#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Použití rostlinných extraktů jako efektivní alternativní terapie zánětů cest dýchacích


Autoři: Karel Šmejkal;  Veronika Rjašková
Působiště autorů: University of Veterinary and Pharmaceutical Sciences Brno, Faculty of Pharmacy, Department of Natural Drugs
Vyšlo v časopise: Čes. slov. Farm., 2016; 65, 139-160
Kategorie: Přehledy a odborná sdělení

Souhrn

Léčivé rostliny jsou s výhodou používány v terapii onemocnění cest dýchacích. Katary horních cest dýchacích patří mezi nemoci spojené se sezónním snížením imunity, proto je často používáno rostlinných drog s nespecifickým imunomodulačním účinkem. Mezi takové rostliny patří např. echinacea (Echinacea purpurea) a americký ženšen (Panax quinquefolius). Ve spojení s antibakteriálně a antisepticky účinnými léčivými rostlinami, jako jsou tymián (Thymus vulgaris) a pelargonium (Pelargonium sidoides) mohou představovat účinnou pomoc při onemocněních cest dýchacích, zkracovat trvání nemoci a snižovat nutnost antibiotické terapie. Prezentovaný text shrnuje základní informace o těchto rostlinách, jejich obsahových látkách, mechanismech účinku a klinických zkouškách potvrzujících efekt a monitorujících případné nežádoucí účinky.

Klíčová slova:
Echinacea purpurea • Panax quinquefolius • Pelargonium sidoides • Thymus vulgaris • katary horních cest dýchacích • imunita


Zdroje

1. Kopřiva F. Diagnostika a léčba bronchitídy u dětí. Pediatr. pro Praxi 2007; 2, 106–108.

2. Pauk N. Terapie akutní bronchitídy. Interní Med. 2011; 13, 327–328.

3. Tan B. K., Vanitha J. Immunomodulatory and antimicrobial effects of some traditional Chinese medicinal herbs: a review. Curr. Med. Chem. 2004; 11, 1423–30.

4. Das S., Bordoloi R., Newar N. A review on immune modulatory effect of some traditional medicinal herbs. J. Pharm. Chem. Biol. Sci. 2014; 2, 33–42.

5. Cundell D. R. Herbal Phytochemicals as Immunomodulators. Curr. Immunol. Rev. 2014; 10, 64–81.

6. Shukla S., Bajpai V. K., Kim M. Plants as potential sources of natural immunomodulators. Rev. Environ. Sci. Bio. 2014; 13, 17–33.

7. Manayi A., Vazirian M., Saeidnia S. Echinacea purpurea: Pharmacology, phytochemistry and analysis methods. Pharmacogn. Rev. 2015; 9, 63–72.

8. Gupta M., Sharma D., Sharma A., Kumari V., Goshain O. P. A review on purple cone flower (Echinacea purpurea L. Moench. J. Pharm. Res. 2012; 8, 4076–4081.

9. Community herbal monograph on Echinacea purpurea (L. Moench, radix) EMA/HMPC/577784/2008. http://www. ema.europa.eu/docs/en_GB/document_library/Herbal_-_Community_herbal_monograph/2011/01/WC500101497.pdf

10. Community herbal monograph on Echinacea angustifolia DC., radix. EMA/HMPC/688216/2008. http://www.ema.europa.eu/ docs/en_GB/document_library/Herbal_-_Community_herbal_monograph/2012/05/WC500127890.pdf

11. Community herbal monograph on Echinacea pallida (Nutt. Nutt., radix) EMEA/HMPC/332350/2008. http://www. ema.europa.eu/docs/en_GB/document_library/Herbal_-_Community_herbal_monograph/2009/12/WC500018248.pdf

12. European Union herbal monograph on Echinacea purpurea (L. Moench, herba recens) EMA/HMPC/48704/2014. http://www.ema.europa.eu/docs/en_GB/document_library/Herbal_-_Community_herbal_monograph/2015/04/WC500185437.pdf

13. Gajalakshmi S., Vijayalakshmi S., Devirajeswari V. Echinaceae puprpurea – a potent immunostimulant. Int. J. Pharm. Sci. Rev. Res. 2012; 14, 47–52.

14. Barnes J., Anderson L. A., Gibbons S., Phillipson J. D. Echinacea species (Echinacea angustifolia (DC. Hell.) Echinacea pallida (Nutt.) Echinacea purpurea (L.) Moench. a review of their chemistry, pharmacology and clinical properties. J. Pharm. Pharmacol. 2005; 57, 929–954.

15. Woelkart K., Dittrich P., Beubler E., Pinl F., Schoop R., Suter A., Bauer R. Pharmacokinetics of the main alkamides after administration of three different Echinacea purpurea preparations in humans. Planta Med. 2008; 74, 651–656.

16. Shekarchi M., Hajimehdipoor H., Khanavi M., Roostaie A. The effects of plant age and harvesting time on chicoric and caftaric acids content of E. purpurea (L. Moench). Iranian J. Pharm. Sci. 2012; 8, 203–208.

17. Thomsen M. O., Frette X. C., Christensen K, B., Christensen L. P., Grevsen K. Seasonal Variations in the Concentrations of Lipophilic Compounds and Phenolic Acids in the Roots of Echinacea purpurea and Echinacea pallida. J. Agric. Food Chem. 2012; 60, 12131–12141.

18. Barrett B. Medicinal properties of Echinacea: a critical review. Phytomedicine 2003; 10, 66–86.

19. Rininger J. A., Kickner S., Chigurupati P., McLean A., Franck Z. Immunopharmacological activity of Echinacea preparations following simulated digestion on murine macrophages and human peripheral blood mononuclear cells. J. Leukocyte Biol. 2000; 68, 503–510.

20. Eichler F., Krüger G. R. F. Effects of non-specific immunostimulants (Echinacin, isoprinosine, and thymus factors. on the infection and antigen expression in herpesvirus–6 exposed human lymphoid cells. In Vivo 1994; 8, 565–576.

21. Bauer R., Remiger P., Jurcic K., Wagner H. Beeinflussung der Phagozytose-Aktivität durch Echinacea-Extrakte. Influence of Echinacea extract on phagocytotic activity. Z. Phytother. 1989; 10, 43–48.

22. Bauer R., Wagner H. Echinacea species as potential immunostimulatory drugs. In: Wagner H., Farnsworth N. R. (eds.) Economic and Medicinal Plant Research. New York: Academic Press Limited 1991; 253–318.

23. Bauer R. Chemistry, analysis and immunological investigations of Echinacea phytopharmaceuticals. In: Wagner H. (ed.) Immunomodulatory Agents from Plants. Basel, Boston, Berlin: Birkhauser Verlag 1999; 41–88.

24. Woelkart, K., Bauer, R. The role of alkamides as an active principle of echinacea. Planta Med. 2007; 73, 615–623.

25. Woelkart K., Xu W., Pei Y., Makriyannis A., Picone R. P., Bauer R. The endocannabinoid system as a target for alkamides from Echinacea angustifolia roots. Planta Med. 2005; 71, 701–705.

26. Karsch-Völk M., Barrett B., Kiefer D., Bauer R., Ardjomand-Woelkart K., Linde K. Echinacea for preventing and treating the common cold. Cochrane Database Syst. Rev. 2014; 2, CD000530.

27. Schapowal A., Klein P., Johnston S. L. Echinacea reduces the risk of recurrent respiratory tract infections and complications: a meta-analysis of randomized controlled trials. Adv. Ther. 2015; 32, 187–200.

28. Baetgen D. Erfolge in der Keuchhusten-Behandlung mit Echinacin®. Successful treatment of whooping cough with Echinacea. Therapiewoche 1984; 34, 5115–5119.

29. Baetgen D. Behandlung der akuten Bronchitis im Kindesalter. Praxisstudie mit einem Immunstimulans aus Echinacea purpurea. Treatment of acute bronchitis in children. A study of an immunostimulatory agent from Echinacea purpurea in a primary care setting. Translator unknown. Therapiewoche Pädiatrie 1988; 1, 65–70.

30. Melchart D., Linde K., Worku F., Bauer R., Wagner H. Immunomodulation with echinacea – a systematic review of controlled clinical trials. Phytomedicine 1994; 1, 245–254.

31. Turner R. B., Riker D. K., Gangemi J. D. Ineffectiveness of Echinacea for prevention of experimental rhinovirus colds. Antimicrob. Agents Chemother. 2000; 44, 1708–1709.

32. Melchart D., Walther E., Linde K., Brandmaier R., Lersch C. Echinacea root extracts for the prevention of upper respiratory tract infections: A double-blind, placebo-controlled randomized trial. Arch. Family Med. 1998; 7, 541–545.

33. Schöneberger D. Einfluß der immunstimulierenden Wirkung von Preßsaft aus Echinacea purpurea auf Verlauf und Schweregrad von Erkältungskrankheiten. The influence of the immunostimulating effects of pressed juice from Echinacea purpurea on the course and severity of cold infections. Forum Immunologie 1992; 8, 18–22.

34. Grimm W., Muller H. H. A randomized controlled trial of the effect of fluid extract of Echinacea purpurea on incidence and severity of colds and respiratory infections. Am. J. Med. 1999; 106, 138–143.

35. Bräunig B., Dorn M., Knick E. Echinaceae purpureae radix: Zur Stärkung der körpereigenen Abwehr bei grippalen Infekten. Strengthening of the endogenous resistence to influenzal infections. Z. Phytother. 1992; 13, 7–13.

36. Melchart D., Linde K. Clinical investigations of Echinacea phytopharmaceuticals. In: Wagner H. (ed.) Immunomodulatory Agents from Plants. Basel, Boston, Berlin: Birkhauser Verlag 1999; 105–118.

37. Brinkeborn R. M., Shah D. V., Degenring F. H. Echinaforce® and other Echinacea fresh plant preparations in the treatment of the common cold. Phytomedicine 1999; 6, 1–6.

38. Schmidt U., Albrecht M., Schenk N. Pflanzliches Immunstimulans senkt Haufigkeit grippaler Infekte. Natur- und Ganzheitsmedizin 1990; 3, 277–281.

39. Cohen H. A., Varsano I., Kahan E., Sarrell E. M., Uziel Y. Effectiveness of an herbal preparation containing echinacea, propolis, and vitamin C in preventing respiratory tract infections in children: a randomized, double-blind, placebo-controlled, multicenter study. Arch. Pediatr. Adolesc. Med. 2004; 158, 217–221.

40. Taylor J. A., Weber W., Standish L., et al. Efficacy and safety of echinacea in treating upper respiratory tract infections in children: a randomized controlled trial. JAMA 2003; 290, 2824–2830.

41. Jawad M., Schoop R., Suter A., Klein P., Eccles R. Safety and efficacy profile of Echinacea purpurea to prevent common cold episodes: a randomized, doubleblind, placebo-controlled trial. Evid Based Complement. Alternat. Med. 2012; 2012, 841315.

42. Gallo M., Sarkar M., Au W., Pietrzak K., Comas B., Smith M, Jaeger T. V., Einarson A., Koren G. Pregnancy outcome following gestational exposure to Echinacea. Arch. Intern. Med. 2000; 160, 3141–3143.

43. Qi L.-W., Wang C.-Z., Yuan C.-S. Ginsenosides from American ginseng: chemical and pharmacological diversity. Phytochemistry 2011; 72, 689–699.

44. Yuan C.-S., Wang C.-Z., Wicks S. M., Qi L.-W. Chemical and pharmacological studies of saponins with a focus on American ginseng. J. Ginseng Res. 2010; 34, 160–167.

45. Do J. H., Lee H. O., Lee S. K., Noh K. B., Lee S. D., Lee K. S. Comparisons of acidic polysaccharide content in various ginseng species and parts. J. Ginseng Res. 1993; 17, 145–147.

46. Liao J.-F., Shen Y.-C., Huang Y.-T. Pharmacology of Polysaccharides from Ginseng Species. Int. J. Biomed. Pharm. Sci. 2012; 6, 63–69.

47. Han B. H., Park M. H., Han Y. N., Woo L. K., Sankawa U., Yahara S., Tanaka O. Degradation of ginseng saponins under mild acidic conditions. Planta Med. 1982; 44: 146–149.

48. Karikura M., Miyase T., Tanizawa H., Taniyama T., Takino Y. Studies on absorption, distribution, excretion and metabolism of ginseng saponins. VI. The decomposition products of ginsenoside Rb2 in the stomach of rats. Chem. Pharm. Bull. 1991; 39, 400–404.

49. Karikura M., Miyase T., Tanizawa H., Taniyama T., Takino Y. Studies on absorption, distribution, excretion and metabolism of ginseng saponins. VII. Comparison of the decomposition modes of ginsenoside-Rb1 and -Rb2 in the digestive tract of rats. Chem. Pharm. Bull. 1991; 39, 2357–2361.

50. Hasegawa H., Sung J. H., Matsumiya S., Uchiyama M. Main ginseng saponin metabolites formed by intestinal bacteria. Planta Med. 1996; 62, 453–457.

51. Wakabayashi C., Murakami K., Hasegawa H., Murata J., Saiki I. An intestinal bacterial metabolite of ginseng protopanaxadiol saponins has the ability to induce apoptosis in tumor cells. Biochem. Biophys. Res. Commun. 1998; 246, 725–730.

52. Suda K., Murakami K., Murata J., Hasegawa H., Saiki I. Induction of apoptosis in Lewis lung carcinoma cells by an intestinal bacterial metabolite resulted from orally administered ginseng protopanaxadiol saponins. J. Trad. Med. 2000; 17, 236–244.

53. Hasegawa H., Suzuki R., Nagaoka T., Tezuka Y., Kadota S., Saiki I. Prevention of growth and metastasis of murine melanoma through enhanced natural killer cytotoxicity by fatty acid conjugate of protopanxatriol. Biol. Pharm. Bull. 2002; 25, 861–866.

54. Hasegawa H., Lee K. S., Nagaoka T., Tezuka Y., Uchiyama M., Kadota S., Saiki I. Pharmacokinetics of ginsenoside deglycosylated by intestinal bacteria and its transformation to biologically active fatty acid ester. Biol. Pharm. Bull. 2000; 23, 298–304.

55. Sievenpiper J. L., Arnason J. T., Leiter L. A., Vuksan V. Decreasing, null and increasing effects of eight popular types of ginseng on acute postprandial glycemic indices in healthy humans: the role of ginsenosides. J. Am. Coll. Nutr. 2004; 23, 248–258.

56. Dascalu A., Sievenpiper J. L., Jenkins A. L., Stavro M. P., Leiter L. A., Arnason T., Vuksan V. Five batches representative of Ontario–grown American ginseng root produce comparable reductions of postprandial glycemia in healthy individuals. Can. J. Physiol. Pharmacol. 2007; 85, 856–864.

57. Kang S., Min H. Ginseng, the ‘Immunity Boost’: The Effects of Panax ginseng on Immune System. J. Ginseng Res. 2012; 36, 354–368.

58. Rivera E., Ekholm Pettersson F., Inganäs M., Paulie S., Grönvik K. O. The Rb1 fraction of ginseng elicits a balanced Th1 and Th2 immune response. Vaccine 2005; 23, 5411–5419.

59. Yang Z., Chen A., Sun H., Ye Y., Fang W. Ginsenoside Rd elicits Th1 and Th2 immune responses to ovalbumin in mice. Vaccine 2007; 25, 161–169.

60. Sun J., Song X., Hu S. Ginsenoside Rg1 and aluminum hydroxide synergistically promote immune responses to ovalbumin in BALB/c mice. Clin. Vaccine Immunol. 2008; 15, 303–307.

61. Song X., Chen J., Sakwiwatkul K., Li R., Hu S. Enhancement of immune responses to influenza vaccine (H3N2). by ginsenoside Re. Int. Immunopharmacol. 2010; 10, 351–356.

62. Song X., Hu S. Adjuvant activities of saponins from traditional Chinese medicinal herbs. Vaccine 2009; 27, 4883–4890.

63. Lee E. J., Ko E., Lee J., Rho S., Ko S., Shin M. K., Min B. I., Hong M. C., Kim S., Y., Bae H. Ginsenoside Rg1 enhances CD4+ T-cell activities and modulates Th1/Th2 differentiation. Int. Immunopharmacol. 2004; 4, 235–244.

64. Lee J. H., Han Y. Ginsenoside Rg1 helps mice resist to disseminated candidiasis by Th1 type differentiation of CD4+ T cell. Int. Immunopharmacol. 2006; 6, 1424–1430.

65. Kenarova B., Neychev H., Hadjiivanova C., Petkov V. D. Immunomodulating activity of ginsenoside Rg1 from Panax ginseng. Jpn. J. Pharmacol. 1990; 54, 447–454.

66. Song X., Zang L., Hu S. Amplified immune response by ginsenoside–based nanoparticles (ginsomes.. Vaccine 2009; 27, 2506–2511.

67. Tung N. H., Quang T. H., Son J. H., Koo J. E., Hong H. J., Koh Y. S., Song G. Y., Kim Y. H. Inhibitory effect of ginsenosides from steamed ginseng-leaves and flowers on the LPS-stimulated IL-12 production in bone marrow-derived dendritic cells. Arch. Pharm. Res. 2011; 34, 681–685.

68. Luo Y. M., Cheng X. J., Yuan W. X. Effects of ginseng root saponins and ginsenoside Rb1 on immunity in cold water swim stress mice and rats. Zhongguo Yao Li Xue Bao 1993; 14, 401–404.

69. Su W., Sun A. J., Xu D. L., Zhang H. Q., Yang L., Yuan L. Y., Jia J. G., Zou Y. Z., Wu Y. L., Wang K. Q., Ge J. B. Inhibiting effects of total saponins of panax ginseng on immune maturation of dendritic cells induced by oxidized-low density lipoprotein. Cell Immunol. 2010; 263, 99–104.

70. Joh E. H., Lee I. A., Jung I. H., Kim D. H. Ginsenoside Rb1 and its metabolite compound K inhibit IRAK-1 activation-the key step of inflammation. Biochem. Pharmacol. 2011; 82, 278–286.

71. Byeon S. E., Lee J., Kim J. H., Yang W. S., Kwak Y. S., Kim S. Y., Choung E. S., Rhee M. H., Cho J. Y. Molecular mechanism of macrophage activation by red ginseng acidic polysaccharide from Korean red ginseng. Mediat. Inflamm. 2012; 2012, ID 732860.

72. Tomoda M., Hirabayashi K., Shimizu N., Gonda, R., Ohara, N. The core structure of ginsenan PA, a phagocytosis-activating polysaccharide from the root of Panax ginseng. Biol. Pharm. Bull. 1994; 17, 1287–1291.

73. Kim K. H., Lee Y. S., Jung I. S., Park S. Y., Chung H. Y., Lee I. R., Yun Y. S. Acidic polysaccharide from Panax ginseng, Ginsan, induces Th1 cell and macrophage cytokines and generates LAK cells in synergy with IL-2. Planta Med. 1998; 64, 110–115.

74. Sonoda Y., Kasahara T., Mukaida N., Shimizu N., Tomoda M., Takeda T. Stimulation of interleukin-8 production by acidic polysaccharides from the root of Panax ginseng. Immunopharmacology 1998; 38, 287–294.

75. Assinewe V., Arnason J., Aubry A., Mullin J., Lemaire I. Extractable polysaccharides of Panax quinquefolius L. North American ginseng. root stimulate TNFα production by alveolar macrophages. Phytomedicine 2002; 9, 398–404.

76. Jang H. I., Shin H. M. Wild Panax ginseng. Panax ginseng C. A. Meyer. protects against methotrexate-induced cell regression by enhancing the immune response in RAW 264.7 macrophages. Am. J. Chin. Med. 2010; 38, 949–960.

77. Azike C. G., Charpentier P. A., Hou J., Pei H., King Lui E. M. The Yin and Yang actions of North American ginseng root in modulating the immune function of macrophages. Chin. Med. 2011; 6, 21.

78. Ahn J. Y., Choi I. S., Shim J. Y., Yun E. K., Yun Y. S., Jeong G., Song J. Y. The immunomodulator ginsan induces resistance to experimental sepsis by inhibiting Toll–like receptor–mediated inflammatory signals. Eur. J. Immunol. 2006; 36, 37–45.

79. Kim D. Y., Yang W. M. Panax ginseng ameliorates airway inflammation in an ovalbumin-sensitized mouse allergic asthma model. J. Ethnopharmacol. 2011; 136, 230–235.

80. Ro J. Y., Ahn Y. S., Kim K. H. Inhibitory effect of ginsenoside on the mediator release in the guinea pig lung mast cells activated by specific antigen-antibody reactions. Int. J. Immunopharmacol. 1998; 20, 625–41.

81. Biondo P. D., Goruk S., Ruth M. R., O’Connell E., Field C. J. Effect of CVT–E002 (COLD-fX. versus a ginsenoside extract on systemic and gut–associated immune function. Int. Immunopharmacol. 2008; 8, 1134–1142.

82. Lee J. H., Shim J. S., Chung M. S., Lim S. T., Kim K. H. Inhibition of pathogen adhesion to host cells by polysaccharides from Panax ginseng. Biosci. Biotechnol. Biochem. 2009; 73, 209–212.

83. Lee J. H., Shim J. S., Lee J. S, Kim M. K., Chung M. S., Kim K. H. Pectin-like acidic polysaccharide from Panax ginseng with selective antiadhesive activity against pathogenic bacteria. Carbohydr. Res. 2006; 341, 1154–1163.

84. Fukuyama N., Shibuya M., Orihara Y. Antimicrobial polyacetylenes from Panax ginseng hairy root culture. Chem. Pharm. Bull. 2012; 60, 377–380.

85. Chan L. Y., Kwok H. H., Chan R. W., Peiris M. J., Mak N. K., Wong R. N., Chan M. C., Yue P. Y. Dual functions of ginsenosides in protecting human endothelial cells against influenza H9N2-induced inflammation and apoptosis. J. Ethnopharmacol. 2011; 137, 1542–1546.

86. Kim J. Y., Kim H. J., Kim H. J. Effect of oral administration of Korean red ginseng on infl uenza A (H1N1. virus infection. J. Ginseng. Res. 2011; 35, 104–110.

87. Quan F. S., Compans R. W., Cho Y. K., Kang S. M. Ginseng and Salviae herbs play a role as immune activators and modulate immune responses during influenza virus infection. Vaccine 2007; 25, 272–282.

88. Kim J., Han B. J., Kim H., Lee J. Y., Joo I., Omer S., Kim Y. S., Han Y. Th1 immunity induction by ginsenoside Re involves in protection of mice against disseminated candidiasis due to Candida albicans. Int. Immunopharmacol. 2012; 14, 481–486.

89. Song X., Zang L., Hu S. Amplified immune response by ginsenoside-based nanoparticles ginsomes. Vaccine 2009; 27, 2306–2311.

