Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

The Anti-inflammatory Potential of Selected Plant-derived Compounds in Respiratory Diseases

Author(s): Joanna Wieczfinska, Przemyslaw Sitarek, Tomasz Kowalczyk, Ewa Skała and Rafal Pawliczak*

Volume 26, Issue 24, 2020

Page: [2876 - 2884] Pages: 9

DOI: 10.2174/1381612826666200406093257

Price: $65

Abstract

Inflammation plays a major role in chronic airway diseases like asthma, COPD, and cystic fibrosis. Inflammation plays a crucial role in the worsening of the lung function resulting in worsening symptoms. The inflammatory process is very complexed, therefore the strategies for developing an effective treatment for inflammatory airway diseases would benefit from the use of natural substances.

Plant products have demonstrated anti-inflammatory properties on various lung disease models and numerous natural plant agents have successfully been used to treat inflammation.

Naturally occurring substances may exert some anti-inflammatory effects by modulating some of the inflammatory pathways. These agents have been used in different cultures for thousands of years and have proven to be relatively safe.

Parthenolide, apocynin, proanthocyanidins, and boswellic acid present different mechanisms of actions - among others, through NF-κB or NADPH oxidase inhibition, therefore showing a wide range of applications in various inflammatory diseases. Moreover, some of them have also antioxidant properties.

This review provides an overview of the anti-inflammatory effects of some of the natural agents and illustrates their great potential as sources of drugs to cover an extensive range of pharmacological effects.

Keywords: Parthenolide, apocynin, proanthocyanidins, boswellic acid, inflammation, airway disease.

[1]
Gohy ST, Hupin C, Pilette C, Ladjemi MZ. Chronic inflammatory airway diseases: the central role of the epithelium revisited. Clin Exp Allergy 2016; 46(4): 529-42.
[http://dx.doi.org/10.1111/cea.12712] [PMID: 27021118]
[2]
Global strategy for asthma management and prevention Available at:. http://www.ginasthma.org
[3]
Global Initiative for Chronic Obstructive Lung Disease (GOLD) Available at:. https://goldcopd.org
[4]
Santana FP, Pinheiro NM, Mernak MI, et al. Evidences of herbal medicine-derived natural products effects in inflammatory lung diseases. Mediators Inflamm 2016; 2016 2348968
[http://dx.doi.org/10.1155/2016/2348968] [PMID: 27445433]
[5]
Murphy DM, O’Byrne PM. Recent advances in the pathophysiology of asthma. Chest 2010; 137(6): 1417-26.
[http://dx.doi.org/10.1378/chest.09-1895] [PMID: 20525652]
[6]
Tiddens H, Silverman M, Bush A. The role of inflammation in airway disease: remodeling. Am J Respir Crit Care Med 2000; 162(2 Pt 2): S7-S10.
[http://dx.doi.org/10.1164/ajrccm.162.supplement_1.maic-2] [PMID: 10934123]
[7]
Aghasafari P, George U, Pidaparti R. A review of inflammatory mechanism in airway diseases. Inflamm Res 2019; 68(1): 59-74.
[http://dx.doi.org/10.1007/s00011-018-1191-2] [PMID: 30306206]
[8]
Sanei H, Asoodeh A, Hamedakbari-Tusi S, et al. Multi-spectroscopic investigations of aspirin and colchicine interactions with human hemoglobin: Binary and ternary systems. J Solution Chem 2011; 40: 1905-31.
[http://dx.doi.org/10.1007/s10953-011-9766-3]
[9]
Christenson SA, Steiling K, van den Berge M, et al. Asthma-COPD overlap. Clinical relevance of genomic signatures of type 2 inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2015; 191(7): 758-66.
[http://dx.doi.org/10.1164/rccm.201408-1458OC] [PMID: 25611785]
[10]
Barnes PJ. Cellular and molecular mechanisms of asthma and COPD. Clin Sci (Lond) 2017; 131(13): 1541-58.
[http://dx.doi.org/10.1042/CS20160487] [PMID: 28659395]
[11]
Uriarte SM. Novel insights related to CF neutrophils. Blood 2014; 124(7): 985-6.
[http://dx.doi.org/10.1182/blood-2014-07-583831] [PMID: 25124777]
[12]
Mousa HA. Prevention and treatment of influenza, influenza-like illness, and common cold by herbal, complementary, and natural therapies. J Evid Based Complementary Altern Med 2017; 22(1): 166-74.
[http://dx.doi.org/10.1177/2156587216641831] [PMID: 27055821]
[13]
Rehman SU, Kim IS, Choi MS, Kim SH, Zhang Y, Yoo HH. Time-dependent inhibition of CYP2C8 and CYP2C19 by Hedera helix extracts, a traditional respiratory herbal medicine. Molecules 2017; 22(7): 22.
[http://dx.doi.org/10.3390/molecules22071241] [PMID: 28737724]
[14]
Suroowan S, Mahomoodally F. Herbal products for common auto-inflammatory disorders - novel approaches. Comb Chem High Throughput Screen 2018; 21(3): 161-74.
[http://dx.doi.org/10.2174/1386207321666180213093449] [PMID: 29436996]
[15]
Guo BJ, Bian ZX, Qiu HC, Wang YT, Wang Y. Biological and clinical implications of herbal medicine and natural products for the treatment of inflammatory bowel disease. Ann N Y Acad Sci 2017; 1401(1): 37-48.
[http://dx.doi.org/10.1111/nyas.13414] [PMID: 28891095]
[16]
Ferlazzo N, Cirmi S, Calapai G, Ventura-Spagnolo E, Gangemi S, Navarra M. Anti-inflammatory activity of Citrus bergamia derivatives: Where do we stand? Molecules 2016; 21(10): 21.
[http://dx.doi.org/10.3390/molecules21101273] [PMID: 27669206]
[17]
Rastogi S, Pandey MM, Rawat AK. Traditional herbs: a remedy for cardiovascular disorders. Phytomedicine 2016; 23(11): 1082-9.
[http://dx.doi.org/10.1016/j.phymed.2015.10.012] [PMID: 26656228]
[18]
Rupani R, Chavez A. Medicinal plants with traditional use: Ethnobotany in the Indian subcontinent. Clin Dermatol 2018; 36(3): 306-9.
[http://dx.doi.org/10.1016/j.clindermatol.2018.03.005] [PMID: 29908572]
[19]
Xu CC, Wang B, Pu YQ, Tao JS, Zhang T. Advances in extraction and analysis of phenolic compounds from plant materials. Chin J Nat Med 2017; 15(10): 721-31.
[http://dx.doi.org/10.1016/S1875-5364(17)30103-6] [PMID: 29103457]
[20]
Brezani V, Smejkal K, Hosek J, Tomasova V. Anti-inflammatory natural prenylated phenolic compounds - potential lead substances. Curr Med Chem 2018; 25(10): 1094-159.
[http://dx.doi.org/10.2174/0929867324666170810161157] [PMID: 28799496]
[21]
Ginovyan M, Petrosyan M, Trchounian A. Antimicrobial activity of some plant materials used in Armenian traditional medicine. BMC Complement Altern Med 2017; 17(1): 50.
[http://dx.doi.org/10.1186/s12906-017-1573-y] [PMID: 28095835]
[22]
Mo EJ, Ahn JH, Jo YH, Kim SB, Hwang BY, Lee MK. Inositol derivatives and phenolic compounds from the roots of Taraxacum coreanum. Molecules 2017; 22(8): 22.
[PMID: 28805750]
[23]
Cota BB, Bertollo CM, de Oliveira DM. Anti-allergic potential of herbs and herbal natural products - activities and patents. Recent Pat Endocr Metab Immune Drug Discov 2013; 7(1): 26-56.
[http://dx.doi.org/10.2174/187221413804660935] [PMID: 22946460]
[24]
Yuan G, Wahlqvist ML, He G, Yang M, Li D. Natural products and anti-inflammatory activity. Asia Pac J Clin Nutr 2006; 15(2): 143-52.
[PMID: 16672197]
[25]
Sencanski M, Radosevic D, Perovic V, et al. Natural products as promising therapeutics for treatment of influenza disease. Curr Pharm Des 2015; 21(38): 5573-88.
[http://dx.doi.org/10.2174/1381612821666151002113426] [PMID: 26429712]
[26]
Hirsch GE, Viecili PRN, de Almeida AS, et al. Natural products with antiplatelet action. Curr Pharm Des 2017; 23(8): 1228-46.
[http://dx.doi.org/10.2174/1381612823666161123151611] [PMID: 27881059]
[27]
Rayan A, Raiyn J, Falah M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS One 2017; 12(11) e0187925
[http://dx.doi.org/10.1371/journal.pone.0187925] [PMID: 29121120]
[28]
Cardoso SM. Special issue: The antioxidant capacities of natural products. Molecules 2019; 24(3): 24.
[http://dx.doi.org/10.3390/molecules24030492] [PMID: 30704064]
[29]
Talib WH, Al Kury LT. Parthenolide inhibits tumor-promoting effects of nicotine in lung cancer by inducing P53 - dependent apoptosis and inhibiting VEGF expression. Biomed Pharmacother 2018; 107: 1488-95.
[http://dx.doi.org/10.1016/j.biopha.2018.08.139] [PMID: 30257366]
[30]
Lin M, Bi H, Yan Y, et al. Parthenolide suppresses non-small cell lung cancer GLC-82 cells growth via B-Raf/MAPK/Erk pathway. Oncotarget 2017; 8(14): 23436-47.
[http://dx.doi.org/10.18632/oncotarget.15584] [PMID: 28423582]
[31]
Chai WM, Lin MZ, Wang YX, et al. Inhibition of tyrosinase by cherimoya pericarp proanthocyanidins: Structural characterization, inhibitory activity and mechanism. Food Res Int 2017; 100(Pt 1): 731-9.
[http://dx.doi.org/10.1016/j.foodres.2017.07.082] [PMID: 28873743]
[32]
Liu Z, Liu X, Sang L, Liu H, Xu Q, Liu Z. Boswellic acid attenuates asthma phenotypes by downregulation of GATA3 via pSTAT6 inhibition in a murine model of asthma. Int J Clin Exp Pathol 2015; 8(1): 236-43.
[PMID: 25755710]
[33]
Zhu GF, Guo HJ, Huang Y, Wu CT, Zhang XF. Eriodictyol, a plant flavonoid, attenuates LPS-induced acute lung injury through its antioxidative and anti-inflammatory activity. Exp Ther Med 2015; 10(6): 2259-66.
[http://dx.doi.org/10.3892/etm.2015.2827] [PMID: 26668626]
[34]
Kuo MY, Liao MF, Chen FL, et al. Luteolin attenuates the pulmonary inflammatory response involves abilities of antioxidation and inhibition of MAPK and NFκB pathways in mice with endotoxin-induced acute lung injury. Food Chem Toxicol 2011; 49(10): 2660-6.
[http://dx.doi.org/10.1016/j.fct.2011.07.012] [PMID: 21782879]
[35]
Takashima K, Matsushima M, Hashimoto K, et al. Protective effects of intratracheally administered quercetin on lipopolysaccharide-induced acute lung injury. Respir Res 2014; 15: 150.
[http://dx.doi.org/10.1186/s12931-014-0150-x] [PMID: 25413579]
[36]
Qiu J, Chi G, Wu Q, Ren Y, Chen C, Feng H. Pretreatment with the compound asperuloside decreases acute lung injury via inhibiting MAPK and NF-κB signaling in a murine model. Int Immunopharmacol 2016; 31: 109-15.
[http://dx.doi.org/10.1016/j.intimp.2015.12.013] [PMID: 26710167]
[37]
Chen X, Yang X, Liu T, et al. Kaempferol regulates MAPKs and NF-κB signaling pathways to attenuate LPS-induced acute lung injury in mice. Int Immunopharmacol 2012; 14(2): 209-16.
[http://dx.doi.org/10.1016/j.intimp.2012.07.007] [PMID: 22835426]
[38]
Carneiro NVQ, Silva HBFD, Silva RRD, et al. Sambucus australis modulates inflammatory response via inhibition of nuclear factor kappa B (NF-kB) in vitro. An Acad Bras Cienc 2019; 91(1) e20170831
[http://dx.doi.org/10.1590/0001-3765201920170831] [PMID: 30916148]
[39]
Saikia M, Retnakumari AP, Anwar S, et al. Heteronemin, a marine natural product, sensitizes acute myeloid leukemia cells towards cytarabine chemotherapy by regulating farnesylation of Ras. Oncotarget 2018; 9(26): 18115-27.
[http://dx.doi.org/10.18632/oncotarget.24771] [PMID: 29719594]
[40]
Akanda MR, Kim IS, Ahn D, et al. Anti-inflammatory and gastroprotective roles of rabdosia inflexa through downregulation of pro-inflammatory cytokines and MAPK/NF-κB signaling pathways. Int J Mol Sci 2018; 19(2): 19.
[http://dx.doi.org/10.3390/ijms19020584] [PMID: 29462911]
[41]
Guo Z. The modification of natural products for medical use. Acta Pharm Sin B 2017; 7(2): 119-36.
[http://dx.doi.org/10.1016/j.apsb.2016.06.003] [PMID: 28303218]
[42]
Abdollahpour N, Soheili V, Saberi MR, Chamani J. Investigation of the interaction between human serum albumin and two drugs as binary and ternary systems. Eur J Drug Metab Pharmacokinet 2016; 41(6): 705-21.
[http://dx.doi.org/10.1007/s13318-015-0297-y] [PMID: 26328807]
[43]
Mokaberi P, Reyhani V, Amiri-Tehranizadeh Z. New insights into the binding behavior of lomefloxacin and human hemoglobin using biophysical techniques: binary and ternary approaches. New J Chem 2019; 43: 8132-45.
[http://dx.doi.org/10.1039/C9NJ01048C]
[44]
Mathema VB, Koh YS, Thakuri BC, Sillanpää M. Parthenolide, a sesquiterpene lactone, expresses multiple anti-cancer and anti-inflammatory activities. Inflammation 2012; 35(2): 560-5.
[http://dx.doi.org/10.1007/s10753-011-9346-0] [PMID: 21603970]
[45]
Wang M, Li Q. Parthenolide could become a promising and stable drug with anti-inflammatory effects. Nat Prod Res 2015; 29(12): 1092-101.
[http://dx.doi.org/10.1080/14786419.2014.981541] [PMID: 25429885]
[46]
Arıkan-Ayyıldız Z, Karaman M, Özbal S, et al. Efficacy of parthenolide on lung histopathology in a murine model of asthma. Allergol Immunopathol (Madr) 2017; 45(1): 63-8.
[http://dx.doi.org/10.1016/j.aller.2016.06.005] [PMID: 27717727]
[47]
Jang YJ, Back MJ, Fu Z, et al. Protective effect of sesquiterpene lactone parthenolide on LPS-induced acute lung injury. Arch Pharm Res 2016; 39(12): 1716-25.
[http://dx.doi.org/10.1007/s12272-016-0716-x] [PMID: 27757770]
[48]
Uchi H, Arrighi JF, Aubry JP, Furue M, Hauser C. The sesquiterpene lactone parthenolide inhibits LPS- but not TNF-alpha-induced maturation of human monocyte-derived dendritic cells by inhibition of the p38 mitogen-activated protein kinase pathway. J Allergy Clin Immunol 2002; 110(2): 269-76.
[http://dx.doi.org/10.1067/mai.2002.126381] [PMID: 12170268]
[49]
Diekmann H, Fischer D. Parthenolide: a novel pharmacological approach to promote nerve regeneration. Neural Regen Res 2016; 11(10): 1566-7.
[http://dx.doi.org/10.4103/1673-5374.193228] [PMID: 27904480]
[50]
Mazor RL, Menendez IY, Ryan MA, Fiedler MA, Wong HR. Sesquiterpene lactones are potent inhibitors of interleukin 8 gene expression in cultured human respiratory epithelium. Cytokine 2000; 12(3): 239-45.
[http://dx.doi.org/10.1006/cyto.1999.0526] [PMID: 10704251]
[51]
de Carvalho LSA, Fontes LBA, Gazolla MC, et al. Parthenolide modulates immune response in cells from C57BL/6 mice induced with experimental autoimmune encephalomyelitis. Planta Med 2017; 83(8): 693-700.
[PMID: 27997959]
[52]
Zhang X, Fan C, Xiao Y, Mao X. Anti-inflammatory and antiosteoclastogenic activities of parthenolide on human periodontal ligament cells in vitro. Evid Based Complement Alternat Med 2014; 2014 546097
[http://dx.doi.org/10.1155/2014/546097] [PMID: 25610476]
[53]
Saadane A, Eastman J, Berger M, Bonfield TL. Parthenolide inhibits ERK and AP-1 which are dysregulated and contribute to excessive IL-8 expression and secretion in cystic fibrosis cells. J Inflamm (Lond) 2011; 8: 26.
[http://dx.doi.org/10.1186/1476-9255-8-26] [PMID: 21992677]
[54]
Freund RRA, Gobrecht P, Fischer D, Arndt HD. Advances in chemistry and bioactivity of parthenolide. Nat Prod Rep 2019.
[PMID: 31763637]
[55]
Kim CY, Kang B, Suh HJ, Choi HS. Parthenolide, a feverfew-derived phytochemical, ameliorates obesity and obesity-induced inflammatory responses via the Nrf2/Keap1 pathway. Pharmacol Res 2019; 145 104259
[http://dx.doi.org/10.1016/j.phrs.2019.104259] [PMID: 31078744]
[56]
Ge W, Liu Z, Sun Y, et al. Design and synthesis of parthenolide-SAHA hybrids for intervention of drug-resistant acute myeloid leukemia. Bioorg Chem 2019; 87: 699-713.
[http://dx.doi.org/10.1016/j.bioorg.2019.03.056] [PMID: 30953889]
[57]
Li X, Cui X, Li Y, Fitz Y, Hsu L, Eichacker PQ. Parthenolide has limited effects on nuclear factor-kappa beta increases and worsens survival in lipopolysaccharide-challenged C57BL/6J mice. Cytokine 2006; 33(6): 299-308.
[http://dx.doi.org/10.1016/j.cyto.2006.03.002] [PMID: 16720096]
[58]
Petrovic SD, Dobric S, Bokonjic D, Niketic M, García-Piñeres A, Merfort I. Evaluation of Tanacetum larvatum for an anti-inflammatory activity and for the protection against indomethacin-induced ulcerogenesis in rats. J Ethnopharmacol 2003; 87(1): 109-13.
[http://dx.doi.org/10.1016/S0378-8741(03)00118-1] [PMID: 12787963]
[59]
Curry EA III, Murry DJ, Yoder C, et al. Phase I dose escalation trial of feverfew with standardized doses of parthenolide in patients with cancer. Invest New Drugs 2004; 22(3): 299-305.
[http://dx.doi.org/10.1023/B:DRUG.0000026256.38560.be] [PMID: 15122077]
[60]
Wiegman CH, Michaeloudes C, Haji G, et al. Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 2015; 136(3): 769-80.
[http://dx.doi.org/10.1016/j.jaci.2015.01.046] [PMID: 25828268]
[61]
Haberzettl P, O’Toole TE, Bhatnagar A, Conklin DJ. Exposure to fine particulate air pollution causes vascular insulin resistance by inducing pulmonary oxidative stress. Environ Health Perspect 2016; 124(12): 1830-9.
[http://dx.doi.org/10.1289/EHP212] [PMID: 27128347]
[62]
Valavanidis A, Vlachogianni T, Fiotakis K, Loridas S. Pulmonary oxidative stress, inflammation and cancer: respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int J Environ Res Public Health 2013; 10(9): 3886-907.
[http://dx.doi.org/10.3390/ijerph10093886] [PMID: 23985773]
[63]
Kim SY, Moon KA, Jo HY, et al. Anti-inflammatory effects of apocynin, an inhibitor of NADPH oxidase, in airway inflammation. Immunol Cell Biol 2012; 90(4): 441-8.
[http://dx.doi.org/10.1038/icb.2011.60] [PMID: 21709687]
[64]
Zhang HX, Liu SJ, Tang XL, et al. H2S Attenuates LPS-induced acute lung injury by reducing oxidative/nitrative stress and inflammation. Cell Physiol Biochem 2016; 40(6): 1603-12.
[http://dx.doi.org/10.1159/000453210] [PMID: 28006762]
[65]
Toblli JE, Cao G, Giani JF, Dominici FP, Angerosa M. Markers of oxidative/nitrosative stress and inflammation in lung tissue of rats exposed to different intravenous iron compounds. Drug Des Devel Ther 2017; 11: 2251-63.
[http://dx.doi.org/10.2147/DDDT.S132612] [PMID: 28814833]
[66]
Wang XL, Li T, Li JH, Miao SY, Xiao XZ. The effects of resveratrol on inflammation and oxidative stress in a rat model of chronic obstructive pulmonary disease. Molecules 2017; 22(9): 22.
[http://dx.doi.org/10.3390/molecules22091529] [PMID: 28895883]
[67]
McGovern TK, Chen M, Allard B, Larsson K, Martin JG, Adner M. Neutrophilic oxidative stress mediates organic dust-induced pulmonary inflammation and airway hyperresponsiveness. Am J Physiol Lung Cell Mol Physiol 2016; 310(2): L155-65.
[http://dx.doi.org/10.1152/ajplung.00172.2015] [PMID: 26545900]
[68]
Ferrari RS, Andrade CF. Oxidative stress and lung ischemia-reperfusion injury. Oxid Med Cell Longev 2015; 2015 590987
[http://dx.doi.org/10.1155/2015/590987] [PMID: 26161240]
[69]
Wieczfinska J, Pawliczak R. Thymic stromal lymphopoietin and apocynin alter the expression of airway remodeling factors in human rhinovirus-infected cells. Immunobiology 2017; 222(8-9): 892-9.
[http://dx.doi.org/10.1016/j.imbio.2017.05.010] [PMID: 28545810]
[70]
Abdelmageed ME, El-Awady MS, Suddek GM. Apocynin ameliorates endotoxin-induced acute lung injury in rats. Int Immunopharmacol 2016; 30: 163-70.
[http://dx.doi.org/10.1016/j.intimp.2015.12.006] [PMID: 26687059]
[71]
Muijsers RB, van Ark I, Folkerts G, et al. Apocynin and 1400 W prevents airway hyperresponsiveness during allergic reactions in mice. Br J Pharmacol 2001; 134(2): 434-40.
[http://dx.doi.org/10.1038/sj.bjp.0704235] [PMID: 11564663]
[72]
Impellizzeri D, Esposito E, Mazzon E, et al. Effect of apocynin, a NADPH oxidase inhibitor, on acute lung inflammation. Biochem Pharmacol 2011; 81(5): 636-48.
[http://dx.doi.org/10.1016/j.bcp.2010.12.006] [PMID: 21147071]
[73]
Pandey A, Kour K, Bani S, et al. Amelioration of adjuvant induced arthritis by apocynin. Phytother Res 2009; 23(10): 1462-8.
[http://dx.doi.org/10.1002/ptr.2803] [PMID: 19288522]
[74]
Stefanska J, Sarniak A, Wlodarczyk A, et al. Apocynin reduces reactive oxygen species concentrations in exhaled breath condensate in asthmatics. Exp Lung Res 2012; 38(2): 90-9.
[http://dx.doi.org/10.3109/01902148.2011.649823] [PMID: 22296407]
[75]
Stefanska J, Sarniak A, Wlodarczyk A, et al. Hydrogen peroxide and nitrite reduction in exhaled breath condensate of COPD patients. Pulm Pharmacol Ther 2012; 25(5): 343-8.
[http://dx.doi.org/10.1016/j.pupt.2012.06.001] [PMID: 22705948]
[76]
Xu L, Li Y, Wan S, Wang Y, Yu P. Protective effects of apocynin nitrone on acute lung injury induced by lipopolysaccharide in rats. Int Immunopharmacol 2014; 20(2): 377-82.
[http://dx.doi.org/10.1016/j.intimp.2014.03.014] [PMID: 24704624]
[77]
Oostwoud LC, Gunasinghe P, Seow HJ, et al. Apocynin and ebselen reduce influenza A virus-induced lung inflammation in cigarette smoke-exposed mice. Sci Rep 2016; 6: 20983.
[http://dx.doi.org/10.1038/srep20983] [PMID: 26877172]
[78]
Orosz Z, Csiszar A, Labinskyy N, et al. Cigarette smoke-induced proinflammatory alterations in the endothelial phenotype: role of NAD(P)H oxidase activation. Am J Physiol Heart Circ Physiol 2007; 292(1): H130-9.
[http://dx.doi.org/10.1152/ajpheart.00599.2006] [PMID: 17213480]
[79]
Stefanska J, Sokolowska M, Sarniak A, et al. Apocynin decreases hydrogen peroxide and nitrate concentrations in exhaled breath in healthy subjects. Pulm Pharmacol Ther 2010; 23(1): 48-54.
[http://dx.doi.org/10.1016/j.pupt.2009.09.003] [PMID: 19786113]
[80]
Kilic T, Parlakpinar H, Taslidere E, et al. Protective and therapeutic effect of apocynin on bleomycin-induced lung fibrosis in rats. Inflammation 2015; 38(3): 1166-80.
[http://dx.doi.org/10.1007/s10753-014-0081-1] [PMID: 25502443]
[81]
Choi SH, Suh GJ, Kwon WY, et al. Apocynin suppressed the nuclear factor-κB pathway and attenuated lung injury in a rat hemorrhagic shock model. J Trauma Acute Care Surg 2017; 82(3): 566-74.
[http://dx.doi.org/10.1097/TA.0000000000001337] [PMID: 28030501]
[82]
Kučera J, Binó L, Štefková K, et al. Apocynin and diphenyleneiodonium induce oxidative stress and modulate PI3K/Akt and MAPK/Erk activity in mouse embryonic stem cells. Oxid Med Cell Longev 2016; 2016 7409196
[http://dx.doi.org/10.1155/2016/7409196] [PMID: 26788250]
[83]
’t Hart BA, Copray S, Philippens I. Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res Int 2014; 2014 298020
[PMID: 25140304]
[84]
Hwang YJ, Nam SJ, Chun W, et al. Anti-inflammatory effects of apocynin on dextran sulfate sodium-induced mouse colitis model. PLoS One 2019; 14(5) e0217642
[http://dx.doi.org/10.1371/journal.pone.0217642] [PMID: 31141554]
[85]
Marín M, Gimeno C, Giner RM, Ríos JL, Máñez S, Recio MAC. Influence of dimerization of apocynin on its effects in experimental colitis. J Agric Food Chem 2017; 65(20): 4083-91.
[http://dx.doi.org/10.1021/acs.jafc.7b00872] [PMID: 28485605]
[86]
Simonyi A, Serfozo P, Lehmidi TM, et al. The neuroprotective effects of apocynin. Front Biosci (Elite Ed) 2012; 4: 2183-93.
[http://dx.doi.org/10.2741/e535] [PMID: 22202030]
[87]
Stefanska J, Pawliczak R. Apocynin: molecular aptitudes. Mediators Inflamm 2008; 2008 106507
[http://dx.doi.org/10.1155/2008/106507] [PMID: 19096513]
[88]
Peters EA, Hiltermann JT, Stolk J. Effect of apocynin on ozone-induced airway hyperresponsiveness to methacholine in asthmatics. Free Radic Biol Med 2001; 31(11): 1442-7.
[http://dx.doi.org/10.1016/S0891-5849(01)00725-0] [PMID: 11728816]
[89]
Coleman SL, Shaw OM. Progress in the understanding of the pathology of allergic asthma and the potential of fruit proanthocyanidins as modulators of airway inflammation. Food Funct 2017; 8(12): 4315-24.
[http://dx.doi.org/10.1039/C7FO00789B] [PMID: 29140397]
[90]
Sun W, Meng J, Wang Z, et al. Proanthocyanidins attenuation of H2O2-induced oxidative damage in tendon-derived stem cells via upregulating Nrf-2 signaling pathway. BioMed Res Int 2017; 2017 7529104
[http://dx.doi.org/10.1155/2017/7529104] [PMID: 29201913]
[91]
Martinez-Micaelo N, González-Abuín N, Ardèvol A, Pinent M, Blay MT. Procyanidins and inflammation: molecular targets and health implications. Biofactors 2012; 38(4): 257-65.
[http://dx.doi.org/10.1002/biof.1019] [PMID: 22505223]
[92]
Agache I, Strasser DS, Klenk A, et al. Serum IL-5 and IL-13 consistently serve as the best predictors for the blood eosinophilia phenotype in adult asthmatics. Allergy 2016; 71(8): 1192-202.
[http://dx.doi.org/10.1111/all.12906] [PMID: 27060452]
[93]
Nyanhanda T, Gould EM, McGhie T, Shaw OM, Harper JL, Hurst RD. Blackcurrant cultivar polyphenolic extracts suppress CCL26 secretion from alveolar epithelial cells. Food Funct 2014; 5(4): 671-7.
[http://dx.doi.org/10.1039/c3fo60568j] [PMID: 24526266]
[94]
Zhou DY, Du Q, Li RR, Huang M, Zhang Q, Wei GZ. Grape seed proanthocyanidin extract attenuates airway inflammation and hyperresponsiveness in a murine model of asthma by downregulating inducible nitric oxide synthase. Planta Med 2011; 77(14): 1575-81.
[http://dx.doi.org/10.1055/s-0030-1270957] [PMID: 21452107]
[95]
Miller M, Cho JY, McElwain K, et al. Corticosteroids prevent myofibroblast accumulation and airway remodeling in mice. Am J Physiol Lung Cell Mol Physiol 2006; 290(1): L162-9.
[http://dx.doi.org/10.1152/ajplung.00252.2005] [PMID: 16344333]
[96]
Katiyar SK. Grape seed proanthocyanidines and skin cancer prevention: inhibition of oxidative stress and protection of immune system. Mol Nutr Food Res 2008; 52(Suppl. 1): S71-6.
[http://dx.doi.org/10.1002/mnfr.200700198] [PMID: 18384090]
[97]
Liu W, Zhao S, Wang J, et al. Grape seed proanthocyanidin extract ameliorates inflammation and adiposity by modulating gut microbiota in high-fat diet mice. Mol Nutr Food Res 2017; 61(9): 61.
[PMID: 28500724]
[98]
Liu W, Xu C, Sun X, et al. Grape seed proanthocyanidin extract protects against perfluorooctanoic acid-induced hepatotoxicity by attenuating inflammatory response, oxidative stress and apoptosis in mice. Toxicol Res (Camb) 2015; 5(1): 224-34.
[http://dx.doi.org/10.1039/C5TX00260E] [PMID: 30090339]
[99]
Aswar UM, Kandhare AD, Mohan V, Thakurdesai PA. Anti-allergic effect of intranasal administration of type-A procyanidin polyphenols based standardized extract of cinnamon bark in ovalbumin sensitized BALB/c mice. Phytother Res 2015; 29(3): 423-33.
[http://dx.doi.org/10.1002/ptr.5269] [PMID: 25504814]
[100]
Li Y, Yu Q, Zhao W, et al. Oligomeric proanthocyanidins attenuate airway inflammation in asthma by inhibiting dendritic cells maturation. Mol Immunol 2017; 91: 209-17.
[http://dx.doi.org/10.1016/j.molimm.2017.09.012] [PMID: 28963930]
[101]
Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol 2008; 8(3): 193-204.
[http://dx.doi.org/10.1038/nri2275] [PMID: 18301423]
[102]
Shenoy SF, Keen CL, Kalgaonkar S, Polagruto JA. Effects of grape seed extract consumption on platelet function in postmenopausal women. Thromb Res 2007; 121(3): 431-2.
[http://dx.doi.org/10.1016/j.thromres.2007.09.004] [PMID: 17950783]
[103]
Ma Q, Kim EY, Lindsay EA, Han O. Bioactive dietary polyphenols inhibit heme iron absorption in a dose-dependent manner in human intestinal Caco-2 cells. J Food Sci 2011; 76(5): H143-50.
[http://dx.doi.org/10.1111/j.1750-3841.2011.02184.x] [PMID: 22417433]
[104]
Asma B, Vicky L, Stephanie D, Yves D, Amy H, Sylvie D. Standardised high dose versus low dose cranberry Proanthocyanidin extracts for the prevention of recurrent urinary tract infection in healthy women [PACCANN]: a double blind randomised controlled trial protocol. BMC Urol 2018; 18(1): 29.
[http://dx.doi.org/10.1186/s12894-018-0342-7] [PMID: 29716563]
[105]
Occhipinti A, Germano A, Maffei ME. Prevention of urinary tract infection with oximacro, a cranberry extract with a high content of A-type proanthocyanidins: A pre-clinical double-blind controlled study. Urol J 2016; 13(2): 2640-9.
[PMID: 27085566]
[106]
Wang MX, Zhao JX, Meng YJ, et al. Acetyl-11-keto-β-boswellic acid inhibits the secretion of cytokines by dendritic cells via the TLR7/8 pathway in an imiquimod-induced psoriasis mouse model and in vitro. Life Sci 2018; 207: 90-104.
[http://dx.doi.org/10.1016/j.lfs.2018.05.044] [PMID: 29859222]
[107]
Ding Y, Qiao Y, Wang M, et al. Enhanced Neuroprotection of Acetyl-11-Keto-β-Boswellic Acid (AKBA)-loaded O-carboxymethyl chitosan nanoparticles through antioxidant and anti-inflammatory pathways. Mol Neurobiol 2016; 53(6): 3842-53.
[http://dx.doi.org/10.1007/s12035-015-9333-9] [PMID: 26162321]
[108]
Safayhi H, Mack T, Sabieraj J, Anazodo MI, Subramanian LR, Ammon HP. Boswellic acids: novel, specific, nonredox inhibitors of 5-lipoxygenase. J Pharmacol Exp Ther 1992; 261(3): 1143-6.
[PMID: 1602379]
[109]
Safayhi H, Sailer ER, Ammon HP. Mechanism of 5-lipoxygenase inhibition by acetyl-11-keto-beta-boswellic acid. Mol Pharmacol 1995; 47(6): 1212-6.
[PMID: 7603462]
[110]
Safayhi H, Rall B, Sailer ER, Ammon HP. Inhibition by boswellic acids of human leukocyte elastase. J Pharmacol Exp Ther 1997; 281(1): 460-3.
[PMID: 9103531]
[111]
Ali EN, Mansour SZ. Boswellic acids extract attenuates pulmonary fibrosis induced by bleomycin and oxidative stress from gamma irradiation in rats. Chin Med 2011; 6: 36.
[http://dx.doi.org/10.1186/1749-8546-6-36] [PMID: 21961991]
[112]
Sailer ER, Subramanian LR, Rall B, Hoernlein RF, Ammon HP, Safayhi H. Acetyl-11-keto-beta-boswellic acid (AKBA): structure requirements for binding and 5-lipoxygenase inhibitory activity. Br J Pharmacol 1996; 117(4): 615-8.
[http://dx.doi.org/10.1111/j.1476-5381.1996.tb15235.x] [PMID: 8646405]
[113]
Gupta I, Gupta V, Parihar A, et al. Effects of Boswellia serrata gum resin in patients with bronchial asthma: results of a double-blind, placebo-controlled, 6-week clinical study. Eur J Med Res 1998; 3(11): 511-4.
[PMID: 9810030]
[114]
Notarnicola A, Maccagnano G, Moretti L, et al. Methylsulfonylmethane and boswellic acids versus glucosamine sulfate in the treatment of knee arthritis: Randomized trial. Int J Immunopathol Pharmacol 2016; 29(1): 140-6.
[http://dx.doi.org/10.1177/0394632015622215] [PMID: 26684635]
[115]
Ammon HP. Boswellic acids and their role in chronic inflammatory diseases. Adv Exp Med Biol 2016; 928: 291-327.
[http://dx.doi.org/10.1007/978-3-319-41334-1_13] [PMID: 27671822]
[116]
Fell CD, Martinez FJ, Liu LX, et al. Clinical predictors of a diagnosis of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2010; 181(8): 832-7.
[http://dx.doi.org/10.1164/rccm.200906-0959OC] [PMID: 20056903]
[117]
Khan MA, Ali R, Parveen R, Najmi AK, Ahmad S. Pharmacological evidences for cytotoxic and antitumor properties of Boswellic acids from Boswellia serrata. J Ethnopharmacol 2016; 191: 315-23.
[http://dx.doi.org/10.1016/j.jep.2016.06.053] [PMID: 27346540]
[118]
Ammon HP. Modulation of the immune system by Boswellia serrata extracts and boswellic acids. Phytomedicine 2010; 17(11): 862-7.
[http://dx.doi.org/10.1016/j.phymed.2010.03.003] [PMID: 20696559]
[119]
Baram SM, Karima S, Shateri S, et al. Functional improvement and immune-inflammatory cytokines profile of ischaemic stroke patients after treatment with boswellic acids: a randomized, double-blind, placebo-controlled, pilot trial. Inflammopharmacology 2019; 27(6): 1101-12.
[http://dx.doi.org/10.1007/s10787-019-00627-z] [PMID: 31407195]
[120]
Roy NK, Deka A, Bordoloi D, et al. The potential role of boswellic acids in cancer prevention and treatment. Cancer Lett 2016; 377(1): 74-86.
[http://dx.doi.org/10.1016/j.canlet.2016.04.017] [PMID: 27091399]
[121]
Roy NK, Parama D, Banik K, et al. An update on pharmacological potential of boswellic acids against chronic diseases. Int J Mol Sci 2019; 20(17): 20.
[http://dx.doi.org/10.3390/ijms20174101] [PMID: 31443458]
[122]
Riva A, Giacomelli L, Togni S, et al. Oral administration of a lecithin-based delivery form of boswellic acids (Casperome®) for the prevention of symptoms of irritable bowel syndrome: a randomized clinical study. Minerva Gastroenterol Dietol 2019; 65(1): 30-5.
[http://dx.doi.org/10.23736/S1121-421X.18.02530-8] [PMID: 30676012]
[123]
Skarke C, Kuczka K, Tausch L, et al. Increased bioavailability of 11-keto-β-boswellic acid following single oral dose frankincense extract administration after a standardized meal in healthy male volunteers: modeling and simulation considerations for evaluating drug exposures. J Clin Pharmacol 2012; 52(10): 1592-600.
[http://dx.doi.org/10.1177/0091270011422811] [PMID: 22167571]
[124]
Yang N, Ray DW, Matthews LC. Current concepts in glucocorticoid resistance. Steroids 2012; 77(11): 1041-9.
[http://dx.doi.org/10.1016/j.steroids.2012.05.007] [PMID: 22728894]
[125]
Barnes PJ. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci (Lond) 1998; 94(6): 557-72.
[http://dx.doi.org/10.1042/cs0940557] [PMID: 9854452]
[126]
Fardet L, Flahault A, Kettaneh A, et al. Corticosteroid-induced clinical adverse events: frequency, risk factors and patient’s opinion. Br J Dermatol 2007; 157(1): 142-8.
[http://dx.doi.org/10.1111/j.1365-2133.2007.07950.x] [PMID: 17501951]
[127]
Ericson-Neilsen W, Kaye AD. Steroids: pharmacology, complications, and practice delivery issues. Ochsner J 2014; 14(2): 203-7.
[PMID: 24940130]
[128]
Allijn IE, Vaessen SF, Quarles van Ufford LC, et al. Head-to-head comparison of anti-inflammatory performance of known natural products in vitro. PLoS One 2016; 11(5) e0155325
[http://dx.doi.org/10.1371/journal.pone.0155325] [PMID: 27163931]
[129]
Li H, Zhou M, Zhao A, Jia W. Traditional Chinese medicine: balancing the gut ecosystem. Phytother Res 2009; 23(9): 1332-5.
[http://dx.doi.org/10.1002/ptr.2590] [PMID: 19253310]
[130]
Lan K, Xie G, Jia W. Towards polypharmacokinetics: pharmacokinetics of multicomponent drugs and herbal medicines using a metabolomics approach. Evid Based Complement Alternat Med 2013; 2013 819147
[http://dx.doi.org/10.1155/2013/819147] [PMID: 23573155]
[131]
Gautam R, Jachak SM. Recent developments in anti-inflammatory natural products. Med Res Rev 2009; 29(5): 767-820.
[http://dx.doi.org/10.1002/med.20156] [PMID: 19378317]
[132]
Cheung RCF, Ng TB, Wong JH, Chen Y, Chan WY. Marine natural products with anti-inflammatory activity. Appl Microbiol Biotechnol 2016; 100(4): 1645-66.
[http://dx.doi.org/10.1007/s00253-015-7244-3] [PMID: 26711278]
[133]
Karimi A, Majlesi M, Rafieian-Kopaei M. Herbal versus synthetic drugs; beliefs and facts. J Nephropharmacol 2015; 4(1): 27-30.
[PMID: 28197471]
[134]
Shakibapour N, Dehghani Sani F, Beigoli S, Sadeghian H, Chamani J. Multi-spectroscopic and molecular modeling studies to reveal the interaction between propyl acridone and calf thymus DNA in the presence of histone H1: binary and ternary approaches. J Biomol Struct Dyn 2019; 37(2): 359-71.
[http://dx.doi.org/10.1080/07391102.2018.1427629] [PMID: 29338579]
[135]
Sohrabi T, Hosseinzadeh M, Beigoli S. Probing the binding of lomefloxacin to a calf thymus DNA-histone H1 complex by multi-spectroscopic and molecular modeling techniques. J Mol Liq 2018; 256: 127-38.
[http://dx.doi.org/10.1016/j.molliq.2018.02.031]
[136]
George P. Concerns regarding the safety and toxicity of medicinal plants - An overview. J Appl Pharm Sci 2011; 01: 40-4.
[137]
Haq I. Safety of medicinal plants. Pak J Med Res 2004; 43: 203-10.
[138]
Jiao R, Liu Y, Gao H, Xiao J, So KF. The anti-oxidant and antitumor properties of plant polysaccharides. Am J Chin Med 2016; 44(3): 463-88.
[http://dx.doi.org/10.1142/S0192415X16500269] [PMID: 27109156]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy