DOI QR코드

DOI QR Code

Cordycepin Suppresses Expression of Diabetes Regulating Genes by Inhibition of Lipopolysaccharide-induced Inflammation in Macrophages

  • Received : 2009.04.13
  • Accepted : 2009.05.04
  • Published : 2009.06.30

Abstract

Background: It has been recently noticed that type 2 diabetes (T2D), one of the most common metabolic diseases, causes a chronic low-grade inflammation and activation of the innate immune system that are closely involved in the pathogenesis of T2D. Cordyceps militaris, a traditional medicinal mushroom, produces a component compound, cordycepin (3'-deoxyadenosine). Cordycepin has been known to have many pharmacological activities including immunological stimulating, anti-cancer, and anti-infection activities. The molecular mechanisms of cordycepin in T2D are not clear. In the present study, we tested the role of cordycepin on the anti-diabetic effect and anti-inflammatory cascades in LPS-stimulated RAW 264.7 cells. Methods: We confirmed the levels of diabetes regulating genes mRNA and protein of cytokines through RT-PCR and western blot analysis and followed by FACS analysis for the surface molecules. Results: Cordycepin inhibited the production of NO and pro-inflammatory cytokines such as IL-$1{\beta}$, IL-6, and TNF-${\alpha}$ in LPS-activated macrophages via suppressing protein expression of pro-inflammatory mediators. T2D regulating genes such as $11{\beta}$-HSD1 and PPAR${\gamma}$ were decreased as well as expression of co-stimulatory molecules such as ICAM-1 and B7-1/-2 were also decreased with the increment of its concentration. In accordance with suppressed pro-inflammatory cytokine production lead to inhibition of diabetic regulating genes in activated macrophages. Cordycepin suppressed NF-${\kappa}B$ activation in LPS-activated macrophages. Conclusion: Based on these observations, cordycepin suppressed T2D regulating genes through the inactivation of NF-${\kappa}B$ dependent inflammatory responses and suggesting that cordycepin will provide potential use as an immunomodulatory agent for treating immunological diseases.

