Next Article in Journal
Autoimmunity and Gastric Cancer
Next Article in Special Issue
Therapeutics in Osteoarthritis Based on an Understanding of Its Molecular Pathogenesis
Previous Article in Journal
Growth Hormone (GH) and Gonadotropin-Releasing Hormone (GnRH) in the Central Nervous System: A Potential Neurological Combinatory Therapy?
Previous Article in Special Issue
Human MHC-II with Shared Epitope Motifs Are Optimal Epstein-Barr Virus Glycoprotein 42 Ligands—Relation to Rheumatoid Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effect of Triptolide in Rheumatoid Arthritis: From Basic Research towards Clinical Translation

1
Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
2
Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
3
School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu 610031, China
4
School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(2), 376; https://doi.org/10.3390/ijms19020376
Submission received: 27 December 2017 / Revised: 21 January 2018 / Accepted: 23 January 2018 / Published: 26 January 2018
(This article belongs to the Special Issue Research of Pathogenesis and Novel Therapeutics in Arthritis)

Abstract

:
Triptolide (TP), a major extract of the herb Tripterygium wilfordii Hook F (TWHF), has been shown to exert potent pharmacological effects, especially an immunosuppressive effect in the treatment of rheumatoid arthritis (RA). However, its multiorgan toxicity prevents it from being widely used in clinical practice. Recently, several attempts are being performed to reduce TP toxicity. In this review, recent progress in the use of TP for RA, including its pharmacological effects and toxicity, is summarized. Meanwhile, strategies relying on chemical structural modifications, innovative delivery systems, and drug combinations to alleviate the disadvantages of TP are also reviewed. Furthermore, we also discuss the challenges and perspectives in their clinical translation.

Graphical Abstract

1. Introduction

Rheumatoid arthritis (RA) is an immune-related disease that generally gives rise to continuous joint destruction, decreased expectancy of life and work ability, considerable disability, and even raised mortality [1]. Disease-modifying anti-rheumatic drugs (DMARDs), such as conventional synthetic DMARDs (csDMARDs) and biological DMARDs (bDMARDs), are currently the most commonly used drugs for treating RA. However, these drugs can not cure RA completely and often bring about severe side effects, such as infection and malignancies. Moreover, bDMARDs have low cost-effectiveness and bring a huge financial burden to the patients. Thus, it is still an imperative mission for researchers to find safer and more cost-effective medications.
Traditional Chinese medicine (TCM), as an important kind of complementary and alternative medicine, is a precious resource for finding cost-efficient drugs, such as artemisinin. As for RA, there are many Chinese herbs with excellent immunosuppressive and anti-inflammatory functions [2]. Tripterygium wilfordii Hook F (TWHF) is a case in point. Tripterygium glycosides, extracted from TWHF, have been widely used to treat RA in China [3]. As the main active ingredient in Tripterygium glycosides, Triptolide (TP, a dierpene triepoxide in chemical structure, see Figure 1) has been considered as a promising anti-RA drug [4]. Increasing experimental evidence has verified its anti-RA effect. TP can significantly alleviate the severity of collagen-induced arthritis (CIA) in rats, with not only a potent anti-inflammatory effect but also the ability to prevent bone destruction [5,6]. Because of its outstanding anti-RA effect, TP has a great application potential in the clinic. Nonetheless, TP also exerts extreme toxicity and has poor water solubility, which impede its clinical application. Fortunately, many promising attempts for its clinical translation have been performed by researchers.
Thus, on the one hand, in order to gain a comprehensive and deep understanding of TP’s pharmacodynamic effect and toxicity in RA, related studies were summarized and reviewed in this paper; on the other hand, we also focused on the clinical translation researches of TP in RA hoping to get a better grasp of the progress in this area and provide proper directions and suggestions for its further study.

2. Effect and Mechanisms of Triptolide (TP) in Rheumatoid Arthritis (RA)

As a chronic immune-mediated inflammatory disease, immune regulatory factors play vital roles in the pathogenesis of RA. Until now, the anti-RA properties of TP in this condition have been attributed to its immunosupressive and antiproliferative effect (Figure 2).

2.1. Regulation of Immunological Functions

2.1.1. Regulation of Immune-Related Cells

T cells are among the key regulators of synovial inflammation in the development of RA, having both stimulatory and inhibitory roles [7] and playing a destructive or a protective role in bone metabolism in a context- and subtype-dependent manner [8]. TP was effective in preventing T cells proliferation [9]. CD4+ T cells play an important role in the induction and development of CIA, and CD8+ T cells might have a suppressive role in the etiology of CIA [10]. Previous studies showed that TP could increase CD8+ cells, while it decreased CD4+ cells in the Peyer’s patch. Therefore, the effect of TP on Peyer’s patch immune cells might partially explain some of the immunosuppressive activities of TP [11,12]. In addition, the overexpression of T cell receptor (TCR) variable gene (V gene) fragments can cause the activation and infiltration of autoreactive T cells. Nevertheless, TP was found to decrease the expression levels of TCR BV15 and TCR BV19. These changes might help explain the effectiveness of TP in the treatment of RA [13].
Th17 cells, a more recently characterized subset of CD4+ T cells, were shown to be more osteoclastogenic [8] and play an important role in the pathogenesis of RA through the production of Th17 signature cytokines [14]. Interleukin (IL)-6 and transforming growth factor (TGF)-β in mice or TGF-β and inflammatory cytokines in human are recognized as crucial factors necessary for the differentiation of naïve T cells into Th17 cells [14,15]. In vivo, TP significantly suppressed the production of Th17 cells from murine splenocytes and purified CD4+ T cells. Importantly, TP could inhibit the transcription of IL-17 mRNA and IL-6-induced phosphorylation of signal transducers and activators of transcription (STAT)3, which is a key signaling molecule involved in the development of Th17 cells. In vitro, TP reduced the production of collagen type II (CII)-specific IL-17 and the percentages of CII-specific IL-17+ CD4+ T cells in draining lymph nodes and spleens in CIA mice [16].
The dendritic cell (DC) is the most potent professional antigen-presenting cell (APC). Immature DCs (iDCs) have the ability to capture and process antigens in inflammatory tissues and undergo phenotypic and functional maturation implying the production of cytokines and chemokines in inflammatory microenvironments. Mature DCs produce multiple chemokines which act as chemoattractants for T cells, B cells, natural killer (NK) cells, and even neutrophils [17,18,19]. Therefore, DC is also regarded as an important target of immunosuppressants. Recently, research indicated that TP treatment inhibited lipopolysaccharide (LPS)-induced phenotypic changes and maturation of DCs [20,21]. TP also prevented the differentiation of immature human monocytes (MoDC) by inhibiting CD1a, CD40, CD80, and CD86 expression and upregulating CD14 expression [22]. In addition, the ability of DCs to stimulate allogeneic T cell responses was also impaired by TP. Furthermore, the production of IL-10 and IL-12 by DCs was modulated after TP treatment [20]. Yan et al. study indicated that TP might induce splenic DCs to CD11clow differentiation, followed by shifting of Th1 to Th2 in vitro [23]. Cao et al. [24] conducted a study to investigate whether TP can inhibit DC-mediated chemoattraction of immune cells, because DC and chemokines are all important mediators in linking innate immunity and adaptive immunity. They found that TP impaired DC-mediated chemoattraction of neutrophils and T cells. Additionally, TP inhibited LPS-induced DC production of chemokines such as macrophage inflammatory protein (MIP)-1α, MIP-1β, monocyte chemoattractant protein (MCP)-1, regulated upon activation normal T cell expressed and secreted (RANTES), and interferon-induced protein 10 (IP-10) via suppression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation and STAT3 phosphorylation. These data provided new insights into TP immunopharmacology.

