Nutr Res Pract.  2022 Jun;16(3):298-313. 10.4162/nrp.2022.16.3.298.

Gynostemma pentaphyllum extract and its active component gypenoside L improve the exercise performance of treadmill-trained mice

Affiliations
  • 1Technology Development Center, BTC Corporation, Ansan 15588, Korea
  • 2Regional Strategic Industry Innovation Center, Hallym University, Chuncheon 24252, Korea

Abstract

BACKGROUND/OBJECTIVES
The effectiveness of natural compounds in improving athletic ability has attracted attention in both sports and research. Gynostemma pentaphyllum (Thunb.) leaves are used to make traditional herbal medicines in Asia. The active components of G.pentaphyllum, dammarane saponins, or gypenosides, possess a range of biological activities. On the other hand, the anti-fatigue effects from G. pentaphyllum extract (GPE) and its effective compound, gypenoside L (GL), remain to be determined.
MATERIALS/METHODS
This study examined the effects of GPE on fatigue and exercise performance in ICR mice. GPE was administered orally to mice for 6 weeks, with or without treadmill training. The biochemical analysis in serum, glycogen content, mRNA, and protein expressions of the liver and muscle were analyzed.
RESULTS
The ExGPE (exercise with 300 mg/kg body weight/day of GPE) mice decreased the fat mass percentage significantly compared to the ExC mice, while the ExGPE showed the greatest lean mass percentage compared to the ExC group. The administration of GPE improved the exercise endurance and capacity in treadmill-trained mice, increased glucose and triglycerides, and decreased the serum creatine kinase and lactate levels after intensive exercise. The muscle glycogen levels were higher in the ExGPE group than the ExC group. GPE increased the level of mitochondrial biogenesis by enhancing the phosphorylation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) protein and the mRNA expression of nuclear respiratory factor 1, mitochondrial DNA, peroxisome proliferatoractivated receptor-δ, superoxide dismutase 2, and by decreasing the lactate dehydrogenase B level in the soleus muscle (SOL). GPE also improved PGC-1α activation in the SOL significantly through AMPK/p38 phosphorylation.
CONCLUSIONS
These results showed that GPE supplementation enhances exercise performance and has anti-fatigue activity. In addition, the underlying molecular mechanism was elucidated. Therefore, GPE is a promising candidate for developing functional foods and enhancing the exercise capacity and anti-fatigue activity.

Keyword

Gynostemma pentaphyllum; exercise; fatigue; glycogen; mitochondrial biogenesis

Figure

  • Fig. 1 Experimental design to examine the effects of GPE, GL, CrM supplementation on exercise adaptation. The animals were assigned randomly to the indicated 6 groups (n = 10 per group in each test). Physical exercise capacity and related assessments were conducted during the test for 6 weeks.GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW/day of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM.

  • Fig. 2 Effects of GPE and GL treatment on treadmill exercise performance. (A) Endurance time to exhaustion and (B) exercise capacity. Values are means ± SEM for n = 10 per group. GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW/day of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM.***P < 0.001 (SC group vs. SGPE or ExC group); #P < 0.05, ##P < 0.01, ###P < 0.001 (ExC group vs. ExGPE, ExGL or ExCrM group).

  • Fig. 3 Effects of GPE and GL treatment on glycogen contents in the liver and muscle of ICR mice. (A) Glycogen content (mg/g liver). (B) Total glycogen in liver (mg). (C) Glycogen content (mg/g GA). (D) Total glycogen in GA (mg). The values are the means ± SEM for n = 10 per group.GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW/day of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM; GA, gastrocnemius muscle.#P < 0.05 (ExC group vs. ExGPE, ExGL or ExCrM group).

  • Fig. 4 Effects of the GPE and GL treatment on the protein and mRNA expression of mitochondrial biogenesis genes in the soleus muscle of ICR mice. (A) Protein levels of phosphorylated PGC-1α and PGC-1α were analyzed using a western blot assay. β-actin served as an internal control. The ratio of p-PGC-1α/PGC-1α was determined (n = 4 per group). The mRNA levels of Nrf1 (B), and mtDNA content (C) were analyzed by qRT-PCR. The target mRNA expression was normalized to Gapdh. The values are means ± SEM for n = 8–10 per group.GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM; p-PGC-1α, phosphorylated peroxisome proliferator-activated receptor γ coactivator 1-alpha; PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-alpha; Nrf1, nuclear respiratory factor 1; mtDNA, mitochondrial DNA; qRT-PCR, quantitative reverse-transcription polymerase chain reaction; Gapdh, glyceraldehyde 3-phosphate dehydrogenase.*P < 0.05, **P < 0.01, ***P < 0.001 (SC group vs. SGPE or ExC group); #P < 0.05, ##P < 0.01, ###P < 0.001 (ExC group vs. ExGPE, ExGL, or ExCrM group).

