THE ANTI-INFLAMMATORY ACTIVITY OF PLANT DERIVED INGREDIENTS: AN ANALYTICAL REVIEW
HTML Full TextTHE ANTI-INFLAMMATORY ACTIVITY OF PLANT DERIVED INGREDIENTS: AN ANALYTICAL REVIEW
Debjani Sarkar
Department of Zoology, A.P.C. Roy Government College, Himanchal Vihar, Matigara, Siliguri, Darjeeling - 734010, West Bengal, India.
ABSTRACT: Inflammation, a protective response, can be dangerous if left untreated. The current synthetic anti-inflammatory drugs available in the market are coupled with various side effects. Plant-derived products can serve as a better alternative being cheap, easily available and having very few side effects. A sizable number of plants having anti-inflammatory activity have been studied. The crude extract of the plant is easy to obtain but it is not possible to indicate the component responsible for a particular property of the extract as it is a mixture of various components. Therein lays the importance of isolating the active principles from the crude extract and studying their mechanism of action. The present study aspires to provide an insight into the current trend of treatment of inflammation using herbal remedies by highlighting the different active principles obtained from different parts of plants, their concentrations used in specific test systems alongside underlining their molecular mode of action.
Keywords: |
Anti-inflammatory, Plant products, Active components, Mechanism of action
INTRODUCTION: Inflammation is defined as the response shown by the body when it is invaded by an antigen or any type of damage (physical, chemical or traumatic) occurs 1. It is a naturally resolving process in which body fluid, proteins, and cells from the blood are transferred to the damaged tissues for their removal and resolution of the area. The inflammatory response is initiated by temporary vasoconstriction which is followed by acute vascular response due to increased blood flow leading to edema 2. The acute cellular response occurs over the next few hours and is characterized by infiltration of neutrophils into the damaged area.
If the damage is severe, chronic cellular response follows and is characterized by appearance of macrophages and lymphocytes. Over a few weeks, normal tissue design is restored. When the immune system is unable to restore the body to the original state because of the presence of a large amount of antigen or inaccessibility of the antigen, persistent inflammation occurs, and treatment becomes essential.
Immune Components Responsible for Inflammation: Both cells and soluble components act as mediators of inflammation. The vasoactive amines, reactive oxygen species, arachidonic acid derivatives, etc. are some of the mediators responsible for the propagation of inflammation. The cells include macrophages, eosinophils, neutrophils, etc. Some of the mediators are described below:
Arachidonic Acid Derivatives: Prostaglandin E2 (PGE2) is responsible for the pain and edema which occurs during inflammation 3. It is formed when cycloxygenase (mainly COX-2, which is inducible and plays an important role; COX-1 being constitutive) acts on arachidonic acid 4. Cycloxygenase, in turn, is activated by nitric oxide (NO), another molecule of great importance, synthesized by inducible nitric oxide synthase (iNOS) 5. The lipoxygenase pathway is also activated during inflammation to produce leukotrienes. Both the pathways activate various kinins, neuro-peptides, and histamine, as well as complement components, along with cytokines and free radicals.
Cytokines: Cytokines are proteins, secreted by white blood cells which play a critical role in inflammation by regulating inflammatory mediators usually secreted by other cells 6. Pro-inflammatory cytokines aggravate inflammation while anti-inflammatory cytokines down-regulate the same. Pro-inflammatory cytokines include IL-1β, IL-6, IL-12 and TNF-a while anti-inflammatory cytokines include IL-10, IL-4, etc. 7
Oxidative Status and Anti-oxidant Enzymes: Generation of reactive oxygen species (ROS) like superoxide, peroxynitrite, nitric oxide, etc. are greatly increased in inflammation causing damage to the cell components 8. To minimize damage, cells have evolved enzymatic (catalase, superoxide dismutase) and non-enzymatic (glutathione) defense mechanisms but are downregulated during inflammation 9.
Signaling Pathways: NF-κB is a transcription factor which plays an important role in inflammation as it controls the expression of NO, COX-2 and pro-inflammatory cytokines 10. It remains in the cytoplasm rendered inactive by IκB. When activated, IκB is phosphorylated and degraded, and NF-κB binds to DNA after entering the nucleus, allowing transcription of iNOS mRNA and various other pro-inflammatory cytokines. The activation of NF-κB occurs via the activation of mitogen-activated protein kinases (MAPKs) which include p38 MAPK (p38), extracellular signal-regulated kinases 1/2 (ERK1/2) and c-jun N-terminal kinase (JNK) 11, 12.
Current Anti-inflammatory Drugs are Associated with Side Effects: The control of inflammation includes synthetic anti-inflammatory drugs that can be given orally. They include the non-steroidal anti-inflammatory drugs (NSAIDs), steroids and disease-modifying antirheumatic drugs (DMARDs) 13. The NSAIDs results in relief of pain by inhibiting cycloxygenases but are associated with side effects like headache, giddiness, abdominal pain, gastric ulcer, abnormal liver function, etc. 13
Other than the general symptoms some drugs are associated with specific symptoms like Flurbiprofen is associated with cogwheel rigidity and Ibuprofen with agranulocytosis and aplastic anemia 14. The major steroids used for the treatment of inflammation are glucocorticoids which reduces bone erosions but cause suppression of T cells, extreme changes in mood, headache, giddiness, extreme changes in mood, seizures, etc. 15 The DMARDs slows down the disease by suppression of the immune system which in itself is a disadvantage. TNF-α blocking agents are associated with increased bacterial infections.
Alternate Anti-Inflammatory Drugs from Natural Flora: As the market available drugs pose danger to the human body 16, the search for a natural product is essential as it would be less toxic but equally effective. Research is currently concentrated in this area and herein lays the importance of India being a treasure trove of medicinal plants by virtue of its being a mega diversity country. Plant products are therefore easily available and are a cheap option in India. The properties of the medicinal plants were well documented in Ancient Indian Literature and practiced in Ayurveda and other traditional forms of treatment.
MATERIALS AND METHODS: The names of the plants having anti-inflammatory properties were identified from a myriad of books on medicinal plants and also searched in an online database by entering the name of the plant along with the word “anti-inflammatory”. The plants having anti-inflammatory properties were then separately searched in Pubmed, Google, Science Direct etc to see whether their active components were isolated and the mechanism of action studied. The information accumulated on the active ingredients and their mode of action were then represented in a table format.
RESULTS AND DISCUSSION:
Active Ingredients Obtained from Plants and Their Mechanism of Action: The plants manifesting anti-inflammatory properties are listed Table 1 along with their mechanism of action. Most of the anti-inflammatory properties of plants are ascribed to phenols derived from the secondary metabolism of plants. Phenolic compounds are grouped into flavonoids, composed of two aromatic rings linked through an oxygen heterocycle, and non-flavonoids, commonly known as phenolic acids 17. Flavonoids possess significant anti-inflammatory activity 18.
Active Ingredients: Some components namely cirsimarin (Cirsium japonicum) 19, eucalyptol (Eucalyptus sp.) 20, allylpyrocatechol (Piper betle) 21, curcumin (Curcuma longa) 22, theanine (Camellia sinensis)23 etc. are obtained from specific one or two plants in which they are present as a major component or alongside other phenols. Most of them have various therapeutic properties. Since inflammation is associated with various diseases like rheumatoid arthritis, neurodegenerative diseases, heart diseases, gastric ulcers, cancer, etc., some of these components were able to relieve the symptoms of various diseases simply by inhibiting the mediators of inflammation or by upregulating the anti-oxidant properties of the organism besides acting on specific targets of the respective diseases Table 1.
Animals/Cell Lines Used: Various strains of rat (Sprague Dawley, Wistar, etc.) and mice (ICR, C57BL/6, BALB/c, etc.) were the common choices as models of inflammation wherein inflammation was induced by lipopolysaccharide, pro-inflam-matory cytokines or other phlogistic agents. Lipopolysaccharide was used in maximum number of cases. The drug of choice is given orally in most cases and then the plasma/tissues/cells are collected for analyzing the effect of the drug. Histological studies of the affected tissues using common stains were done in many cases to prove the efficacy of the herbal drugs. For in-vitro studies, mouse peritoneal cells or cell lines were used. RAW264.7 is the general choice for study of the anti-inflammatory activity of the plant product (though other cell lines like THP-1 and U937 were also used) while more specific cell types (BV2 microglial cells, human umbilical vein endothelial cells, human lung cancer cell, etc.) were chosen for studying the anti-inflammatory effect related to a particular disease Table 1.
TABLE 1: PLANTS HAVING ANTI-INFLAMMATORY PROPERTIES AND THEIR MECHANISM OF ACTION
Plant-derived product | Part of plant | Test
system(s) |
Concentration | Molecular mechanism(s) of
action |
Caffeic acid, S-allyl cysteine, and uracil from Allium sativum | Bulb | HR-1 hairless mice | 5-20 μM | Inhibited DCFDA induced ROS production. Inhibited degradation of type1 procollagen and expression of MMP 3 and 9. Uracil inhibited COX-2 and Inos. Others inhibited NF-κB and AP-1 activity24 |
S-allyl cysteine | Wistar rats
C2C12 myotubes |
25-100 mg/kg b.w.
0.01- 0.1 mM |
Improved spatial recognition memory. Inhibited MDA and increased SOD, catalase and GSH levels. Inhibited glial fibrillary acidic protein, IL-6, IL-1β, Nox, NF-κB, IL-1 β and TLR4 activity. Upregulated Nrf2 activity25, 26 | |
Alliin | 3T3-L1 adipocytes | 100 μmol/L | Inhibited LPS induced IL-6, MCP-1, Egr-1. Inhibited phosphorylation of ERK1/2 upregulation of genes involved in immune response27 | |
Thiacremonone | ICR/(APP/ PS1) transgenic mice: attenuated cognitive impairments. BV-2 microglia cells. Cortical neuronal cells from Sprague Dawley rats | 1-10 mg/kg b.w.,
10 mg/L 1-5 μg/ml 1-5μg/ml |
Inhibited ROS and NO, iNOS, COX-2 and GFAP expression. Inhibited amyloid β, NF-κB and ERK activity. Inhibited BACE level brains and augmented peroxiredoxin 6 (PRDX6)28, 29 | |
Hirsutenone from Alnus sp. | Bark | Human keratinocytes HEK001
RAW264.7 cells |
1 – 10 μM
IC50: 14.08 μM
|
Reduced NO, IL-1 β, IL-8, CCL17 and scavenged free radicals. Reduced activation of TLR-4, NF-κB and ERK phosphorylation30, 31 |
Antcin A from Antrodia camphorata | Fruiting bodies | Human lung cancer cell A549 | 10 mol/L | Induced nuclear migration of glucocorticoid receptor. Successfully docked to the binding cavity of GR32 |
Antrocamphin A | RAW264.7 cells | 1-20 μg/ml,
20 mg/ ml |
Inhibited NO, iNOS, COX-2 and superoxide. Inhibited platelet aggregation. Inhibited NF- κB activation 33, 34 | |
Arctigenin from Arctium lappa | Seed | C57BL/6 mice peritoneal macrophages C57BL/6 mice | 5 mg/kg b.w.
0-20 μM 25-50 mg/kg b.w. |
Inhibited IL-1β, IL-6, TNF-α, MIP-2, MCP-1, MAdCAM-1, ICAM-1, and VCAM-1 increased IL-10 and CD204 expression. Decreased MDA level but increased SOD and GSH levels. Inhibited PI3K, AKT, IKKβ, NF-κB and MAP kinase activity35, 36 |
3-acetyl-11-keto-β-boswellic acid from Boswellia serrata | Gum resin | KBM-5, RAW 264.7, H1299, HEK A293, HCC4 and MDA 1986 cells, Albino rats | 10-50 μM
125-250 mg/kg |
Inhibited COX-2, IL-6, and TNF-α. Reduced TNF-α induced NF- κB. Blocked NF- κB activation and increased dopamine levels 37, 38 |
Cajaninstilbene acid from Cajanus cajan | Leaves | RAW264.7 cells
U937, Zebrafish Larvae |
25 -100 μg/ml
10-50μM 1.25-5 μM |
Activated PPARγ. Inhibited NO, iNOS, IL-6, and TNF-α. Inhibited migration of neutrophils and macrophages. Inhibit NF-κB and MAP kinase activation 39, 40 |
Epigallocatechin-3-gallate (green tea) from Camellia sinensis | Leaves | Fibroblast cells
BALB/c mice: reduced OVA-induced nasal rubbing and sneezing |
10-100 mM
25-100mg/kg b.w. |
Inhibited COX-2, TNF-α, IL-4, IL-1β,
and IL-6 41, 42 |
Theaflavin (black tea) | C57BL/6 mice
U937 and RAW264.7 cells Gingival keratinocytes: enhanced tight junction integrity |
20-40 mg/kg bw
6.25-50 mM/7.9-125 mg/ml 31.25-250 mg/ml |
Inhibited NF-κB, JNK, and p38 activity. Inhibited TNF-α, IL-1β, IL-6, chemokine (C-X-C) ligand 8, MMP-3, MMP-8 and MMP-9 43, 44 | |
Thearubigin (black tea) | Mice: inhibited 2,4,6-trinitrobenzene sulfonic acid (TBNS) induced colitis | 40-100 mg/kg b.w. | Inhibited TBNS induced NO, iNOS. Inhibited NF-κB activation 45 | |
Theanine | ICR mice CD-1 mice: reduced TPA-induced erythema, vascular permeability increase, epidermal and dermal hyperplasia, | 20- 40 mg/kg
5-250 mM |
Decreased alanine aminotransferase, aspartate aminotransferase, SOD and MDA levels. Reduced neutrophil infiltration and PECAM-1. Decreased IL-1β, TNF-α, COX-2, iNOS, MMP-3 IFN-γ and increased IL-10. Decreased NF-κB activity 23, 46 | |
Capsaicin from Capsicum sp. | Fruit | Human THP-1 cells | 10 μM | Decreased LPS induced IL-1β, TNF-α and Il-6. Increased LXRα expression through PPARγ pathway and inhibiting NF-κB activity47 |
Cirsimarin from Cirsium japonicum | Aerial part | RAW 264.7 cells
|
10-40 μM | Inhibited LPS induced NO, iNOS, COX-2, PGE2, TNF- α and IL-6. Downregulated phosphorylation of JAK and MAPK and nuclear translocation of interferon regulatory factor 3.19 |
Colchicine from Colchicum autumnale | Bulb like corms | Recurrent aphthous stomatitis (RAS) patients | 0.5 mg | Decrease in level of neutrophil-lymphocyte ratio, white cell count and red cell distribution width. No change in mean platelet volume, Hb levels 48 |
Corynoline from Corydalis bungeana | Whole herb | RAW264.7 cells
Human umbilical vein endothelial cells |
10 -100μg/ml,
1-4 μM 1-4 μM |
Inhibited LPS-induced NO, iNOS, COX-2, IL-6, IL-10, TNF-α, IL-1β, VCAM-1 and ICAM-1. Increased expression of Nrf2, quinone oxidoreductase 1 and HO-1. Inhibited NF-κB, p38 and ERK49-51 |
Cudratricuxanthone A from Cudrania tricuspidata | Root | RINm5F
C57BL/6 mice |
1- 10 µM
0.058-0.294 mg/kg b. w. |
Inhibited ROS, NO, iNOS, IL-6, TNF-α and NF-κB. Reduced phosphorylation of STAT-1, 3 and 5. Inhibited glucose-stimulated insulin secretion. Reduced cecal ligation and puncture (CLP)-induced creatinine, blood urea nitrogen (BUN), lipid peroxidation52 |
Curcumin from Curcuma longa | Rhizome | BV2 microglial cells
C57BL/6 mice RAW264.7, THP-1 cells Wistar rats |
10-50 μM
10 mg/kg 5 - 15 μM 0.15 -0.6 mg/ml |
Inhibited NO, iNOS, TNF-α, IL-6 and IL-1β. Improved ovalbumin-induced total WBC, PLA2, TP, IgE, IL-4, IFN-γ, IFN-γ/IL-4 ratio, SOD, thiol. Inhibited NF-κB, ERK, p38 pathway and p13K/AKT pathway22, 53, 54 |
Eucaluptol (1,8 cineole) from Eucalyptus sp | Leaves | Murine alveolar macrophage cell line MH-S (CRL 2019)
C57BL/6 and TRPM8 mice HEK293T cells |
0-100% vol/vol
30-300 mg/kg b.w. |
Reduced IL-1α, IL-1β, TNF-α, IFN-γ and IL-6. Downregulated TREM -1 and NLRP3 pattern recognition receptors and enhanced phagocytic activity. Inhibited JNK1/2 and ERK1/2 activity 20, 55 |
Ginkgetin from Gikngo biloba | Leaves | Mouse bone marrow-derived mast cells, ICR mice: TPA induced ear edema | 1.6- 50 µM
20—80 µg /ear/treatment |
Reduced PGD2, PGE2, COX-2 and IL-1β. Inhibited leukotriene and β-hexoseaminidase56, 57
|
Alantolactone and isoalantolactone from Inula helenium, | roots | HaCat cells | 0.6-2.4 μg/ml | Inhibited TNF-α-induced activation of NF-κB and the expression of TNF-α, IL-1, and IL-4.58 |
Scopariusols from Isodon scoparius | Aerial parts | RAW264.7 cells | IC50 value of 0.6 μmol·L-1 | Inhibited LPS induced NO production59 |
Kalopanaxsaponin A from Kalopanax pictus | Stem bark | ICR mice: inhibited weight reduction, colon shortening, inflammation and thickening
peritoneal macrophages Murine BV2 microglial cell |
10-20 mg/kg b.w.
5-10 μM 100-300 μM |
Inhibited LPS-induced iNOS, COX-2 IL-1β, TNF-α and IL-6, ROS and upregulated IL-10. Inhibited activation of IRAK-1, NF-κB, JNK, ERK and p38 and upregulated HO-1 60, 61 |
Momordin from Kochia scoparia | Fruits | RAW 264.7 cells | 7.5-30 μg/ml
5-80 μM |
Reduced NO, iNOS, COX-2, PGE2, IL-6 and TNF-α and NF-κ 62, 63 |
Thymoquinone from Nigella sativa | Seed | BV2 microglial cells
Wistar rats: Reduced neutrophil accumulation in pleural space Human neutrophils C57BL/6: Reduced EtOH–induced gastritis, RAW264.7, and human monocyte-like U937 |
10 µM
10-50 mg/kg b.w. 0.1-2.5 μg/ml 0- 25 µM
|
Inhibited NO, iNOS, COX-2, TNF-α, IL-6, IL-1β IL-12p40/70, superoxide ion, CCL12 /MCP-5, CCL2/ MCP-1, and G-CSF, Cxcl10. Inhibited IRAK-1, AP-1, PKC, MAPK, and NF-κB. Impaired the phosphorylation on Ser-304 and Ser-328 of p47(PHOX), a cytosolic subunit of the NADPH oxidase64-66 |
Ginsenoside from Panax ginseng | Roots | Human airway epithelial cells
A549 cells, Intestinal epithelial cells (H-29); RAW264.7 cells Sprague Dawley rats |
50 μM
900 nM 1-50 μM 20mg/kg b.w. |
Reduced IL-1β, TNF-α, IL-6, NO, COX-2, ROS, MDA and caspase 3. Increased GSH, p-AKT/AKT, GSK 3β, and reduced p21WAF1/CIP1 and p53 proteins. Inhibition of NF- κB activity 67-69 |
Picroliv isolated from Picrorhiza kurroa
|
Roots and rhizomes
|
KBM 5 /H1299/A293 cells
BALB/c mice |
50- 150 μg/ml.
12.5 mg/kg b.w.
|
Downregulated IL-1β, TNF-α, and NF-κB pathway. Suppressed IAP1, Bcl-2, Bcl-xL, survivin, and TRAF2, cyclin D1 and COX-2, VEGF, ICAM-1, and MMP-9. Inhibited MPO, MDA expression and increased SOD levels 70,71 |
Allylpyrocatechol from Piper betle | Leaves | Sprague Dawley rats: reduced paw edema.
RAW 264.7 cells |
10 mg/kg bw/ 20 mg/kg b.w.
2.5 and 5 mg/ml |
Inhibited NO and iNOS. Inhibited IL-12p40, TNF-a, IL-6 COX-2 and PGE2. Inhibited NF-κB
activation 21, 72 |
Platycodin D from Platycodon granduflorum
|
Roots
|
Sprague Dawley rats
C57BL/6 mice bovine mammary epithelial cells (bMEC) |
10-30 mg/kg b.w.
20-80 mg/kg.b.w. 5-20 μM |
Attenuated cigarette smoke-induced/LPS induced MDA, NO, IL-6, TNF-α, and IL-1β. Decreased serum alanine transaminase, aspartate transaminase and total bilirubin. Attenuated cigarette smoke-induced pathological changes in lung. Down-regulated MD-2 and CD14. Attenuated TLR4, MyD88 and TRAF-6, NF-κB and upregulated LXRα expression 73-75 |
Cirsitakaoside from Premna szemaoensis | Stem and leaves | C57BL/6J mice
Mouse peritoneal macrophages and RAW264.7 cells |
50mg/kg
0-75 μM |
Suppressed LPS induced activation of IL-1B, TNF-α, IL-6. Inhibited iNOS and COX-2 mRNA. Inhibited NF-KB and MAPK pathway 76 |
Punicalagin from Punica granatum | Fruit | ICR mice: prevented LPS induced memory impairment
Astrocytes and microglial BV-2 cells. Bovine endometrial epithelial cells (bEEc) |
1.5 mg/kg b.w.
1 μg/ml 5-20 μg/ml |
Inhibits LPS induced iNOS, COX-2, production of ROS, NO, TNF-α, IL-6, IL-8, and IL-1β. Suppress NF-κB, p38, JNK and ERK activity. Downregulated APP and BACE1 expression 77, 78 |
Tanshinone IIA from Salvia miltiorrhiza | Root | Human vascular endothelial cells (HUVEC)
C57B/L mice RAW264.7 cells |
0-20 μM
30-60 mg/kg b.w. 10 μM |
Attenuated IL-1β, TNF-α, COX-2, fractalkine/CX3CL1, VCAM-1, ICAM-1, GMCSF and AP-1. Reduced total cholesterol, triglyceride, LDL, VLDL, MDA. Inhibited adhesion of THP-1 monocytes. Inhibited ERK1/2 and NF-κB activity 79-81 |
Cryptotanshinone | A7r5 cells
CD-1 mice: alleviate memory decline in Aβ1-42-injected mice |
10-7 to 10-5 M
1-10 mg/kg b.w. |
Reduced expression of NO iNOS COX-2 via ERβ-dependent and NF-kB pathway 82, 83 | |
Salvianolic acid B | Wistar rats: improved cerebral ischemic injury PC-12 cells.
Sprague Dawley rats: Primary cortical neurons were obtained and LPS induced depression was reduced. C57BL/6: suppressed cartilage destruction and slowed osteoarthritis development Human OA chondrocytes |
30 mg/kg
200-800 ng/ml 20 mg/kg b.w. 25 mg/kg b.w. 25-100 μM |
Reduced chemical and oxygen-glucose deprivation and reoxygenation (OCD/R) IL-1β, IL-6, TNF-α, NO, PGE2, INOS, COX-2, MMP-13 and ADAMTS-5. Inhibited TLR4/MyD88/TRAF6 signaling ameliorated NeuN release. Induced autophagy and decreased NLRP3 inflammasome activation. Inhibited NF-κB activity 84-86 | |
Saurolactum from Saururus chinensis
|
Roots | RAW 264.7 cell | 1 – 10 μM | Inhibited LPS induced IL-1b, IL-6, TNF-α, iNOS, COX- 2, PGE2. Inhibited activation of NF-κB, phosphorylation of ERK, JNK and p38 kinase87 |
Sauchinone | Roots | Microglial BV-2 cells and cultured astrocytes. Human osteoarthritis chondrocytes | 1 – 10 μM
2.5-20 μg/ml
|
Inhibited generation of ROS, NO, iNOS, COX-2, PGE2. Inhibited MMP-3 and MMP-13 release 88, 89 Inhibited β-secretase, STAT3, and NF- κB |
Schisandrin B from Schisandra chinensis | Fruits | BV2 microglial cells. Human umbilical vein endothelial cells (HUVECs) | 12.5 -50 μM
10 -40 μM |
Inhibited LPS induced TNF-α, IL-6, IL-1β,IL-8, ICAM-1, VCAM-1, and PGE2. Downregulated NF-κB upregulated the expression of PPAR-γ, Nrf2 and HO-1 90, 91 |
Schisandrin A | RAW 264.7 cells | 50 -200 μM | Inhibited Lps induced NO and PGE2 by inhibiting iNOS and COX-2 respectively. Reduced TNF-α and IL-1β. Inhibited NF-κB, MAPK, and PI3K/Akt activity. Upregulated Nrf2 and HO-1 92 | |
Schisandrin C | Human dental pulp cells (HDPC) | 10 -20 μM | Inhibited LPS induced TNF-α, IL-1b, ICAM-1, VCAM-1.MMP-2 and -9, NO production, ROS formation, NF-κB. Upregulated SOD, HO-1, peroxisome proliferator-activated receptor-gamma coactivator 1-α, p-Akt and Nrf2 93 | |
Gomisin N | Fruits | Human periodontal ligament cells (HPDLC). C57BL/6 mice: Attenuates Ethanol-Induced Hepatic Steatosis and Liver Injury SK-HEp-1 cells
HepG2 cells |
0-50 μM
20 mg/kg b.w. 5-20 μM 50-100 μM |
Inhibited TNF-α induced IL-6, IL-8, chemokine ligand CCL 2, and CCL20. Inhibited NF-κB, ERK and JNK pathway. Decreased MDA and CYP2E1 and increased SOD, catalase, GSH, and GPX levels. Enhances sirtuin1 (SIRT1) and phosphorylated AMP-kinase (AMPK)94, 95 |
Gomisin A | C57BL/6J peritoneal macrophages | 0.01-1 ng/ml | Inhibited LPS induced COX-2, iNOS, IL-6, TNF-α and NO. Inhibited receptor-interacting protein 2 (RIP2) and NF-κB activity96 | |
Taraxasterol from Taraxacum officinale | Aerial part and root | Wistar rats: suppressed Freunds Complete Adjuvant induced paw swelling and arthritis index, human umbilical vein endothelial cells | 2-8 mg/kg b.w.
5-18μg/ml |
Inhibited FCA/LPS induced TNF-α, IL-8, IL-1β, NO, iNOS, COX, PGE2, and RANKL, Increased serum OPG production. Decreased NF-κB and increased expression of LXRα97, 98 |
Triptolide from Tripterygium wilfordii | Leaves | BALB/c mice
Sprague Dawley rats MH7A cells |
5-15 μg/kg b.w.
0.2mg/kg b.w. 0.5-2 μM |
Reduced LPS/ TNF-α induced leukocyte, myeloperoxidase activity, edema of the lung,
Inhibited TNF-α, IL-1β, and IL-6, IL-8, iNOS, COX-2 Inhibited expression of TLR4 and NF-κB, ERK and JNK activity. Attenuated brain infarction volume, death of neuronal cells, Bcl2, Bax, caspase3 99-101 |
Zingerone from Zinziber officinale | Rhizome | C57BL/6 mice
SW1353 cell line Wistar rats |
10-40 mg/kg b.w.
10-40 µM 10-50 mg/kg b.w. |
Suppressed chemically induced BUN, creatinine, lactate dehydrogenase, and reduced inflammatory cytokines TNF-α, IL-6, IL-8, and IL-1 β. Inhibited TLR4, MyD88, TRIF expression and NF-κB, p38 and JNK activation 102-104 |
Gingerol | RAW264.7 cells | 50-300 µg/ml | Inhibited LPS induced NO, PGE2, TNF-α, IL-1β, and IL-6. Inhibited iNOS and COX-2 mRNA expression. Inhibited NF-κB activation 105 | |
6-Shogaol | BV2 microglial cells | Inhibited LPS-induced TNF-α, IL-1β, IL-6, and PGE2. Inhibited NF-κB and increased PPAR-γ 106 |
ADAMTS: disintegrin and metalloproteinase with thrombospondin motifs, Akt: Protein kinase B, AMPK: AMP-activated protein kinase, AP: activator protein, APP: Amyloid precursor protein, BACE: beta-site APP Cleaving Enzyme, BUN: blood urea nitrogen, CCL: chemokine ligand, CD: cluster of differentiation. COX-2: cycloxygenase2, CLP: cecal ligation and puncture, CXCL: chemokine (C-X-C motif) ligand, CYP: Cytochrome 450, DCFDA: 2',7'-dichlorodihydrofluorescein diacetate, Egr: early growth response proteins, ERβ: Estrogen receptor β, ERK: extracellular signal regulated kinase, GFAP: Glial fibrillary acidic protein, GMCSF: granulocyte macrophage colony stimulating factor, GSH: glutathione, GSK: glycogen synthase kinase, GR: glucocorticoid receptor, HO: heam oxygenase, IAP: Inhibitors of apoptosis proteins, ICAM: intercellular adhesion molecule, IFN: interferon, IKK: IκB kinase, IL: interleukin, iNOS: inducible nitric oxide synthase, IRAK: interleukin-1 receptor-associated kinase, JNK: Jun N terminal kinase, LPS: lipopolysaccharide, LXR: Liver X receptor, MadCAM: mucosal vascular addressin cell adhesion molecule, MAPK: mitogen activated protein kinase, MCP: monocyte chemoattractant protein, MD2: lymphocyte antigen 96, MDA: malondialdehyde, MIP: macrophage inflammatory protein, MMP: matrix metalloproteinase, Myd88: Myeloid differentiation primary response 88, NeuN: neuronal nuclei, Hexaribonucleotide Binding Protein, NF- κB: Nuclear factor kappa B, NLRP: Leucine rich Repeat and Pyrin domain containing, NO: nitric oxide, NOX: NADPH oxidase, Nrf: nuclear factor (erythroid derived), OCD/R: oxygen glucose deprivation and reoxygenation, OPG: osteoprotegerin, P21Waf1: Cip1/Kip1 family of cyclin-dependent kinase inhibitors, PECAM: platelet endothelial cell adhesion molecule, PGE2: Prostaglandin E2, PI3k: phosphatidylinositol 3-kinase,PKC: protein kianse C, PPAR: Peroxisome proliferator-activated receptor, PRDX: Peroxiredoxin, RIP: receptor interacting protein, ROS: Reactive oxygen species, RANKL: Receptor activator of nuclear factor kappa-Β ligand, SIRT: Sirtuin, SOD: superoxide dismutase, STAT: signal transducer and activator of transcription, TLR: toll like receptor, TBNS: 2,4,6-trinitrobenzene sulfonic acid, TNF: tumor necrosis factor, TP: thromboxane, TPA: 12-O-tetradecanoylphorbol- 13-acetate, TRAF: Tumor necrosis factor receptor–associated factor, TREM: Triggering receptor expressed on myeloid cells, VCAM: vascular cell adhesion molecule, VEGF: Vascular endothelial growth factor.
Mechanism of Action: The anti-inflammatory property of many of the active plant components was exhibited by their ability to bring down the common mediators of inflammation like NO, iNOS and pro-inflammatory cytokines like TNF-α, IL-1β, IL-6, IL-12p40, etc. Chemokines such as RANTES, MCP-1, etc. were also inhibited by certain plant products. The other important mediators of inflammation such as cycloxygenase-2 (COX-2), prostaglandins and leukotrienes, the chosen targets of the existing anti-inflammatory drugs, were also downregulated by majority of compounds derived from plants. Reduction in ROS, lipid peroxidation along with upregulation of the enzymatic (superoxide dismutase, catalase, etc.) and non-enzymatic (glutathione, etc.) defense systems were also tested as markers of anti-inflammatory activity. Inhibition of neutrophil infiltration reduced expression of cell adhesion proteins (ICAM, VCAM, PECAM, etc.) and metallo-proteins (MMP-3, 8, 9, etc.) were also taken into account. All the above-mentioned events were mainly attributed to the downregulation of signaling pathways like NF- κB pathway by the naturally derived plant products. The MAP kinase pathway (p38, ERK, and JNK pathway) was also inhibited and Nrf2 pathway activated to bring about a remission of inflammation Table 1.
CONCLUSION: Therapies of inflammation must inhibit one or more of the pro-inflammatory mediators released from different cells without hampering the normal functions of the body. As the currently available drugs are yet to be perfected, researchers are looking into traditional and folk medicine in the pursuit of devising a more improved drug. The present study focuses on the role of traditional plants and their phytoconstituents in reducing inflammation along with their molecular mechanisms of action. Most of the plant's products exert their effect by reducing the known mediators of inflammation such as NO, TNF-α, IL-6, ROS, COX-2, and prostaglandins. These reductions were due to the inhibition of the signaling pathways like NF-κB pathway or MAP kinase pathways. This review attempts to present a collage of the different anti-inflammatory constituents from plants that would enable researchers to use this study as a foundation for their future experiments towards devising a more potent medicine for the treatment of inflammation.
ACKNOWLEDGEMENT: I hereby acknowledge my Ph.D. guide, Dept. of Pharmacology, IPGME & R, for introducing me to the field of medicinal plants with anti-inflammatory properties. I also acknowledge Officer in charge, Acharya Prafulla Chandra Roy Govt. College, and my departmental colleagues for giving me support to carry on my work.
CONFLICTS OF INTEREST: There are no conflicts of interest.
REFERENCES:
- Goldsby RA, Kindt TJ, Osborne BA and Kuby J: Leukocyte migration and inflammation, immunology. WH Freeman and Company, New York, Edition 5th, 2003: 338-60.
- Roitt I, Brostoff, J and Male D: Cell migration and Inflammation, Immunology. Gower Medical Publications, New York, Edition 6th, 2001: 47-64.
- Ricciotti E and FitzGerald GA: Prostaglandins and inflammation. Arteriosclerosis, Thrombosis and Vascular Biology 2011; 31: 986-00.
- Wang MT, Honn KV and Nie D: Cycloxygenases, prostanoids and tumor progression. Cancer Metastasis Reviews 2007; 26: 525-34.
- Andrew PJ and Mayer B: Enzymatic function of nitric oxide synthases. Cardiovascular Research 1999; 43: 521-31.
- Dinarello CA: Historical insights into cytokines. European Journal of Immunology 2007; 37: 34-45.
- Feghali CA and Wright TM: Cytokines in acute and chronic inflammation. Frontiers in Biosci 1997; 2: 1-26.
- Lugrin J, Rosenblatt-Velin N, Parapanov R and Liaudet L: The role of oxidative stress during inflammatory processes. Biological Chemistry 2014; 395: 203-30.
- Pisochi AM and Pop A: The role of antioxidants in the chemistry of oxidative stress: A review. European Journal of Medicinal Chemistry 2015; 97: 55-74.
- Lawrence T: The Nuclear factor NF-kappa B in inflammation. Cold Spring Harbor Perspectives in Biology 2009; 1: 1-10.
- Guha, M and Mackman N: LPS induction of gene expression in human monocytes. Cellular Signalling 2001; 13: 85-94.
- Garrington TP and Johnson L: Organization and regulation of mitogen-activated protein kinase signaling pathways. Current Opinion in Cell Biology 1999; 11: 211-8.
- Payan DG and Katzung BG: Non-steroidal anti-inflammatory drugs; Non-opoid Analgesics; Drugs used in Gout. Basic and Clinical Pharmacology, Prentice-Hall International, London, Edition 6th, 1995: 536-59.
- Gryfe CI: Letter: Agranulocytosis and aplastic anemia possibly due to ibuprofen. Canadian Medical Association Journal 1976; 114: 877.
- Oray M, Abu Samra K, Ebrahimiadib N, Meese H and Foster CS: Long-term side effects of glucocorticoids. Expert Opinion on Drug Safety 2016; 15: 457-65.
- Mckellar G, Madhok R and Singh, G: The problem with NSAIDs: What data to believe? Current Pain and Headache Reports 2007; 11: 423-27.
- Bravo L: Polyphenols: Chemistry, dietary sources, metabolism and nutritional significance. Nutrition Reviews 1998; 56: 317-33.
- Handa SS, Chawla AS and Sharma AK: Plants with anti-inflammatory activity. Fitoterapia 1992; LXIII: 3-31.
- Han HS, Shin JS, Lee SB, Park JC and Lee KT: Cirsimarin, a flavone glucoside from the aerial part of Cirsium japonicum ussuriense (Regel) Kitam. ex Ohwi, suppresses the JAK/STAT and IRF-3 signaling pathway in LPS-stimulated RAW 264.7 macrophages. Chemico- Biological Interactions 2018; 293: 38-47.
- Yadav N and Chandra H: Suppression of inflammatory and infection responses in lung macrophages by eucalyptus oil and its constituent 1,8-cineole: Role of pattern recognition receptors TREM-1 and NLRP3, the MAP kinase regulator MKP-1, and NFκB. PLoS One 2017; 12: e0188232.
- Sarkar D, Saha P, Gamre S, Bhattacharjee S, Hariharan C, Ganguly S, Sen R, Mandal G, Chattopadhyay S, Majumdar S and Chatterjee M: Anti-inflammatory effect of allylpyrocatechol in LPS-induced macrophages is mediated by suppression of iNOS and COX-2 via the NF-kappaB pathway. International Immunopharmacology 2008; 8: 1264-71.
- Cianciulli A, Calvello R, Porro C, Trotta, T, Salvatore R and Panaro MA: PI3k/Akt signalling pathway plays a crucial role in the anti-inflammatory effects of curcumin in LPS-activated microglia. Int Immunopharmacology 2016; 36: 282-90.
- Wang D, Gao Q, Zhao G, Kan Z, Wang X, Wang H, Huang J, Wang T, Qian F, Ho CT and Wang Y: Protective effect and mechanism of Theanine on lipopolysaccharide-induced inflammation and acute liver injury in mice. Journal of Agricultural and Food Chemistry 2018; 66: 7674-83.
- Kim SR, Jung YR, An HJ, Kim DH, Jang EJ, Choi YJ, Moon KM, Park MH, Park CH, Chung KW, Bae HR, Choi YW, Kim ND and Chung HY: Anti-wrinkle and anti-inflammatory effects of active garlic components and the inhibition of MMPs via NF-κB signaling. PLoS One 2013; 8: e73877.
- Zarezadeh M, Baluchnejadmojarad T, Kiasalari Z, Afshin-Majd S and Roghani M: Garlic active constituent s-allyl cysteine protects against lipopolysaccharide-induced cognitive deficits in the rat: Possible involved mechanisms. European Journal of Pharmacology 2017; 795: 13-21.
- Dutt V, Saini V, Gupta P, Kaur N, Bala M, Gujar R, Grewal A, Gupta S, Dua A and Mittal A: S-allyl cysteine inhibits TNFα-induced skeletal muscle wasting through suppressing proteolysis and expression of inflammatory molecules. Biochimica et Biophysica Acta General Subjects 2018; 1862: 895-06.
- Quintero-Fabián S, Ortuño-Sahagún, D, Vázquez-Carrera M and López-Roa RI: Alliin, a garlic (Allium sativum) compound, prevents LPS-induced inflammation in 3T3-L1 adipocytes. Mediators of Inflammation 2013; 381815.
- Lin GH, Lee YJ, Choi DY, Han SB, Jung JK, Hwang BY, Moon DC, Kim Y, Lee MK, Oh KW, Jeong HS, Leem JY, Shin, HK, Lee JH and Hong JT: Anti-amyloidogenic effect of thiacremonone through anti-inflammation in-vitro and in-vivo Journal of Alzheimers Disease 2012; 29: 659-76.
- Yun HM, Jin P, Park KR, Hwang J, Jeong HS, Kim EC, Jung JK, Oh KW, Hwang BY, Han SB and Hong JT: Thiacremonone potentiates anti-oxidant effects to improve memory dysfunction in an APP/PS1 transgenic mice model. Molecular Neurobiology 2016; 53: 2409-20.
- Lee CS, Jang ER, Kim YJ, Lee MS, Seo SJ and Lee MW: Hirusutenone inhibits lipopolysaccharide activated NF-κB induced inflammatory mediator production by suppressing Toll-like receptor 4 and ERK activation. International Immunopharmacology 2010; 10: 520-5.
- Yin J, Yoon SH, Ahn HS and Lee M.W: Inhibitory activity of allergic contact dermatitis and atopic dermatitis-like skin in BALB/c mouse through oral administration of fermented barks of Alnus sibirica. Molecules 2018; 23: E450.
- Chen YC, Liu YL, Li FY, Chang CI, Wang SY, Lee KY, Li SL, Chen YP, Jinn TR and Tzen JT: Antcin A, a steroid-like compound from Antrodia camphorata, exerts anti-inflammatory effect via mimicking glucocorticoids. Acta Pharmacologica Sinica 2011; 32: 904-11.
- Hsieh YH, Chu FH, Wang YS, Chien SC, Chang ST, Shaw JF, Chen CY, Hsiao WW, Kuo YH and Wang SY: Antrocamphin A, an anti-inflammatory principal from the fruiting body of Taiwanofungus camphoratus, and its mechanisms. Journal of Agricultural and Food Chemistry 2010; 58: 3153-8.
- Lee CL, Huang CH, Wang HC, Chuang DW, Wu MJ, Wang SY, Hwang TL, Wu CC, Chen YL, Chang FR and Wu YC: First total synthesis of antrocamphin A and its analogs as anti-inflammatory and anti-platelet aggregation agents. Organic and Biomolecular Chemi 2011; 9: 70-3.
- Hyam SR, Lee IA, Gu W, Kim KA, Jeong JJ, Jang SE, Han MJ and Kim DH: Arctigenin ameliorates inflammation in-vitro and in-vivo by inhibiting the PI3K/AKT pathway and polarizing M1 macrophages to M2-like macrophages. European Journal of Pharmacology 2013; 708: 21-9.
- Wu X, Yang Y, Dou Y, Ye J, Bian D, Wei Z, Tong B, Kong L, Xia Y and Dai Y: Arctigenin but not arctiin acts as the major effective constituent of Arctium lappa fruit for attenuating colonic inflammatory response induced by dextran sulfate sodium in mice. International Immunopharmacology 2014; 23: 505-15.
- Takada Y, Ichikawa H, Badmaev V and Aggarwal BB: Acetyl-11-keto-beta-boswellic acid potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis by suppressing NF-κB and NF-κB regulated gene expression. Journal of Immunology 2006; 176: 3127-40.
- Ameen AM, Elkazaz AY, Mohammad HMF and Barakat BM: Anti-inflammatory and neuroprotective activity of boswellic acids in rotenone parkinsonian rats. Canadian J of Physiology and Pharmacology 2017; 95: 819-29.
- Schuster R, Holzer W, Doerfler H, Weckwerth W, Viernstein H, Okonogi S and Mueller M: Cajanus cajan- a source of PPARγ activators leading to anti-inflammatory and cytotoxic effects. Food and Function 2016; 7: 3798-06.
- Huang MY, Lin J, Lu K, Xu HG, Geng ZZ, Sun PH and Chen WM: Anti-inflammatory effects of cajaninstilbene acid and its derivatives. Journal of Agricultural and Food Chemistry 2016; 64: 2893-00.
- Wu YR, Choi HJ, Kang YG, Kim JK and Shin JW: In vitro study on anti-inflammatory effects of epigallocatechin-3-gallate-loaded nano- and microscale particles. International Journal of Nanomedicine 2017; 12: 7007-13.
- Fu M, Fu S, Ni S, Zou L, Liu Y and Hong T: Anti-inflammatory effect of epigallocatechin gallate in a mouse model of ovalbumin-induced allergic rhinitis. International Immunopharmacology 2017; 49: 102-8.
- Wu Y, Jin F, Wang Y, Li F, Wang L, Wang Q, Ren Z and Wang Y: In-vitro and in-vivo anti-inflammatory effects of theaflavin-3, 3'-digallate on lipopolysaccharide-induced inflammation. European Journal of Pharmacology 2017; 794: 52-60.
- Ben Lagha A and Grenier D: Black tea theaflavins attenuate Porphyromonas gingivalis virulence properties, modulate gingival keratinocyte tight junction integrity and exert anti-inflammatory activity. Journal of Periodontal Research 2017; 52: 458-70.
- Maity S, Ukil A, Karmakar S, Datta N, Chaudhuri T, Vedasiromoni JR, Ganguly DK and Das PK: Thearubigin, the major polyphenol of black tea, ameliorates mucosal injury in trinitrobenzene sulfonic acid-induced colitis. European Journal of Pharmacology 2003; 470: 103-12.
- Zeng WJ, Tan Z, Lai XF, Xu YN, Mai CL, Zhang J, Lin ZJ, Liu XG, Sun SL and Zhou LJ: Topical delivery of l-theanine ameliorates TPA-induced acute skin inflammation via downregulating endothelial PECAM-1 and neutrophil infiltration and activation. Chemico- Biological Interactions 2018; 284: 69-79.
- Tang J, Luo K, Li Y, Chen Q, Tang D, Wang D and Xiao J: Capsaicin attenuates LPS-induced inflammatory cytokine production by upregulation of LXRα. International Immunopharmacology 2015; 28: 264-9.
- Seçkin HY, Bütün I, Baş Y, Takcı Z and Kalkan G: Effects of colchicine treatment on mean platelet volume and the inflammatory markers in recurrent aphthous stomatitis. The Journal of Dermatological Treatment 2016; 27: 389-91.
- Dong ZB, Zhang YH, Zhao BJ, Li C, Tian G, Niu B, Qi H, Feng L and Shao JG: Screening for anti-inflammatory components from Corydalis bungeana based on macrophage binding combined with HPLC. BMC Complementary and Alternative Medicine 2015; 15: 363.
- Yang C, Zhang C, Wang Z, Tang Z, Kuang H and Kong AN: Corynoline isolated from Corydalis bungeana exhibits anti-inflammatory effects via modulation of Nfr2 and MAPKs. Molecules 2016; 21: 975.
- Liu B, Su K, Wang J, Wang J, Xin Z, Li F and Fu Y: Corynoline exhibits anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells through activating Nrf2. Inflammation 2018; 41: 1640-7.
- Lee DS and Jeong GS: Cudratricusxanthone A protects pancreatic beta cells from cytokines-mediated toxicity through the inhibition of NF-κB and STAT pathways. International Immunopharmacology 2014; 21: 26-33.
- Ma F, Liu F, Ding L, You M, Yue H, Zhou Y and Hou Y: Anti-inflammatory effects of curcumin are associated with down-regulating microRNA-155 in LPS-treated macrophages and mice. Pharmaceutical Biology 2017; 55: 1263-73.
- Shakeri F and Boskabady MH: Anti-inflammatory, antioxidant, and immunomodulatory effects of curcumin in ovalbumin-sensitized rat. Biofactors 2017; 43: 567-76.
- Caceres AI, Liu B, Jabba SV, Achanta S, Morris JB and Jordt SE: Transient receptor potential cation channel subfamily M Member 8 channels mediate the anti-inflammatory effects of eucalyptol. British Journal of Pharmacology 2017; 174: 867-79.
- Son JK, Son MJ, Lee E, Moon TC, Son KH, Kim CH, Kim HP, Kang SS and Chang HW: Ginkgetin, a biflavone from from Ginko biloba leaves, inhibits cycloxygenases -2 and 5-lipoxygenase in mouse bone marrow derived mast cells. Biological and Pharmaceutical Bulletin 2005; 28: 2181-4.
- Lim H, Son KH, Chang HW, Kang SS and Kim HP: Effects of anti-inflammatory biflavonoid, ginkgetin, on chronic skin inflammation. Biological and Pharmaceutical Bulletin 2006; 29: 1046-9.
- Wang, Q, Gao S, Wu GZ, Yang N, Zu XP, Li WC, Xie N, Zhang RR, Li CW, Hu ZL and Zhang WD: Total sesquiterpene lactones isolated from Inula helenium attenuates 2, 4-dinitrochlorobenzene-induced atopic dermatitis-like skin lesions in mice. Phytomedicine 2018; 46: 78-84.
- Jiang HY, Li XN, Sun HD, Zhang HB and Puno PT: Scopariusols L-T, nine new ent-kaurane diterpenoids isolated from Isodon scoparius. Chinese Journal of Natural Medicine 2018; 16: 456-64.
- Joh EH and Kim DH: Kalopanaxsaponin A ameliorates experimental colitis in mice by inhibiting IRAK-1 activation in the NF-κB and MAPK pathways. British Journal of Pharmacology 2011; 162: 1731-42.
- Jeong YH, Hyun JW, Kim Van Le T, Kim DH and Kim HS: Kalopanaxsaponin A exerts anti-inflammatory effects in lipopolysaccharide-stimulated microglia via inhibition of JNK and NF-κB/AP-1 pathways. Biomolecules and Therapeutics (Seoul) 2013; 21: 332-7.
- Shin KM, Kim YH, Park WS, Kang I, Ha J, Choi JW, Park HJ and Lee KT: Inhibition of methanol extract from the fruits of Kochia scoparia on lipopolysaccharide-induced nitric oxide, prostaglandin E2 and tumor necrosis factor production from murine macrophage RAW 264.7 cells. Biological and Pharmaceutical Bulletin 2004; 27: 538-43.
- Yoo SR, Jeong SJ, Lee NR, Shin HK and Seo CS: Quantification analysis and in-vitro anti-inflammatory effects of 20-Hydroxyecdysone, momordinic, and oleanolic acid from the fructus of Kochia scoparia. Pharmacognosy Magazine 2017; 13: 339-44.
- Taka E, Mazzio EA, Goodman CB, Redmon N, Redmon N, Flores-Rozas H, Reams R, Darling-Reed S and Soliman KF: Anti-inflammatory effects of thymoquinone in activated BV-2 microglial cells. Journal of Neuro-immunology 2015; 286: 5-12.
- Boudiaf K, Hurtado-Nedelec M, Belambri SA, Marie JC, Derradji Y, Benboubetra M, El-Benna J and Dang PM: Thymoquinone strongly inhibits fMLF-induced neutrophil functions and exhibits anti-inflammatory properties in- vivo. Biochemical Pharmacology 2016; 104: 62-73.
- Hossen MJ, Yang WS, Kim D, Aravinthan A, Kim JH and Cho JY: Thymoquinone: An IRAK1 inhibitor with in-vivo and in-vitro anti-inflammatory activities. Science Reports 2017; 7: 42995.
- Lee IS, Uh I, Kim KS, Kim KH, Park J, Kim Y, Jung JH, Jung HJ and Jang HJ: Anti-inflammatory effects of ginsenoside Rg3 via NF-κB pathway in A549 cells and human asthmatic lung tissue. Journal of Immunology Research 2016; 2016: 7521601.
- Ahn S, Siddiqi MH, Aceituno VC, Simu SY and Yang DC: Suppression of MAPKs/NF-κB activation induces intestinal anti-inflammatory action of Ginsenoside Rf in HT-29 and RAW264.7 cells. Immunological Investigation 2016; 45: 439-49.
- Zhang Y, Zhang Z, Wang H, Cai N, Zhou S, Zhao Y, Chen X, Zheng S, Si Q and Zhang W: Neuroprotective effect of ginsenoside Rg1 prevents cognitive impairment induced by isoflurane anesthesia in aged rats via antioxidant, anti-inflammatory and anti-apoptotic effects mediated by the PI3K/AKT/GSK-3β pathway. Molecular Medicine Reports 2016; 14: 2778-84.
- Anand P, Kunnumakkara AB, Harikumar KB, Ahn KS, Badmaev V and Aggarwal BB: Modification of cysteine residue in p65 subunit of nuclear factor- κB by picroliv suppress NF- κB regulated gene products and potentiates apoptosis. Cancer Research 2008; 68: 8861-70.
- Zhang DK, Yu JJ, Li YM, Wei LN, Yu Y, Feng YH and Wang X: A Picrorhiza kurroa derivative, picroliv, attenuates the development of dextran-sulfate-sodium-induced colitis in mice. Mediators of Inflammation 2012; 2012: 751629.
- De S, Kundu S, Chatterjee U, Chattopadhyay S and Chatterjee M: Allylpyrocatechol attenuates methotrexate-induced hepatotoxicity in a collagen-induced model of arthritis. Free Radical Research 2018; 52: 698-11.
- Wu JT, Yang GW, Qi CH, Zhou L, Hu JG and Wang MS: Anti-inflammatory activity of platycodin D on alcohol-induced fatty liver rats via TLR4-Myd88-NF-κB signal path. African Journal of Traditional, Complementary and Alternative Medicine 2016; 13: 176-83.
- Gao W, Guo Y and Yang H: Platycodin D protects against cigarette smoke-induced lung inflammation in mice. International Immunopharmacology 2017; 47: 53-8.
- Wang Y, Zhang X, Wei Z, Wang J, Zhang Y, Shi M, Yang Z and Fu Y: Platycodin D suppressed LPS-induced inflammatory response by activating LXRα in LPS-stimulated primary bovine mammary epithelial cells. European Journal of Pharmacol 2017; 814: 138-43.
- Zhu H, Pu D, Di Q, Zhao X, Ji F, Li H, Zhao Z, Gao J, Xiao W and Chen W: Cirsitakaoside isolated from Premna szemaoensis reduces LPS-induced inflammatory responses in-vitro and in-vivo. International Immunopharmacology 2018; 59: 384-90.
- Kim YE, Hwang CJ, Lee HP, Kim CS, Son DJ, Ham YW, Hellström M, Han SB, Kim HS, Park EK and Hong JT: Inhibitory effect of punicalagin on lipopolysaccharide-induced neuroinflammation, oxidative stress and memory impairment via inhibition of nuclear factor-kappa B. Neuropharmacology 2017; 117: 21-32.
- Lyu A, Chen JJ, Wang HC, Yu XH, Zhang ZC, Gong P, Jiang LS and Liu FH: Punicalagin protects bovine endometrial epithelial cells against lipopolysaccharide-induced inflammatory injury. Journal of Zhejiang University Science B 2017; 18: 481-91.
- Chang CC, Chu CF, Wang CN, Wu HT, Bi KW, Pang JH and Huang ST: The anti-atherosclerotic effect of tanshinone IIA is associated with the inhibition of TNF-α-induced VCAM-1, ICAM-1 and CX3CL1 expression. Phytomedicine 2014; 21: 207-16.
- Liu X, Guo CY, Ma XJ, Wu CF, Zhang Y, Sun MY, Pan YT and Yin HJ: Anti-inflammatory effects of tanshinone IIA on atherosclerostic vessels of ovariectomized ApoE mice are mediated by estrogen receptor activation and through the ERK signaling pathway. Cellular Physiology and Biochemistry 2015; 35: 1744-55.
- Fan G, Jiang X, Wu X, Fordjour PA, Miao L, Zhang H, Zhu Y and Gao X: Anti-inflammatory activity of tanshinone IIA in LPS-stimulated RAW264.7 macrophages via miRNAs and TLR4-NF-κB pathway. Inflammation 2016; 39: 375-84.
- Oche B, Chen L, Ma YK, Yang Y, Li CX, Geng X, Qiu LZ, Gao XM and Wang H: Cryptotanshinone and wogonin up-regulate eNOS in vascular endothelial cells via ERα and down-regulate iNOS in LPS stimulated vascular smooth muscle cells via ERβ. Archives of Pharmacal Research 2016; 39: 249-58.
- Maione F, Piccolo M, De Vita S, Chini MG, Cristiano C, De Caro C, Lippiello P, Miniaci MC, Santamaria R, Irace C, De Feo V, Calignano A, Mascolo N and Bifulco G: Downregulation of pro-inflammatory pathways by tanshinone IIA and cryptotanshinone in a non-genetic mouse model of Alzheimer's disease. Pharmacological Research 2018; 129: 482-90.
- Wang Y, Chen G, Yu X, Li Y, Zhang L, He Z, Zhang N, Yang X, Zhao Y, Li N and Qiu H: Salvianolic acid B ameliorates cerebral ischemia/reperfusion injury through inhibiting TLR4/MyD88 signaling pathway. Inflammation 2016; 39: 1503-13.
- Jiang P, Guo Y, Dang R, Yang M, Liao D, Li H, Sun Z, Feng Q and Xu P: Salvianolic acid B protects against lipopolysaccharide-induced behavioral deficits and neuroinflammatory response: involvement of autophagy and NLRP3 inflammasomes. Journal of Neuroinflammation 2017; 14: 239.
- Lou Y, Wang C, Zheng W, Tang Q, Chen Y, Zhang X, Guo X and Wang J: Salvianolic acid B inhibits IL-1β-induced inflammatory cytokine production in human osteoarthritis chondrocytes and has a protective effect in a mouse osteoarthritis model. Int Immunopharmacology 2017; 46: 31-7.
- Lee SU, Choi YH, Kim YS, Min YK, Rhee M and Kim SH: Anti-resoprtive saurolactum exhibits in vitro anti-inflammatory activity via ERK-NF-κB pathway. International Immunopharmacology 2010; 10: 298-03.
- Song SY, Jung YY, Hwang CJ, Lee HP, Sok CH, Kim JH, Lee SM, Seo HO, Hyun BK, Choi DY, Han SB, Ham YW, Hwang BY and Hong JT: Inhibitory effect of ent-Sauchinone on amyloidogenesis via inhibition of STAT3-mediated NF-κB activation in cultured astrocytes and microglial BV-2 cells. Journal of Neuroinflammation 2014; 11: 118.
- Gao Y, Zhao H and Li Y: Sauchinone prevents IL-1β-induced inflammatory response in human chondrocytes. Journal of Biochemical and Molecular Toxicology 2018; 32: 22033.
- Liu N, Zheng JX, Zhuang YS, Zhou ZK, Zhao JH and Yang L: Anti-Inflammatory Effects of Schisandrin B on LPS-Stimulated BV2 Microglia via Activating PPAR-γ. Inflammation 2017; 40: 1006-11.
- Lin Q, Qin X, Shi M, Qin Z, Meng Y, Qin Z and Guo S: Schisandrin B inhibits LPS-induced inflammatory response in human umbilical vein endothelial cells by activating Nrf2. International Immunopharmacology 2017; 49:142-7.
- Kwon DH, Cha HJ, Choi EO, Leem SH, Kim GY, Moon SK, Chang YC, Yun SJ, Hwang HJ, Kim BW, Kim WJ and Choi YH: Schisandrin A suppresses lipopoly-saccharide-induced inflammation and oxidative stress in RAW 264.7 macrophages by suppressing the NF-κB, MAPKs and PI3K/Akt pathways and activating Nrf2/HO-1 signaling. International Journal of Molecular Medicine 2018; 41: 264-74.
- Takanche JS, Lee YH, Kim JS, Kim JE, Han SH, Lee SW and Yi HK: Anti-inflammatory and antioxidant properties of Schisandrin C promote mitochondrial biogenesis in human dental pulp cells. International Endodontic Journal 2018; 51: 438-47.
- Hosokawa Y, Hosokawa I, Shindo S, Ozaki K and Matsuo T: Gomisin N decreases inflammatory cytokine production in human periodontal ligament cells. Inflammation 2017; 40: 360-5.
- Nagappan A, Jung DY, Kim JH, Lee H and Jung MH: Gomisin N alleviates ethanol-induced liver injury through ameliorating lipid metabolism and oxidative stress. International Journal of Molecular Sciences 2018; 19: E2601.
- Jeong HJ, Han NR, Kim KY, Choi IS and Kim HM: Gomisin A decreases the LPS-induced expression of iNOS and COX-2 and activation of RIP2/NF-κB in mouse peritoneal macrophages. Immunopharmacology and Immunotoxicology 2014; 36:195-201.
- Wang S, Wang Y, Liu X, Guan L, Yu L and Zhang X: Anti-inflammatory and anti-arthritic effects of taraxasterol on adjuvant-induced arthritis in rats. Journal of Ethnopharmacology 2016; 187: 42-8.
- Zheng F, Dong X and Meng X: Anti-inflammatory effects of Taraxasterol on LPS-stimulated human umbilical vein endothelial cells. Inflammation 2018; 41: 1755-61.
- Wei D and Huang Z: Anti-inflammatory effects of triptolide in LPS-induced acute lung injury in mice. Inflammation 2014; 37: 1307-16.
- Bai S, Hu Z, Yang Y, Yin Y, Li W, Wu L and Fang M: Anti-inflammatory and neuroprotective effects of Triptolide via the NF-κB signaling pathway in a rat MCAO model. Anatomical Record (Hoboken) 2016; 299: 256-66.
- Su Z, Sun H, Ao M and Zhao C: Atomic force microscopy study of the anti-inflammatory effects of Triptolide on rheumatoid arthritis fibroblast-like synoviocytes. Microscopy and Microanalysis 2017; 23: 1002-12.
- Song J, Fan HJ, Li H, Ding H, Lv Q and Hou SK: Zingerone ameliorates lipopolysaccharide-induced acute kidney injury by inhibiting Toll-like receptor 4 signaling pathway. European J of Pharmacology 2016; 772: 108-14.
- Ruangsuriya J, Budprom P, Viriyakhasem N, Kongdang P, Chokchaitaweesuk C, Sirikaew N, Chomdej S, Nganvongpanit K and Ongchai S: Suppression of cartilage degradation by Zingerone involving the p38 and JNK MAPK signaling pathway. Plant Medica 2017; 83: 268-76.
- Alibakhshi T, Khodayar MJ, Khorsandi L, Rashno M and Zeidooni L: Protective effects of zingerone on oxidative stress and inflammation in cisplatin-induced rat nephrotoxicity. Biomedicine and Pharmacotherapy 2018; 105: 225-32.
- Liang N, Sang Y, Liu W, Yu W and Wang X: Anti-Inflammatory effects of Gingerol on lipopolysaccharide-stimulated RAW 264.7 cells by inhibiting NF-κB signaling pathway. Inflammation 2018; 41: 835-45.
- Han Q, Yuan Q, Meng X, Huo J, Bao Y and Xie G: 6-Shogaol attenuates LPS-induced inflammation in BV2 microglia cells by activating PPAR-γ. Oncotarget 2017; 8: 42001-6.
How to cite this article:
Sarkar D: The anti-inflammatory activity of plant derived ingredients: an analytical review. Int J Pharm Sci & Res 2020; 11(2): 496-06. doi: 10.13040/IJPSR.0975-8232.11(2).496-06.
All © 2013 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
1
496-506
780
1484
English
IJPSR
D. Sarkar
Department of Zoology, A.P.C. Roy Government College, Himanchal Vihar, Matigara, Siliguri, Darjeeling, West Bengal, India.
debjani_sarkar2002@yahoo.co.in
25 April 2019
06 August 2019
01 September 2019
10.13040/IJPSR.0975-8232.11(2).496-06
01 February 2020