Keywords

References

  1. Hotamisligil G, Arner P, Caro J, Atkinson R, Spiegelman B: Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95;2409-2415, 1995 https://doi.org/10.1172/JCI117936
  2. Ogden C, Carroll M, Curtin L, McDowell M, Tabak C, Flegal K: Prevalence of overweight and obesity in the United States, 1999-2004. Am Med Assoc 295;1549-1555, 2006 https://doi.org/10.1001/jama.295.13.1549
  3. Li W, Zheng H, Bukuru J, De Kimpe N: Natural medicines used in the traditional Chinese medical system for therapy of diabetes mellitus. J Ethnopharmacol 92;1-21, 2004 https://doi.org/10.1016/j.jep.2003.12.031
  4. DeFronzo R: Pathogenesis of NIDDM. A balanced overview. Diabetes care 15;318-368, 1992 https://doi.org/10.2337/diacare.15.3.318
  5. sj$\ddot{o}$holm A, Nystr$\ddot{o}$m T: Endothelial inflammation in insulin resistance. The Lancet 365;610-612, 2005
  6. Ross JA, Auger MJ, In Burke B, Lewis CE: The biology of the macrophage. In: Burke B, Lewis CE, editors. The macrophage. 2nd ed. Oxford: Oxford Medical Publications; p1-72, 2002
  7. Kinne R, Brauer R, Stuhlmuller B, Palombo-Kinne E, Burmester G: Macrophages in rheumatoid arthritis. Arthritis Research 2;189-202, 2000 https://doi.org/10.1186/ar86
  8. Gai G, Jin S, Wang B, Li Y, Li C: The efficacy of Cordyceps militaris capsules in treatment of chronic bronchitis in comparison with Jinshuibao capsules. Chin J New Drugs 13;169-171, 2004
  9. Paterson R: Cordyceps: a traditional Chinese medicine and another fungal therapeutic biofactory? Phytochemistry 69;1469-1495, 2008 https://doi.org/10.1016/j.phytochem.2008.01.027
  10. Yoo H, Shin J, Cho J, Son C, Lee Y, Park S, Cho C: Effects of Cordyceps militaris extract on angiogenesis and tumor growth. Acta Pharmacologica Sinica 25;657-665, 2004
  11. Kim H, Shrestha B, Lim S, Yoon D, Chang W, Shin D, Han S, Park S, Park J, Park H: Cordycepin inhibits lipopolysaccharide- induced inflammation by the suppression of NF-kappaB through Akt and p38 inhibition in RAW 264.7 macrophage cells. Eur J Pharmacol 545;192-199, 2006 https://doi.org/10.1016/j.ejphar.2006.06.047
  12. Yun Y, Han S, Lee S, Ko S, Lee C, Ha N, Kim K: Anti-diabetic effects of CCCA, CMESS, and cordycepin from Cordyceps militaris and the immune responses in streptozotocin- induced diabetic mice. Nat Pro Sci 9;291-298, 2003
  13. Cho M, Lee D, Kim M, Sung J, Ham S: Antimutagenicity and cytotoxicity of cordycepin isolated from Cordyceps militaris. Food Sci Biotechnol 12;472-475, 2003
  14. Majumder N, Dey R, Mathur R, Datta S, Maitra M, Ghosh S, Saha B, Majumdar S: An unusual pro-inflammatory role of interleukin-10 induced by arabinosylated lipoarabinomannan in murine peritoneal macrophages. Glycoconjugate J 23;675-686, 2006 https://doi.org/10.1007/s10719-006-9017-9
  15. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends in immunology 25;677-686, 2004 https://doi.org/10.1016/j.it.2004.09.015
  16. Furchgott R, Cherry P, Zawadzki J, Jothianandan D: Endothelial cells as mediators of vasodilation of arteries. J Cardiovasc Pharmacol 6;S336-343, 1984 https://doi.org/10.1097/00005344-198406002-00008
  17. Rausch-Fan X, Matejka M: From plaque formation to periodontal disease, is there a role for nitric oxide? Eur J Clin Invest 31;833-835, 2008 https://doi.org/10.1046/j.1365-2362.2001.00903.x
  18. Wei X, Charles I, Smith A, Ure J, Feng G, Huang F, Xu D, Muller W, Moncada S, Liew F: Altered immune responses in mice lacking inducible nitric oxide synthase. Nature 375;408-411, 1995 https://doi.org/10.1038/375408a0
  19. Haddad JJ, Safieh-Garabedian B, Saad$\acute{e}$ NE, Kanaan SA, Land SC: Chemioxyexcitation (delta pO2/ROS)-dependent release of IL-1 beta, IL-6 and TNF-alpha: evidence of cytokines as oxygen-sensitive mediators in the alveolar epithelium. Cytokine 13;138-147, 2001 https://doi.org/10.1006/cyto.2000.0789
  20. Moestrup S, Moller H: CD163: a regulated hemoglobin scavenger receptor with a role in the anti-inflammatory response. Annals of Medicine 36;347-354, 2004 https://doi.org/10.1080/07853890410033171
  21. Hotamisligil G, Shargill N, Spiegelman B: Adipose expression of tumor necrosis factor-alpha: direct role in obesity- linked insulin resistance. Science 259;87-91, 1993 https://doi.org/10.1126/science.7678183
  22. Rotter V, Nagaev I, Smith U: Interleukin-6 (IL-6) Induces Insulin Resistance in 3T3-L1 Adipocytes and Is, Like IL-8 and Tumor Necrosis Factor-α, Overexpressed in Human Fat Cells from Insulin-resistant Subjects. J Bio Chem 278;45777-45784, 2003 https://doi.org/10.1074/jbc.M301977200
  23. Uysal KT, Wiesbrock SM, Hotamisligil GS: Functional analysis of tumor necrosis factor (TNF) receptors in TNF-alpha-mediated insulin resistance in genetic obesity. Endocrinology 139;4832-4838, 1998 https://doi.org/10.1210/en.139.12.4832
  24. Xu H, Barnes G, Yang Q, Tan G, Yang D, Chou C, Sole J, Nichols A, Ross J, Tartaglia L: Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112;1821-1830, 2003
  25. Bujalska I, Kumar S, Stewart P: Does central obesity reflect Cushing's disease of the omentum? Lancet (British edition) 349;1210-1213, 1997 https://doi.org/10.1016/S0140-6736(96)11222-8
  26. Tomlison J, Stewart P: 11$\beta$-Hydroxysteroid dehydrogenase type I as a therapeutic target in the metabolic syndrome. Drug Discov Today 2;93-96, 2005 https://doi.org/10.1016/j.ddmec.2005.05.008
  27. Wu H, Ghosh S, Perrard X, Feng L, Garcia G, Perrard J, Sweeney J, Peterson L, Chan L, Smith C: T-cell accumulation and regulated on activation, normal T cell expressed secreted upregulation in adipose tissue in obesity. Circulation 115;1029, 2007 https://doi.org/10.1161/CIRCULATIONAHA.106.638379
  28. Issemann I, Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347;645-650, 1990 https://doi.org/10.1038/347645a0
  29. Marx N, Sukhova G, Murphy C, Libby P, Plutzky J: Macrophages in human atheroma contain ppar$\gamma$ differentiation- dependent peroxisomal proliferator-activated receptor $\gamma$ (PPAR$\gamma$) expression and reduction of MMP-9 activity through PPAR$\gamma$ activation in mononuclear phagocytes in vitro. ASIP 153;17-23, 1998
  30. Nicklin M, Hughes D, Barton J, Ure J, Duff G: Arterial inflammation in mice lacking the interleukin 1 receptor antagonist gene. J Exp Med 191;303-312, 2000 https://doi.org/10.1084/jem.191.2.303
  31. Horai R, Saijo S, Tanioka H, Nakae S, Sudo K, Okahara A, Ikuse T, Asano M, Iwakura Y: Development of chronic inflammatory arthropathy resembling rheumatoid arthritis in interleukin 1 receptor antagonist-deficient mice. J Exp Med 191;313-320, 2000 https://doi.org/10.1084/jem.191.2.313
  32. Yamshchikov V, Mishina M, Cominelli F: A possible role of IL-1ra 3’-untranslated region in modulation of protein production. Cytokine 17;98-107, 2002
  33. Beach L, Ross J: Cordycepin. An inhibitor of newly synthesized globin messenger RNA. J Bio Chem 253;2628-2632, 1978
  34. Dazzi F, D'andrea E, Biasi G, De Silvestro G, Gaidano G, Schena M, Tison T, Vianello F, Girolami A, Caligaris-Cappio F: Failure of B cells of chronic lymphocytic leukemia in presenting soluble and alloantigens. Clin Immunol Immunopathol 75;26-32, 1995 https://doi.org/10.1006/clin.1995.1048
  35. Deeths MJ, Mescher MF: ICAM-1 and B7-1 provide similar but distinct costimulation for CD8+ T cells, while CD4+ T cells are poorly costimulated by ICAM-1. Eur J Immunol 29;45-53, 1999 https://doi.org/10.1002/(SICI)1521-4141(199901)29:01<45::AID-IMMU45>3.0.CO;2-I

Cited by

  1. 전립선암 세포주인 PC-3에서 cordycepin에 의해 유도된 세포 내 칼슘농도 변화와 미토콘드리아 기능 상실을 통한 세포사멸 유도 vol.21, pp.3, 2011, https://doi.org/10.5352/jls.2011.21.3.451
  2. The ethyl acetate extract of Cordyceps militaris inhibits IgE-mediated allergic responses in mast cells and passive cutaneous anaphylaxis reaction in mice vol.135, pp.2, 2009, https://doi.org/10.1016/j.jep.2011.03.030
  3. Cordycepin-induced apoptosis and autophagy in breast cancer cells are independent of the estrogen receptor vol.257, pp.2, 2009, https://doi.org/10.1016/j.taap.2011.08.030
  4. Medicinal mushrooms in prevention and control of diabetes mellitus vol.56, pp.1, 2009, https://doi.org/10.1007/s13225-012-0187-4
  5. Triacetyl-3-hydroxyphenyladenosine, a derivative of cordycepin, attenuates atherosclerosis in apolipoprotein E-knockout mice vol.237, pp.11, 2009, https://doi.org/10.1258/ebm.2012.011401
  6. Effect of Egg White Combined with Chalcanthite on Lipopolysaccharide induced Inflammatory Cytokine Expression in RAW 264.7 cells vol.15, pp.1, 2009, https://doi.org/10.3831/kpi.2012.15.1.007
  7. Cordycepin: pharmacological properties and their relevant mechanisms vol.2, pp.2, 2009, https://doi.org/10.5667/tang.2012.0016
  8. Prevention of disuse osteoporosis in rats by Cordyceps sinensis extract vol.23, pp.9, 2009, https://doi.org/10.1007/s00198-011-1842-4
  9. Bioactive metabolites from macrofungi: ethnopharmacology, biological activities and chemistry vol.62, pp.1, 2009, https://doi.org/10.1007/s13225-013-0265-2
  10. Cordycepin에 의한 LNCap 인체 전립선 암세포의 apoptosis 및 G2/M arrest 유발 vol.24, pp.1, 2009, https://doi.org/10.5352/jls.2014.24.1.92
  11. Cordycepin prevented IL-β-induced expression of inflammatory mediators in human osteoarthritis chondrocytes vol.38, pp.7, 2009, https://doi.org/10.1007/s00264-013-2219-4
  12. Optimization of fermentation process of Cordyceps militaris and antitumor activities of polysaccharides in vitro vol.22, pp.4, 2014, https://doi.org/10.1016/j.jfda.2014.01.028
  13. Optimization of Ultrasonic-Assisted Extraction of Cordycepin from Cordyceps militaris Using Orthogonal Experimental Design vol.19, pp.12, 2009, https://doi.org/10.3390/molecules191220808
  14. Hypoglycemic Activity through a Novel Combination of Fruiting Body and Mycelia of Cordyceps militaris in High-Fat Diet-Induced Type 2 Diabetes Mellitus Mice vol.2015, pp.None, 2009, https://doi.org/10.1155/2015/723190
  15. Cordycepin Decreases Compound Action Potential Conduction of Frog Sciatic Nerve In Vitro Involving Ca 2+ -Dependent Mechanisms vol.2015, pp.None, 2015, https://doi.org/10.1155/2015/927817
  16. Effects of Cordycepin on the Microglia-Overactivation-Induced Impairments of Growth and Development of Hippocampal Cultured Neurons vol.10, pp.5, 2009, https://doi.org/10.1371/journal.pone.0125902
  17. N6-(2-Hydroxyethyl)adenosine in the Medicinal Mushroom Cordyceps cicadae Attenuates Lipopolysaccharide-Stimulated Pro-inflammatory Responses by Suppressing TLR4-Mediated NF-κB Signali vol.78, pp.10, 2009, https://doi.org/10.1021/acs.jnatprod.5b00573
  18. Dietary Phytochemicals: Natural Swords Combating Inflammation and Oxidation-Mediated Degenerative Diseases vol.2016, pp.None, 2009, https://doi.org/10.1155/2016/5137431
  19. Cordycepin Prevents Bone Loss through Inhibiting Osteoclastogenesis by Scavenging ROS Generation vol.8, pp.4, 2016, https://doi.org/10.3390/nu8040231
  20. Cordycepin induces apoptosis in human liver cancer HepG2 cells through extrinsic and intrinsic signaling pathways vol.12, pp.2, 2016, https://doi.org/10.3892/ol.2016.4706
  21. Modulation Effects of Cordycepin on Voltage-Gated Sodium Channels in Rat Hippocampal CA1 Pyramidal Neurons in the Presence/Absence of Oxygen vol.2017, pp.None, 2009, https://doi.org/10.1155/2017/2459053
  22. Development of Cordycepin Formulations for Preclinical and Clinical Studies vol.18, pp.8, 2009, https://doi.org/10.1208/s12249-017-0795-0
  23. Immune-enhancing activity of C. militaris fermented with Pediococcus pentosaceus (GRC-ON89A) in CY-induced immunosuppressed model vol.18, pp.None, 2018, https://doi.org/10.1186/s12906-018-2133-9
  24. Enrichment of cordycepin for cosmeceutical applications: culture systems and strategies vol.103, pp.4, 2009, https://doi.org/10.1007/s00253-019-09623-3
  25. The protective effect of Cordycepin on diabetic nephropathy through autophagy induction in vivo and in vitro vol.51, pp.10, 2019, https://doi.org/10.1007/s11255-019-02241-y
  26. Ubiquitin fold modifier 1 activates NF-κB pathway by down-regulating LZAP expression in the macrophage of diabetic mouse model vol.40, pp.1, 2009, https://doi.org/10.1042/bsr20191672
  27. Cordycepin exhibits a suppressive effect on T cells through inhibiting TCR signaling cascade in CFA-induced inflammation mice model vol.42, pp.2, 2009, https://doi.org/10.1080/08923973.2020.1728310
  28. The novel application of cordycepin in maintaining stem cell pluripotency and increasing iPS cell generation efficiency vol.10, pp.None, 2009, https://doi.org/10.1038/s41598-020-59154-5
  29. Anti‐inflammatory effects of cordycepin: A review vol.35, pp.3, 2021, https://doi.org/10.1002/ptr.6890
  30. A Systematic Review of the Biological Effects of Cordycepin vol.26, pp.19, 2009, https://doi.org/10.3390/molecules26195886