  • Fig. 5 Effects of the GPE and GL treatment on the mRNA expression of PGC-1α targeted genes in the soleus muscle of ICR mice. The mRNA levels of Ldhb (A), Ppard (B), and Sod2 (C) were analyzed by qRT-PCR. The target mRNA expression was normalized to Gapdh. Values are means ± SEM for n = 4–6 per group.GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW/day of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM; PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-alpha; qRT-PCR, quantitative reverse-transcription polymerase chain reaction; Ldhb, lactate dehydrogenase B; Ppard, peroxisome proliferator-activated receptor-δ; Sod2, superoxide dismutase 2; Gapdh, glyceraldehyde 3-phosphate dehydrogenase.*P < 0.05 (SC group vs. SGPE or ExC group); #P < 0.05, ##P < 0.01 (ExC group vs. ExGPE, ExGL, or ExCrM group).

  • Fig. 6 Effects of the GPE and GL treatment on PGC-1α activation through AMPK/p38 phosphorylation in soleus muscle of ICR mice. The protein levels of phosphorylated AMPK and AMPK (A) and phosphorylated p38, and p38 (B) were analyzed by western blot assay. β-actin served as an internal control. The ratio of p-AMPK/AMPK and p-p38/p38 were determined. The values are the means ± SEM for n = 4 per group.GPE, G. pentaphyllum extract; GL, gypenoside L; BW, body weight; CrM, creatine monohydrate; SC, sedentary with vehicle; SGPE, sedentary with 300 mg/kg BW/day of GPE; ExC, exercise with vehicle; ExGPE, exercise with 300 mg/kg BW/day of GPE; ExGL, exercise with 7 mg/kg BW/day of GL; ExCrM, exercise with 75 mg/kg BW/day of CrM; PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-alpha; p-AMPK, phosphorylated adenosine monophosphate-activated protein kinase; AMPK, adenosine monophosphate-activated protein kinase; p-p38, phosphorylated p38 MAP kinase; p38, p38 MAP kinase.*P < 0.05 (SC group vs. SGPE or ExC group); #P < 0.05, ##P < 0.01, ###P < 0.001 (ExC group vs. ExGPE, ExGL, or ExCrM group).

  • Fig. 7 Summary of the effects of the G. pentaphyllum extract on the exercise endurance and energy metabolism.GPE, G. pentaphyllum extract; p38, p38 MAP kinase; AMPK, adenosine monophosphate-activated protein kinase; PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-alpha; LDH B, lactate dehydrogenase B; LDH A, lactate dehydrogenase A; PPARδ, peroxisome proliferator-activated receptor-δ; NRF-1, nuclear respiratory factor 1; SOD2, superoxide dismutase 2; TFAM, transcription factor A mitochondrial; mtDNA, mitochondrial DNA.


