Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2132 word(s) 2132 2021-11-05 04:07:26 |
2 format correction Meta information modification 2132 2021-11-17 03:10:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lin, L. Xanthones, A Promising Anti-Inflammatory Scaffold. Encyclopedia. Available online: https://encyclopedia.pub/entry/16045 (accessed on 29 March 2024).
Lin L. Xanthones, A Promising Anti-Inflammatory Scaffold. Encyclopedia. Available at: https://encyclopedia.pub/entry/16045. Accessed March 29, 2024.
Lin, Li-Gen. "Xanthones, A Promising Anti-Inflammatory Scaffold" Encyclopedia, https://encyclopedia.pub/entry/16045 (accessed March 29, 2024).
Lin, L. (2021, November 16). Xanthones, A Promising Anti-Inflammatory Scaffold. In Encyclopedia. https://encyclopedia.pub/entry/16045
Lin, Li-Gen. "Xanthones, A Promising Anti-Inflammatory Scaffold." Encyclopedia. Web. 16 November, 2021.
Xanthones, A Promising Anti-Inflammatory Scaffold
Edit

Inflammation is the body’s self-protective response to multiple stimulus, from external harmful substances to internal danger signals released after trauma or cell dysfunction. Many diseases are considered to be related to inflammation, such as cancer, metabolic disorders, aging, and neurodegenerative diseases.  Xanthones, a unique scaffold with a 9H-Xanthen-9-one core structure, widely exist in natural sources. Till now, over 250 xanthones were isolated and identified in plants from the families Gentianaceae and Hypericaceae. Many xanthones have been disclosed with anti-inflammatory properties on different models, either in vitro or in vivo.

xanthones anti-inflammation drug likeness SwissADME

1. Introduction

Inflammation is a kind of active defense reaction of organisms to external stimulations, such as infectious microorganisms, or internal processes, such as tissue injury, cell death, and cancer [1][2][3]. However, long-term low-grade inflammation leads to many human diseases, including aging, metabolic disorders, cancer, and neurodegenerative diseases [4][5][6][7]. Thus, the discovery of anti-inflammatory medicines has been and is continuing to be one of the hotspots of pharmaceutical research.
Currently, anti-inflammatory therapy mainly includes non-steroidal anti-inflammatory drugs (NSAIDS) and glucocorticoids, both of which possess various side effects, such as cardiotoxicity, hepatotoxicity, and immunological dysfunction [8][9]. Natural products have attracted increasingly more attention due to their safety and effectiveness [10]. Emerging evidence indicates that natural products always function as multi-component and multi-target patterns [11]. Naturally occurring anti-inflammatory compounds might be promising candidates for the treatment of enteritis, arthritis, and skin inflammation. Xanthones were firstly isolated in 1855 by a German scientist pursuing research on dysentery and then named by the Greek word for yellow, xanthos [12]. Xanthones possess a unique 9H-Xanthen-9-one scaffold (Figure 1), which mainly occurs in the plants of the families Gentianaceae and Hypericaceae, as well as some fungi and lichens [13]. Several types of xanthones have been identified, including simple oxygenated xanthones, xanthone glycosides, prenylated xanthones, xanthonolignoids, and miscellaneous [14]. The studies of xanthone are provoking not only due to the structural diversity but also a variety of pharmacological activities. Many xanthones have been reported with potent anti-inflammatory properties [15][16][17][18][19].
Figure 1. The core structure of xanthone.

2. Xanthones with Anti-Inflammatory Properties

Using the keywords xanthone and inflammation, we collected data from Google Scholar, Web of Science, Scopus, and Pubmed. A total of 44 xanthones were found with anti-inflammatory properties, containing 6 simple oxygenated xanthones (16), 2 xanthone glycosides (7, 8), 33 prenylated xanthones (941), and 3 xanthonolignoids (4244) (Figure 2). Many models, either in vitro or in vivo, have been recruited to evaluate the anti-inflammatory properties of xanthones. To organize the review, the xanthones were classified based on bioassays (Table 1).
Figure 2. Structures of xanthones with anti-inflammatory activity. The symbol Ac represents an acetyl group.
Table 1. Xanthones with anti-inflammatory activity.

Model/Method

No

Dose

Outcomes

Ref.

LPS-stimulated RAW264.7 macrophages

1

10 μmol/L

Suppressed the phosphorylation of IKK-β, Akt, and p65

[20]

2

10 μmol/L

Inhibited the production of IL-6 and TNF-α

[20]

4, 7

25, 50 μg/mL

Suppressed the generation of TNF-α and ICAM-1

[21]

6, 1518, 21, 2628, 37

3, 10, 30, 100 μmol/L

Downregulated mRNA expressions of iNOS and COX-2

[22]

9

50 μmol/L

Suppressed iNOS, COX-2, inhibited TNF-α, IL-1β, IL-6, IκB-α

[23]

12

1, 2, 5, 10 μmol/L

Induced HO-1 expression and increased HO-1 activity, inhibited TNF-α, IL-1β

[24]

19

5, 10, 20 μmol/L

Inhibited NO production and IL-6 secretion

[25]

22

11.72 ± 1.16 μmol/L

Inhibited NO production

[26]

30

20, 40, 60 μmol/L

Inhibited the production of NO, iNOS, TNF-α, IL-6, and IL-1β

[27]

33

6.25 μmol/L

Suppressed NO production

[28]

38

50 μg/mL

Inhibited COX-1, COX-2 and 5-LOX-mediated LTB4 formation

[29]

40

11.3 ± 1.7 μmol/L

Inhibited NO production

[30]

41

18.0 ± 1.8 μmol/L

Inhibited NO production

[30]

LPS/IFN?-stimulated RAW264.7 macrophages

20

3.125–25 ?mol/L

Suppressed IL-6, IL-12, and TNF-?

[31]

39

10 μmol/L

Decreased NO production

[32]

Human neutrophils

3, 7, 42, 43

1000 μg/mL

Inhibited WST-1 by NADPH oxidase

[33]

23, 24, 29, 31, 32, 34, 35

10 μg/mL

Inhibited superoxide anion generation and elastase release

[34]

CD3 synovial cells

7

100 μg/mL

Downregulation of TNF-α, IL-1β, and IFN-γ

[35]

Lung of septic mice

10, 30, 100 mg/kg

Upregulated the expression and activity of HO-1

[36]

Carrageenan-induced mechanical hyperalgesia Wistar rats

100 μg/paw

Inhibited TNF-α level through CINC-1/epinephrine/PKA pathway

[37]

MC 3T3-E1 cell line

10, 20, 30, 40 μmol/L

Alleviated oxidative stress by activating the BMP2/Smad-1 signaling pathway

[38]

HFLS-RA cells

10

10 μg/mL

Inhibited nuclear translocation of p65

[39]

AA rats

10

2.5−10 μg/mL

Inhibited fibrous hyperplasia, synovial angiogenesis, cartilage

[39]

Peripheral LPS-induced neuroinflammation in C57BL/6J mice

10

40 mg/kg

Reduced brain levels of IL-6 and COX-2

[40]

Established CIA in DBA/1J mice

10

10, 40 mg/kg

Reduced the levels of anti-collagen IgG2a and autoantibodies in serum and the production of LIX/CXCL5, IP-10/CXCL10, MIG/CXCL9, RANTES/CCL5, IL-6 and IL-33 in joints

[41]

Ovalbumin-induced allergic asthma mice

9, 10

10, 30 mg/kg

Increased Th2 cytokine

[42]

3T3-L1 cells

10, 19

10 μmol/L

Inhibited PPARγ and NFR2 through NF-κB

[43]

Acetic acid-induced mice

5

10, 20 mg/kg

Reduced paw edema

[44]

EPP-induced ear edema

10, 13, 14, 25, 36

1 mg/kg

Inhibited edema

[45]

LPS-induced adipose tissue inflammation mice

10

10 mg/kg

Reduced macrophage content and shifted pro-inflammatory macrophage polarization

[12]

19

20 mg/kg

Reduced macrophage content through inhibiting MAPKs and NF-κB activation

[25]

Macrophages are a major component of the mononuclear phagocyte system [46]. Monocytes migrate into various tissues and transform into macrophages. Macrophages play a critical role in the initiation, maintenance, and resolution of inflammation. Lipopolysaccharide (LPS), a component of the Gram-negative bacterial cell wall, has been widely used to induce an inflammatory response in macrophages [47]. The LPS-stimulated RAW264.7 macrophage model is an effective tool for anti-inflammatory drug screening and anti-inflammatory mechanism investigation. Using a LPS-induced RAW264.7 macrophage model, 3,4,5,6-tetrahydroxyxanthone (4) and mangiferin (7) were found to suppress the generation of TNF-α and intercellular adhesion molecule-1 (ICAM-1) [21]. Six xanthones, including 1,3,6,7-tetrahydroxy-8-prenylxanthone (19) [25], β-mangostin (22) [26], nagostenone F (30) [27], inophinnin (33) [28], garcinoxanghone B (40) [30], and garcinoxanthone C (41) [30], were reported to reduce NO production in LPS-stimulated RAW264.7 macrophage. 6′-O-acetyl mangiferin (OAM) (8), an acetylated xanthone C-glucoside, was reported to suppress iNOS and COX-2 expression, thereby inhibiting the levels of TNF-α, IL-1β, and IL-6 in LPS-stimulated RAW264.7 cells. Furthermore, OAM inhibited the LPS-induced phosphorylation of c-Jun N-terminal kinases (JNK), extracellular signal-regulated kinase (ERK), and p38, which led to the blockade of nuclear factor-κB (NF-κB) and inhibitor κB (IκB)-α activation [23]. Cudratricusxanthone A (12), isolated from the roots of Cudrania tricuspidata Bureau (Moraceae), was found to induce heme oxygenase-1 (HO-1) expression at a non-cytotoxic concentration (1–10 μmol/L) in LPS-treated RAW264.7 macrophages, which in turn suppressed PGE2, NO, TNF-?, and IL-1β production [24]. Dual arachidonate 5-lipoxygenase (5-LOX) and COX inhibitors are potential new drugs to treat inflammation. 6-Dihydroxy-5-methoxy-4′,5′-dihydro-4′,4′,5′-trimethylfurano-(2′,3′:3,4)-xanthone (38) inhibited COX-1 and COX-2 production, and 5-LOX mediated leukotriene B4 (LTB4) formation in LPS-induced RAW264.7 macrophages [29]. Among the xanthones isolated from the roots of Cratoxylum formosum ssp. pruniflorum, several compounds (6, 1518, 21, 2628, and 37) showed anti-inflammatory activity in LPS-induced RAW 264.7 macrophages [22]. Dulxisxanthone F (21) was found to downregulate the mRNA expression of iNOS and COX-2 in dose-dependent manners, and 5,9-dihydroxy-8-methoxy-2,2-dimethyl-7-(3-methylbut-2-enyl)-2H,6H-pyrano-[3,2b]-xanthone (27) only inhibited the mRNA expression of iNOS but not COX-2. Two xanthones, ellidifolin (1) and swerchirin (2), were isolated from Swertia chiraytia [20], which were found to inhibit the production of pro-inflammatory cytokines IL-6 and TNF-? in LPS-stimulated RAW264.7 macrophages; furthermore, ellidifolin (1) inhibited the production of PGE2 by suppressing the phosphorylation of JNK, ERK, and p38 MAPKs (mitogen-activated protein kinases).
Interferon γ (IFN?) is a dimerized soluble cytokine, and aberrant IFN? expression is related to a number of inflammatory and autoimmune diseases [48]. LPS plus IFN? stimulation caused the increase of TNF receptor associated factor family member-associated NF-κB activator binding kinase 1 (TBK1) expression, p50/p65 nuclear translocation, and activation of NF-κB in RAW264.7 macrophages, 1,3,5,7-tetrahydroxy-8-prenylxanthone (20), reversed the above changes to suppress the production of IL-6, IL-12, and TNF-? [32]. In IFN? plus LPS-induced RAW264.7 macrophages, hyperxanthone E (39) was reported to decrease NO production [32].
The major role of neutrophils in the host defense is to eliminate invading microorganisms [49]. In neutrophils, N-formylmethionyl-leucyl-phenylalanine (fMLP) is a powerful activator of polymorphonuclear and mononuclear phagocytes, and the effects of fMLP on neutrophil activity can be inhibited by pertussis toxin [50]. The neutrophil-mediated inflammatory response is regarded as a multi-step process involving the initial adhesion of circulating neutrophils to activated vascular endothelium [51]. In fMLP/CB-stimulated human neutrophils, several gambogic acid analogs (23, 24, 29, 31, 32, 34, and 35) inhibited superoxide anion generation and elastase release [34]. Several xanthons (3, 9, 42, and 43) were isolated from the twigs of Hypericum oblongifolium wall, which showed anti-inflammatory activity in isolated human neutrophils [33].
CD3 synovial cells are suggested to play an important role in RA development and therefore are a perfect model in the search for new anti-arthritic drugs. Mangiferin (7) downregulated TNF-α, IL-1β, and IFN-γ expression in TNF-α-stimulated CD3 synovial cells from rheumatoid arthritis (RA) patients, which indicated that mangiferin could be a potent candidate for the treatment of RA [35].
Sepsis is a major cause of death worldwide [52]. Infection-induced inflammation is strongly regulated by many endogenous negative feedback mechanisms that modulate the intensity of inflammation, promote its eventual resolution, and return it back to homeostasis. Mangiferin (7) dose-dependently upregulated the expression and activity of HO-1 in the lung from septic mice [36].
Carrageenan is a pro-inflammatory agent used as a tool to induce inflammatory hyperalgesia in rats and mice [53]. The carrageenan-induced peripheral inflammatory pain model is widely used because it resembles inflammatory pain susceptible to both steroidal and nonsteroidal anti-inflammatory drugs [54]. Local administration of mangiferin (7) prevented inflammatory mechanical hyperalgesia induced by carrageenan in rats, which depended on the inhibition of TNF-α production/release and the CINC1 (cytokine-induced neutrophil chemoattractant 1)/epinephrine/PKA (protein kinase A) pathway [37].
MC3T3 is an osteoblast precursor cell line derived from Mus musculus (mouse), which is one of the most convenient and physiologically relevant systems for the study of transcriptional control in calvarial osteoblasts [55]. Dexamethasone is a known synthetic glucocorticoid, which induces sodium-dependent vitamin C transporter in MC3T3-E1 cells [56]. Bone morphogenetic protein 2 (BMP2) plays a role in postnatal bone formation, mediated by activating ligand-bound Small Mothers Against Decapentaplegic (SMAD) family members [20]. Mangiferin (7) attenuated dexamethasone-induced injury and inflammation in MC3T3-E1 cells by activating the BMP2/Smad-1 signaling pathway [38].
HFLS-RA is a human fibroblast-like synoviocyte with high proliferating ability and susceptibility. HFLS-RA cell is an excellent cellular model for studying synoviocyte physiology in relation to the development and treatment of RA [57]. α-Mangostin (10) (10 μg/mL) was found to suppress the expression and activation of key proteins in the NF-κB pathway and inhibit the nuclear translocation of p65 in HFLS-RA cells [39].
Adjuvant-induced arthritis (AA) is evaluated by paw edema, arthritis score, and hematological parameters. α-Mangostin (10) protected joints from rats suffering from AA, indicated by attenuated paw swelling, reduced inflammatory cell infiltration, decreased secretion of IL-1β and TNF-α in serum, and inhibition of NF-κB activation in synovia [39].
The presence of neuroinflammation is a common feature of dementia [58]. Reactive microgliosis, oxidative damage, and mitochondrial dysfunction are associated with the pathogenesis of all types of neurodegenerative dementia, such as Parkinson’s disease dementia (PDD), frontotemporal dementia (FTD), Alzheimer’s disease (AD), and Lewy body dementia (LBD). Peripheral LPS-induced neuroinflammation in C57bl/6J mice has been used to evaluate neuroinflammation and neurodegeneration as an adjuvant therapeutic strategy. α-Mangostin (10) reduced the levels of proinflammatory cytokine IL-6, COX-2, and 18 kDa translocator protein (TSPO) in the brain from LPS-induced neuroinflammation in C57BL/6J mice, which was considered as an adjuvant treatment in preclinical models of AD, PD, and multiple sclerosis [40].
RA is a long-term autoimmune disease in which the body’s immune system mistakenly attacks the joints; RA causes pain, stiffness, and swelling in the joints [59]. α-Mangostin (10) decreased the clinical score at both doses (10 and 40 mg/kg) and decreased the histopathological score at the high dose in collagen-induced arthritis (CIA) in DBA/1J mice [41].
Asthma is a chronic inflammatory disease of the airways characterized by reversible airway obstruction, airway hyperreactivity (AHR), and remodeling of the airways [60]. Allergic asthma is associated with excessive T helper type 2 (Th 2) cell activation and AHR [57].

3. Comparison of the Drug Likeness of Anti-Inflammatory Xanthones with Marketed Drugs

Swiss Institute of Bioinformatics provides SwissADME to calculate molecular descriptors of the identified anti-inflammatory xanthones [61]. For each compound, the following descriptors were calculated: Molecular weight (MW); number of stereogenic centers; number of hydrogen bond acceptors (HBA) and donors (HBD), described as the electrostatic bond between a hydrogen and a lone pair of electrons; number of rotatable bonds (RB); number of rings; fraction of sp3 carbons (Fsp3) defined as the ratio of sp3 hybridized carbons over the total number of carbons; and fraction of aromatic heavy atoms (Far), defined as the number of aromatic heavy atoms divided by the total number of heavy atoms [62] (Figure 3).
Figure 3. Mean values of MW (A), stereogenic centers (B), Log P (C), HBA (D), HBD (E), PSA (F), and rotatable bond (G) for anti-inflammatory xanthone derivatives (red), natural products (black), natural derivatives (light grey), synthetic (dark blue), assumed synthetic (dark grey), natural product type macrocyte (light blue), and natural products polycyclic (yellow).

4. Compliance of Xanthones with the Rules of Drug Likeness

In order to quickly eliminate lead candidates that have poor physicochemical properties for oral bioavailability, the five rules of drug likeness have been widely adopted in the pharmaceutical industry, which helps to predict the in vivo behavior of potential drugs [63]. The biophysicochemical properties and molecular descriptors of the anti-inflammatory xanthone derivatives were framed as different rules of compliance. Most anti-inflammation xanthone derivatives appear to have a good drug likeness.

References

  1. Artis, D.; Spits, H. The biology of innate lymphoid cells. Nature 2015, 517, 293–301.
  2. Rock, K.L.; Lai, J.J.; Kono, H. Innate and adaptive immune responses to cell death. Immunol. Rev. 2011, 243, 191–205.
  3. Pedraza-Alva, G.; Pérez-Martínez, L.; Valdez-Hernández, L.; Meza-Sosa, K.F.; Ando-Kuri, M. Negative regulation of the inflammasome: Keeping inflammation under control. Immunol. Rev. 2015, 265, 231–257.
  4. Wentworth, J.M.; Naselli, G.; Brown, W.A.; Doyle, L.; Phipson, B.; Smyth, G.K.; Wabitsch, M.; O’Brien, P.E.; Harrison, L.C. Pro-inflammatory CD11c+ CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes 2010, 59, 1648–1656.
  5. Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 2007, 117, 175–184.
  6. Vegeto, E.; Benedusi, V.; Maggi, A. Estrogen anti-inflammatory activity in brain: A therapeutic opportunity for menopause and neurodegenerative diseases. Front. Neuroendocrin. 2008, 29, 507–519.
  7. Navab, M.; Gharavi, N.; Watson, A.D. Inflammation and metabolic disorders. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 459–464.
  8. Bally, M.; Dendukuri, N.; Rich, B.; Nadeau, L.; Helin-Salmivaara, A.; Garbe, E.; Brophy, J.M. Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data. Brit. Med. J. 2017, 357, j1909.
  9. Sriuttha, P.; Sirichanchuen, B.; Permsuwan, U. Hepatotoxicity of nonsteroidal anti-inflammatory drugs: A systematic review of randomized controlled trials. Int. J. Hepatol. 2018, 2018, 5253623.
  10. Pal, S.; Pal, P.B.; Das, J.; Sil, P.C. Involvement of both intrinsic and extrinsic pathways in hepatoprotection of arjunolic acid against cadmium induced acute damage in vitro. Toxicology 2011, 283, 129–139.
  11. Manna, P.; Das, J.; Ghosh, J.; Sil, P.C. Contribution of type 1 diabetes to rat liver dysfunction and cellular damage via activation of NOS, PARP, IκBα/NF-κB, MAPKs, and mitochondria-dependent pathways: Prophylactic role of arjunolic acid. Free Radic. Biol. Med. 2010, 48, 1465–1484.
  12. Masters, K.S.; Brase, S. Xanthones from fungi, lichens, and bacteria: The natural products and their synthesis. Chem. Rev. 2012, 112, 3717–3776.
  13. Cardona, M.L.; Fernández, I.; Pedro, J.R.; Serrano, A. Xanthones from Hypericum reflexum. Phytochemistry 1990, 29, 3003–3006.
  14. Peres, V.; Nagem, T.J. Trioxygenated naturally occurring xanthones. Phytochemistry 1997, 44, 191–214.
  15. Pant, N.; Jain, D.; Bhakuni, R. Phytochemicals from genus Swertia and their biological activities. Ind. J. Chem. 2000, 39, 565–586.
  16. Jantan, I.; Saputri, F.C. Benzophenones and xanthones from Garcinia cantleyana var. cantleyana and their inhibitory activities on human low-density lipoprotein oxidation and platelet aggregation. Phytochemistry 2012, 80, 58–63.
  17. Chin, Y.-W.; Jung, H.-A.; Chai, H.; Keller, W.J.; Kinghorn, A.D. Xanthones with quinone reductase-inducing activity from the fruits of Garcinia mangostana (Mangosteen). Phytochemistry 2008, 69, 754–758.
  18. Louh, G.N.; Lannang, A.M.; Mbazoa, C.D.; Tangmouo, J.G.; Komguem, J.; Castilho, P.; Ngninzeko, F.N.; Qamar, N.; Lontsi, D.; Choudhary, M.I. Polyanxanthone A, B and C, three xanthones from the wood trunk of Garcinia polyantha Oliv. Phytochemistry 2008, 69, 1013–1017.
  19. Chhetri, D.; Parajuli, P.; Subba, G. Antidiabetic plants used by Sikkim and Darjeeling Himalayan tribes, India. J. Ethnopharmacol. 2005, 99, 199–202.
  20. Langenfeld, E.; Kong, Y.; Langenfeld, J. Bone morphogenetic protein 2 stimulation of tumor growth involves the activation of Smad-1/5. Oncogene 2006, 25, 685.
  21. Chang, T.; Neelakandan, C.; Define, L.; Alexander, T.; Kyu, T. Effects of glucose on cell viability and antioxidant and anti-inflammatory properties of phytochemicals and phytochemically modified membranes. J. Phys. Chem. B 2014, 118, 11993–12001.
  22. Boonnak, N.; Chantrapromma, S.; Tewtrakul, S.; Sudsai, T. Inhibition of nitric oxide production in lipopolysaccharide-activated RAW264. 7 macrophages by isolated xanthones from the roots of Cratoxylum formosum ssp. pruniflorum. Arch. Pharm. Res. 2014, 37, 1329–1335.
  23. Jang, J.-H.; Lee, K.-H.; Jung, H.-K.; Sim, M.-O.; Kim, T.-M.; Woo, K.-W.; An, B.-K.; Cho, J.-H.; Cho, H.-W. Anti-inflammatory effects of 6′-O-acetyl mangiferin from Iris rossii Baker via NF-κb signal blocking in lipopolysaccharide-stimulated RAW 264.7 cells. Chem.-Biol. Interact. 2016, 257, 54–60.
  24. Jeong, G.; Lee, D. Yc Cudratricusxanthone A from Cudrania tricuspidata suppresses pro-inflammatory mediators through expression of anti-inflammatory heme oxygenase-1 in RAW264.7 macrophages. Int. Immunopharmacol. 2009, 9, 241–246.
  25. Li, D.; Liu, Q.; Sun, W.; Chen, X.; Wang, Y.; Sun, Y.; Lin, L. 1, 3, 6, 7-Tetrahydroxy-8-prenylxanthone ameliorates inflammatory responses resulting from the paracrine interaction of adipocytes and macrophages. Br. J. Pharmacol. 2018, 175, 1590–1606.
  26. Karunakaran, T.; Ee, G.C.L.; Ismail, I.S.; Mohd Nor, S.M.; Zamakshshari, N.H. Acetyl-and O-alkyl-derivatives of β-mangostin from Garcinia mangostana and their anti-inflammatory activities. Nat. Prod. Res. 2018, 32, 1390–1394.
  27. Cho, B.O.; Ryu, H.W.; So, Y.; Chang, W.L.; Chang, H.J.; Hong, S.Y.; Yong, W.J.; Park, J.C.; Jeong, I.Y. Anti-Inflammatory effect of mangostenone F in lipopolysaccharide-stimulated RAW264.7 macrophages by suppressing NF-κB and MAPK activation. Biomol. Ther. 2014, 22, 288–294.
  28. Ee, G.C.L.; Mah, S.H.; Rahmani, M.; Yun, T.Y.; Teh, S.S.; Yang, M.L. A new furanoxanthone from the stem bark of Calophyllum inophyllum. Lett. Org. Chem. 2011, 13, 956–960.
  29. Crockett, S.L.; Poller, B.; Tabanca, N.; Pferschywenzig, E.M.; Kunert, O.; Wedge, D.E.; Bucar, F. Bioactive xanthones from the roots of Hypericum perforatum (common St John’s wort). J. Sci. Food Agric. 2011, 91, 428–434.
  30. Liu, Q.; Li, D.; Wang, A.; Dong, Z.; Yin, S.; Zhang, Q.; Ye, Y.; Li, L.; Lin, L. Nitric oxide inhibitory xanthones from the pericarps of Garcinia mangostana. Phytochemistry 2016, 131, 115–123.
  31. Zhang, D.D.; Hong, Z.; Lao, Y.Z.; Rong, W.; Xu, J.W.; Ferid, M.; Bian, K.; Xu, H.X. Anti-inflammatory effect of 1,3,5,7-tetrahydroxy-8-isoprenylxanthone isolated from twigs of Garcinia esculentaon stimulated macrophage. Mediat. Inflamm. 2015, 2015, 11–24.
  32. Zhang, H.; Zhang, D.D.; Lao, Y.Z.; Fu, W.W.; Liang, S.; Yuan, Q.H.; Yang, L.; Xu, H.X. Cytotoxic and anti-inflammatory prenylated benzoylphloroglucinols and xanthones from the twigs of Garcinia esculenta. J. Nat. Prod. 2014, 77, 2148–2149.
  33. Ali, M.; Arfan, M.; Ahmad, M.; Singh, K.; Anis, I.; Ahmad, H.; Choudhary, M.I.; Shah, M.R. Anti-inflammatory xanthones from the twigs of Hypericum oblongifolium wall. Planta Med. 2011, 77, 2013–2018.
  34. Yen, C.T.; Nakagawagoto, K.; Hwang, T.; Morrisnatschke, S.L.; Bastow, K.F.; Wu, Y.C.; Lee, K.H. Design and synthesis of gambogic acid analogs as potent cytotoxic and anti-inflammatory agents. Bioorg. Med. Chem. Lett. 2012, 22, 4018–4022.
  35. Kokotkiewicz, A.; Luczkiewicz, M.; Pawlowska, J.; Luczkiewicz, P.; Sowinski, P.; Witkowski, J.M.; Bryl, E.; Bucinski, A. Isolation of xanthone and benzophenone derivatives from Cyclopia genistoides (L.) Vent. (honeybush) and their pro-apoptotic activity on synoviocytes from patients with rheumatoid arthritis. Fitoterapia 2013, 90, 199–208.
  36. Gong, X.; Zhang, L.; Jiang, R.; Ye, M.; Yin, X.; Wan, J. Anti-inflammatory effects of mangiferin on sepsis-induced lung injury in mice via up-regulation of heme oxygenase-1. J. Nutr. Biochem. 2013, 24, 1173–1181.
  37. Rocha, L.W.; Bonet, I.J.M.; Tambeli, C.H.; De-Faria, F.M.; Parada, C.A. Local administration of mangiferin prevents experimental inflammatory mechanical hyperalgesia through CINC-1/epinephrine/PKA pathway and TNF-α inhibition. Eur. J. Pharmacol. 2018, 830, 87–94.
  38. Ding, L.Z.; Teng, X.; Zhang, Z.B.; Zheng, C.J.; Chen, S.H. Mangiferin inhibits apoptosis and oxidative stress via BMP2/Smad-1 signaling in dexamethasone-induced MC3T3-E1 cells. Int. J. Mol. Med. 2018, 41, 2517–2526.
  39. Zuo, J.; Yin, Q.; Wang, Y.W.; Li, Y.; Lu, L.M.; Xiao, Z.G.; Wang, G.D.; Luan, J.J. Inhibition of NF-κB pathway in fibroblast-like synoviocytes by α-mangostin implicated in protective effects on joints in rats suffering from adjuvant-induced arthritis. Int. Immunopharmacol. 2018, 56, 78–89.
  40. Nava, C.M.; Acero, G.; Pedraza-Chaverri, J.; Fragoso, G.; Govezensky, T.; Gevorkian, G. Alpha-mangostin attenuates brain inflammation induced by peripheral lipopolysaccharide administration in C57BL/6J mice. J. Neuroimmunol. 2016, 297, 20–27.
  41. Herrera-Aco, D.R.; Medina-Campos, O.N.; Pedraza-Chaverri, J.; Sciutto-Conde, E.; Rosas-Salgado, G.; Fragoso-González, G. Alpha-mangostin: Anti-inflammatory and antioxidant effects on established collagen-induced arthritis in DBA/1J mice. Food Chem. Toxicol. 2019, 124, 300–315.
  42. Jang, H.Y.; Kwon, O.K.; Oh, S.R.; Lee, H.K.; Ahnab, K.S. Mangosteen xanthones mitigate ovalbumin-induced airway inflammation in a mouse model of asthma. Food Chem. Toxicol. 2012, 50, 4042–4051.
  43. Shen, Q.; Chitchumroonchokchai, C.; Thomas, J.L.; Gushchina, L.V.; Disilvestro, D.; Failla, M.L.; Ziouzenkova, O. Adipocyte reporter assays: Application for identification of anti-inflammatory and antioxidant properties of mangostin xanthones. Mol. Nutr. Food Res. 2014, 58, 239–247.
  44. Moreira, M.E.; Pereira, R.G.; Dias Silva, M.J.; Dias, D.F.; Gontijo, V.S.; Giustipaiva, A.; Veloso, M.P.; Doriguetto, A.C.; Nagem, T.J.; dos Santos, M.H. Analgesic and anti-inflammatory activities of the 2,8-dihydroxy-1,6-dimethoxyxanthone from Haploclathra paniculata (Mart) Benth (Guttiferae). J. Med. Food 2014, 17, 686–693.
  45. Panthong, K.; Hutadilok-Towatana, N.; Panthong, A. Cowaxanthone F, a new tetraoxygenated xanthone, and other antiinflammatory and antioxidant compounds from Garcinia cowa. Cheminform 2010, 41, 281–286.
  46. ujiwara, N.; Kobayashi, K. Macrophages in inflammation. Curr. Drug Targets Inflamm. Allergy 2005, 4, 281–286.
  47. Poltorak, A.; He, X.; Smirnova, I.; Liu, M.-Y.; Van Huffel, C.; Du, X.; Birdwell, D.; Alejos, E.; Silva, M.; Galanos, C. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 1998, 282, 2085–2088.
  48. Gray, P.W.; Goeddel, D.V. Structure of the human immune interferon gene. Nature 1982, 298, 859–863.
  49. Witko-Sarsat, V.; Rieu, P.; Descamps-Latscha, B.; Lesavre, P.; Halbwachs-Mecarelli, L. Neutrophils: molecules, functions and pathophysiological aspects. Lab. Invest. 2000, 80, 617–624.
  50. Panaro, M.; Mitolo, V. Cellular responses to FMLP challenging: A mini-review. Immunopharmacol. Immunotoxicol. 1999, 21, 397–419.
  51. Faurschou, M.; Borregaard, N. Neutrophil granules and secretory vesicles in inflammation. Microbes Infect. 2003, 5, 1317–1327.
  52. Lever, A.; Mackenzie, I. Sepsis: definition, epidemiology, and diagnosis. Brit. Med. J. 2007, 335, 879–883.
  53. Levy, L. Carrageenan paw edema in the mouse. Life Sci. 1969, 8, 601–606.
  54. Sammons, M.J.; Raval, P.; Davey, P.T.; Rogers, D.; Parsons, A.A.; Bingham, S. Carrageenan-induced thermal hyperalgesia in the mouse: role of nerve growth factor and the mitogen-activated protein kinase pathway. Brain Res. 2000, 876, 48–54.
  55. Kodama, H.-a.; Amagai, Y.; Sudo, H.; Kasai, S.; Yamamoto, S. Establishment of a clonal osteogenic cell line from newborn mouse calvaria. Japanese J. Oral Biol. 1981, 23, 899–901.
  56. Fujita, I.; Hirano, J.; Itoh, N.; Nakanishi, T.; Tanaka, K. Dexamethasone induces sodium-dependant vitamin C transporter in a mouse osteoblastic cell line MC3T3-E1. Br. J. Nutr. 2001, 86, 145–149.
  57. Liu, F.-L.; Chen, C.-H.; Chu, S.-J.; Chen, J.-H.; Lai, J.-H.; Sytwu, H.-K.; Chang, D.-M. Interleukin (IL)-23 p19 expression induced by IL-1 β in human fibroblast-like synoviocytes with rheumatoid arthritis via active nuclear factor-κ B and AP-1 dependent pathway. Rheumatology 2007, 46, 1266–1273.
  58. Pasqualetti, G.; Brooks, D.J.; Edison, P. The role of neuroinflammation in dementias. Curr. Neurol. Neurosci. Rep. 2015, 15, 17–24.
  59. Firestein, G.S. Evolving concepts of rheumatoid arthritis. Nature 2003, 423, 356–361.
  60. Choi, M.S.; Park, S.; Choi, T.; Lee, G.; Haam, K.-K.; Hong, M.-C.; Min, B.-I.; Bae, H. Bee venom ameliorates ovalbumin induced allergic asthma via modulating CD4+ CD25+ regulatory T cells in mice. Cytokine 2013, 61, 256–265.
  61. Loureiro, D.R.; Soares, J.X.; Costa, J.C.; Magalhães, Á.F.; Azevedo, C.M.; Pinto, M.M.; Afonso, C.M. Structures, activities and drug-likeness of anti-infective xanthone derivatives isolated from the marine environment: A review. Molecules 2019, 24, 243.
  62. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686.
  63. Veber, D.F.; Johnson, S.R.; Cheng, H.-Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615–2623.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 527
Revisions: 2 times (View History)
Update Date: 17 Nov 2021
1000/1000