Antidiabetic Activity of a Flavonoid-Rich Extract From Sophora davidii (Franch.) Skeels in KK-Ay Mice via Activation of AMP-Activated Protein Kinase

The present study was undertaken to investigate the hypoglycemic activity and potential mechanisms of action of a flavonoid-rich extract from Sophora davidii (Franch.) Skeels (SD-FRE) through in vitro and in vivo studies. Four main flavonoids of SD-FRE namely apigenin, maackiain, leachianone A and leachianone B were purified and identified. In vitro, SD-FRE significantly promoted the translocation and expression of glucose transporter 4 (GLUT4) in L6 cells, which was significantly inhibited by Compound C (AMPK inhibitor), but not by Wortmannin (PI3K inhibitor) or Gö6983 (PKC inhibitor). These results indicated that SD-FRE enhanced GLUT4 expression and translocation to the plasma membrane via the AMPK pathway and finally resulted in an increase of glucose uptake. In vivo, using a spontaneously type 2 diabetic model, KK-Ay mice received intragastric administration of SD-FRE for 4 weeks. As a consequence, SD-FRE significantly alleviated the hyperglycemia, glucose intolerance, insulin resistance and hyperlipidemia in these mice. Hepatic steatosis, islet hypertrophy and larger adipocyte size were observed in KK-Ay mice. However, these pathological changes were effectively relieved by SD-FRE treatment. SD-FRE promoted GLUT4 expression and activated AMPK phosphorylation in insulin target tissues (muscle, adipose tissue and liver) of KK-Ay mice, thus facilitating glucose utilization to ameliorate insulin resistance. Regulation of ACC phosphorylation and PPARγ were also involved in the antidiabetic effects of SD-FRE. Taken together, these findings indicated that SD-FRE has the potential to alleviate type 2 diabetes.

[1]  Mi Huang,et al.  Activity of Isoliensinine in Improving the Symptoms of Type 2 Diabetic Mice via Activation of AMP-Activated Kinase and Regulation of PPARγ. , 2017, Journal of agricultural and food chemistry.

[2]  Mi Huang,et al.  Hypoglycemic Activity and the Potential Mechanism of the Flavonoid Rich Extract from Sophora tonkinensis Gagnep. in KK-Ay Mice , 2016, Front. Pharmacol..

[3]  Md. Asiful Islam,et al.  Updates on Managing Type 2 Diabetes Mellitus with Natural Products: Towards Antidiabetic Drug Development. , 2016, Current medicinal chemistry.

[4]  M. Prentki,et al.  AMP-activated protein kinase and coordination of hepatic fatty acid metabolism of starved/carbohydrate-refed rats. , 2005, American journal of physiology. Endocrinology and metabolism.

[5]  T. Nishizaki,et al.  Diacylglycerol promotes GLUT4 translocation to the cell surface in a PKCε-dependent and PKCλ/ι and -ζ-independent manner. , 2013, Life sciences.

[6]  M. Lampson,et al.  Demonstration of insulin-responsive trafficking of GLUT4 and vpTR in fibroblasts. , 2000, Journal of cell science.

[7]  J. Shaw,et al.  IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. , 2018, Diabetes research and clinical practice.

[8]  Zheng-Tao Wang,et al.  Natural Flavonoids as Potential Herbal Medication for the Treatment of Diabetes Mellitus and its Complications , 2015, Natural product communications.

[9]  Yabin Yang,et al.  Antioxidant activity and chemical constituents of edible flower of Sophora viciifolia. , 2011, Food chemistry.

[10]  A. Albiston,et al.  GLUT4 associated proteins as therapeutic targets for diabetes. , 2011, Recent patents on endocrine, metabolic & immune drug discovery.

[11]  F. Thong,et al.  Turning signals on and off: GLUT4 traffic in the insulin-signaling highway. , 2005, Physiology.

[12]  E. Bonora,et al.  Homeostasis model assessment closely mirrors the glucose clamp technique in the assessment of insulin sensitivity: studies in subjects with various degrees of glucose tolerance and insulin sensitivity. , 2000, Diabetes care.

[13]  A. Kitabchi,et al.  Management of type 2 diabetes: evolving strategies for the treatment of patients with type 2 diabetes. , 2011, Metabolism: clinical and experimental.

[14]  Jinhua Shen,et al.  Antidiabetic Activity of Ergosterol from Pleurotus Ostreatus in KK‐Ay Mice with Spontaneous Type 2 Diabetes Mellitus , 2018, Molecular nutrition & food research.

[15]  M. Prentki,et al.  Type 2 diabetes across generations: from pathophysiology to prevention and management , 2011, The Lancet.

[16]  S. Kahn,et al.  Mechanisms linking obesity to insulin resistance and type 2 diabetes , 2006, Nature.

[17]  A. Sugawara,et al.  Transcription Suppression of Peroxisome Proliferator-Activated Receptor γ2 Gene Expression by Tumor Necrosis Factor α via an Inhibition of CCAAT/ Enhancer-binding Protein δ during the Early Stage of Adipocyte Differentiation , 2004 .

[18]  Gaochao Zhou,et al.  AMPK: an emerging drug target for diabetes and the metabolic syndrome. , 2009, Cell metabolism.

[19]  Koji Yamada,et al.  Cistanche salsa Extract Enhanced Antibody Production in Human Lymph Node Lymphocytes , 2008 .

[20]  M. Czech,et al.  The GLUT4 glucose transporter. , 2007, Cell metabolism.

[21]  Furong Zhao,et al.  Chemical study of the Chinese medicine Pi Han Yao. , 2016, Biomedical reports.

[22]  M. Sabran,et al.  The anti-obesity effects of Lactobacillus casei strain Shirota versus Orlistat on high fat diet-induced obese rats , 2015, Food & nutrition research.

[23]  Chunguo Wang,et al.  QiDiTangShen Granules Activate Renal Nutrient-Sensing Associated Autophagy in db/db Mice , 2019, Front. Physiol..

[24]  Alan R. Saltiel,et al.  Regulation of glucose transport by insulin: traffic control of GLUT4 , 2012, Nature Reviews Molecular Cell Biology.

[25]  L. Tecott,et al.  Up-Regulation of Peroxisome Proliferator-Activated Receptors (PPAR-α) and PPAR-γ Messenger Ribonucleic Acid Expression in the Liver in Murine Obesity: Troglitazone Induces Expression of PPAR-γ-Responsive Adipose Tissue-Specific Genes in the Liver of Obese Diabetic Mice* * This work was supported by , 2000, Endocrinology.

[26]  R. Aggarwal,et al.  Spectrum of malabsorption syndrome among adults & factors differentiating celiac disease & tropical malabsorption , 2012, The Indian journal of medical research.

[27]  D. Hardie Printed in U.S.A. Copyright © 2003 by The Endocrine Society doi: 10.1210/en.2003-0982 Minireview: The AMP-Activated Protein Kinase Cascade: The Key Sensor of Cellular Energy Status , 2022 .

[28]  L. Szablewski Glucose and Lipid Metabolism , 2011 .

[29]  V. Ramachandran,et al.  Glucose uptake through translocation and activation of GLUT4 in PI3K/Akt signaling pathway by asiatic acid in diabetic rats , 2015, Human & experimental toxicology.

[30]  Y. Shirataki,et al.  Two flavanones in Sophora leachiano and some related structures , 1990 .

[31]  H. Itokawa,et al.  Studies on a Novel p-Coumaroyl Glucoside of Apigenin and on Other Flavonoids isolated from Patchouli (Labiatae) , 1981 .

[32]  H. A. Lee,et al.  Jicama (Pachyrhizus erosus) extract increases insulin sensitivity and regulates hepatic glucose in C57BL/Ksj-db/db mice , 2015, Journal of clinical biochemistry and nutrition.

[33]  M. Esmaeili,et al.  Pancreatic BCell Protective Effect of Rutin and Ap igenin Isolated from Teucrium Polium , 2009 .

[34]  Haixia Chen,et al.  Network Pharmacology Studies on the Bioactive Compounds and Action Mechanisms of Natural Products for the Treatment of Diabetes Mellitus: A Review , 2017, Front. Pharmacol..

[35]  U. Boelsterli,et al.  Diabetic KKAy mice exhibit increased hepatic PPARgamma1 gene expression and develop hepatic steatosis upon chronic treatment with antidiabetic thiazolidinediones. , 2001, Journal of hepatology.

[36]  P. Ramarao,et al.  Animal models in type 2 diabetes research: an overview. , 2007, The Indian journal of medical research.

[37]  Jinhua Shen,et al.  Ethanolic Extract of Folium Sennae Mediates the Glucose Uptake of L6 Cells by GLUT4 and Ca2+ , 2018, Molecules.

[38]  B. Sepodes,et al.  Antihyperglycaemic and protective effects of flavonoids on streptozotocin–induced diabetic rats , 2010, Phytotherapy research : PTR.

[39]  Jinhua Shen,et al.  Neferine Promotes GLUT4 Expression and Fusion With the Plasma Membrane to Induce Glucose Uptake in L6 Cells , 2019, Front. Pharmacol..

[40]  R. B. Narayanan,et al.  Cinnamic acid, from the bark of Cinnamomum cassia, regulates glucose transport via activation of GLUT4 on L6 myotubes in a phosphatidylinositol 3‐kinase‐independent manner , 2009, Journal of diabetes.

[41]  F. Gonzalez,et al.  Adipocyte-specific Gene Expression and Adipogenic Steatosis in the Mouse Liver Due to Peroxisome Proliferator-activated Receptor γ1 (PPARγ1) Overexpression* , 2003, The Journal of Biological Chemistry.

[42]  D. James,et al.  Variations in the requirement for v-SNAREs in GLUT4 trafficking in adipocytes , 2009, Journal of Cell Science.

[43]  Michael Lehrke,et al.  The Many Faces of PPARγ , 2005, Cell.

[44]  C. Kahn,et al.  Insulin signalling and the regulation of glucose and lipid metabolism , 2001, Nature.

[45]  Mi Huang,et al.  Anti-diabetic activity of a polyphenol-rich extract from Phellinus igniarius in KK-Ay mice with spontaneous type 2 diabetes mellitus. , 2018, Food & function.

[46]  C. Kahn,et al.  Targeted disruption of the glucose transporter 4 selectively in muscle causes insulin resistance and glucose intolerance , 2000, Nature Medicine.

[47]  Z. Shen,et al.  Berberine inhibits mouse insulin gene promoter through activation of AMP activated protein kinase and may exert beneficial effect on pancreatic β-cell. , 2012, European journal of pharmacology.

[48]  Y. Yoshikawa,et al.  Morphological characterization of systemic changes in KK-Ay mice as an animal model of type 2 diabetes. , 2013, In vivo.

[49]  Y. Barak,et al.  Role for PPARγ in obesity‐induced hepatic steatosis as determined by hepatocyte‐ and macrophage‐specific conditional knockouts , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[50]  T. Bengtsson,et al.  alpha1A-adrenoceptors activate glucose uptake in L6 muscle cells through a phospholipase C-, phosphatidylinositol-3 kinase-, and atypical protein kinase C-dependent pathway. , 2005, Endocrinology.

[51]  Myung‐Sook Choi,et al.  Apigenin Ameliorates Dyslipidemia, Hepatic Steatosis and Insulin Resistance by Modulating Metabolic and Transcriptional Profiles in the Liver of High-Fat Diet-Induced Obese Mice , 2016, Nutrients.

[52]  Demetrios Vavvas,et al.  Malonyl-CoA, fuel sensing, and insulin resistance. , 1999, American journal of physiology. Endocrinology and metabolism.

[53]  G. Aliev,et al.  New Therapeutic Property of Dimebon as a Neuroprotective Agent. , 2016, Current medicinal chemistry.

[54]  Michiho Ito,et al.  Trypanocidal Flavonoids from Sophora flavescens , 2003 .

[55]  G. Shulman,et al.  Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver , 2001, Nature.

[56]  J. Borén,et al.  Fatty liver, insulin resistance, and dyslipidemia , 2008, Current diabetes reports.