2.1.2. Regulation of Immune-Related Inflammatory Mediators

As RA is a complicated disease caused by a variety of factors, the inflammatory response has been considered as the main protracted cause of RA. The process of inflammation is usually tightly regulated by both mediators that initiate and maintain inflammation and mediators that shut the process down [25]. In states of chronic inflammation, an imbalance between the two types of mediators leaves inflammation unchecked, which leads to cellular damage. Previous studies have demonstrated that proinflammatory cytokines and chemokines produced by infiltrating immune cells and synoviocytes are implicated in the pathogenesis of RA. Meanwhile, plenty of cytokines and chemokines are also found in the synovial fluid of RA patients [26]. These cytokines and chemokines play an essential role in synovitis, pannus formation, and joint destruction caused by RA [27,28,29,30]. Previous studies showed that TP could lower the level of tumour necrosis factor (TNF)-α, IL-1β, IL-6, nuclear factor (NF)-κB, and cyclooxygenase (COX)-2 in ankle joints and serum in CIA rats [5,31]. Meanwhile, in LPS-induced mouse macrophages, TP could induce the reduction of toll-like receptor 4 (TLR4) proteins and of TIR-domain-containing adapter-inducing interferon-β (TRIF) adapter proteins in the MyD88-independent pathway of TLR4, confirming that both MyD88- and TRIF-mediated NF-κB activation might be suppressed by TP [32]. Moreover, TP decreased C-C chemokine receptor type 5 (CCR5) protein and mRNA levels in synovial tissue of adjuvant-induced arthritis (AIA) rats [33]. Except for CCR5, the overexpression of MCP-1, MIP-1α, and RANTES were also downregulated in TP-treated AIA rats [34]. Additionally, TP could inhibit prostaglandin (PG) E [2] production via a selective suppression of the production and gene expression of COX-2 in CIA rats [35]. Simultaneously, Wang et al. reported that TP could inhibit the production of nitric oxide (NO) by decreasing inducible NO synthase gene transcription [36]. Triggering receptor expressed on myeloid cells (TREM)-1 is a member of the Ig superfamily, and its activation can result in an inflammatory reaction [37,38]. We learned that the expression of TREM-1 could be activated by TLR through LPS, which could further lead to the production of proinflammatory cytokines via the NF-κB pathway [39,40]. Our study indicated that TP could significantly inhibit TREM-1 expressions in CIA rats, as well as decrease the production of TREM-1 in LPS-stimulated U937 cells, which demonstrated that TP could modulate the TREM-1 signaling pathway to inhibit the inflammatory response in RA [5]. TP suppressed TNF-α-induced expression of the IL-1β, IL-6, and IL-8 in fibroblast-like synoviocytes (FLSs) [41]. Treatment with TP also decreased the activation of matrix metalloproteinase (MMP)-3, MMP-9, MMP-13, and the cytoskeleton rearrangement of RA FLSs [42,43]. Moreover, TP not only decreased the IL-1α-induced production of proMMP-1 and 3, but also suppressed their messenger RNA (mRNA) levels in human RA FLSs. Conversely, the expression of tissue inhibitors of metalloproteinases (TIMPs) 1 and 2 induced by IL-1α was augmented by TP in the synovial cells [44]. In phorbol 12-myristate 13-acetate (PMA)-stimulated RA, the expression of IL-18 and IL-18 receptor (IL-18R) at protein and gene levels FLSs were also reduced by TP [45].
While some cytokines initiate and maintain the inflammatory process, others dampen it. The two best studied anti-inflammatory cytokines are IL-10 and IL-4. These cytokines cooperate to inhibit the production of inflammatory cytokines in vitro [46,47]. Xu et al. reported that TP could enhance the expression of IL-10 in regulatory T cells (Tregs) and further suppress osteoclast formation and bone resorption [6], and in vivo data revealed that the level of IL-10 was increased in the TP treatment group compared with the CIA group [13].

2.1.3. Regulation of Immune-Related Angiogenesis

In the development of RA, blood vessel proliferation is common because of the influence of angiogenesis factors and angiogenic activators, like vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, and hepatocyte growth factor in the inflamed and hypoxic environment. Angiogenesis is indispensable in perpetuating immune and inflammatory responses and can foster the infiltration of inflammatory cells into the joints, resulting in synovial hyperplasia and progressive bone destruction [48,49,50,51]. Previous studies suggested that TP could markedly reduce the capillary and the small, medium, and large vessel density in synovial membrane tissues of inflamed joints, and inhibit the expression of VEGF in the sera of CIA rats. The levels of VEGF, vascular endothelial growth factor receptor (VEGFR), Angiopoietin (Ang)-1, Ang-2, and IL-17 in the supernatants of human RA FLSs and human umbilical vein endothelial cells (HUVEC) were also decreased after TP treatment. These results implied that TP might possess an anti-angiogenic effect in RA both in vivo and in in vitro assay systems [52,53].

2.1.4. Regulation of Immune-Related Bone Homeostasis

As an autoimmune disease characterized by inflammation and bone loss, bone homeostasis, which involves bone formation mediated by osteoblasts and bone resorption regulated by osteoclasts, is disrupted in the pathological condition of RA. The bone loss and joint destruction are mediated by immunological insults by various immune cells and inflammatory cytokines. The bone destruction that occurs in RA is also regulated by the receptor activator of nuclear factor-κB (RANK) and its ligand (RANKL), simultaneously [8]. Liu et al. found that TP could upregulate the bone mineral density (BMD), bone volume fraction, and trabecular thickness of inflamed joints and downregulate the trabecular separation, which suggests a protective role of TP on the volume and quality of the preserved trabecular bone despite joint inflammation [54]. Meanwhile, TP could significantly reduce the expression of RANKL and RANK, enhance the level of osteoprotegerin (OPG) in joints and sera of CIA rats, as well as decrease RANKL and RANK level and increase OPG production in the coculture system of human FLSs and peripheral blood mononuclear cells (PBMCs), which further revealed that TP might attenuate RA in part by preventing bone destruction, and inhibit osteoclast formation by regulating the RANKL–RANK–OPG signaling pathway [54]. Another study showed that the protective effects of TP on the joint destruction seen in RA might be associated with its inhibitory effect on the aggression of RA FLSs by blocking c-Jun N-terminal kinase (JNK) activation [42]. Furthermore, Tregs secrete cytokines like IL-10 and TGF-β1 that appear to play a key role in suppressing the differentiation of osteoclasts and the resorption of bone [55]. Research by Xu et al. indicated that TP could enhance the expression of IL-10 and TGF-β1 secreted by Tregs in vitro, which further inhibit osteoclast formation and bone resorption [6]. In another study, TP was found be able to reverse TNF-α-associated suppression of osteoblast differentiation, suggesting that TP might have a positive effect on bone remodeling [56].

2.2. Regulation of Cell Proliferation

Accumulating research suggests that FLSs contribute to synovial inflammation and joint destruction [57,58,59]. They play a crucial part in the initial stages of synovitis through the local production of proinflammatory cytokines and small-molecule mediators of inflammation [7,59]. TP could inhibit the proliferation of FLSs, arrest the cycle of FLSs, and induce apoptosis of FLSs [41,60]. In addition, the migration of FLSs to the cartilage and bone is regarded as a critical process in cartilage destruction in RA [59]. Yang et al. demonstrated that TP could suppress the migration and invasion of RA FLSs by partially blocking the phosphorylation of the JNK pathway [42].
Macrophages are found in the synovial membrane and are central effectors of synovitis. Macrophages act through the release of cytokines such as TNF-α and IL-1 [7]. TP treatment could result in macrophage apoptosis, while no obvious necrosis occurred [61]. The level of TNF-α in LPS-induced macrophages could be decreased by TP [62].

3. Mechanisms of TP Toxicity

Despite TP remarkable effect on RA, an increasing number of studies demonstrated that TP could induce toxicity, including hepatotoxicity, nephrotoxicity, reproductive toxicity, and so on.

3.1. Hepatotoxicity

To evaluate the liver injury effect of TP, the serum activities of alanine transaminase (ALT), aspartate transaminase (AST), and lactic dehydrogenase (LDH) were used as biochemical markers. One study on C57BL/6 mice reported the time-dependent hepatotoxicity of TP, accompanied by an increasing trend of AST and ALT in the serum at 6 and 12 h, a peak at 24 h after TP (600 mg/kg) administration, and a decrease after 24 h [63]. Another study showed that ALT, AST, and LDH activities in serum were multiplied by 9.1, 9.8, and 3.0, respectively, which occurred in BALB/C mice treated only with TP (1.0 mg/kg) but not in control groups [64]. Additionally, the livers of TP-treated (0.5 mg/kg) mice showed hyperemic, mottled, fragile, and fuzzy structures, hepatocytes' nuclei displayed pyknosis and ruptures, and cytoplasmic staining was uneven with slight cell damage [65]. In contrast, after giving TP (0.1, 0.3 mg/kg) through intravenous administration once daily for 14 days, AST activity in the serum of Wistar rats significantly decreased as the TP dose increased, but there was no significant change in ALT [66]. Moreover, TP (200–400 μg/kg, 28 days) induced mitochondrial membrane depolarization in female Sprague Dawley (SD) rats, resulting in liver damage with microvesicular steatosis and hyperlactacidaemia, and was accompanied by an augmentation in reactive oxygen species (ROS) [67]. In addition, an abnormal immune response can induce organ or tissue damage influenced by CD4+ T cells such as Th17 and Tregs. Recently, Wang et al. reported that TP (500 μg/kg for 24 h) elevated the Th17/Treg ratio, which was positively correlated with ALT and AST in the serum, as well as acute liver injury of female C57BL/6 mice [63]. Recently, Yang and her colleagues found that the intragastric administration of TP (400 μg/kg body weight, 28 days) increased serum total bile acid and ALP levels and suppressed hepatic gluconeogenesis in Wistar rats, indicating that TP induced hepatotoxicity, and this hepatotoxicity was related to the sirtuin (Sirt1)/farnesoid X receptor (FXR) signaling pathway [68]. Simultaneously, Lu et al. suggested that TP could cause hepatotoxicity by reducing substrate affinity, activity, and expression of the CYP450 isoforms 3A, 2C9, 2C19, and 2E1 [69].

3.2. Nephrotoxicity

To estimate the nephrotoxicity of TP, blood urea nitrogen (BUN) and creatinine (Cr), which are important biochemical parameters in the serum, were used. Yang et al. reported that TP could cause a significant reduction of renal function characterized by a remarkable upregulation of Cr and BUN concentrations. Research about the relationship between TP-induced nephrotoxicity and oxidative stress indicated that TP caused serious oxidative stress after a single dose of 1 mg/kg in male SD rats, decreased the activities of renal superoxide dismutase (SOD) and glutathione (GSH), increased the level of malondialdehyde (MDA) and BUN, and caused structural damage [70]. In the meantime, TP induced severe damage in the renal structure, characterized by tubular epithelial cell detachment, necrosis, and tubular obstruction [71]. Furthermore, renal glomeruli were hyperemic, swelling, scattered, and necrotic after TP treatment [65].

3.3. Reproductive Toxicity

Except for hepatotoxicity and nephrotoxicity, toxicity for the reproductive system and an antifertility effect were also obvious. In female reproductive toxicity studies, TP caused prolonged estrous cycles and reduced the relative weights of the ovary and uterus [72]. In male reproductive toxicity studies, after treating with TP, the testis and epididymis weights were severely decreased. The cauda epididymis sperm content and motility even decreased to zero [73]. Studies have demonstrated that TP toxicity to the reproduction system emerged mainly through a disruption of the normal androgen and estrogen signaling [74]. Estrogen synthesis enzymes, aromatase and steroidogenic regulatory protein, play important roles in estradiol synthesis and estrogen signaling. TP could disrupt the expression of these three key proteins leading to estradiol synthesis reduction and reproductive dysfunction [75]. Intracellular ROS, glutathione peroxidase (GPx), and SOD are very important for testosterone generation. Studies found that TP had an influence on ROS, GPx, and SOD resulting in testosterone reduction. It was also found that TP could induce direct cytotoxicity in Leydig cells [76].

3.4. Further Toxicity

It is widely known that TP could cause reproductive toxicity, liver damage, and renal injury. However, TP could also lead to damage in other organs. TP acute poisoning could cause acute myocardial damage, such as myocardium swelling, denaturation, cytolysis, and contraction band necrosis. This toxicological effect of TP might be closely related to mitochondria and cell membrane functions [77]. Furthermore, there was also injury to the spleen after long-term TP administration. As an inflammation inhibitor, a long-time usage of TP could cause immunotoxicity in the spleen. Increased spleen index, spleen volume, and spleen weight could be seen in impaired spleens [66]. Gastrointestinal tract symptoms, such as nausea, anorexia, vomiting, diarrhea, gastrointestinal ulcers, and bleeding, were also a result of adverse reactions to TP [78]. In the meantime, TP could induce hematologic toxicity. In hepatic P450-deficient mice, the total number of platelets (PLT) and the number of white blood cells were reduced after TP treatment (0.5, 1.0 mg/kg). TP also decreased the absolute number and percent of lymphocytes, while it increased the absolute number and percent of neutrophils to a concentration of 1.0 mg/kg. There was no difference in the levels of red blood cells (RBC) or hemoglobin (Hb) after TP treatment [79]. Scientists confirmed that P450s was responsible for the metabolism of TP in the liver. P450s deficiency might cause an increase in the bioavailability and toxicity of TP [79]. In the study of Liu et al., TP (200 and 400 mg/kg/day for 28 days) showed a reduced toxicity and a higher metabolic rate in male SD rats linked to CYP3A2 which was the main metabolic isozyme in male rats, revealing the importance of CYP3A2 on the sex-based differences in TP toxicity [80]. Although there was no clear explanation of the effects of TP toxicity on RA, this research provided novel directions for further studies on TP toxicity.

4. Translational Research of TP

As mentioned above, the potent immunosuppressive and antiproliferative effects make TP a promising drug for clinical RA therapy. At the same time, its high toxicity as well as its poor water solubility greatly hinder TP’s clinical applications [73,81]. In order to improve the characteristics of TP, strategies relying on chemical structural modifications, innovative delivery systems, and drug combinations are increasingly employed by researchers [65,82,83].

4.1. Chemical Structural Modifications of TP

Many drugs like TP exert excellent therapeutic effects while simultaneously causing dramatic toxicity and displaying poor water solubility. Certain chemical properties of a compound can be changed by modifying its chemical structure. These modifications may be employed to increase water solubility or decrease the toxicity of a drug, thus making it available for clinical use. Over the past decades, several TP analogs (Table 1) have been developed and evaluated, mainly including (5R)-5-hydroxytriptolide (LLDT-8) [84], PG490-88 [85], LLDT-67 [86], LLDT-288 [87], and so on. Among these derivatives, LLDT-8 has comparable immunosuppressive and anti-inflammatory functions and a much lower toxicity compared to TP [88]. Its effects on RA have been proved by preclinical tests and Phase I clinical trials in RA patients [88,89]. With regard to its mechanism of action, LLDT-8 is thought to inhibit the activation of macrophages and regulate T cells proliferation and function [90,91].

4.2. Innovative Delivery System

Drugs with poor solubility in water have trouble dissolving in the gastrointestinal tract, engendering a low bioavailability. Some innovative delivery systems, like those obtained through nanotechnology and microemulsions, can be employed to enhance the delivery efficiency of medications [94,95]. Hence, studies of TP delivered by liposomes, nanoparticles, solid lipid nanoparticles, and microemulsions are summarized below and listed in Table 2.

4.2.1. Liposomes

Chen et al. [96] developed a TP-loaded liposome hydrogel patch (TP-LHP) which was proved to improve the bioavailability of TP because of its stable and long-term release. Similar to TP, TP-LHP showed significant efficacy in CIA rats. Moreover, TP was delivered transdermally in this study, which can avoid the first-pass effects on the liver and abate gastrointestinal toxicity.

4.2.2. Nanoparticles

Nanocarriers can reduce the side effects and increase the delivery efficiency of many drugs. Poly-γ-glutamic acid (γ-PGA) has been reported to be a promising drug carrier. Zhang and his colleagues created a nanodrug carrier system called γ-PGA-l-PAE-TP (PPT) by wrapping TP in a poly-γ-glutamicacid-grafted l-phenylalanine ethylester copolymer. PPT demonstrated controlled release behavior. This research indicated that PPT could alleviate free TP toxicity on murine macrophage RAW264.7 cells and normal C57/B6 mice. The nanodrug carrier system showed broad application prospects in RA treatment [97].
Poly(d,l-lacticacid) nanoparticles were used as TP carrier by Liu group. They fabricated TP-loaded poly(d,l-lacticacid) nanoparticles (TP-PLA-NPs) through the spontaneous emulsification solvent diffusion method with modifications. This delivery system caused TP to be burst-released initially and slow-released subsequently. In vivo tests demonstrated the significant inhibition effect of TP-PLA-NPs on AIA rats [98]. Furthermore, another study demonstrated that TP-PLA-NPs could effectively lower renal toxicity in rats [99].

4.2.3. Solid Lipid Nanoparticles

Solid lipid nanoparticles (SLNs) were introduced as an innovative drug delivery system at the beginning of the 1990s. This system has become a promising alternative to liposomes, polymeric nanoparticles, and so on because of its merits, like nontoxicity, excellent biocompatibility, as well as large-scale production possibilities [106]. The solid matrix of SLNs can protect the loaded drug from degrading in the gastrointestinal tract [78,107]. SLNs can be employed in both topical application and oral administration. Mei et al. [100,101,102] found that SLNs could efficiently promote TP penetration into the skin. Furthermore, they also confirmed the anti-inflammatory effect of SLNs on carrageenan-induced rats as well as AIA rats, with improved safety and minimized toxicity compared to TP. Another research group compared the toxicokinetics and tissue distribution of TP-SLN versus free TP in rats, and the results suggested that TP-SLN enhanced TP absorption, with a slow release which may contribute to boost TP efficacy. Tissue distribution results showed that TP-SLN was more distributed in the lung and spleen than in plasma, liver, kidney, and testes. This explained why TP-SLN could mitigate the genital toxicity of TP [103].

4.2.4. Microemulsions

Microemulsions are increasingly used for the transdermal delivery of drugs because of their several advantages, such as enhanced efficacy in transdermal applications over conventional formulations, elevated drug solubility, and ease of manufacturing [108]. A previous study prepared TP-loaded microemulsions and proved that they could penetrate in vitro through the mouse skin without obvious irritation to the skin [109]. Furthermore, Xu et al. [105] developed a kind of TP-loaded hydrogel-thickened microemulsion (TP-MTH) to treat RA through transdermal delivery. They testified its good effects without apparent local and systemic toxicities.

4.3. Drug Combinations

In the clinic, it has been found that drug combinations could be a good choice to solve drug toxicity. Drug combinations use several drugs that interact with multiple targets in the molecular networks of a disease and, in practice, may achieve better efficacy and lower toxicity than monotherapies. Thus, drug combinations can produce a synergistic effect without increased toxicity [110]. To solve TP toxicity, scientists have already found some drug, such as glycyrrhetinic acid and silymarin, which could produce a synergistic therapeutic effect, detoxication, or both.

4.3.1. Glycyrrhetinic Acid

During the process of RA treatment, Licorice (Glycyrrhiza glabra L.) was often used combined with TWHF or TWHF preparations to reduce the latter’s adverse effects. Glycyrrhizin (GL) was considered a main active component of Licorice. Research showed that a combination of GL and TP could reduce the side effects of TP. The detoxifying effect of GL on TP was considered inseparable from GL’s selective influence on cytochrome P4503A (CYP3A). CYP3A, a major Phase I xenobiotic metabolizing enzyme, is responsible for regulating the metabolism of TP in the liver, avoiding the accumulation of TP [111]. By activating CYP3A, GL could accelerate the metabolism of TP and reduce the body exposure to TP. This suggested a significant protective action against chronic liver injury in rats [82]. In addition, many studies have reported that both GL and TP have an anti-inflammatory effect [112,113]. Furthermore, GL combined with TP produced a synergistic anti-inflammatory effect [114].
GL dissolves in water and transforms into glycyrrhetinic acid (GA), which is an important active ingredient with pharmacological properties [115]. Pharmacokinetic studies found that an extensive accumulation of TP in the liver caused liver damage [116]. This kind of liver damage could be reduced by the combination of GA and TP. The possible mechanism is that GA could reduce TP accumulation by promoting TP hepatic metabolic clearance. Several studies proved that GA could promote TP hepatic metabolic clearance, and this action was closely related to P-glycoprotein (P-gp) [117,118,119].

4.3.2. Silymarin

The excessive release of inflammatory mediators could lead to immunological injury. TP combined with silymarin produced synergistic anti-inflammatory effects when treating inflammatory diseases like RA [35]. Silymarin is an active ingredient of Silybum marianum and it was reported to have various pharmacological functions. Silymarin was often used, alone or as a major component of various pharmaceutical preparations, as a hepatoprotective agent clinically. Additionally, silymarin has also exhibited protective effects against inflammation [120]. Short-term oral administration of silymarin exerted protective effects on TP-induced liver injury. The combination of silymarin and TP could produce a synergistic immunosuppressive effect by reducing the excessive expression of proinflammatory cytokines and inhibiting inflammatory signaling [121].

5. Discussion and Further Perspectives

Here, in this review, we examined the research on the pharmacodynamic effects, toxicity, and clinical translation of TP in RA. An increasing number of preclinical studies have testified the immunosuppressant, anti-inflammatory, and antiproliferative effects of TP which scientifically explain its good clinical effect on RA. Additionally, TP toxicity in RA is also increasingly studied. By analyzing a series of reports, we speculated that the potential primary effect of TP in RA might be achieved via its immunosuppressive property. As RA is a systemic disease, the effective and toxic mechanisms of TP in RA still need deep investigation. Perhaps, bioinformatic methods rising recently can be exploited to explore TP pharmacodynamics and toxicological mechanisms from a more systematic point of view.
In terms of the clinical translation of TP, several problems should be raised here. Firstly, we found that several derivatives of TP were synthesized and proven to possess effects comparable to those of TP and are even currently able to enter clinical trials. However, most of the derivatives are studied for cancer with only one of them used to treat RA. Furthermore, only LLDT-8 is still awaiting the outcomes of the clinical tests, although it showed promising anti-RA effects in preclinical studies. In addition, with regard to innovative delivery systems, targeted drug deliveries are becoming more and more popular because of their specific targeting of certain organs or cells. Nevertheless, the current targeted delivery systems of TP are mostly renal-targeted and tumor-targeted. For example, 3,5-dipentadecyloxybenzamidine hydrochloride (TRX-20)-modified liposomes [122], PF-A299–585 [123], 2-Glucosamine [124], and lysozyme [125] were reported to specifically deliver TP to the kidney. Carbonic anhydrase IX (CA IX) [126], AS1411 [127], and nanoformulations coated with folate [128] were used to specifically deliver TP to lung cancer, pancreatic cancer, and hepatocellular carcinoma cells, respectively. Investigations using targeted delivery system for TP to treat RA are still scarce. Thus, more research is needed to advance the application of TP in RA.

Acknowledgments

This study was supported by the Hong Kong Baptist University Strategic Development Fund (SDF15-0324-P02(b)).

Author Contributions

Danping Fan wrote the manuscript; Qingqing Guo, Jiawen Shen, Kang Zheng, and Cheng Lu contributed to the literature research for the manuscript ; Ge Zhang and Aiping Lu revised the manuscript; Xiaojuan He revised and approved the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burmester, G.R.; Pope, J.E. Novel treatment strategies in rheumatoid arthritis. Lancet 2017, 389, 2338–2348. [Google Scholar] [CrossRef]
  2. Lu, S.; Wang, Q.; Li, G.; Sun, S.; Guo, Y.; Kuang, H. The treatment of rheumatoid arthritis using Chinese medicinal plants: From pharmacology to potential molecular mechanisms. J. Ethnopharmacol. 2015, 176, 177–206. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, J.; Chu, Y.; Zhou, X. Inhibitory effect of Triperygium wilfordii polyglucoside on dipeptidyl peptidase I in vivo and in vitro. Biomed. Pharmacother. 2017, 96, 466–470. [Google Scholar] [CrossRef] [PubMed]
  4. Han, R.; Rostami-Yazdi, M.; Gerdes, S.; Mrowietz, U. Triptolide in the treatment of psoriasis and other immune-mediated inflammatory diseases. Br. J. Clin. Pharmacol. 2012, 74, 424–436. [Google Scholar] [CrossRef] [PubMed]
  5. Fan, D.; He, X.; Bian, Y.; Guo, Q.; Zheng, K.; Zhao, Y.; Lu, C.; Liu, B.; Xu, X.; Zhang, G. Triptolide Modulates TREM-1 Signal Pathway to Inhibit the Inflammatory Response in Rheumatoid Arthritis. Int. J. Mol. Sci. 2016, 17, 498. [Google Scholar] [CrossRef] [PubMed]
  6. Xu, H.; Zhao, H.; Lu, C. Triptolide Inhibits Osteoclast Differentiation and Bone Resorption In Vitro via Enhancing the Production of IL-10 and TGF-β1 by Regulatory T Cells. Mediat. Inflamm. 2016, 2016, 8048170. [Google Scholar] [CrossRef] [PubMed]
  7. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [PubMed]
  8. Jung, S.M.; Kim, K.W.; Yang, C.W.; Park, S.H.; Ju, J.H. Cytokine-mediated bone destruction in rheumatoid arthritis. J. Immunol. Res. 2014, 2014, 263625. [Google Scholar] [CrossRef] [PubMed]
  9. Chan, M.A.; Kohlmeier, J.E.; Branden, M.; Jung, M.; Benedict, S.H. Triptolide is more effective in preventing T cell proliferation and interferon-gamma production than is FK506. Phytother. Res. 1999, 13, 464–467. [Google Scholar] [CrossRef]
  10. Mellado, M.; Martinez-Munoz, L.; Cascio, G.; Lucas, P.; Pablos, J.L.; Rodriguez-Frade, J.M. T Cell Migration in Rheumatoid Arthritis. Front. Immunol. 2015, 6, 384. [Google Scholar] [CrossRef] [PubMed]
  11. Xiao, C.; Zhao, L.; Liu, Z.; Lu, C.; Zhao, N.; Yang, D.; Chen, S.; Tang, J.C.; Chan, A.; Lu, A.P. The effect of triptolide on CD4+ and CD8+ cells in the Peyer’s patch of DA rats with collagen induced arthritis. Nat. Prod. Res. 2009, 23, 1699–1706. [Google Scholar] [CrossRef] [PubMed]
  12. Zhou, J.; Xiao, C.; Zhao, L.; Jia, H.; Zhao, N.; Lu, C.; Yang, D.; Tang, J.C.; Chan, A.S.; Lu, A.P. The effect of triptolide on CD4+ and CD8+ cells in Peyer’s patch of SD rats with collagen induced arthritis. Int. Immunopharmacol. 2006, 6, 198–203. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, J.; Wang, A.; Zeng, H.; Liu, L.; Jiang, W.; Zhu, Y.; Xu, Y. Effect of triptolide on T-cell receptor beta variable gene mRNA expression in rats with collagen-induced arthritis. Anal. Rec. 2012, 295, 922–927. [Google Scholar] [CrossRef] [PubMed]
  14. Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and type 17 helper T cells. N. Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef] [PubMed]
  15. Volpe, E.; Servant, N.; Zollinger, R.; Bogiatzi, S.I.; Hupe, P.; Barillot, E.; Soumelis, V. A critical function for transforming growth factor-β, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses. Nat. Immunol. 2008, 9, 650–657. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, Y.; Jia, L.; Wu, C.Y. Triptolide inhibits the differentiation of Th17 cells and suppresses collagen-induced arthritis. Scand. J. Immunol. 2008, 68, 383–390. [Google Scholar] [CrossRef] [PubMed]
  17. Morelli, A.E.; Thomson, A.W. Dendritic cells: Regulators of alloimmunity and opportunities for tolerance induction. Immunol. Rev. 2003, 196, 125–146. [Google Scholar] [CrossRef] [PubMed]
  18. Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef] [PubMed]
  19. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of dendritic cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, Y.; Chen, Y.; Lamb, J.R.; Tam, P.K. Triptolide, a component of Chinese herbal medicine, modulates the functional phenotype of dendritic cells. Transplantation 2007, 84, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
  21. Chen, X.; Murakami, T.; Oppenheim, J.J.; Howard, O.M. Triptolide, a constituent of immunosuppressive Chinese herbal medicine, is a potent suppressor of dendritic-cell maturation and trafficking. Blood 2005, 106, 2409–2416. [Google Scholar] [CrossRef] [PubMed]
  22. Zhu, K.J.; Shen, Q.Y.; Cheng, H.; Mao, X.H.; Lao, L.M.; Hao, G.L. Triptolide affects the differentiation, maturation and function of human dendritic cells. Int. Immunopharmacol. 2005, 5, 1415–1426. [Google Scholar] [CrossRef] [PubMed]
  23. Yan, Y.H.; Shang, P.Z.; Lu, Q.J.; Wu, X. Triptolide regulates T cell-mediated immunity via induction of CD11c(low) dendritic cell differentiation. Food Chem. Toxicol. 2012, 50, 2560–2564. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, Q.; Chen, T.; Chen, G.; Li, N.; Wang, J.; Ma, P.; Cao, X. Immunosuppressant triptolide inhibits dendritic cell-mediated chemoattraction of neutrophils and T cells through inhibiting Stat3 phosphorylation and NF-κB activation. Biochem. Biophys. Res. Commun. 2006, 345, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  25. Choy, E.H.; Panayi, G.S. Cytokine pathways and joint inflammation in rheumatoid arthritis. N. Engl. J. Med. 2001, 344, 907–916. [Google Scholar] [CrossRef] [PubMed]
  26. Iwamoto, T.; Okamoto, H.; Toyama, Y.; Momohara, S. Molecular aspects of rheumatoid arthritis: Chemokines in the joints of patients. FEBS J. 2008, 275, 4448–4455. [Google Scholar] [CrossRef] [PubMed]
  27. Kim, W.U.; Kwok, S.K.; Hong, K.H.; Yoo, S.A.; Kong, J.S.; Choe, J.; Cho, C.S. Soluble Fas ligand inhibits angiogenesis in rheumatoid arthritis. Arthritis Res. Ther. 2007, 9, R42. [Google Scholar] [CrossRef] [PubMed]
  28. Harris, E.D., Jr. Rheumatoid arthritis. Pathophysiology and implications for therapy. N. Engl. J. Med. 1990, 322, 1277–1289. [Google Scholar] [PubMed]
  29. Sivalingam, S.P.; Thumboo, J.; Vasoo, S.; Thio, S.T.; Tse, C.; Fong, K.Y. In vivo pro- and anti-inflammatory cytokines in normal and patients with rheumatoid arthritis. Ann. Acad. Med. Singap. 2007, 36, 96–99. [Google Scholar] [PubMed]
  30. Koch, A.E. Angiogenesis as a target in rheumatoid arthritis. Ann. Rheum. Dis. 2003, 62, ii60–ii67. [Google Scholar] [CrossRef] [PubMed]
  31. Xiao, C.; Zhou, J.; He, Y.; Jia, H.; Zhao, L.; Zhao, N.; Lu, A. Effects of triptolide from Radix Tripterygium wilfordii (Leigongteng) on cartilage cytokines and transcription factor NF-κB: A study on induced arthritis in rats. Chin. Med. 2009, 4, 13. [Google Scholar] [CrossRef] [PubMed]
  32. Premkumar, V.; Dey, M.; Dorn, R.; Raskin, I. MyD88-dependent and independent pathways of Toll-Like Receptors are engaged in biological activity of Triptolide in ligand-stimulated macrophages. BMC Chem. Biol. 2010, 10, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Yifan, W.; Dengming, W.; Zheng, L.; Yanping, L.; Junkan, S. Triptolide inhibits CCR5 expressed in synovial tissue of rat adjuvant-induced arthritis. Pharmacol. Rep. 2007, 59, 795–799. [Google Scholar] [PubMed]
  34. Wang, Y.; Wei, D.; Lai, Z.; Le, Y. Triptolide inhibits CC chemokines expressed in rat adjuvant-induced arthritis. Int. Immunopharmacol. 2006, 6, 1825–1832. [Google Scholar] [CrossRef] [PubMed]
  35. Lin, N.; Liu, C.; Xiao, C.; Jia, H.; Imada, K.; Wu, H.; Ito, A. Triptolide, a diterpenoid triepoxide, suppresses inflammation and cartilage destruction in collagen-induced arthritis mice. Biochem. Pharmacol. 2007, 73, 136–146. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, B.; Ma, L.; Tao, X.; Lipsky, P.E. Triptolide, an active component of the Chinese herbal remedy Tripterygium wilfordii Hook F, inhibits production of nitric oxide by decreasing inducible nitric oxide synthase gene transcription. Arthritis Rheum. 2004, 50, 2995–3003. [Google Scholar] [CrossRef] [PubMed]
  37. Bleharski, J.R.; Kiessler, V.; Buonsanti, C.; Sieling, P.A.; Stenger, S.; Colonna, M.; Modlin, R.L. A role for triggering receptor expressed on myeloid cells-1 in host defense during the early-induced and adaptive phases of the immune response. J. Immunol. 2003, 170, 3812–3818. [Google Scholar] [CrossRef] [PubMed]
  38. Bouchon, A.; Dietrich, J.; Colonna, M. Cutting edge: Inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J. Immunol. 2000, 164, 4991–4995. [Google Scholar] [CrossRef] [PubMed]
  39. Choi, S.T.; Kang, E.J.; Ha, Y.J.; Song, J.S. Levels of plasma-soluble triggering receptor expressed on myeloid cells-1 (sTREM-1) are correlated with disease activity in rheumatoid arthritis. J. Rheumatol. 2012, 39, 933–938. [Google Scholar] [CrossRef] [PubMed]
  40. Fortin, C.F.; Lesur, O.; Fulop, T., Jr. Effects of TREM-1 activation in human neutrophils: Activation of signaling pathways, recruitment into lipid rafts and association with TLR4. Int. Immunol. 2007, 19, 41–50. [Google Scholar] [CrossRef] [PubMed]
  41. Su, Z.; Sun, H.; Ao, M.; Zhao, C. Atomic Force Microscopy Study of the Anti-inflammatory Effects of Triptolide on Rheumatoid Arthritis Fibroblast-like Synoviocytes. Microsc. Microanal. 2017, 23, 1002–1012. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, Y.; Ye, Y.; Qiu, Q.; Xiao, Y.; Huang, M.; Shi, M.; Liang, L.; Yang, X.; Xu, H. Triptolide inhibits the migration and invasion of rheumatoid fibroblast-like synoviocytes by blocking the activation of the JNK MAPK pathway. Int. Immunopharmacol. 2016, 41, 8–16. [Google Scholar] [CrossRef] [PubMed]
  43. Liacini, A.; Sylvester, J.; Zafarullah, M. Triptolide suppresses proinflammatory cytokine-induced matrix metalloproteinase and aggrecanase-1 gene expression in chondrocytes. Biochem. Biophys. Res. Commun. 2005, 327, 320–327. [Google Scholar] [CrossRef] [PubMed]
  44. Lin, N.; Sato, T.; 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. 2001, 44, 2193–2200. [Google Scholar] [PubMed]
  45. Lu, Y.; Wang, W.J.; Leng, J.H.; Cheng, L.F.; Feng, L.; Yao, H.P. Inhibitory effect of triptolide on interleukin-18 and its receptor in rheumatoid arthritis synovial fibroblasts. Inflamm. Res. 2008, 57, 260–265. [Google Scholar] [CrossRef] [PubMed]
  46. Van Roon, J.A.; van Roy, J.L.; Gmelig-Meyling, F.H.; Lafeber, F.P.; Bijlsma, J.W. Prevention and reversal of cartilage degradation in rheumatoid arthritis by interleukin-10 and interleukin-4. Arthritis Rheum. 1996, 39, 829–835. [Google Scholar] [CrossRef] [PubMed]
  47. Sugiyama, E.; Kuroda, A.; Taki, H.; Ikemoto, M.; Hori, T.; Yamashita, N.; Maruyama, M.; Kobayashi, M. Interleukin 10 cooperates with interleukin 4 to suppress inflammatory cytokine production by freshly prepared adherent rheumatoid synovial cells. J. Rheumatol. 1995, 22, 2020–2026. [Google Scholar] [PubMed]
  48. Szekanecz, Z.; Koch, A.E. Vascular involvement in rheumatic diseases: “vascular rheumatology”. Arthritis Res. Ther. 2008, 10, 224. [Google Scholar] [CrossRef] [PubMed]
  49. Lainer-Carr, D.; Brahn, E. Angiogenesis inhibition as a therapeutic approach for inflammatory synovitis. Nat. Clin. Pract. Rheumatol. 2007, 3, 434–442. [Google Scholar] [CrossRef] [PubMed]
  50. Koch, A.E. Review: Angiogenesis: Implications for rheumatoid arthritis. Arthritis Rheum. 1998, 41, 951–962. [Google Scholar] [CrossRef]
  51. Veale, D.J.; Fearon, U. Inhibition of angiogenic pathways in rheumatoid arthritis: Potential for therapeutic targeting. Best Pract. Res. Clin. Rheumatol. 2006, 20, 941–947. [Google Scholar] [CrossRef] [PubMed]
  52. Kong, X.; Zhang, Y.; Liu, C.; Guo, W.; Li, X.; Su, X.; Wan, H.; Sun, Y.; Lin, N. Anti-angiogenic effect of triptolide in rheumatoid arthritis by targeting angiogenic cascade. PLoS ONE 2013, 8, e77513. [Google Scholar] [CrossRef] [PubMed]
  53. He, M.F.; Huang, Y.H.; Wu, L.W.; Ge, W.; Shaw, P.C.; But, P.P. Triptolide functions as a potent angiogenesis inhibitor. Int. J. Cancer 2010, 126, 266–278. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, C.; Zhang, Y.; Kong, X.; Zhu, L.; Pang, J.; Xu, Y.; Chen, W.; Zhan, H.; Lu, A.; 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. Altern. Med. 2013, 2013, 626038. [Google Scholar] [CrossRef] [PubMed]
  55. Luo, C.Y.; Wang, L.; Sun, C.; Li, D.J. Estrogen enhances the functions of CD4+CD25+Foxp3+ regulatory T cells that suppress osteoclast differentiation and bone resorption in vitro. Cell. Mol. Immunol. 2011, 8, 50–58. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, S.P.; Wang, G.D.; Du, X.J.; Wan, G.; Wu, J.T.; Miao, L.B.; Liang, Q.D. Triptolide inhibits the function of TNF-α in osteoblast differentiation by inhibiting the NF-κB signaling pathway. Exp. Ther. Med. 2017, 14, 2235–2240. [Google Scholar] [CrossRef] [PubMed]
  57. Cooles, F.A.; Isaacs, J.D. Pathophysiology of rheumatoid arthritis. Curr. Opin. Rheumatol. 2011, 23, 233–240. [Google Scholar] [CrossRef] [PubMed]
  58. Noss, E.H.; Brenner, M.B. The role and therapeutic implications of fibroblast-like synoviocytes in inflammation and cartilage erosion in rheumatoid arthritis. Immunol. Rev. 2008, 223, 252–270. [Google Scholar] [CrossRef] [PubMed]
  59. Bartok, B.; Firestein, G.S. Fibroblast-like synoviocytes: Key effector cells in rheumatoid arthritis. Immunol. Rev. 2010, 233, 233–255. [Google Scholar] [CrossRef] [PubMed]
  60. Kusunoki, N.; Yamazaki, R.; Kitasato, H.; Beppu, M.; Aoki, H.; Kawai, S. Triptolide, an active compound identified in a traditional Chinese herb, induces apoptosis of rheumatoid synovial fibroblasts. BMC Pharmacol. 2004, 4, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Bao, X.; Cui, J.; Wu, Y.; Han, X.; Gao, C.; Hua, Z.; Shen, P. The roles of endogenous reactive oxygen species and nitric oxide in triptolide-induced apoptotic cell death in macrophages. J. Mol. Med. 2007, 85, 85–98. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, F.; Bai, X.J.; Hu, D.; Li, Z.F.; Liu, K.J. Effect of triptolide on secretion of inflammatory cellular factors TNF-α and IL-8 in peritoneal macrophages of mice activated by lipopolysaccharide. World J. Emerg. Med. 2010, 1, 70–74. [Google Scholar] [PubMed]
  63. Wang, X.; Jiang, Z.; Cao, W.; Yuan, Z.; Sun, L.; Zhang, L. Th17/Treg imbalance in triptolide-induced liver injury. Fitoterapia 2014, 93, 245–251. [Google Scholar] [CrossRef] [PubMed]
  64. Li, J.; Shen, F.; Guan, C.; Wang, W.; Sun, X.; Fu, X.; Huang, M.; Jin, J.; Huang, Z. Activation of Nrf2 protects against triptolide-induced hepatotoxicity. PLoS ONE 2014, 9, e100685. [Google Scholar] [CrossRef] [PubMed]
  65. Lu, J.; Jiang, F.; Lu, A.; Zhang, G. Linkers Having a Crucial Role in Antibody-Drug Conjugates. Int. J. Mol. Sci. 2016, 17, 561. [Google Scholar] [CrossRef] [PubMed]
  66. Xu, L.; Qiu, Y.; Xu, H.; Ao, W.; Lam, W.; Yang, X. Acute and subacute toxicity studies on triptolide and triptolide-loaded polymeric micelles following intravenous administration in rodents. Food Chem. Toxicol. 2013, 57, 371–379. [Google Scholar] [CrossRef] [PubMed]
  67. Fu, Q.; Huang, X.; Shu, B.; Xue, M.; Zhang, P.; Wang, T.; Liu, L.; Jiang, Z.; Zhang, L. Inhibition of mitochondrial respiratory chain is involved in triptolide-induced liver injury. Fitoterapia 2011, 82, 1241–1248. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, J.; Sun, L.; Wang, L.; Hassan, H.M.; Wang, X.; Hylemon, P.B.; Wang, T.; Zhou, H.; Zhang, L.; Jiang, Z. Activation of Sirt1/FXR Signaling Pathway Attenuates Triptolide-Induced Hepatotoxicity in Rats. Front. Pharmacol. 2017, 8, 260. [Google Scholar] [CrossRef] [PubMed]
  69. Lu, Y.; Xie, T.; Zhang, Y.; Zhou, F.; Ruan, J.; Zhu, W.; Zhu, H.; Feng, Z.; Zhou, X. Triptolide Induces hepatotoxicity via inhibition of CYP450s in Rat liver microsomes. BMC Complement. Altern. Med. 2017, 17, 15. [Google Scholar] [CrossRef] [PubMed]
  70. Yang, F.; Ren, L.; Zhuo, L.; Ananda, S.; Liu, L. Involvement of oxidative stress in the mechanism of triptolide-induced acute nephrotoxicity in rats. Exp. Toxicol. Pathol. 2012, 64, 905–911. [Google Scholar] [CrossRef] [PubMed]
  71. Yang, F.; Zhuo, L.; Ananda, S.; Sun, T.; Li, S.; Liu, L. Role of reactive oxygen species in triptolide-induced apoptosis of renal tubular cells and renal injury in rats. J. Huazhong Univ. Sci. Technol. 2011, 31, 335–341. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, J.; Jiang, Z.; Liu, L.; Zhang, Y.; Zhang, S.; Xiao, J.; Ma, M.; Zhang, L. Triptolide induces adverse effect on reproductive parameters of female Sprague-Dawley rats. Drug Chem. Toxicol. 2011, 34, 1–7. [Google Scholar] [CrossRef] [PubMed]
  73. Ni, B.; Jiang, Z.; Huang, X.; Xu, F.; Zhang, R.; Zhang, Z.; Tian, Y.; Wang, T.; Zhu, T.; Liu, J.; et al. Male Reproductive Toxicity and Toxicokinetics of Triptolide in Rats. Arzneimittelforschung 2008, 58, 673–680. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, J.; Liu, L.; Mu, X.; Jiang, Z.; Zhang, L. Effect of triptolide on estradiol release from cultured rat granulosa cells. Endocr. J. 2012, 59, 473–481. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, J.; Jiang, Z.; Mu, X.; Wen, J.; Su, Y.; Zhang, L. Effect of triptolide on progesterone production from cultured rat granulosa cells. Arzneimittelforschung 2012, 62, 301–306. [Google Scholar] [CrossRef] [PubMed]
  76. Guo, W.; Hu, S.; Elgehama, A.; Shao, F.; Ren, R.; Liu, W.; Zhang, W.; Wang, X.; Tan, R.; Xu, Q.; et al. Fumigaclavine C ameliorates dextran sulfate sodium-induced murine experimental colitis via NLRP3 inflammasome inhibition. J. Pharmacol. Sci. 2015, 129, 101–106. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, H.; Huang, G.Z.; Zheng, N.; Liu, L. Injury of myocadium of rats by acute triptolide poisoning. ICI World J. 2010, 24, 460–465. [Google Scholar]
  78. Zhang, C.; Gu, C.; Peng, F.; Liu, W.; Wan, J.; Xu, H.; Lam, W.C.; Yang, X. Preparation and Optimization of Triptolide-Loaded Solid Lipid Nanoparticles for Oral Delivery with Reduced Gastric Irritation. Molecules 2013, 18, 13340–13356. [Google Scholar] [CrossRef] [PubMed]
  79. Xue, X.; Gong, L.; Qi, X.; Wu, Y.; Xing, G.; Yao, J.; Luan, Y.; Xiao, Y.; Li, Y.; Wu, X.; et al. Knockout of hepatic P450 reductase aggravates triptolide-induced toxicity. Toxicol. Lett. 2011, 205, 47–54. [Google Scholar] [CrossRef] [PubMed]
  80. Liu, L.; Jiang, Z.; Liu, J.; Huang, X.; Wang, T.; Liu, J.; Zhang, Y.; Zhou, Z.; Guo, J.; Yang, L.; et al. Sex differences in subacute toxicity and hepatic microsomal metabolism of triptolide in rats. Toxicology 2010, 271, 57–63. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, Q. Triptolide and its expanding multiple pharmacological functions. Int. Immunopharmacol. 2011, 11, 377–383. [Google Scholar] [CrossRef] [PubMed]
  82. Tai, T.; Huang, X.; Su, Y.; Ji, J.; Su, Y.; Jiang, Z.; Zhang, L. Glycyrrhizin accelerates the metabolism of triptolide through induction of CYP3A in rats. J. Ethnopharmacol. 2014, 152, 358–363. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, C.; Sun, P.P.; Guo, H.T.; Liu, Y.; Li, J.; He, X.J.; Lu, A.P. Corrigendum: Safety Profiles of Tripterygium wilfordii Hook F: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2017, 8, 59. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, L.; Xu, Y.; Fu, L.; Li, Y.; Lou, L. (5R)-5-hydroxytriptolide (LLDT-8), a novel immunosuppressant in clinical trials, exhibits potent antitumor activity via transcription inhibition. Cancer Lett. 2012, 324, 75–82. [Google Scholar] [CrossRef] [PubMed]
  85. Fidler, J.M.; Li, K.; Chung, C.; Wei, K.; Ross, J.A.; Gao, M.; Rosen, G.D. PG490-88, a derivative of triptolide, causes tumor regression and sensitizes tumors to chemotherapy. Mol. Cancer Ther. 2003, 2, 855–862. [Google Scholar] [PubMed]
  86. Wu, D.D.; Huang, L.; Zhang, L.; Wu, L.-Y.; Li, Y.-C.; Feng, L. LLDT-67 attenuates MPTP-induced neurotoxicity in mice by up-regulating NGF expression. Acta Pharmacol. Sin. 2012, 33, 1187–1194. [Google Scholar] [CrossRef] [PubMed]
  87. Xu, H.; Fan, X.; Zhang, G.; Liu, X.; Li, Z.; Li, Y.; Jiang, B. LLDT-288, a novel triptolide analogue exhibits potent antitumor activity in vitro and in vivo. Biomed. Pharmacother. 2017, 93, 1004–1009. [Google Scholar] [CrossRef] [PubMed]
  88. Tang, W.; Zuo, J.P. Immunosuppressant discovery from Tripterygium wilfordii Hook f: The novel triptolide analog (5R)-5-hydroxytriptolide (LLDT-8). Acta Pharmacol. Sin. 2012, 33, 1112–1118. [Google Scholar] [CrossRef] [PubMed]
  89. Zhou, R.; Tang, W.; Ren, Y.-X.; He, P.-L.; Zhang, F.; Shi, L.-P.; Fu, Y.-F.; Li, Y.-C.; Ono, S.; Fujiwara, H.; et al. (5R)-5-hydroxytriptolide attenuated collagen-induced arthritis in DBA/1 mice via suppressing interferon-γ production and its related signaling. J. Pharmacol. Exp. Ther. 2006, 318, 35–44. [Google Scholar] [CrossRef] [PubMed]
  90. Zhou, R.; Tang, W.; He, P.-L.; Li, X.-Y.; Yang, Y.-F.; Li, Y.-C.; Geng, J.-G.; Zuo, J.-P. Inhibition of inducible nitric-oxide synthase expression by (5R)-5-hydroxytriptolide in interferon-γ- and bacterial lipopolysaccharide-stimulated macrophages. J. Pharmacol. Exp. Ther. 2006, 316, 121–128. [Google Scholar] [CrossRef] [PubMed]
  91. Fu, Y.F.; Ni, J.; Zhong, X.-G.; Tang, W.; Zhou, R.; Zhou, Y.; Dong, J.-R.; He, P.-L.; Wan, H.; Li, Y.-C.; et al. (5R)-5-hydroxytriptolide (LLDT-8), a novel triptolide derivative, prevents experimental autoimmune encephalomyelitis via inhibiting T cell activation. J. Neuroimmunol. 2006, 175, 142–151. [Google Scholar] [CrossRef] [PubMed]
  92. Banerjee, S.; Modi, S.; McGinn, O.; Zhao, X.; Dudeja, V.; Ramakrishnan, S.; Saluja, A.K. Impaired Synthesis of Stromal Components in Response to Minnelide Improves Vascular Function, Drug Delivery, and Survival in Pancreatic Cancer. Clin. Cancer Res. 2016, 22, 415–425. [Google Scholar] [CrossRef] [PubMed]
  93. Carter, B.Z.; Shi, Y.; Fidler, J.M.; Chen, R.; Ling, X.; Plunkett, W.; Andreeff, M. MRx102, a triptolide derivative, has potent antileukemic activity in vitro and in a murine model of AML. Leukemia 2012, 26, 443–450. [Google Scholar] [CrossRef] [PubMed]
  94. Lin, C.H.; Chen, C.H.; Lin, Z.C.; Fang, J.Y. Recent advances in oral delivery of drugs and bioactive natural products using solid lipid nanoparticles as the carriers. J. Food Drug Anal. 2017, 25, 219–234. [Google Scholar] [CrossRef] [PubMed]
  95. Callender, S.P.; Mathews, J.A.; Kobernyk, K.; Wettig, S.D. Microemulsion utility in pharmaceuticals: Implications for multi-drug delivery. Int. J. Pharm. 2017, 526, 425–442. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, G.; Hao, B.; Ju, D.; