Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review)

  • Authors:
    • Xinqiang Song
    • Yu Zhang
    • Erqin Dai
  • View Affiliations

  • Published online on: April 2, 2020     https://doi.org/10.3892/mmr.2020.11052
  • Pages: 2303-2310
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Celastrol and triptolide, chemical compounds isolated from Tripterygium wilfordii hook (also known as thunder god vine), are effective against rheumatoid arthritis (RA). Celastrol targets numerous signaling pathways involving NF‑κB, endoplasmic reticulum Ca2+‑ATPase, myeloid differentiation factor 2, toll‑like receptor 4, pro‑inflammatory chemokines, DNA damage, cell cycle arrest and apoptosis. Triptolide, inhibits NF‑κB, the receptor activator of NF‑κB (RANK)/RANK ligand/osteoprotegerin signaling pathway, cyclooxygenase‑2, matrix metalloproteases and cytokines. The present review examined the chemistry and bioavailability of celastrol and triptolide, and their molecular targets in treating RA. Clinical studies have demonstrated that T. wilfordii has several promising bioactivities, but its multi‑target toxicity has restricted its application. Thus, dosage control and structural modification of T. wilfordii are required to reduce the toxicity. In this review, future directions for research into these promising natural products are discussed.

Introduction

Rheumatoid arthritis (RA) is a complicated disease caused by environmental and genetic factors that involves a hyperactive immune system and synovial inflammation in multiple joints, which if left untreated irreversibly destroys joints and leads to severe disability (1). The common drugs used to treat RA, including methotrexate, cannot prevent the disease and cause severe complications in many patients (2); therefore, the development of safer, cost-effective therapeutics is required.

RA involves the action of pro-inflammatory cytokines and chemokines produced by synoviocytes and infiltrating immune cells (3), including interleukin (IL)-1, IL-2, IL-3, IL-4, IL-6, IL-8, IL-17, interferon (IFN)-α and IFN-β, tumor necrosis factor (TNF)-α, transforming growth factor β, granulocyte-macrophage colony-stimulating factor and macrophage inflammatory protein (MIP)-3α (2,47). These cytokines and chemokines are able to activate NF-κB, upregulate the expressions of cyclooxygenase-2 (COX-2) and nitric oxide synthase, and promote the production of prostaglandin E2 (PGE2) and nitric oxide. Moreover, these changes contribute to synovial inflammation accompanied by arthrosis, swelling, hyperplasia, angiogenesis, bone destruction and arthritic decay (8,9). Natural products that target these molecules are less likely to induce adverse effects may be used as therapeutic agents owing to their high therapeutic potential (2,1013). For example, bioactive natural compounds extracted from plants, such as polyphenolic compounds, commonly exert multiple therapeutic effects (1417).

Tripterygium wilfordii hook (also known as thunder god vine) is a common plant species, which has been used for a variety of purposes in traditional Chinese medicine (TCM) (18). Previous studies in the TCM literature suggest that T. wilfordii can treat several autoimmune and inflammatory conditions including RA, and improvements to its efficacy and safety have been made (14,19).

Currently, >380 secondary metabolites have been isolated from T. wilfordii, and ≥350 of these are structurally diverse terpenoids with a wide range of pharmacological activities, including anti-inflammatory, immunosuppressive and antineoplastic effects (20). It has been shown that two diterpenoid tri-epoxides, celastrol and triptolide (Fig. 1), are primarily responsible for the anti-inflammatory and immunosuppressive effects of T. wilfordii preparations (21,22). Furthermore, these two compounds are the most abundant and most pharmacologically active of the metabolites found in T. wilfordii extracts (23).

The present study critically reviews the chemistry, bioavailability, bioactivities, multi-target toxicities and molecular targets of celastrol and triptolide for the treatment of RA (Fig. 2; Table I). The present review also discussing future directions for research into the aforementioned promising natural products.

Table I.

Molecular targets, signaling pathways and potential biological effects of celastrol and triptolide in the treatment of rheumatoid arthritis.

Table I.

Molecular targets, signaling pathways and potential biological effects of celastrol and triptolide in the treatment of rheumatoid arthritis.

A, Celastrol
Molecular targetsSignaling pathwaysPotential biological effects(Refs.)
IκB kinaseNF-κBNF-κB function is regulated via rapid degradation of its endogenous inhibitory molecule IκB(3239)
Ca2+-ATPaseCa2+ signalingAlter Ca2+ signaling pathways to downregulate inflammatory response genes(4044)
Cyclins with CDKsDNA damage, cell cycle arrest and apoptosisInhibit cell cycle progression by blocking the association of cyclins with cyclin-dependent kinases(33,4547)
MD2 or TLR4Interaction between LPS and the TLR4/MD2 complexBlock the most upstream step in TLR4 activation, maybe targets MD2.(4851)
Pro-inflammatory chemokinesReduces expression levels of RANTES, MCP-1, MIP-1α and GRO/KC, as well as the pro-inflammatory cytokines tumor necrosis factor-α and IL-1βInhibit leukocyte migration into joints(4,5,5155)

B, Triptolide

Molecular targetsSignaling pathwaysPotential biological effects(Refs.)

IκB kinaseNF-κBRapid degradation of its endogenous inhibitory molecule IκB(5664)
RANKLRANKL/RANK/OPG signalingReduce the number of osteoclasts in areas of bone destruction by downregulating RANKL and RANK, and upregulating OPG(6568)
COX-2 and matrix metalloproteinasesNF-κBDownregulate COX-2 and PGE2, and alleviate LPS-induced inflammation(6979)
CytokinesCytokines pathwayInhibit cytokines80–83
VEGFInfiltration of the synovial membraneInhibit several downstream effects of IL1-β, including cell adhesion of human FLS, upregulate several angiogenic activators and activate MAPK signaling(5,8486)

[i] MD2, myeloid differentiation factor 2; TLR4, Toll-like receptor 4; RANK, receptor activator of NF-κB; RANKL, RANK ligand; OPG, osteoprotegerin; COX-2, cyclooxygenase-2; IL, interleukin; IκB, inhibitor of NF-κB; RANTES, regulated upon activation, normally T cell expressed and secreted chemokine protein; VEGF, vascular endothelial growth factor; MAPK, mitogen-activated protein kinase; PGE2, prostaglandin E2; GRO/KC, growth-related oncogene/keratinocyte chemoattractant; MCP-1, monocyte chemotactic protein-1; MIP-1α, macrophage inflammatory protein-3α. LPS, lipopolysaccharide.

Metabolism and bioavailability

Oral administration of triptolide is recommended for to treat inflammation, autoimmune diseases and tumors (24). In rats, triptolide at a dose of 1 mg/kg exhibits a bioavailability of 81.6% after intravenous injection and 63.9% after oral administration (25). Moreover, it achieves a maximum concentration of 293.19±24.43 ng/ml within ~10 min. Triptolide distributes into the liver, followed by plasma, kidney, lung, spleen, heart and testicular tissue, and is quickly excreted through the biliary, urinary and fecal routes with a half-life of 0.42 h (26). Moreover, the metabolism and bioavailability of triptolide is not fully understood as clinical trials investigating the use of triptolide to treat RA have not been performed (27,28).

Celastrol has low aqueous solubility (13.25±0.83 µg/ml at 37°C) and poor intestinal absorption, which results in low oral bioavailability and limits its clinical application (29). To overcome these constraints, the compound can be delivered using solid lipid nanoparticles, liposomes, micelles or nanostructured lipid carriers (30,31).

Molecular targets of celastrol

Celastrol inhibits activation of NF-κB by targeting inhibitor of NF-κB (IκB) kinase

NF-κB regulates the transcription of numerous genes that are involved in immune, inflammatory and anti-apoptotic responses (32). Furthermore, the function of NF-κB is regulated by rapid degradation of its endogenous inhibitory molecule IκB. Inflammatory stimuli, such as cytokines, initiate a signaling cascade that leads to the activation of two IκB kinases (IKK), IKK-1 and IKK-2, which then phosphorylate IκB at two N-terminal serine residues (33). The IKK complex is expressed in fibroblast-like synoviocytes (FLS) and is activated by IL-1 and TNF-α (3436). These studies demonstrated that celastrol suppresses NF-κB activation by inhibiting IKK activity, possibly by targeting cysteine 179 in the activation loop of IKK (34,35,37). Moreover, celastrol inhibits the activation of NF-κB by targeting IκB kinase, which is not specific in the RA (38). It has been revealed that the NF-κB signaling pathway is involved in a number of diseases, including RA, systemic lupus erythematosus and ankylosing spondylitis (32,3437). Moreover, myeloid differentiation factor 2 (MD2) and Toll-like receptor 4 (TLR4) are associated with RA, thus it is likely that celastrol targets MD2 (39).

Celastrol inhibits endoplasmic reticulum (ER) Ca2+-ATPase

In RA, Ca2+ signaling mediates the expression of pro-inflammatory cytokines through autoreactive T- and B-lymphocytes after autoantigen stimulation (40). Various pumps maintain Ca2+ homeostasis, among which is the ER Ca2+-ATPase, which can be inhibited by celastrol (41). Therefore, celastrol may alter Ca2+ signaling pathways to downregulate inflammatory response genes (42) and promote Ca2+-mediated autophagic cell death (43,44), as shown by studies in RA-FLS. Thus, it is speculated that resistance of FLS to apoptosis may be a characteristic of RA.

Celastrol induces DNA damage, cell cycle arrest and apoptosis

Celastrol blocks RA-FLS at G2/M phase (45), which may be a potential mechanism for its ability to inhibit proliferation and induce apoptosis. It has also been demonstrated that celastrol inhibits cell cycle progression by blocking the association of cyclins with cyclin-dependent kinases (33). Furthermore, celastrol leads to an increase in cell division cycle protein 2 homolog (Cdc2) phosphorylation and downregulation of Cdc2 and Cyclin-b1, which may reduce the number of Cdk1-Cyclin-b1 complexes and arrest cells at the G2/M phase (45). Celastrol also increases phosphorylation of Cdc25, which may contribute to G2/M phase arrest (46).

It has been shown that celastrol activates cleaved caspases 3 and 9, as well as cleaved poly (ADP-ribose) polymerase, downregulates FasR and increases the Bax/Bcl-2 ratio (46). Therefore, celastrol may induce apoptosis in RA-FLS, which express a variety of death-inducing surface receptors of the TNF receptor family such as Fas/CD95, TNF-related apoptosis-inducing ligand-receptor (TRAIL-R1), TRAIL-R2 and TNF receptor 1 (47).

Celastrol targets MD2 and inhibits TLR4 activation

TLR4 exists as a complex with a co-receptor, MD2, in the plasma membrane of various immune cells (48). Celastrol blocks the most upstream step in TLR4 activation (49), and thus it likely targets MD2. Moreover, celastrol may function similar to the anti-inflammatory phytochemicals sulforaphane and caffeic acid phenethyl ester, which interfere with the interaction between lipopolysaccharide (LPS) and the TLR4/MD2 complex (50,51). Furthermore, it is speculated that celastrol may have intracellular targets, including MD2 and TLR4 (48,49).

Celastrol modulates pro-inflammatory chemokines

Chemokines are a superfamily of cytokines that are associated with cell migration and recruitment to sites of inflammation (5). Chemokines are categorized into four groups, CXC, CX3C, CC and C, based on the location of conserved cysteine residues (21). In inflammatory disorders such as RA, chemokines bind to their receptors leading to leukocyte trafficking into the joints, where leukocytes exacerbate inflammation and lead to pannus formation and tissue damage (4). Moreover, several chemokines have been implicated in RA and experimental arthritis, including T cell specific protein RANTES [RANTES; also known as C-C motif chemokine 5 (CCL5)], monocyte chemotactic protein-1 (MCP-1; also known as CCL2), MIP-1α (also known as CCL3) and growth-related oncogene/keratinocyte chemoattractant (GRO/KC) (5153). It has been revealed that treating arthritic rats with celastrol significantly reduces expression levels of RANTES, MCP-1, MIP-1α and GRO/KC, as well as the pro-inflammatory cytokines TNF-α and IL-1β (54), and this may inhibit leukocyte migration into joints (55).

Molecular targets of triptolide

Triptolide inhibits the NF-κB pathway

The NF-κB family comprises Rel domain-containing proteins that regulate inflammatory and immune responses (56). In resting cells, these proteins are retained in the cytosol by a group of inhibitory proteins, such as IκBα (57). Upon activation, IKKs, including IKK-1 and IKK-2, phosphorylate IκBα, which is subsequently ubiquitinated and destroyed by the proteasome. This liberates NF-κB to translocate into the nucleus, where it activates several genes associated with RA (56). It has been shown that triptolide regulates IKK-1 and IKK-2 activities induced by various stimuli (57). The purified T. wilfordii component PG490 inhibits both IKK-1 and IKK2 activities with similar potency (58). As NF-κB transcription factors upregulate the expression of several genes involved in inflammatory responses, the targeting of components of NF-κB signaling is a major therapeutic strategy for treating autoimmune diseases (59).

Similar to NF-κB, activating protein-1 (AP-1) transcription factors, comprising Jun and Fos family proteins, regulate cell proliferation, transformation and death, and may be potential therapeutic targets for the control of inflammation (6062). Triptolide also inhibits mitogen-activated protein kinase (MAPK)/AP-1 signaling pathways, effectively suppressing MAP kinases, including JNK, p38 and ERK activities (59). Therefore, triptolide is a promising candidate immunomodulatory drug for autoimmune disorder therapy (63,64).

Triptolide alters RANKL/RANK/OPG signaling

Osteoclasts are the primary bone resorptive cells, and are located mainly in the synovial inflammatory tissue; RANKL stimulates osteoclast-mediated bone destruction in RA by binding to its receptor RANK (65). Under physiological conditions, osteoblasts and activated T cells express RANKL, which binds to RANK on osteoclasts to trigger osteoclast maturation and bone resorption. Osteoblasts counteract the action of osteoclasts in the balance between bone formation and destruction; osteoblasts express osteoprotegerin (OPG), which ‘mops up’ RANKL and prevents it from binding to RANK, thus inhibiting bone resorption (66). However, under pathological conditions such as RA, this balance is shifted toward bone destruction (67). In mice with collagen-induced arthritis, triptolide significantly reduces the number of osteoclasts in areas of bone destruction by downregulating RANKL and RANK, and upregulating OPG (68).

Triptolide inhibits COX-2 and matrix metalloproteinases (MMPs)

It has been revealed that injury, tumorigenesis and invasion from the joint into multiple organs upregulate COX-2 via NF-κB to produce prostaglandins, which induce inflammation and increase capillary permeability (69). Moreover, triptolide downregulates COX-2 and PGE2, thus alleviating LPS-induced inflammation (70).

MMPs participate in tumorigenesis, tumor metastasis and inflammatory diseases such as RA (57,7175). In human synovial fibroblasts and mouse macrophages, triptolide inhibits IL-1α-induced phosphorylation of MMP-1 and LPS-induced phosphorylation of MMP-3. By inhibiting MMP-3 and MMP-13, triptolide slows the degradation of extracellular matrix and cartilage degeneration in mice with collagen-induced arthritis, as well as in primary human osteoarthritis and bovine chondrocytes (26,7679).

Triptolide inhibits cytokines

Antigen-presenting cells produce IL-12 and IL-23, which are heterodimeric cytokines sharing a p40 subunit; these cytokines stimulate the generation and functions of T helper (Th)1 and Th17 cells, respectively. These cytokines are involved in the pathogenesis of several autoimmune disorders, including RA and systemic lupus erythematosus (80). Triptolide downregulates p40, in part, by activating the expression and phosphorylation of CCAAT/enhancer binding protein-α (C/EBPα) via the kinases ERK1/2 and AKT-glycogen synthase kinase 3β (81). This phosphorylation allows C/EBPα to bind antagonistically to the p40 promoter (81). Furthermore, programmed cell death 5 enhances the ability of triptolide to induce FLS apoptosis in RA, and therefore may be a potential therapeutic target in RA (29,82,83).

Triptolide targets vascular endothelial growth factor (VEGF)

VEGF-driven angiogenesis promotes RA progress by allowing inflammatory cell infiltration of the synovial membrane (5). Triptolide prevents the formation of new blood vessels in vitro and in vivo, and it inhibits several downstream effects of IL-1β, including cell adhesion of human FLS in RA and human umbilical vein endothelial cells (HUVECs) (84). Furthermore, triptolide upregulates several angiogenic activators, including TNF-α, IL-17, VEGF, VEGFR, Angiopoietin (Ang)-1, Ang-2 and Tie2, and activates the MAPK signaling pathway involving phosphorylated (p)-ERK, p-p38 and p-JNK (85). Moreover, triptolide disrupts tube formation in HUVECs on Matrigel, and suppresses VEGF-induced chemotactic migration of HUVECs and human FLS in RA (86).

Adverse effects of T. wilfordii

T. wilfordii is a Chinese herb that has been traditionally used in clinics for RA treatment (3). Numerous preclinical studies have demonstrated that extracts from T. wilfordii roots inhibit the expression levels of RA-related inflammatory factors secreted by macrophages, lymphocytes, synovial fibroblasts and chondrocytes (8791). Moreover, T. wilfordii suppresses lymphocytes and synovial fibroblast proliferation by inducing apoptosis (46). The anti-angiogenesis property of synovial fibroblasts has been shown in a previous study (83). Although T. wilfordii has several promising bioactivities in vivo and in vitro, its multi-target toxicity has restricted its clinical application (88). Data from the China Food and Drug Administration catalogue at least 633 instances of adverse reactions (53 severe) involving reproductive organ, liver and renal toxicity. Furthermore, clinical studies have concluded that T. wilfordii can damage the digestive system, including liver injury and stomachache, as well as the endocrine and reproductive systems (30,32,92). Moreover, 271 patients with RA have reported digestive tract symptoms and irregular menstruation. As a compound of T. wilfordii, triptolide-induced toxicity was shown to be dependent on dosage and administration times (30,93). To avoid toxicity, previous studies have attempted to alter the dosage and structure, and to assess its compatibility with other drugs (34). For example, Freag et al (94) developed self-assembled celastrol phytosomal nanocarriers (celastrol-PHY) to improve celastrol solubility and oral bioavailability; these were confirmed through an in vitro release study and a pharmacokinetic study in rabbits (94). Structural modification and alternation of triptolide can produce derivatives of triptolide with lower toxicity and relative higher activity. Apart from structural alterations, the development of novel triptolide delivery systems is a valuable strategy to improve water solubility, and the efficiency of absorption and metabolism, and to reduce toxicity (95).

Conclusions

Celastrol and triptolide from T. wilfordii are effective against RA; they target numerous signaling pathways, proteases and cytokines. The present review examined the chemistry and bioavailability of celastrol and triptolide, and their molecular targets in treating RA, which may be potential effective drugs. However, owing to the strong toxicity of T. wilfordii, novel approaches are required for the safe application of this TCM. These may include investigating new triptolide formulations or its combination with other drugs. Furthermore, defining early toxicity markers, investigating dosage ranges for different target organs and establishing a toxicity warning system are required.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. U1804179), The Key Scientific and Technological Projects in Henan Province (grant no. 202102310190), The Henan Science and Technology Innovation Team, Investigation on Plant Resources in Dabie Mountains and The Study and Utilization of Active Components of Special Plants (grant no. 2017083), and The Nanhu Scholars Program for Young Scholars of Xinyang Normal University (grant no. 2018001).

Availability of data and materials

Not applicable.

Authors' contributions

XS conceived and designed the study, and wrote the manuscript. YZ and ED analyzed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Deane KD and Holers VM: The natural history of rheumatoid arthritis. Clin Ther. 41:1256–1269. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Burmester GR and Pope JE: Novel treatment strategies in rheumatoid arthritis. Lancet. 389:2338–2348. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Ridgley LA, Anderson AE and Pratt AG: What are the dominant cytokines in early rheumatoid arthritis? Curr Opin Rheumatol. 30:207–214. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Noack M and Miossec P: Selected cytokine pathways in rheumatoid arthritis. Semin Immunopathol. 39:365–383. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Kang S, Tanaka T, Narazaki M and Kishimoto T: Targeting interleukin-6 signaling in clinic. Immunity. 50:1007–1023. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Siouti E and Andreakos E: The many facets of macrophages in rheumatoid arthritis. Biochem Pharmacol. 165:152–169. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Yasuda K, Takeuchi Y and Hirota K: The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol. 41:283–297. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Arleevskaya MI, Larionova RV, Brooks WH, Bettacchioli E and Renaudineau Y: Toll-like receptors, infections, and rheumatoid arthritis. Clin Rev Allergy Immunol. May 29–2019.(Epub ahead of print).

9 

Alam J, Jantan I and Bukhari SNA: Rheumatoid arthritis: Recent advances on its etiology, role of cytokines and pharmacotherapy. Biomed Pharmacother. 92:615–633. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Silvagni E, Di Battista M, Bonifacio AF, Zucchi D, Governato G and Scirè CA: One year in review 2019: Novelties in the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 37:519–534. 2019.PubMed/NCBI

11 

Conigliaro P, Triggianese P, De Martino E, Fonti GL, Chimenti MS, Sunzini F, Viola A, Canofari C and Perricone R: Challenges in the treatment of rheumatoid arthritis. Autoimmun Rev. 18:706–713. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Cecchi I, Arias de la Rosa I, Menegatti E, Roccatello D, Collantes-Estevez E, Lopez-Pedrera C and Barbarroja N: Neutrophils: Novel key players in rheumatoid arthritis. Current and future therapeutic targets. Autoimmun Rev. 17:1138–1149. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Cheung TT and McInnes IB: Future therapeutic targets in rheumatoid arthritis? Semin Immunopathol. 39:487–500. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Hou W, Liu B and Xu H: Triptolide: Medicinal chemistry, chemical biology and clinical progress. Eur J Med Chem. 176:378–392. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Dong Y, Chen H, Gao J, Liu Y, Li J and Wang J: Bioactive ingredients in Chinese herbal medicines that target non-coding RNAs: Promising new choices for disease treatment. Front Pharmacol. 10:5152019. View Article : Google Scholar : PubMed/NCBI

16 

Huang Y, Ma S, Wang Y, Yan R, Wang S, Liu N, Chen B, Chen J and Liu L: The role of traditional Chinese herbal medicines and bioactive ingredients on ion channels: A brief review and prospect. CNS Neurol Disord Drug Targets. 18:257–265. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Dong Y, Wang P, Feng X, Li B, Wang Z and Li H: The role of Chinese herbal medicines and bioactive ingredients targeting myocardial KCa and KATP Channels in cardiovascular diseases. Curr Pharm Des. 23:1070–1076. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Lv H, Jiang L, Zhu M, Li Y, Luo M, Jiang P, Tong S, Zhang H and Yan J: The genus Tripterygium: A phytochemistry and pharmacological review. Fitoterapia. 137:1041902019. View Article : Google Scholar : PubMed/NCBI

19 

Venkatesha SH, Dudics S, Astry B and Moudgil KD: Control of autoimmune inflammation by celastrol, a natural triterpenoid. Pathog Dis. 74(pii): ftw0592016. View Article : Google Scholar : PubMed/NCBI

20 

Tu L, Su P, Zhang Z, Gao L, Wang J, Hu T, Zhou J, Zhang Y, Zhao Y, Liu Y, et al: Genome of Tripterygium wilfordii and identification of cytochrome P450 involved in triptolide biosynthesis. Nat Commun. 11:9712020. View Article : Google Scholar : PubMed/NCBI

21 

Lin N, Sato T and Ito A: Triptolide, a novel diterpenoid triepoxide from Tripterygium wilfordii Hook. f., suppresses the production and gene expression of pro-matrix metalloproteinases 1 and 3 and augments those of tissue inhibitors of metalloproteinases 1 and 2 in human synovial fibroblasts. Arthritis Rheum. 44:2193–2200. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Astry B, Venkatesha SH, Laurence A, Christensen-Quick A, Garzino-Demo A, Frieman MB, O'Shea JJ and Moudgil KD: Celastrol, a Chinese herbal compound, controls autoimmune inflammation by altering the balance of pathogenic and regulatory T cells in the target organ. Clin Immunol. 157:228–238. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Di YM, Zhou ZW, Guang Li C and Zhou SF: Current and future therapeutic targets of rheumatoid arthritis. Antiinflamm Antiallergy Agents Med Chem. 10:92–120. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Liu J, Zhou X, Chen XY and Zhong DF: Excretion of [3H]triptolide and its metabolites in rats after oral administration. Acta Pharmacol Sin. 35:549–554. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Liu Q: Triptolide and its expanding multiple pharmacological functions. Int Immunopharmacol. 11:377–383. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Li XJ, Jiang ZZ and Zhang LY: Triptolide: Progress on research in pharmacodynamics and toxicology. J Ethnopharmacol. 155:67–79. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Cheng Y, Chen G, Wang L, Kong J, Pan J, Xi Y, Shen F and Huang Z: Triptolide-induced mitochondrial damage dysregulates fatty acid metabolism in mouse sertoli cells. Toxicol Lett. 292:136–150. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Xi C, Peng S, Wu Z, Zhou Q and Zhou J: Toxicity of triptolide and the molecular mechanisms involved. Biomed Pharmacother. 90:531–541. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Song J, Shi F, Zhang Z, Zhu F, Xue J, Tan X, Zhang L and Jia X: Formulation and evaluation of celastrol-loaded liposomes. Molecules. 16:7880–7892. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Qi J, Lu Y and Wu W: Absorption, disposition and pharmacokinetics of solid lipid nanoparticles. Curr Drug Metab. 13:418–428. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Peng X, Wang J, Song H, Cui D, Li L, Li J, Lin L, Zhou J and Liu Y: Optimized preparation of celastrol-loaded polymeric nanomicelles using rotatable central composite design and response surface methodology. J Biomed Nanotechnol. 8:491–499. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Cascao R, Fonseca JE and Moita LF: Celastrol: A spectrum of treatment opportunities in chronic diseases. Front Med (Lausanne). 4:692017. View Article : Google Scholar : PubMed/NCBI

33 

Venkatesha SH and Moudgil KD: Celastrol and its role in controlling chronic diseases. Adv Exp Med Biol. 928:267–289. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Shen YF, Zhang X, Wang Y, Cao FF, Uzan G, Peng B and Zhang DH: Celastrol targets IRAKs to block Toll-like receptor 4-mediated nuclear factor-κB activation. J Integr Med. 14:203–208. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Lee JH, Koo TH, Yoon H, Jung HS, Jin HZ, Lee K, Hong YS and Lee JJ: Inhibition of NF-κB activation through targeting I kappa B kinase by celastrol, a quinone methide triterpenoid. Biochem Pharmacol. 72:1311–1321. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Mercurio F, Zhu H, Murray BW, Shevchenko A, Bennett BL, Li J, Young DB, Barbosa M, Mann M, Manning A and Rao A: IKK-1 and IKK-2: Cytokine-activated IkappaB kinases essential for NF-kappaB activation. Science. 278:860–866. 1997. View Article : Google Scholar : PubMed/NCBI

37 

Salminen A, Lehtonen M, Paimela T and Kaarniranta K: Celastrol: Molecular targets of thunder God vine. Biochem Biophys Res Commun. 394:439–442. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Rigoglou S and Papavassiliou AG: The NF-κB signalling pathway in osteoarthritis. Int J Biochem Cell Biol. 45:2580–2584. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Samarpita S, Kim JY, Rasool MK and Kim KS: Investigation of toll-like receptor (TLR) 4 inhibitor TAK-242 as a new potential anti-rheumatoid arthritis drug. Arthritis Res Ther. 22:162020. View Article : Google Scholar : PubMed/NCBI

40 

Berridge MJ: Calcium signalling remodelling and disease. Biochem Soc Trans. 40:297–309. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Clapham DE: Calcium signaling. Cell. 131:1047–1058. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Wong VKW, Qiu C, Xu SW, Law BYK, Zeng W, Wang H, Michelangeli F, Dias IRSR, Qu YQ, Chan TW, et al: Ca2+ signalling plays a role in celastrol-mediated suppression of synovial fibroblasts of rheumatoid arthritis patients and experimental arthritis in rats. Br J Pharmacol. 176:2922–2944. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Yoo SA, Park BH, Park GS, Koh HS, Lee MS, Ryu SH, Miyazawa K, Park SH, Cho CS and Kim WU: Calcineurin is expressed and plays a critical role in inflammatory arthritis. J Immunol. 177:2681–2690. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Villalobo A, Ishida H, Vogel HJ and Berchtold MW: Calmodulin as a protein linker and a regulator of adaptor/scaffold proteins. Biochim Biophys Acta Mol Cell Res. 1865:507–521. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Xu Z, Wu G, Wei X, Chen X, Wang Y and Chen L: Celastrol induced DNA damage, cell cycle arrest, and apoptosis in human rheumatoid fibroblast-like synovial cells. Am J Chin Med. 41:615–628. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Fan XX, Li N, Wu JL, Zhou YL, He JX, Liu L and Leung EL: Celastrol induces apoptosis in gefitinib-resistant non-small cell lung cancer cells via caspases-dependent pathways and Hsp90 client protein degradation. Molecules. 19:3508–3522. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Xu LM, Zheng YJ, Wang Y, Yang Y, Cao FF, Peng B, Xu XF, An HZ, Zheng AX, Zhang DH, et al: Celastrol inhibits lung infiltration in differential syndrome animal models by reducing TNF-α and ICAM-1 levels while preserving differentiation in ATRA-induced acute promyelocytic leukemia cells. PLoS One. 9:e1051312014. View Article : Google Scholar : PubMed/NCBI

48 

Fang Z, He D, Yu B, Liu F, Zuo J, Li Y, Lin Q, Zhou X and Wang Q: High-throughput study of the effects of celastrol on activated fibroblast-like synoviocytes from patients with rheumatoid arthritis. Genes (Basel). 8(pii): E2212017. View Article : Google Scholar : PubMed/NCBI

49 

Mukherjee S, Huda S and Sinha Babu SP: Toll-like receptor polymorphism in host immune response to infectious diseases: A review. Scand J Immunol. 90:e127712019. View Article : Google Scholar : PubMed/NCBI

50 

Zhang X, Wang Y, Ge HY, Gu YJ, Cao FF, Yang CX, Uzan G, Peng B and Zhang DH: Celastrol reverses palmitic acid (PA)-caused TLR4-MD2 activation-dependent insulin resistance via disrupting MD2-related cellular binding to PA. J Cell Physiol. 233:6814–6824. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Khan MA, Khurana N, Ahmed RS, Umar S, Md G Sarwar AH, Alam Q, Kamal MA and Ashraf GM: Chemokines: A potential therapeutic target to suppress autoimmune arthritis. Curr Pharm Des. 25:2937–2946. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Eustace AD, McNaughton EF, King S, Kehoe O, Kungl A, Mattey D, Nobbs AH, Williams N and Middleton J: Soluble syndecan-3 binds chemokines, reduces leukocyte migration in vitro and ameliorates disease severity in models of rheumatoid arthritis. Arthritis Res Ther. 21:1722019. View Article : Google Scholar : PubMed/NCBI

53 

Bahlas S, Damiati L, Dandachi N, Sait H, Alsefri M and Pushparaj PN: Rapid immunoprofiling of cytokines, chemokines and growth factors in patients with active rheumatoid arthritis using luminex multiple analyte profiling technology for precision medicine. Clin Exp Rheumatol. 37:112–119. 2019.PubMed/NCBI

54 

Lee JY, Lee BH, Kim ND and Lee JY: Celastrol blocks binding of lipopolysaccharides to a Toll-like receptor4/myeloid differentiation factor2 complex in a thiol-dependent manner. J Ethnopharmacol. 172:254–260. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Li G, Liu D, Zhang Y, Qian Y, Zhang H, Guo S, Sunagawa M, Hisamitsu T and Liu Y: Celastrol inhibits lipopolysaccharide-stimulated rheumatoid fibroblast-like synoviocyte invasion through suppression of TLR4/NF-κB-mediated matrix metalloproteinase-9 expression. PLoS One. 8:e689052013. View Article : Google Scholar : PubMed/NCBI

56 

Venkatesha SH, Astry B, Nanjundaiah SM, Yu H and Moudgil KD: Suppression of autoimmune arthritis by celastrus-derived celastrol through modulation of pro-inflammatory chemokines. Bioorg Med Chem. 20:5229–5234. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Li GQ, Liu D, Zhang Y, Qian YY, Zhu YD, Guo SY, Sunagawa M, Hisamitsu T and Liu YQ: Anti-invasive effects of celastrol in hypoxia-induced fibroblast-like synoviocyte through suppressing of HIF-1α/CXCR4 signaling pathway. Int Immunopharmacol. 17:1028–1036. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Park B, Sung B, Yadav VR, Chaturvedi MM and Aggarwal BB: Triptolide, histone acetyltransferase inhibitor, suppresses growth and chemosensitizes leukemic cells through inhibition of gene expression regulated by TNF-TNFR1-TRADD-TRAF2-NIK-TAK1-IKK pathway. Biochem Pharmacol. 82:1134–1144. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Yang Y, Ye Y, Qiu Q, Xiao Y, Huang M, Shi M, Liang L, Yang X and Xu H: Triptolide inhibits the migration and invasion of rheumatoid fibroblast-like synoviocytes by blocking the activation of the JNK MAPK pathway. Int Immunopharmacol. 41:8–16. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Fan D, He X, Bian Y, Guo Q, Zheng K, Zhao Y, Lu C, Liu B, Xu X, Zhang G and Lu A: Triptolide modulates TREM-1 signal pathway to inhibit the inflammatory response in rheumatoid arthritis. Int J Mol Sci. 17:4982016. View Article : Google Scholar : PubMed/NCBI

61 

Ho LJ, Chang WL, Chen A, Chao P and Lai JH: Differential immunomodulatory effects by Tripterygium wilfordii Hook f-derived refined extract PG27 and its purified component PG490 (triptolide) in human peripheral blood T cells: Potential therapeutics for arthritis and possible mechanisms explaining in part Chinese herbal theory ‘Junn-Chenn-Zuou-SS’. J Transl Med. 11:2942013. View Article : Google Scholar : PubMed/NCBI

62 

Ruland J: Return to homeostasis: Downregulation of NF-κB responses. Nat Immunol. 12:709–714. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Kanarek N and Ben-Neriah Y: Regulation of NF-κB by ubiquitination and degradation of the IκBs. Immunol Rev. 246:77–94. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Criswell LA: Gene discovery in rheumatoid arthritis highlights the CD40/NF-kappaB signaling pathway in disease pathogenesis. Immunol Rev. 233:55–61. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Schonthaler HB, Guinea-Viniegra J and Wagner EF: Targeting inflammation by modulating the Jun/AP-1 pathway. Ann Rheum Dis. 70 (Suppl 1):i109–i112. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Xiao C, Zhou J, He Y, Jia H, Zhao L, Zhao N and Lu A: Effects of triptolide from radix Tripterygium wilfordii (Leigongteng) on cartilage cytokines and transcription factor NF-kappaB: A study on induced arthritis in rats. Chin Med. 4:132009. View Article : Google Scholar : PubMed/NCBI

67 

Bezerra MC, Carvalho JF, Prokopowitsch AS and Pereira RM: RANK, RANKL and osteoprotegerin in arthritic bone loss. Braz J Med Biol Res. 38:161–170. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Ho TY, Santora K, Chen JC, Frankshun AL and Bagnell CA: Effects of relaxin and estrogens on bone remodeling markers, receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG), in rat adjuvant-induced arthritis. Bone. 48:1346–1353. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Geusens P: The role of RANK ligand/osteoprotegerin in rheumatoid arthritis. Ther Adv Musculoskelet Dis. 4:225–233. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Liu Q, Chen T, Chen G, Shu X, Sun A, Ma P, Lu L and Cao X: Triptolide impairs dendritic cell migration by inhibiting CCR7 and COX-2 expression through PI3-K/Akt and NF-kappaB pathways. Mol Immunol. 44:2686–2696. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Liu C, Zhang Y, Kong X, Zhu L, Pang J, Xu Y, Chen W, Zhan H, Lu A and Lin N: Triptolide prevents bone destruction in the collagen-induced arthritis model of rheumatoid arthritis by targeting RANKL/RANK/OPG signal pathway. Evid Based Complement Alternat Med. 2013:6260382013.PubMed/NCBI

72 

Brinker AM, Ma J, Lipsky PE and Raskin I: Medicinal chemistry and pharmacology of genus Tripterygium (Celastraceae). Phytochemistry. 68:732–766. 2007. View Article : Google Scholar : PubMed/NCBI

73 

Xue M, McKelvey K, Shen K, Minhas N, March L, Park SY and Jackson CJ: Endogenous MMP-9 and not MMP-2 promotes rheumatoid synovial fibroblast survival, inflammation and cartilage degradation. Rheumatology (Oxford). 53:2270–2279. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Geng Y, Blanco FJ, Cornelisson M and Lotz M: Regulation of cyclooxygenase-2 expression in normal human articular chondrocytes. J Immunol. 155:796–801. 1995.PubMed/NCBI

75 

Maekawa K, Yoshikawa N, Du J, Nishida S, Kitasato H, Okamoto K, Tanaka H, Mizushima Y and Kawai S: The molecular mechanism of inhibition of interleukin-1beta-induced cyclooxygenase-2 expression in human synovial cells by Tripterygium wilfordii Hook F extract. Inflamm Res. 48:575–581. 1999. View Article : Google Scholar : PubMed/NCBI

76 

Flower RJ: The development of COX2 inhibitors. Nat Rev Drug Discov. 2:179–191. 2003. View Article : Google Scholar : PubMed/NCBI

77 

Geng Y, Fang M, Wang J, Yu H, Hu Z, Yew DT and Chen W: Triptolide down-regulates COX-2 expression and PGE2 release by suppressing the activity of NF-κB and MAP kinases in lipopolysaccharide-treated PC12 cells. Phytother Res. 26:337–343. 2012.PubMed/NCBI

78 

Ma J, Dey M, Yang H, Poulev A, Pouleva R, Dorn R, Lipsky PE, Kennelly EJ and Raskin I: Anti-inflammatory and immunosuppressive compounds from Tripterygium wilfordii. Phytochemistry. 68:1172–1178. 2007. View Article : Google Scholar : PubMed/NCBI

79 

Liacini A, Sylvester J and Zafarullah M: Triptolide suppresses proinflammatory cytokine-induced matrix metalloproteinase and aggrecanase-1 gene expression in chondrocytes. Biochem Biophys Res Commun. 327:320–327. 2005. View Article : Google Scholar : PubMed/NCBI

80 

Lin N, Liu C, Xiao C, Jia H, Imada K, Wu H and Ito A: Triptolide, a diterpenoid triepoxide, suppresses inflammation and cartilage destruction in collagen-induced arthritis mice. Biochem Pharmacol. 73:136–146. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Zhang Y and Ma X: Triptolide inhibits IL-12/IL-23 expression in APCs via CCAAT/enhancer-binding protein alpha. J Immunol. 184:3866–3877. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Jiang J, Wang N, Guan Z and Houshan LV: Programmed cell death 5 factor enhances triptolide-induced fibroblast-like synoviocyte apoptosis of rheumatoid arthritis. Artif Cells Blood Substit Immobil Biotechnol. 38:38–42. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Tasneem S, Liu B, Li B, Choudhary MI and Wang W: Molecular pharmacology of inflammation: Medicinal plants as anti-inflammatory agents. Pharmacol Res. 139:126–140. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Ziaei S and Halaby R: Immunosuppressive, anti-inflammatory and anti-cancer properties of triptolide: A mini review. Avicenna J Phytomed. 6:149–164. 2016.PubMed/NCBI

85 

Kong X, Zhang Y, Liu C, Guo W, Li X, Su X, Wan H, Sun Y and Lin N: Anti-angiogenic effect of triptolide in rheumatoid arthritis by targeting angiogenic cascade. PLoS One. 8:e775132013. View Article : Google Scholar : PubMed/NCBI

86 

Zhang W, Li F and Gao W: Tripterygium wilfordii inhibiting angiogenesis for rheumatoid arthritis treatment. J Natl Med Assoc. 109:142–148. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Ramgolam V, Ang SG, Lai YH, Loh CS and Yap HK: Traditional Chinese medicines as immunosuppressive agents. Ann Acad Med Singapore. 29:11–16. 2000.PubMed/NCBI

88 

Cameron M, Gagnier JJ and Chrubasik S: Herbal therapy for treating rheumatoid arthritis. Cochrane Database Syst Rev. CD0029482011.PubMed/NCBI

89 

Lipsky PE and Tao XL: A potential new treatment for rheumatoid arthritis: Thunder god vine. Semin Arthritis Rheum. 26:713–723. 1997. View Article : Google Scholar : PubMed/NCBI

90 

Lv QW, Zhang W, Shi Q, Zheng WJ, Li X, Chen H, Wu QJ, Jiang WL, Li HB, Gong L, et al: Comparison of Tripterygium wilfordii Hook F with methotrexate in the treatment of active rheumatoid arthritis (TRIFRA): A randomised, controlled clinical trial. Ann Rheum Dis. 74:1078–1086. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Tao X, Younger J, Fan FZ, Wang B and Lipsky PE: Benefit of an extract of Tripterygium Wilfordii Hook F in patients with rheumatoid arthritis: A double-blind, placebo-controlled study. Arthritis Rheum. 46:1735–1743. 2002. View Article : Google Scholar : PubMed/NCBI

92 

Zhao Q, Liu F, Cheng Y, Xiao XR, Hu DD, Tang YM, Bao WM, Yang JH, Jiang T, Hu JP, et al: Celastrol protects from cholestatic liver injury through modulation of SIRT1-FXR signaling. Mol Cell Proteomics. 18:520–533. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Zhang Y, Jiang Z, Xue M, Zhang S, Wang Y and Zhang L: Toxicogenomic analysis of the gene expression changes in rat liver after a 28-day oral Tripterygium wilfordii multiglycoside exposure. J Ethnopharmacol. 141:170–177. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Freag MS, Saleh WM and Abdallah OY: Self-assembled phospholipid-based phytosomal nanocarriers as promising platforms for improving oral bioavailability of the anticancer celastrol. Int J Pharm. 535:18–26. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Xu H and Liu B: Triptolide-targeted delivery methods. Eur J Med Chem. 164:342–351. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2020
Volume 21 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song X, Zhang Y and Dai E: Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review). Mol Med Rep 21: 2303-2310, 2020
APA
Song, X., Zhang, Y., & Dai, E. (2020). Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review). Molecular Medicine Reports, 21, 2303-2310. https://doi.org/10.3892/mmr.2020.11052
MLA
Song, X., Zhang, Y., Dai, E."Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review)". Molecular Medicine Reports 21.6 (2020): 2303-2310.
Chicago
Song, X., Zhang, Y., Dai, E."Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review)". Molecular Medicine Reports 21, no. 6 (2020): 2303-2310. https://doi.org/10.3892/mmr.2020.11052