90. Song X., Chen J., Sakwiwatkul K., Li R., Hu S. Enhancement of immune responses to influenza vaccine (H3N2. by ginsenoside Re. Int. Immunopharmacol. 2010; 10, 351–356.

91. Scaglione F., Cattaneo G., Alessandria M., Cogo R. Efficacy and safety of the standardised Ginseng extract G115 for potentiating vaccination against the influenza syndrome and protection against the common cold. Drugs Exp. Clin. Res. 1996; 22, 65–72.

92. Sun K., Wang C. S., Guo J., Horie Y., Fang S. P., Wang F., et al. Protective effects of ginsenoside Rb1, ginsenoside Rg1, and notoginsenoside R1 on lipopolysaccharide-induced microcirculatory disturbance in rat mesentery. Life Sci. 2007; 81, 509–518.

93. Park E. K., Shin Y. W., Lee H. U., Kim S. S., Lee Y. C., Lee B. Y., et al. Inhibitory effect of ginsenoside Rb1 and compound K on NO and prostaglandin E2 biosynthesis of RAW 264.7 cells induced by lipopolysaccharide. Biol. Pharm. Bull. 2005; 28, 652–656.

94. Smolinsk A. T., Pestka J. J. Modulation of lipopolysaccharide-induced proinflammatory cytokine production in vitro and in vivo by the herbal constituents apigenin, chamomile, ginsenoside Rb 1. ginseng, and parthenolide feverfew. Food Chem. Toxicol. 2003; 41, 1381–1390.

95. Senchina D. S., Hallam J. E., Cheney D. J. Multidisciplinary perspectives on mechanisms of activity of popular immune-enhancing herbal supplements used by athletes. Front. Biol. 2013; 8, 78–100.

96. Engels H. J., Fahlman M. M., Wirth J. C. Effects of ginseng on secretory IgA, performance, and recovery from interval exercise. Med. Sci. Sports Exerc. 2003; 35, 690–696.

97. Gaffney B. T., Hügel H. M., Rich P. A. The effects of Eleutherococcus senticosus and Panax ginseng on steroidal hormone indices of stress and lymphocyte subset numbers in endurance athletes. Life Sci. 2001; 70, 431–442.

98. Hsu M. Effect of American ginseng supplementation on cytokines and oxidative stress following an acute downhill running. Med. Sci. Sports Exerc. 2010; 42, S788.

99. Jung H., Kang H., Chang Y., Kim T. Effect of Panax ginseng supplementation on muscle damage and inflammation after treadmill running in humans. Med. Sci. Sports Exerc. 2011; 43(Suppl 1), S432.

100. Park S., Jung H., Hong S., Kang Y., Lee G., Kim S., Kang H., Lee C. Effects of red ginseng intake on interleukin-6 (IL-6) and cortisol responses after high-intensity exercise. Med. Sci. Sports Exerc. 2008; 40(Suppl), S432.

101. Biondo P., McCargar L., Harber V., Field C. A randomized controlled trial of ginseng supplementation on the immune response to a moderate exercise stress protocol in non-athletic women. Open Nutr. J. 2010; 4, 1–10.

102. Biondo P. D., Robbins S. J., Walsh J. D., McCargar L. J., Harber V. J., Field C. J. A randomized controlled crossover trial of the effect of ginseng consumption on the immune response to moderate exercise in healthy sedentary men. Appl. Physiol. Nutr. Metab. 2008; 33, 966–975.

103. Lau W., Liang M., Sokmen B., Spalding T., Quezada L., Chuang W. Effect of Panax notoginseng (Chinese ginseng) and cycling exercise on IL-6 and cortisol in untrained non-diabetic men. Med. Sci. Sports Exerc. 2011; 43(Suppl 1), S432.

104. Scaglione F., Ferrara F., Dugnani S., Falchi M., Santoro G., Fraschini F. Immunomodulatory effects of two extracts of Panax ginseng C.A. Meyer. Drugs Exp. Clin. Res. 1990; 16, 537–542.

105. Scaglione, F., Weiser, K., Alessandria, M. Effects of the Standardised Ginseng Extract G115® in Patients with Chronic Bronchitis A Nonblinded, Randomised, Comparative Pilot Study. Clin. Drug Invest. 2001; 21, 41–45.

106. McElhaney J. E., Gravenstein S., Cole S. K., Davidson E., O’Neill D., Petitjean S., et al. A placebo-controlled trial of a proprietary extract of North American ginseng (CVT-E002) to prevent acute respiratory illness in institutionalized older adults. J. Am. Geriatr. Soc. 2004; 52, 13–19.

107. McElhaney J. E., Goel V., Toane B., Hooten J., Shan J. J. Efficacy of COLD-fX in the prevention of respiratory symptoms in community-dwelling adults: a randomized, double-blinded, placebo controlled trial. J. Altern. Complement. Med 2006; 12, 153–157.

108. Predy G. N., Goel V., Lovlin R., Donner A., Stitt L., Basu T. K. Efficacy of an extract of North American ginseng containing poly-furanosyl-pyranosyl-saccharides for preventing upper respiratory tract infections: a randomized controlled trial. Canadian Med. Assoc. J. 2005; 173, 1043–1048.

109. Vohra S., Johnston B. C., Laycock K. L., Midodzi W. K., Dhunnoo I., Harris E., Baydala L. Safety and tolerability of North American ginseng extract in the treatment of pediatric upper respiratory tract infection: a phase II randomized, controlled trial of 2 dosing schedules. Pediatrics 2008; 122, e402–410.

110. Seida J. K., Durec T., Kuhle S. North American (Panax quinquefolius) and Asian ginseng (Panax ginseng) preparations for prevention of the common cold inhealthy adults: a systematic review. Evid. Based Complement. Alternat. Med. 2011; 2011, 282151.

111. Moyo M., van Staden J. Medicinal properties and conservation of Pelargonium sidoides DC. J. Ethnopharmacol. 2014; 152, 243–255.

112. Navrátilová Z. Léčivý muškát Pelargonium sidoides. Prakt. Lék. 2012; 8, 290–292.

113. Kolodziej H. Pelargonium reniforme and Pelargonium sidoides: their botany, chemistry and medicinal use. In: Lis Balchin M (ed.) Geranium and Pelargonium. In Medicinal and Aromatic Plants – Industrial Profiles. New York, London: Taylor & Francis 2002; 262–290

114. Colling J., Groenewald J.-H., Makunga N. P. Genetic alteration for increased coumarin production lead to metabolic changes in the medicinally important Pelargonium sidoides DC (Geraniaceae). Metab. Eng. 2010; 12, 561–572.

115. Brendler T., van Wyk B.-E. A historical, scientific and commercial perspective on the medicinal use of Pelargonium sidoides (Geraniaceae). J. Ethnopharmacol. 2008; 119, 420–433.

116. Lewu F. B., Adebola P. O., Afolayan A. J. Commercial harvesting of Pelargonium sidoides in the Eastern Cape, South Africa: striking a balance between resources conservation and livelihoods. J. Arid. Environ. 2007; 70, 380–388.

117. Hutchings A., Scott A. H., Lewis G., Cunningham A. Zulu Medicinal Plants. Pietermaritzburg: Natal University Press 1996.

118. Community herbal monograph on Pelargonium sidoides DC and/or Pelargonium reniforme Curt., radix. EMA/HMPC/ 560961/2010. http://www.ema.europa.eu/docs/en_GB/document _library/Herbal_-_Community_herbal_monograph/2013/02/WC 500138439.pdf

119. Kolodziej H. Fascinating metabolic pools of Pelargonium sidoides and Pelargonium reniforme, traditional and phytomedicinal sources of the herbal medicine Umckaloabo®. Phytomedicine 2007; 14, 9–17.

120. Theisen L. L., Muller C. P. EPs®7630 (Umckaloabo®), an extract from Pelargonium sidoides roots, exerts anti-influenza virus activity in vitro and in vivo. Antiviral Res. 2012; 94, 147–156.

121. Michaelis M., Doerr H. W., Cinatl J. Jr. Investigation of the influence of EPs® 7630, a herbal drug preparation from Pelargonium sidoides, on replication of a broad panel of respiratory viruses. Phytomedicine 2011; 18, 384–386.

122. Mativandlela S. P. N., Meyer J. J. M., Hussein A. A., Lall N. Antitubercular activity of compounds isolated from Pelargonium sidoides. Pharm. Biol. 2007; 45, 645–650.

123. Beil W., Kilian P. EPs®7630, an extract from Pelargonium sidoides roots inhibits adherence of Helicobacter pylori to gastric epithelial cells. Phytomedicine 2007; 14, 5–8.

124. Conrad A., Jung I., Tioua D., Lalleman C., Carrapatoso F., Engels I., Daschner F. D., Frank U. Extract of Pelargonium sidoides (EPs®7630) inhibits the interactions of group A-streptococci and host epithelia in vitro. Phytomedicine 2007; 14, 52–59.

125. Kayser O., Kolodziej H., Kinderlen A. F. Immunomodulatory principles of Pelargonium sidoides. Phytother. Res. 2001; 15, 122–126.

126. Thäle C., Kiderlen A. F., Kolodziej H. Anti-infective mode of action of EPs®7630 at the molecular level. Planta Med. 2008; 74, 675–681.

127. Kolodziej H., Kinderlen A. F. In vitro evaluation of antibacterial and immunomodulatory acitvities of Pelargonium reinforme, Pelargonium sidoides and the related herbal drug preparation EPs®7630. Phytomedicine 2007; 14, 18–26.

128. Kolodziej H., Burmeister A., Trun W., Radtke O. A., Kinderlen A. F., Ito H., Hatano T., Yoshida T., Foo L. L. Tannins and related compounds induce nitric oxide synthase and cytokines genes expression in Leishmania major – infected macrophage-like RAW 264.7 cells. Bioorg. Med. Chem. 2005; 13, 6470–6476.

129. Radtke O. A., Kinderlen A. F., Kayser O., Kolodziej H. Gene expression profiles of inducible nitric oxide synthase and cytokines in Leishmania major – infected macrophage-like RAW 264.7 cells treated with gallic acid. Planta Med. 2004; 70, 924–928.

130. Kamin W., Ilyenko L. I., Malek F. A., Kieser M. Treatment of acute bronchitis with EPss 7630: randomized, controlled trial in children and adolescents. Pediatr. Int. 2012; 54, 219–226.

131. Kamin W., Maydannik V. G., Malek F. A., Kieser M. Efficacy and tolerability of EPss 7630 in patients (aged 6–18 years old) with acute bronchitis: a randomized, double-blind, placebo-controlled clinical dose-finding study. Acta Paediatr. 2010; 99, 537–543.

132. Matthys H., Funk P. EPSs 7630 improves acute bronchitic symptoms and shortens time to remission: Results of a randomised, double-blind, placebo-controlled, multicentre trial. Planta Med. 2008; 74, 686–692.

133. Chuchalin A. G., Berman B., Lehmacher W. Treatment of acute bronchitis in adults with a Pelargonium sidoides preparation (EPss 7630) a randomised, double-blind controlled trial. Explore 2005; 1, 437–445.

134. Bachert C., Schapowal A., Funk P., Kieser M. Treatment of acute rhinosinusitis with the preparation from Pelargonium sidoides EPss 7630: a randomized, double-blind, placebo-controlled trial. Rhinology 2009; 47, 51–58.

135. Tahan F., Yaman M. Can the Pelargonium sidoides root extract EPss 7630 prevent asthma attacks during viral infections of the upper respiratory trac tin children? Phytomedicine 2013; 20, 148–150.

136. Bereznoy V. V., Riley D. S., Wasmer G., Heger M. Efficacy of extract of Pelargonium sidoides in children with acute non-group A betaemolytic streptococcus tonsillopharyngitis: a randomized, double-blind, placebo-controlled trial. Altern. Ther. Health Med. 2003, 9, 68–79.

137. Timmer A., Günther J., Rücker G., Motschall E., Antes G., Kern W. V. Pelargonium sidoides extract for acute respiratory tract infections. Cochrane Database Syst. Rev. 2008; 3, CD006323.

138. Leugnerová G. 2008. Thymus vulgaris L. – mateřídouška obecná, tymián/dúška tymiánová. Dostupné na: http://botany.cz/ cs/thymus-vulgaris/, datum citace 25.1.2016

139. Basch E., Ulbricht C., Hammerness P., Bevins A., Sollars D. Thyme (Thymus vulgaris L.). Thymol. J. Herb. Pharmacother. 2004; 4, 49–67.

140. Community herbal monograph on Thymus vulgaris L. and Thymus zygis L., herba. EMA/HMPC/342332/2013. http:// www. ema.europa.eu/docs/en_GB/document_library/Herbal_Community_herbal_monograph/2014/06/WC500167812.pdf

141. Blumenthal M. (ed.) The Complete German Commission E Monographs: Therapeutic Guide to Herbal Medicines. Boston: Integrative Medicine Communications 1998.

142. Czygan F. C., Hiller K. Thymi herba. In: Wichtl M (ed.) Herbal drugs and Phytopharmaceuticals. Medpharm Scientific Publishers 2004; 607–610.

143. Bruneton J. Pharmacognosy, Phytochemistry, Medicinal Plants. 2nd ed. Secaucus, NJ: Lavoisier Publishing 1999.

144. Spiewak R., Skorska C., Dutkiewics J. Occupational airborne contact dermatitis caused by thyme dust. Contact Dermatitis 2001; 44, 235–239.

145. Manou I., Bouillard L., Devleeschouwer M. J., et al. Evaluation of the preservative properties of Thymus vulgaris essentials oil in topically applied formulations under a challenge test. J. Appl. Microbiol. 1998; 84, 368–376.

146. Twetman S., Petersson L. G. Interdental caries incidence and progression in relation to mutant streptococci suppression after chlorhexidine-thymol varnic treatments in schoolchildren. Acta Odontol. Scand. 1999; 57, 144–148.

147. Shapiro S., Guggenheim B. The action of thymol on oral bacteria. Oral Microbiol. Immunol. 1995; 10, 241–246.

148. Bakkali F., Averbeck S., Averbeck D., Idaomar M. Biological effects of essential oils – A review. Food Chem. Toxicol. 2008; 46, 446–475.

149. Di Pasqua R., Betts G., Hoskins N., Edwards M., Ercolini D., Mauriello G. Membrane toxicity of antimicrobial compounds from essential oils. J. Agric. Food Chem. 2007; 55, 4863–4870.

150. Ipek E., Zeytinoglu H., Okay S., Tuylu B. A., Kurcuoglu M., Baser K. H. C. Genotoxicity and antigenotoxicity of Origanum oil and carvacrol evaluated by Ames Salmonella/microsomal test. Food Chem. 2005; 93, 551–556.

151. Franzios G., Mirotsou M., Hatziapostolou E., Kral J., Scouras Z. G., Mavragani-Tsipidou P. Insecticidal and genotoxic activities of mint essential oils. J. Agric. Food Chem. 1997; 45, 2690–2694.

152. Santana-Rios G., Orner G. A., Amantana A., Provost C., Wu S. Y., Dashwood R. H. Potent antimutagenic activity of white tea in comparison with green tea in the Salmonella assay. Mutat. Res. 2001; 495, 61–74.

153. Cal K. Skin penetration of terpenes from essential oils and topical vehicles. Planta Med. 2006; 72, 311–316.

154. Radulovic N. S., Blagojevic P. D., Stojanovic-Radic Z. Z., Stojanovic N. M. Antimicrobial Plant Metabolites: Structural Diversity and Mechanism of Action. Curr. Med. Chem. 2013; 20, 932–952.

155. Friedman M. Chemistry and multibeneficial bioactivities of carvacrol (4-isopropyl-2-methylphenol., a component of essential oils produced by aromatic plants and spices. J. Agric. Food Chem. 2014; 62, 7652–7670.

156. Langeveld W. T., Veldhuizen E. J. A., Burt S. A. Synergy between essential oil components and antibiotics: A review. Cr. Rev. Microbiol. 2014; 40, 76–94.

157. Bassolé I. H. N., Juliani H. R. Essential oils in combination and their antimicrobial properties. Molecules 2012; 17, 3989–4006.

158. Nowotarska S., Nowotarski K., Friedman M. Effect of structure on the interactions between five natural antimicrobial compounds and phospholipids of bacterial cell membrane on model monolayers. Molecules 2014; 19, 7497–7515.

159. Helander I. M., Alakomi H., Latva-Kala K., Mattila-Sandholm T., Pol I., Smid E. J., Gorris L. G. M., von Wright A. Characterization of the action of selected essential oil components on Gram-negative bacteria. J. Agric. Food Chem. 1998; 46, 3590–3595.

160. Lambert R. J. W., Skandamis P. N., Coote P. J., Nychas G. J. A study of the minimum inhibitory concentration and mode of action of oregano essential oil, thymol and carvacrol. J. Appl. Microbiol. 2001; 91, 453–462.

161. Walsh S. E., Maillard J. Y., Russell A. D., Catrenich C. E., Charbonneau D. L., Bartolo R. G. Activity and mechanisms of action of selected biocidal agents on Gram-positive and negative bacteria. J. Appl. Microbiol. 2003; 94, 240–247.

162. Horváth G., Kovács K., Kocsis B., Kustos I. Effect of thyme (Thymus vulgaris L.) essential oil and its main constituents on the outer membrane protein composition of Erwinia strains studied with microfluid chip technology. Chromatographia 2009; 70, 1645–1650.

163. Di Pasqua R., Mamone G., Ferranti P., Ercolini D., Mauriello G. Changes in the proteome of Salmonella enterica serovar Thompson as stress adaptation to sublethal concentrations of thymol. Proteomics 2010; 10, 1040–1049.

164. Liu Y., Song X., He J., Zheng X., Wu H. Synthetic derivatives of chrysin and their biological activities. Med. Chem. Res. 2014; 23, 555–563.

165. Cushnie T. P., Lamb A. J. Antimicrobial activity of flavonoids. Int. J. Antimicrob. Agents 2005; 26, 343–356.

166. Sithisarn P., Michaelis M., Schubert-Zsilavecz M., Cinatl J. Differential antiviral and anti-inflammatory mechanisms of the flavonoids biochanin A and baicalein in H5N1 influenza A virus-infected cells. Antiviral Res. 2013; 97, 41–48.

167. Reiter M., Brandt W. Relaxant effects on tracheal and ileal smooth muscles of the guinea pig. Arzneimittelforschung 1985; 35, 408–414.

168. Meister A., Bernhardt G., Christoffel V., et al. Antispasmodic activity of Thymus vulgaris extract on the isolated guinea-pig trachea: discrimination between drug and ethanol effects. Planta Med. 1999; 65, 512–516.

169. Keyhanmanesh R., Boskabady M. H. Relaxant effects of differrent fractions from Thymus vulgaris on guinea-pig tracheal chains. Biol. Res. 2012; 45, 67–73.

170. van den Broucke C. O., Lemli J. A. Pharmacological and chemical investigation of thyme liquid extracts. Planta Med. 1981; 41, 129–135.

171. Marsik P., Kokoska L., Landa P., Nepovim A., Soudek P., Vanek T. In vitro inhibitory effects of thymol and quinones of Nigella sativa seeds on cyclooxygenase-1- and -2-catalyzed prostaglandin E2 biosyntheses. Planta Med. 2005; 71, 739–742.

172. de Cássia da Silveira e Sá R., Andrade L. N., de Sousa D. P.  A Review on Anti-Inflammatory Activity of Monoterpenes. Molecules 2013; 18, 1227–1254.

173. Landa P., Kokoska L., Pribylova M., Vanek T., Marsik P. In vitro Anti-inflammatory Activity of Carvacrol: Inhibitory Effect on COX–2 Catalyzed Prostaglandin E2 biosynthesis. Arch. Pharm. Res. 2009; 32, 75–78.

174. Patil R. H., Babu R. L., Naveen Kumar M., Kiran Kumar K. M., Hegde S. M., Nagesh R., Ramesh G. T., Sharma S. C. Anti-Inflammatory Effect of Apigenin on LPS-Induced Pro-Inflammatory Mediators and AP-1 Factors in Human Lung Epithelial Cells. Inflammation 2016; 39, 138–147.

175. Arango D., Diosa-Toro, M., Rojas-Hernandez L. S., Cooperstone J. L., Schwartz S. J., Mo X., Jiang J., Schmittgen T. D., Doseff A. I. Dietary apigenin reduces LPS-induced expression of miR-155 restoring immune balance during inflammation. Mol. Nutr. Food Res. 2015; 59, 763–772.

176. Seo H.-S., Sikder M. A., Lee H. J., Ryu J., Lee C. J. Apigenin inhibits tumor necrosis factor-α-induced production and gene expression of mucin through regulating nuclear factor-kappa B signaling pathway in airway epithelial cells. Biomol. Therapeutics 2014; 22, 525–531.

177. Kritas S. K., Saggini A., Varvara G., Murmura G., Caraffa A., Antinolfi P., Toniato E., Pantalone A., Neri G., Frydas S., et al. Luteolin inhibits mast cell-mediated allergic inflammation. J. Biol. Reg. Homeostat. Agents 2013; 27, 955–959.

178. Kaas P. J. Atemwegsinfekte im Kindesalter: Anwendungsbeobachtung bestätigt Effizienz einer Thymian zubereitung. Natura Med. 2003; 18, 2–4.

179. Kemmerich B., Eberhardt R., Stammer H. Efficacy and tolerability of a fluid extract combination of thyme herb and ivy leaves and matched placebo in adults suffering from acute bronchitis with productive cough. A prospective, double-blind, placebo-controlled clinical trial. Arzneimittelforschung 2006; 56, 652–660.

180. Kemmerich B. Evaluation of efficacy and tolerability of a fixed combination of dry extracts of thyme herb and primrose root in adults suffering from acute bronchitis with productive cough. A prospective, double-blind, placebo-controlled multicentre clinical trial. Arzneimittelforschung 2007; 57, 607–615.

181. Ernst E., März R., Sieder C. A controlled multi-centre study of herbal versus synthetic secretolytic drugs for acute bronchitis. Phytomedicine 1997; 4, 287–293.

182. Gruenwald J., Graubaum H. J., Busch R. Efficacy and tolerability of a fixed combination of thyme and primrose root in patients with acute bronchitis. A double-blind, randomized, placebo-controlled clinical trial. Arzneimittelforschung 2005; 55, 669–676.

183. Ismail C., Willer G., Steindl H. Bronchipret bei akuter Bronchitis. Schweiz. Zschr. Ganzheitsmedizin. 2003; 15, 171–175.

184. Lorenzi S., Placucci F., Vincezi C., et al. Allergic contact dermatitis due to thymol. Contact Dermatitis 1995; 33, 439–440.

185. Zava D. T., Dollbaum C. M., Blen M. Estrogen and progestin aktivity of foods, herbs, and spices. Proc. Soc. Exp. Biol. Med. 1998; 217, 369–378.

Štítky
Farmacie Farmakologie

Článek vyšel v časopise

Česká a slovenská farmacie

Číslo 4

2016 Číslo 4
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#