人参皂苷Rg1、Rb1的药效及作用机制研究进展

您所在的位置:网站首页 人参茎叶总皂苷的作用和功效 人参皂苷Rg1、Rb1的药效及作用机制研究进展

人参皂苷Rg1、Rb1的药效及作用机制研究进展

2024-07-16 12:16| 来源: 网络整理| 查看: 265

李蕾, 谢丽娟, 王国明, 等. 人参、西洋参不同部位提取物中14种皂苷含量比较[J]. 人参研究, 2018, 30(3): 11-13. DOI: 10.19403/j.cnki.1671-1521.2018.03.003http://dx.doi.org/10.19403/j.cnki.1671-1521.2018.03.003.

LI L, XIE L J, WANG G M, et al. Comparison of 14 saponin contents in extracts from different parts of ginseng and American ginseng [J]. Ginseng Research, 2018, 30(3): 11-13. DOI: 10.19403/j.cnki.1671-1521.2018.03.003 (Chhttp://dx.doi.org/10.19403/j.cnki.1671-1521.2018.03.003(Ch).

张于, 程伟. 中西医治疗阿尔兹海默病的研究现状[J]. 中医临床研究, 2012, 4(13): 115-117. DOI: 10.3969/j.issn.1674-7860. 2012.13.069http://dx.doi.org/10.3969/j.issn.1674-7860.2012.13.069.

ZHANG Y, CHENG W. Research status on treating AD in the integrative medicine [J]. Clinical Journal of Chinese Medicine, 2012, 4(13): 115-117. DOI:10.3969/j.issn.1674-7860.2012.13.069 (Chhttp://dx.doi.org/10.3969/j.issn.1674-7860.2012.13.069(Ch).

QUAN Q K, WANG J, LI X, et al. Ginsenoside Rg1 decreases Aβ1–42 level by upregulating PPARγ and IDE expression in the hippocampus of a rat model of Alzheimer's disease [J]. PloS One, 2013, 8(3): e59155. DOI: 10.1371/journal.pone.0059155http://dx.doi.org/10.1371/journal.pone.0059155.

FANG F, CHEN X C, HUANG T W, et al. Multi-faced neuroprotective effects of ginsenoside Rg1 in an Alzheimer mouse model [J]. Biochim Biophys Acta, 2012, 1822(2): 286-292. DOI: 10.1016/j.bbadis.2011.10.004http://dx.doi.org/10.1016/j.bbadis.2011.10.004.

CHEN L M, LIN Z Y, ZHU Y G, et al. Ginsenoside Rg1 attenuates β-amyloid generation via suppressing PPARγ-regulated BACE1 activity in N2a-APP695 cells [J]. European Journal of Pharmacology, 2012, 675(1): 15-21. DOI: 10.1016/j.ejphar.2011.11.039http://dx.doi.org/10.1016/j.ejphar.2011.11.039.

HUANG T W, FANG F, CHEN L M, et al. Ginsenoside Rg1 attenuates oligomeric Aβ1-42-induced mitochondrial dysfunction [J]. Current Alzheimer Research, 2012, 9(3): 388-395. DOI: 10.2174/156720512800107636http://dx.doi.org/10.2174/156720512800107636.

LI W, CHU Y Q, ZHANG L, et al. Ginsenoside Rg1 prevents SK-N-SH neuroblastoma cell apoptosis induced by supernatant from Aβ1-40-stimulated THP-1 monocytes [J]. Brain Research Bulletin, 2012, 88(5): 501-506. DOI: 10.1016/j.brainresbull.2012.05.002http://dx.doi.org/10.1016/j.brainresbull.2012.05.002.

WU J Y, YANG H Y, ZHAO Q W, et al. Ginsenoside Rg1 exerts a protective effect against Aβ25-35-induced toxicity in primary cultured rat cortical neurons through the NF-κB/NO pathway [J]. International Journal of Molecular Medicine, 2016, 37(3): 781-788. DOI: 10.3892/ijmm.2016.2485http://dx.doi.org/10.3892/ijmm.2016.2485.

GONG L, LI S L, LI H, et al. Ginsenoside Rg1 protects primary cultured rat hippocampal neurons from cell apoptosis induced by β-amyloid protein [J]. Pharmaceutical Biology, 2011, 49(5): 501-507. DOI: 10.3109/13880209.2010.521514http://dx.doi.org/10.3109/13880209.2010.521514.

WU J Y, PAN Z F, WANG Z Q, et al. Ginsenoside Rg1 protection against β-amyloid peptide-induced neuronal apoptosis via estrogen receptor α and glucocorticoid receptor-dependent anti-protein nitration pathway [J]. Neuropharmacology, 2012, 63(3): 349-361. DOI: 10.1016/j.neuropharm.2012.04.005http://dx.doi.org/10.1016/j.neuropharm.2012.04.005.

ZHAO B S, LIU Y, GAO X Y, et al. Effects of ginsenoside Rg1 on the expression of toll-like receptor 3, 4 and their signalling transduction factors in the NG108-15 murine neuroglial cell line [J]. Molecules, 2014, 19(10): 16925-16936. DOI: 10.3390/molecules191016925http://dx.doi.org/10.3390/molecules191016925.

GU W J, LIU D, ZHANG M R, et al. Effect of ginsenoside Rb1 on insulin signal transduction pathway in hippocampal neurons of high-glucose-fed rats [J]. China Journal of Chinese Materia Medica, 2014, 39(6): 1064-1068.

ZHAO R P, ZHANG Z X, SONG Y J, et al. Implication of phosphatidylinositol-3 kinase/Akt/glycogen synthase kinase-3β pathway in ginsenoside Rb1’s attenuation of beta-amyloid-induced neurotoxicity and Tau phosphorylation [J]. Journal of Ethnopharmacology, 2011, 133(3): 1109-1116. DOI: 10.1016/j.jep.2010.11.054http://dx.doi.org/10.1016/j.jep.2010.11.054.

QIAN Y H, HAN H, HU X D, et al. Protective effect of ginsenoside Rb1 on beta-amyloid protein(1-42)-induced neurotoxicity in cortical neurons [J]. Neurological Research, 2009, 31(7): 663-667. DOI: 10.1179/174313209X385572http://dx.doi.org/10.1179/174313209X385572.

WANG Y H, DU G H. Ginsenoside Rg1 inhibits beta-secretase activity in vitro and protects against Abeta- induced cytotoxicity in PC12 cells [J]. Journal of Asian Natural Products Research, 2009, 11(7): 604-612. DOI: 10.1080/10286020902843152http://dx.doi.org/10.1080/10286020902843152.

LIANG H, LIU L F, LIU J, et al. Ginsenoside Rg1 protects against neurodegeneration by inducing neurite outgrowth in cultured hippocampal neurons [J]. Neural Regeneration Research, 2016, 11(2): 319-325. DOI: 10.4103/1673-5374.177741http://dx.doi.org/10.4103/1673-5374.177741.

WANG Q, SUN L H, JIA W, et al. Comparison of ginsenosides Rg1 and Rb1 for their effects on improving scopolamine-induced learning and memory impairment in mice [J]. Phytotherapy Research, 2010, 24(12): 1748-1754. DOI: 10.1002/ptr.3130http://dx.doi.org/10.1002/ptr.3130.

ZHAO H H, DI J, LIU W S, et al. Involvement of GSK3 and PP2A in ginsenoside Rb1’s attenuation of aluminum-induced Tau hyperphosphorylation [J]. Behavioural Brain Research, 2013, 241(1): 228-234. DOI: 10.1016/j.bbr.2012.11.037http://dx.doi.org/10.1016/j.bbr.2012.11.037.

CHEN X C, HUANG T W, ZHANG J, et al. Involvement of calpain and p25 of CDK5 pathway in ginsenoside Rb1’s attenuation of beta-amyloid peptide25-35-induced Tau hyperphosphorylation in cortical neurons [J]. Brain Research, 2008, 1200: 99-106. DOI: 10.1016/j.brainres.2007.12.029http://dx.doi.org/10.1016/j.brainres.2007.12.029.

WANG Y, FENG Y, FU Q Y, et al. Panax notoginsenoside Rb1 ameliorates Alzheimer’s disease by upregulating brain-derived neurotrophic factor and downregulating Tau protein expression [J]. Experimental & Therapeutic Medicine, 2013, 6(3): 826-830. DOI: 10.3892/etm.2013.1215http://dx.doi.org/10.3892/etm.2013.1215.

李玺, 刘颖, 袁海峰, 等. 人参皂苷Rg1对冈田酸所致大鼠脑片Tau蛋白磷酸化的影响[J]. 中西医结合学报, 2010, 8(10): 955-960. DOI: 10.3736/jcim20101007http://dx.doi.org/10.3736/jcim20101007.

LI X, LIU Y, YUAN H F, et al. Effects of gensenoside Rg1 on Tau protein phosphorylation induced by okadaic acid in rat brain slices [J]. Journal of Chinese Integrative Medicine, 2010, 8(10): 955-960. DOI: 10.3736/jcim20101007 (Chhttp://dx.doi.org/10.3736/jcim20101007(Ch).

李玺, 张欣, 袁海峰, 等. 人参皂苷Rg1对AD模型大鼠脑片P-Tau, caspase-3表达的影响[J].中国中药杂志, 2010, 35(3): 369-372. DOI: 10.4268/cjcmm20100325http://dx.doi.org/10.4268/cjcmm20100325.

LI X, ZHANG X, YUAN H F, et al. Experimental research on effect of gensenoside Rg1 on expressions of P-Tau and caspase-3 in brain slices from AD model rats [J]. China Journal of Chinese Materia Medica, 2010, 35(3): 369-372. DOI: 10.4268/cjcmm20100325 (Chhttp://dx.doi.org/10.4268/cjcmm20100325(Ch).

ZHOU T T, ZU G, WANG X, et al. Immunomodulatory and neuroprotective effects of ginsenoside Rg1 in the MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) -induced mouse model of Parkinson’s disease [J]. International Immunopharmacology, 2015, 29(2): 334-343. DOI: 10.1016/j.intimp.2015.10.032http://dx.doi.org/10.1016/j.intimp.2015.10.032.

ZHOU T T, ZU G, ZHANG X G, et al. Neuroprotective effects of ginsenoside Rg1 through the Wnt/β-catenin signaling pathway in both in vivo and in vitro models of Parkinson’s disease [J]. Neuropharmacology, 2016, 101: 480-489. DOI: 10.1016/j.neuropharm.2015.10.024http://dx.doi.org/10.1016/j.neuropharm.2015.10.024.

HENG Y, ZHANG Q S, MU Z, et al. Ginsenoside Rg1 attenuates motor impairment and neuroinflammation in the MPTP-probenecid-induced Parkinsonism mouse model by targeting α-synuclein abnormalities in the substantia nigra [J]. Toxicology Letters, 2016, 243: 7-21. DOI: 10.1016/j.toxlet.2015.12.005http://dx.doi.org/10.1016/j.toxlet.2015.12.005.

REN X F, CHEN L, GAO X Q, et al. Glucocorticoid receptor is involved in the neuroprotective effect of ginsenoside Rg1 against inflammation-induced dopaminergic neuronal degeneration in substantia nigra [J]. Journal of Steroid Biochemistry & Molecular Biology, 2016, 155: 94-103. DOI: 10.1016/j.jsbmb.2015.09.040http://dx.doi.org/10.1016/j.jsbmb.2015.09.040.

XU L, CHEN W F, WONG M S. Ginsenoside Rg1 protects dopaminergic neurons in a rat model of Parkinson’s disease through the IGF-I receptor signalling pathway [J]. British Journal of Pharmacology, 2010, 158(3): 738-748. DOI: 10.1111/j.1476-5381.2009.00361.xhttp://dx.doi.org/10.1111/j.1476-5381.2009.00361.x.

XU H M, JIANG H, WANG J, et al. Rg1 protects the MPP-treated MES23.5 cells via attenuating DMT1 up-regulation and cellular iron uptake [J]. Neuropharmacology, 2010, 58(2): 488-494. DOI: 10.1016/j.neuropharm.2009.09.002http://dx.doi.org/10.1016/j.neuropharm.2009.09.002.

WANG J, XU H M, YANG H D, et al. Rg1 reduces nigral iron levels of MPTP-treated C57BL6 mice by regulating certain iron transport proteins [J]. Neurochemistry International, 2009, 54(1): 43-48. DOI: 10.1016/j.neuint.2008.10.003http://dx.doi.org/10.1016/j.neuint.2008.10.003.

ZHANG Y L, LIU Y, KANG X P, et al. Ginsenoside Rb1 confers neuroprotection via promotion of glutamate transporters in a mouse model of Parkinson’s disease [J]. Neuropharmacology, 2017, 131: 223-237. DOI: 10.1016/j.neuropharm.2017.12.012http://dx.doi.org/10.1016/j.neuropharm.2017.12.012.

YANG Y D, LI X, ZHANG L M, et al. Ginsenoside Rg1 suppressed infammation and neuron apoptosis by activating PPAR γ/HO-1 in hippocampus in rat model of cerebral ischemia-reperfusion injury [J]. International Journal of Clinical & Experimental Pathology, 2015, 8(3): 2484-2494.

XIE C L, LI J H, WANG W W, et al. Neuroprotective effect of ginsenoside-Rg1 on cerebral ischemia/reperfusion injury in rats by downregulating protease-activated receptor-1 expression [J]. Life Sciences, 2015, 121: 145-151. DOI: 10.1016/j.lfs.2014.12.002http://dx.doi.org/10.1016/j.lfs.2014.12.002.

王巧云, 刘凤, 吴峰阶, 等. 人参皂苷Rg1对局灶性脑缺血再灌注损伤大鼠海马p-ERK1/2与p-JNK表达的影响[J]. 中国中西医结合杂志, 2013, 33(2): 229-234.

WANG Q Y, LIU F, WU F J, et al. Effects of ginsenoside Rg1 on the expressions of p-eRK1/2 and p-JNK in local cerebral ischemia/reperfusion injury rats [J]. Chinese Journal of Integrated Traditional & Western Medicine, 2013, 33(2): 229-234(Ch).

WANG L, ZHAO H, ZHAI Z Z, et al. Protective effect and mechanism of ginsenoside Rg1 in cerebral ischaemia-reperfusion injury in mice [J]. Biomedicine & Pharmacotherapy, 2018, 99:876-882. DOI: 10.1016/j.biopha.2018.01.136http://dx.doi.org/10.1016/j.biopha.2018.01.136.

ZHU J, JIANG Y, WU L, et al. Suppression of local inflammation contributes to the neuroprotective effect of ginsenoside Rb1 in rats with cerebral ischemia [J]. Neuroscience, 2012, 202(2): 342-351. DOI: 10.1016/j.neuroscience.2011.11.070http://dx.doi.org/10.1016/j.neuroscience.2011.11.070.

DONG X, ZHENG L, LU S J, et al. Neuroprotective effects of pretreatment of ginsenoside Rb1 on severe cerebral ischemia-induced injuries in aged mice: Involvement of anti-oxidant signaling [J]. Geriatrics & Gerontology International, 2017, 17(2):338-345. DOI: 10.1111/ggi.12699http://dx.doi.org/10.1111/ggi.12699.

WANG S Y, LI M H, GUO Y, et al. Effects of Panax notoginseng ginsenoside Rb1 on abnormal hippocampal microenvironment in rats [J]. Journal of Ethnopharmacology, 2017, 202: 138-146. DOI: 10.1016/j.jep.2017.01.005http://dx.doi.org/10.1016/j.jep.2017.01.005.

黄小平, 王蓓, 邱咏园, 等. 黄芪甲苷、人参皂苷Rg1、Rb1和三七皂苷R1抗小鼠脑缺血再灌注氧化应激损伤和促进能量代谢的配伍研究[J]. 湖南中医药大学学报, 2014, 34(7): 5-11. DOI: 10.3969/j.issn.1674-070X.2014.07.002.005.07http://dx.doi.org/10.3969/j.issn.1674-070X.2014.07.002.005.07.

HUANG X P, WANG B, QIU Y Y, et al. The combination study of astragaloside Ⅳ, ginsenosides Rg1, Rb1 and notoginsenoside R1 on antagonizing oxidative stress injury and promoting energy metabolism after ischemia-reperfusion in mice [J]. Journal of Traditional Chinese Medicine University of Hunan, 2014, 34(7): 5-11. DOI: 10.3969/j.issn.1674-070X.2014.07.002.005.07(Chhttp://dx.doi.org/10.3969/j.issn.1674-070X.2014.07.002.005.07(Ch).

柯大智, 王红宁, 陈地龙, 等. 人参皂苷Rg1抑制白血病细胞K562增殖并诱导分化的分子机制[J]. 第三军医大学学报, 2014, 36(2): 125-129. DOI:10.16016/j.1000-5404.2014.02.003http://dx.doi.org/10.16016/j.1000-5404.2014.02.003.

KE D Z, WANG H N,CHEN D L, et al. Molecular mechanism of ginsenoside Rg1 in inhibiting proliferation and inducing differentiation in chronic myelocytic leukemia K562 cells [J]. Acta Academiae Medicinae Militaris Tertiae, 2014, 36(2): 125-129. DOI:10.16016/j.1000-5404.2014.02.003 (Chhttp://dx.doi.org/10.16016/j.1000-5404.2014.02.003(Ch).

王国红. 人参皂甙Rg1、肉桂酸、丹参酮ⅡA及其组合对人成骨肉瘤MG-63细胞终末分化的诱导研究[D]. 厦门:厦门大学,2005.

WANG G H. Study on the Terminal Differentiation of Human Osteosarcoma MG-63 Cells Induced by Ginsenoside Rg1, Cinnamic Acid, TanshinoneⅡA and Their Combinations [D]. Xiamen: Xiamen University, 2005(Ch).

张龙江, 周二付. 人参皂苷Rg1对结肠癌肿瘤体外和体内的增殖抑制作用[J]. 世界华人消化杂志, 2014(30): 4599-4603. DOI: 10.11569/wcjd.v22.i30.4599http://dx.doi.org/10.11569/wcjd.v22.i30.4599.

ZHANG L J, ZHOU E F. In vitro and in vivo inhibitory effects of ginsenoside Rg1 on proliferation of colon cancer cells [J]. World Chinese Journal of Digestology, 2014(30): 4599-4603. DOI: 10.11569/wcjd.v22.i30.4599 (Chhttp://dx.doi.org/10.11569/wcjd.v22.i30.4599(Ch).

王红宁, 左国伟, 陈地龙, 等. 人参皂苷Rb1、Rg1、Re对白血病细胞株KG1α增殖的影响[J]. 生物技术, 2010, 20(2): 56-58. DOI:10.16519/j.cnki.1004-311x.2010.02.031http://dx.doi.org/10.16519/j.cnki.1004-311x.2010.02.031.

WANG H N, ZUO G W, CHEN D L, et al. Effects of ginsenoside Rb1, Rg1, Re on proliferation of KG1α cells [J]. Biotechnology, 2010, 20(2): 56-58. DOI:10.16519/j.cnki.1004-311x.2010.02.031 (Chhttp://dx.doi.org/10.16519/j.cnki.1004-311x.2010.02.031(Ch).

崔金刚, 王晓燕, 熊敏琪, 等. 人参皂苷Rg1与Rb1在心肌缺血复合肿瘤病理环境下的作用研究[J]. 上海中医药大学学报, 2014, 28(4): 58-63. DOI:10.16306/j.1008-861x.2014.04.020http://dx.doi.org/10.16306/j.1008-861x.2014.04.020.

CUI J G, WANG X Y, XIONG M Q, et al. Effects of ginsenosides Rg1 and Rb1 in myocardial ischemia complex pathological environment of neoplasia [J]. Journal of Shanghai University of Traditional Chinese Medicine, 2014, 28(4): 58-63. DOI:10.16306/j.1008-861x.2014.04.020(Chhttp://dx.doi.org/10.16306/j.1008-861x.2014.04.020(Ch).

刘嘉. 肿瘤多药耐药机制及其逆转方案的研究进展[J]. 中国肿瘤外科杂志, 2010, 2(3): 174-178. DOI: 10.3969/j.issn.1674-4136.2010.03.012http://dx.doi.org/10.3969/j.issn.1674-4136.2010.03.012.

LIU J. Advances in research on multidrug resistance mechanism of tumor and its reversal program [J]. Chinese Journal of Surgical Oncology, 2010, 2(3): 174-178. DOI: 10.3969/j.issn.1674-4136.2010.03.012(Chhttp://dx.doi.org/10.3969/j.issn.1674-4136.2010.03.012(Ch).

王婷. 三七总皂苷Rb1对肿瘤耐药逆转作用的实验研究[D].昆明: 昆明医学院,2011.

WANG T. PNS-Rb1 Reverse Effect on Tumor Multidrugresistance in vivo Study [D]. Kunming: Kunming Medical College, 2011(Ch).

WANG Y, LIU Y, ZHANG X Y, et al. Ginsenoside Rg1 regulates innate immune responses in macrophages through differentially modulating the NF-κB and PI3K/Akt/mTOR pathways [J]. International Immunopharmacology, 2014, 23(1): 77-84. DOI: 10.1016/j.intimp.2014.07.028http://dx.doi.org/10.1016/j.intimp.2014.07.028.

武文华. 人参皂苷Rg1在裸鼠抗肿瘤治疗中免疫调节作用的研究[D].长春:吉林大学,2017.

WU W H. Immunomodulatory Effects of Ginsenoside Rg1 in Nude Mice with Antitumor Therapy [D]. Changchun: Jilin University, 2017.

DE S B G, MENASCHE G, FISCHER A. Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules [J]. Nature Reviews Immunology, 2010, 10(8): 568-579. DOI: 10.1038/nri2803http://dx.doi.org/10.1038/nri2803.

金岩, 曲婷婷, 柳越冬, 等. 人参皂苷Rb1、Rg1与5-氟脲嘧啶对地塞米松诱导S180荷瘤小鼠脾淋巴细胞凋亡影响的实验研究[J]. 中华中医药学刊, 2006,24(7): 1272-1273. DOI:10.13193/j.archtcm.2006.07.90.jiny.038http://dx.doi.org/10.13193/j.archtcm.2006.07.90.jiny.038.

JIN Y, QU T T, LIU Y D, et al. Effect of ginsenoside Rb1, Rg1 and 5-fluorouracil on apoptosis of spleen lymphocytes in S180 tumor-bearing mice induced by dexamethasone [J]. Chinese Archives of Traditional Chinese Medicine, 2006,24(7): 1272-1273. DOI: 10.13193/j.archtcm.2006.07.90.jiny.038(Chhttp://dx.doi.org/10.13193/j.archtcm.2006.07.90.jiny.038(Ch).

曲婷婷, 金岩, 柳越冬, 等. 人参皂苷Rb1、Rg1与5-氟脲嘧啶对荷瘤小鼠免疫功能的影响[J]. 中医研究, 2006,19(5): 16-18.

QU T T, JIN Y, LIU Y D, et al. Effects of ginsenoside Rb1, Rg1 and 5-fluorouracil on immune function of tumor-bearing mice [J]. Traditional Chinese Medicinal Research, 2006,19(5): 16-18(Ch).

LI J, WEI Q, ZUO G W, et al. Ginsenoside Rg1 induces apoptosis through inhibition of the EpoR-mediated JAK2/STAT5 signalling pathway in the TF-1/Epo human leukemia cell line [J]. Asian Pacific Journal of Cancer Prevention Apjcp, 2014, 15(6): 2453-2459. DOI:10.7314/APJCP.2014.15.6.2453http://dx.doi.org/10.7314/APJCP.2014.15.6.2453.

HWANG J W, OH J H, YOO H S, et al. Mountain ginseng extract exhibits antilung cancer activity by inhibiting the nuclear translocation of NF-κB [J]. American Journal of Chinese Medicine, 2012, 40(1): 187-202. DOI: 10.1142/S0192415X12500152http://dx.doi.org/10.1142/S0192415X12500152.

YU M L, YU X B, GUO D H, et al. Ginsenoside Rg1 attenuates invasion and migration by inhibiting transforming growth factor-β1-induced epithelial to mesenchymal transition in HepG2 cells [J]. Molecular Medicine Reports, 2015, 11(4): 3167-3173. DOI: 10.3892/mmr.2014.3098http://dx.doi.org/10.3892/mmr.2014.3098.

LI L I, WANG Y, BENQUAN Q I, et al. Suppression of PMA-induced tumor cell invasion and migration by ginsenoside Rg1 via the inhibition of NF-κB-dependent MMP-9 expression [J]. Oncology Reports, 2014, 32(5): 1779-1786. DOI: 10.3892/or.2014.3422http://dx.doi.org/10.3892/or.2014.3422.

NADKARNI P, CHEPURNY O G, HOLZ G G. Chapter two—regulation of glucose homeostasis by GLP-1 [J]. Progress in Molecular Biology & Translational Science, 2014, 121(121): 23-65. DOI: 10.1016/B978-0-12-800101-1.00002-8http://dx.doi.org/10.1016/B978-0-12-800101-1.00002-8.

RONDAS D, D’HERTOG W, OVERBERGH L, et al. Glucagon-like peptide-1: Modulator of β-cell dysfunction and death [J]. Diabetes Obesity & Metabolism, 2013, 15(s3): 185-192. DOI: 10.1111/dom.12165http://dx.doi.org/10.1111/dom.12165.

LIU C, ZHANG M, HU M Y, et al. Increased glucagon-like peptide-1 secretion may be involved in antidiabetic effects of ginsenosides [J]. Journal of Endocrinology, 2013, 217(2): 185-196. DOI: 10.1530/JOE-12-0502http://dx.doi.org/10.1530/JOE-12-0502.

YU X Z, YE L F, ZHANG H, et al. Ginsenoside Rb1 ameliorates liver fat accumulation by upregulating perilipin expression in adipose tissue of db/db obese mice [J]. Journal of Ginseng Research, 2015, 39(3): 199-205. DOI: 10.1016/j.jgr.2014.11.004http://dx.doi.org/10.1016/j.jgr.2014.11.004.

AKIYAMA N, AKIYAMA Y, KATO H, et al. Pharmacological evaluation of adipose dysfunction via 11β-hydroxysteroid dehydrogenase type 1 in the development of diabetes in diet-induced obese mice with cortisone pellet implantation [J]. Journal of Pharmacology & Experimental Therapeutics, 2014, 349(1): 66-74. DOI: 10.1124/jpet.113.210716http://dx.doi.org/10.1124/jpet.113.210716.

SONG B, DING L, ZHANG H, et al. Ginsenoside Rb1 increases insulin sensitivity through suppressing 11β-hydroxysteroid dehydrogenase type I [J]. American Journal of Translational Research, 2017, 9(3): 1049-1057.

LECLERCQ I A, SILVA MORAIS A D, SCHROYEN B, et al. Insulin resistance in hepatocytes and sinusoidal liver cells: Mechanisms and consequences [J]. Journal of Hepatology, 2007, 47(1): 142-156. DOI: 10.1016/j.jhep.2007.04.002http://dx.doi.org/10.1016/j.jhep.2007.04.002.

COOL B, ZINKER B, CHIOU W, et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome [J]. Cell Metabolism, 2006, 3(6): 403-416. DOI: 10.1016/j.cmet.2006.05.005http://dx.doi.org/10.1016/j.cmet.2006.05.005.

ZHANG B B, ZHOU G C, LI C. AMPK: An emerging drug target for diabetes and the metabolic syndrome [J]. Cell Metabolism, 2009, 9(5): 407-416. DOI : 10.1016/j.cmet.2009.03.012http://dx.doi.org/10.1016/j.cmet.2009.03.012.

LIU Q, ZHANG F G, ZHANG W S, et al. Ginsenoside Rg1 inhibits glucagon-induced hepatic gluconeogenesis through Akt-FoxO1 interaction [J]. Science Foundation in China, 2017, 7(4): 4001-4012. DOI: 10.7150/thno.18788http://dx.doi.org/10.7150/thno.18788.

尚文斌, 郭超, 赵娟,等. 人参皂苷Rb1通过上调脂肪组织葡萄糖转运体促进葡萄糖消耗[J]. 中国中药杂志, 2014, 39(22): 4448-4452. DOI: 10.4268/cjcmm20142232http://dx.doi.org/10.4268/cjcmm20142232.

SHANG W B, GUO C, ZHAO J, et al. Ginsenoside Rb1 upregulates expressions of GLUTs to promote glucose consumption in adiopcytes [J]. China Journal of Chinese Materia Medica, 2014, 39(22): 4448-4452. DOI: 10.4268/cjcmm20142232 (Chhttp://dx.doi.org/10.4268/cjcmm20142232(Ch).

DYER J, WOOD I S, PALEJWALA A, et al. Expression of monosaccharide transporters in intestine of diabetic humans [J]. The American Journal of Physiology-Gastrointestinal and Liver Physiology, 2002, 282(2): G241. DOI: 10.1152/ajpgi.00310.2001http://dx.doi.org/10.1152/ajpgi.00310.2001.

DOMINGUEZ R J A, CHIRASANI V R, HERMANN K, et al. Regulation of intestinal SGLT1 by catestatin in hyperleptinemic type 2 diabetic mice [J]. Laboratory Investigation, 2016, 96(1): 98-111. DOI: 10.1038/labinvest.2015.129http://dx.doi.org/10.1038/labinvest.2015.129.

WANG C W, SU S C, HUANG S F, et al. An essential role of cAMP response element binding protein in ginsenoside Rg1-mediated inhibition of Na+/glucose cotransporter 1 gene expression [J]. Molecular Pharmacology, 2015, 88(6): 1072-1083. DOI: 10.1124/mol.114.097352http://dx.doi.org/10.1124/mol.114.097352.

LACKEY D E, OLEFSKY J M. Regulation of metabolism by the innate immune system [J]. Nature Reviews Endocrinology, 2015, 12(1): 15-28. DOI: 10.1038/nrendo.2015.189http://dx.doi.org/10.1038/nrendo.2015.189.

WU Y Z, YU Y H, SZABO A, et al. Central inflammation and leptin resistance are attenuated by ginsenoside Rb1 treatment in obese mice fed a high-fat diet [J]. PloS One, 2014, 9(3): e92618. DOI: 10.1371/journal.pone.0092618http://dx.doi.org/10.1371/journal.pone.0092618.

PARK B K, LEE E A, KIM H Y, et al. Fatty liver and insulin resistance in the liver-specific knockout mice of mitogen inducible gene-6 [J]. Journal of Diabetes Research, 2016: 1-9. DOI: 10.1155/2016/1632061http://dx.doi.org/10.1155/2016/1632061.

HUANG C N, WANG C J, LEE Y J, et al. Active subfractions of abelmoschus esculentus substantially prevent free fatty acid-induced beta cell apoptosis via inhibiting dipeptidyl peptidase-4 [J]. PLoS One, 2017, 12(7): e0180285. DOI: 10.1371/journal.pone.0180285http://dx.doi.org/10.1371/journal.pone.0180285.

TIAN W, CHEN L, ZHANG L, et al. Effects of ginsenoside Rg1 on glucose metabolism and liver injury in streptozotocin-induced type 2 diabetic rats [J]. Genetics & Molecular Research, 2017, 16(1): gmr16019463. DOI: 10.4238/gmr16019463http://dx.doi.org/10.4238/gmr16019463.

李鑫. 人参皂苷Rg1对2型糖尿病大鼠肝损伤保护作用[J]. 中国公共卫生, 2015, 31(5): 612-614. DOI: 10.11847/zgggws2015-31-05-21http://dx.doi.org/10.11847/zgggws2015-31-05-21.

LI X. Protective effect of ginsenoside Rg1 on liver injury in rats with type 2 diabetes [J]. Chinese Journal of Public Health, 2015, 31(5): 612-614. DOI: 10.11847/zgggws2015-31-05-21(Chhttp://dx.doi.org/10.11847/zgggws2015-31-05-21(Ch).

HUANG Q, WANG T, YANG L, et al. Ginsenoside Rb2 alleviates hepatic lipid accumulation by restoring autophagy via induction of sirt1 and activation of AMPK [J]. International Journal of Molecular Sciences, 2017, 18(5):1063. DOI: 10.3390/ijms18051063http://dx.doi.org/10.3390/ijms18051063.

赵宗江, 张学凯, 张新雪, 等. Rg1、Rb1对糖尿病肾病大鼠肾脏保护作用及其对肾组织TGF-β1 mRNA与蛋白表达的影响[J]. 北京中医药大学学报, 2008, 31(6): 373-377.

ZHAO Z J, ZHANG X K, ZHANG X X, et al. Protective effect of Rg1 and Rb1 on kidney in rats with diabetic nephropathy and their influences on expressions of TGF-β1 mRNA and protein of renal tissue [J]. Journal of Beijing University of Traditional Chinese Medicine, 2008, 31(6): 373-377(Ch).

张丽娜, 陈晖, 尤冠巧, 等. 人参皂甙Rg1对糖尿病肾病大鼠肾皮质中MMP-2、9和Ⅳ型胶原表达的影响[J]. 世界最新医学信息文摘, 2015, 15(60): 103-104. DOI:10.3969/j.issn.1671-3141.2015.60.093http://dx.doi.org/10.3969/j.issn.1671-3141.2015.60.093.

ZHANG L N, CHEN H, YOU G Q, et al. Effects of ginsenoside Rg1 on the expression of MMP-2, 9 and IV collagen in renal cortex of diabetic nephropathy rats [J]. World Latest Medicine Information, 2015, 15(60): 103-104. DOI: 10.3969/j.issn.1671-3141.2015.60.093(Chhttp://dx.doi.org/10.3969/j.issn.1671-3141.2015.60.093(Ch).

张丽娜, 谢席胜, 左川, 等. 人参皂甙Rg1对糖尿病肾病大鼠TNF-α、MCP-1表达的影响[J]. 四川大学学报(医学版), 2009, 40(3): 466-471.

ZHANG L N, XIE X S, ZUO C, et al. Effect of ginsenoside Rgl on the expression of TNF-α and MCP-1 in rats with diabetic nephropathy [J]. Journal of Sichuan University(Medical Science Edition), 2009, 40(3): 466-471(Ch).

YU H T, ZHEN J, YANG Y, et al. Ginsenoside Rg1 ameliorates diabetic cardiomyopathy by inhibiting endoplasmic reticulum stress-induced apoptosis in a streptozotocin-induced diabetes rat model [J]. Journal of Cellular & Molecular Medicine, 2016, 20(4): 623-631. DOI: 10.1111/jcmm.12739http://dx.doi.org/10.1111/jcmm.12739.

PUTAALA J, LIEBKIND R, GORDIN D, et al. Diabetes mellitus and ischemic stroke in the young: Clinical features and long-term prognosis [J]. Neurology, 2011, 76(21): 1831-1837. DOI: 10.1212/WNL.0b013e31821cccc2http://dx.doi.org/10.1212/WNL.0b013e31821cccc2.

SHEN J, ZHAO Z, SHANG W, et al. Ginsenoside Rg1 nanoparticle penetrating the blood-brain barrier to improve the cerebral function of diabetic rats complicated with cerebral infarction [J]. International Journal of Nanomedicine, 2017, 12: 6477-6486. DOI: 10.2147/IJN.S139602http://dx.doi.org/10.2147/IJN.S139602.

LIU D, ZHANG H, GU W J, et al. Ginsenoside Rb1 protects hippocampal neurons from high glucose-induced neurotoxicity by inhibiting GSK3β-mediated CHOP induction [J]. Molecular Medicine Reports, 2014, 9(4): 1434-1438. DOI: 10.3892/mmr.2014.1958http://dx.doi.org/10.3892/mmr.2014.1958.



【本文地址】


今日新闻


推荐新闻


CopyRight 2018-2019 办公设备维修网 版权所有 豫ICP备15022753号-3