Reference

1. Matsukawa T, Motojima H, Sato Y, Takahashi S, Villareal MO, Isoda H. Upregulation of skeletal muscle PGC-1α through the elevation of cyclic AMP levels by cyanidin-3-glucoside enhances exercise performance. Sci Rep. 2017; 7:44799. PMID: 28317895.
2. Safdar A, Little JP, Stokl AJ, Hettinga BP, Akhtar M, Tarnopolsky MA. Exercise increases mitochondrial PGC-1alpha content and promotes nuclear-mitochondrial cross-talk to coordinate mitochondrial biogenesis. J Biol Chem. 2011; 286:10605–10617. PMID: 21245132.
3. Wang H, Li C, Wu X, Lou X. Effects of Gynostemma pentaphyllum (Thunb.) Makino polysaccharides supplementation on exercise tolerance and oxidative stress induced by exhaustive exercise in rats. Afr J Agric Res. 2012; 7:2632–2638.
4. Lin-Na S, Yong-Xiu S. Effects of polysaccharides from Gynostemma pentaphyllum (Thunb.), Makino on physical fatigue. Afr J Tradit Complement Altern Med. 2014; 11:112–117. PMID: 25371572.
5. Shang X, Chao Y, Zhang Y, Lu C, Xu C, Niu W. Immunomodulatory and antioxidant effects of polysaccharides from Gynostemma pentaphyllum Makino in immunosuppressed mice. Molecules. 2016; 21:1085.
6. Quan Y, Qian MZ. Effect and mechanism of gypenoside on the inflammatory molecular expression in high-fat induced atherosclerosis rats. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2010; 30:403–406. PMID: 20669679.
7. Cai H, Liang Q, Ge G. Gypenoside attenuates beta amyloid-induced inflammation in N9 microglial cells via SOCS1 signaling. Neural Plast. 2016; 2016:6362707. PMID: 27213058.
8. Liou CJ, Huang WC, Kuo ML, Yang RC, Shen JJ. Long-term oral administration of Gynostemma pentaphyllum extract attenuates airway inflammation and Th2 cell activities in ovalbumin-sensitized mice. Food Chem Toxicol. 2010; 48:2592–2598. PMID: 20558230.
9. Müller C, Gardemann A, Keilhoff G, Peter D, Wiswedel I, Schild L. Prevention of free fatty acid-induced lipid accumulation, oxidative stress, and cell death in primary hepatocyte cultures by a Gynostemma pentaphyllum extract. Phytomedicine. 2012; 19:395–401. PMID: 22381945.
10. Gou SH, Huang HF, Chen XY, Liu J, He M, Ma YY, Zhao XN, Zhang Y, Ni JM. Lipid-lowering, hepatoprotective, and atheroprotective effects of the mixture Hong-Qu and gypenosides in hyperlipidemia with NAFLD rats. J Chin Med Assoc. 2016; 79:111–121. PMID: 26842974.
11. la Cour B, Mølgaard P, Yi Z. Traditional Chinese medicine in treatment of hyperlipidaemia. J Ethnopharmacol. 1995; 46:125–129. PMID: 7650951.
12. Wang M, Wang F, Wang Y, Ma X, Zhao M, Zhao C. Metabonomics study of the therapeutic mechanism of Gynostemma pentaphyllum and atorvastatin for hyperlipidemia in rats. PLoS One. 2013; 8:e78731. PMID: 24223845.
13. Lee HS, Lim SM, Jung JI, Kim SM, Lee JK, Kim YH, Cha KM, Oh TK, Moon JM, Kim TY, et al. Gynostemma pentaphyllum extract ameliorates high-fat diet-induced obesity in C57BL/6N mice by upregulating SIRT1. Nutrients. 2019; 11:2475.
14. Huyen VT, Phan DV, Thang P, Hoa NK, Ostenson CG. Gynostemma pentaphyllum tea improves insulin sensitivity in type 2 diabetic patients. J Nutr Metab. 2013; 2013:765383. PMID: 23431428.
15. Keilhoff G, Esser T, Titze M, Ebmeyer U, Schild L. Gynostemma pentaphyllum is neuroprotective in a rat model of cardiopulmonary resuscitation. Exp Ther Med. 2017; 14:6034–6046. PMID: 29250141.
16. Li Y, Lin W, Huang J, Xie Y, Ma W. Anti-cancer effects of Gynostemma pentaphyllum (Thunb.) Makino (Jiaogulan). Chin Med. 2016; 11:43. PMID: 27708693.
17. Kao TH, Huang SC, Inbaraj BS, Chen BH. Determination of flavonoids and saponins in Gynostemma pentaphyllum (Thunb.) Makino by liquid chromatography-mass spectrometry. Anal Chim Acta. 2008; 626:200–211. PMID: 18790122.
18. Xie Z, Liu W, Huang H, Slavin M, Zhao Y, Whent M, Blackford J, Lutterodt H, Zhou H, Chen P, et al. Chemical composition of five commercial Gynostemma pentaphyllum samples and their radical scavenging, antiproliferative, and anti-inflammatory properties. J Agric Food Chem. 2010; 58:11243–11249. PMID: 20939605.
19. Lin CC, Huang PC, Lin JM. Antioxidant and hepatoprotective effects of Anoectochilus formosanus and Gynostemma pentaphyllum . Am J Chin Med. 2000; 28:87–96. PMID: 10794120.
20. Liu J, Zhang L, Ren Y, Gao Y, Kang L, Qiao Q. Anticancer and immunoregulatory activity of Gynostemma pentaphyllum polysaccharides in H22 tumor-bearing mice. Int J Biol Macromol. 2014; 69:1–4. PMID: 24832985.
21. Attawish A, Chivapat S, Phadungpat S, Bansiddhi J, Techadamrongsin Y, Mitrijit O, Chaorai B, Chavalittumrong P. Chronic toxicity of Gynostemma pentaphyllum . Fitoterapia. 2004; 75:539–551. PMID: 15351107.
22. Chi AP, Chen JP, Wang ZZ, Xiong ZY, Li QX. Morphological and structural characterization of a polysaccharide from Gynostemma pentaphyllum Makino and its anti-exercise fatigue activity. Carbohydr Polym. 2008; 74:868–874.
23. Yang X, Zhao Y, Yang Y, Ruan Y. Isolation and characterization of immunostimulatory polysaccharide from an herb tea, Gynostemma pentaphyllum Makino. J Agric Food Chem. 2008; 56:6905–6909. PMID: 18636735.
24. Ding YJ, Tang KJ, Li FL, Hu QL. Effects of gypenosides from Gynostemma pentaphyllum supplementation on exercise-induced fatigue in mice. Afr J Agric Res. 2010; 5:707–711.
25. Tadaishi M, Miura S, Kai Y, Kano Y, Oishi Y, Ezaki O. Skeletal muscle-specific expression of PGC-1α-b, an exercise-responsive isoform, increases exercise capacity and peak oxygen uptake. PLoS One. 2011; 6:e28290. PMID: 22174785.
26. Kim YH, Kim SM, Lee JK, Jo SK, Kim HJ, Cha KM, Lim CY, Moon JM, Kim TY, Kim EJ. Efficacy of Gynostemma pentaphyllum extract in anti-obesity therapy. Rec Nat Prod. 2020; 14:116–128.
27. Kim YH, Jung JI, Jeon YE, Kim SM, Oh TK, Lee J, Moon JM, Kim TY, Kim EJ. Gynostemma pentaphyllum extract and gypenoside L enhance skeletal muscle differentiation and mitochondrial metabolism by activating the PGC-1α pathway in C2C12 myotubes. Nutr Res Pract. 2021; 15:e45.
28. Hearris MA, Hammond KM, Fell JM, Morton JP. Regulation of muscle glycogen metabolism during exercise: implications for endurance performance and training adaptations. Nutrients. 2018; 10:298.
29. Jensen J, Rustad PI, Kolnes AJ, Lai YC. The role of skeletal muscle glycogen breakdown for regulation of insulin sensitivity by exercise. Front Physiol. 2011; 2:112. PMID: 22232606.
30. Cheng CF, Ku HC, Lin H. PGC-1α as a pivotal factor in lipid and metabolic regulation. Int J Mol Sci. 2018; 19:3447.
31. Kang C, Li Ji L. Role of PGC-1α signaling in skeletal muscle health and disease. Ann N Y Acad Sci. 2012; 1271:110–117. PMID: 23050972.
32. Wright DC, Han DH, Garcia-Roves PM, Geiger PC, Jones TE, Holloszy JO. Exercise-induced mitochondrial biogenesis begins before the increase in muscle PGC-1α expression. J Biol Chem. 2007; 282:194–199. PMID: 17099248.
33. Puigserver P, Rhee J, Lin J, Wu Z, Yoon JC, Zhang CY, Krauss S, Mootha VK, Lowell BB, Spiegelman BM. Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator-1. Mol Cell. 2001; 8:971–982. PMID: 11741533.
34. Jäger S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proc Natl Acad Sci U S A. 2007; 104:12017–12022. PMID: 17609368.
35. Wan JJ, Qin Z, Wang PY, Sun Y, Liu X. Muscle fatigue: general understanding and treatment. Exp Mol Med. 2017; 49:e384. PMID: 28983090.
36. Facey A, Irving R, Dilworth L. Overview of lactate metabolism and the implications for athletes. J Sports Sci Med. 2013; l:42–46.
37. Hirabara SM, Silveira LR, Abdulkader FR, Alberici LC, Procopio J, Carvalho CR, Pithon-Curi TC, Curi R. Role of fatty acids in the transition from anaerobic to aerobic metabolism in skeletal muscle during exercise. Cell Biochem Funct. 2006; 24:475–481. PMID: 16924590.
38. DeFronzo RA. Lilly lecture 1987. The triumvirate: beta-cell, muscle, liver. A collusion responsible for NIDDM. Diabetes. 1988; 37:667–687. PMID: 3289989.
39. Liu Y, Liu C. Antifatigue and increasing exercise performance of Actinidia arguta crude alkaloids in mice. J Food Drug Anal. 2016; 24:738–745. PMID: 28911611.
40. Martin WH 3rd, Dalsky GP, Hurley BF, Matthews DE, Bier DM, Hagberg JM, Rogers MA, King DS, Holloszy JO. Effect of endurance training on plasma free fatty acid turnover and oxidation during exercise. Am J Physiol. 1993; 265:E708–E714. PMID: 8238496.
41. Horowitz JF, Klein S. Lipid metabolism during endurance exercise. Am J Clin Nutr. 2000; 72:558S–563S. PMID: 10919960.
42. Li F, Li J, Li S, Guo S, Li P. Modulatory effects of Chinese herbal medicines on energy metabolism in ischemic heart diseases. Front Pharmacol. 2020; 11:995. PMID: 32719602.
43. Kang D, Hamasaki N. Mitochondrial transcription factor A in the maintenance of mitochondrial DNA: overview of its multiple roles. Ann N Y Acad Sci. 2005; 1042:101–108. PMID: 15965051.
44. Rowe GC, Jiang A, Arany Z. PGC-1 coactivators in cardiac development and disease. Circ Res. 2010; 107:825–838. PMID: 20884884.
45. Nalbandian M, Takeda M. Lactate as a signaling molecule that regulates exercise induced adaptations. Biology (Basel). 2016; 5:38.
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr