214-217 hypoxia inducible factor 1 subunit alpha Homo sapiens 38-48 31939450-3 2019 Results: Under hypoxia, expression of HIF-1alpha and VEGF in human gastric cancer BGC823 cells showed an increasing trend, and that was remarkably lower in experiment group than in the control group after applying Rg3, which was obvious at 12 and 24 h (P < 0.05). ginsenoside Rg3 214-217 vascular endothelial growth factor A Homo sapiens 53-57 31939450-4 2019 Conclusion: Rg3 can inhibit expression of HIF-1alpha and VEGF in human gastric cancer cells and may influence abdominal implantation metastasis of gastric cancer through inhibiting its expression. ginsenoside Rg3 12-15 hypoxia inducible factor 1 subunit alpha Homo sapiens 42-52 31939450-4 2019 Conclusion: Rg3 can inhibit expression of HIF-1alpha and VEGF in human gastric cancer cells and may influence abdominal implantation metastasis of gastric cancer through inhibiting its expression. ginsenoside Rg3 12-15 vascular endothelial growth factor A Homo sapiens 57-61 29773855-4 2018 Furthermore, the increased corticotropin releasing hormone, corticosterone and adrenocorticotropic hormone were blocked by GRg3 (20 and 40 mg/kg, i.g). ginsenoside Rg3 123-127 corticotropin releasing hormone Rattus norvegicus 27-58 23717160-7 2013 In comparison of the protective activity between ginsenoside-Rb2 and its two hydrolytic products [20(S)- and 20(R)-ginsenoside-Rg3], 20(S)-ginsenoside-Rg3, but not 20(R)-ginsenoside-Rg3, elicited a partial protection against HVJ. ginsenoside Rg3 135-154 RB transcriptional corepressor like 2 Mus musculus 61-64 23874757-2 2013 In this study, Ginsenoside Rg3/Poly(l-lactide) (G-Rg3/PLLA) electrospun fibrous scaffolds covering on the full-thickness skin excisions location was designed to suppress the hypertrophic scar formation in vivo. ginsenoside Rg3 15-30 TLE family member 3, transcriptional corepressor Homo sapiens 48-53 22471932-3 2012 However, information on the anti-platelet activity of ginsenoside-Rp1 (G-Rp1), a stable derivative of ginsenoside-Rg3, is scarce. ginsenoside Rg3 102-117 RP1, axonemal microtubule associated Rattus norvegicus 66-69 14969341-4 2004 The transformed ginsenoside Rg3 and delta20-ginsenoside Rg3 were metabolized to ginsenoside Rh2 and delta20-ginsenoside Rh2, respectively, by human fecal microflora. ginsenoside Rg3 16-31 Rh associated glycoprotein Homo sapiens 92-95 22975507-7 2012 Morphological activation of microglia and Iba1 protein expression by systemic LPS injection were reduced with ginsenoside Rg3 (30 mg/kg) treatment. ginsenoside Rg3 110-125 induction of brown adipocytes 1 Mus musculus 42-46 14969341-4 2004 The transformed ginsenoside Rg3 and delta20-ginsenoside Rg3 were metabolized to ginsenoside Rh2 and delta20-ginsenoside Rh2, respectively, by human fecal microflora. ginsenoside Rg3 16-31 Rh associated glycoprotein Homo sapiens 120-123 14969341-7 2004 potently transformed ginsenoside Rg3 to ginsenoside Rh2. ginsenoside Rg3 21-36 Rh associated glycoprotein Homo sapiens 52-55 11824558-1 2002 When ginsenoside Rg3 was anaerobically incubated with human fecal microflora, all specimens metabolized ginsenoside Rg3 to ginsenoside Rh2 and protopanaxadiol. ginsenoside Rg3 5-20 Rh associated glycoprotein Homo sapiens 135-138 11824558-3 2002 20(S)-ginsenoside Rg3 was quickly transformed to 20(S)-ginsenoside Rh2 or 20(S)-protopanaxadiol in an amount 19-fold that compared with the transformation of 20(R)-ginsenoside Rg3 to 20(R)-ginsenoside Rh2 or 20(R)-protopanaxadiol. ginsenoside Rg3 2-21 Rh associated glycoprotein Homo sapiens 67-70 11824558-3 2002 20(S)-ginsenoside Rg3 was quickly transformed to 20(S)-ginsenoside Rh2 or 20(S)-protopanaxadiol in an amount 19-fold that compared with the transformation of 20(R)-ginsenoside Rg3 to 20(R)-ginsenoside Rh2 or 20(R)-protopanaxadiol. ginsenoside Rg3 2-21 Rh associated glycoprotein Homo sapiens 201-204 11824558-5 2002 metabolized ginsenoside Rg3 to protopanaxadiol via ginsenoside Rh2. ginsenoside Rg3 12-27 Rh associated glycoprotein Homo sapiens 63-66 11824558-7 2002 metabolized ginsenoside Rg3 to ginsenoside Rh2 alone. ginsenoside Rg3 12-27 Rh associated glycoprotein Homo sapiens 43-46 11824558-8 2002 Among ginsenoside Rg3 and its metabolites, 20(S)-protopanaxadiol and 20(S)-ginsenoside Rh2 exhibited the most potent cytotoxicity against tumor cell lines, 20(S)- and 20(R)-protopanaxadiols potently inhibited the growth of Helicobacter pylori, and 20(S)-ginsenoside Rh2 inhibited H+/K+ ATPase of rat stomach. ginsenoside Rg3 6-21 Rh associated glycoprotein Homo sapiens 87-90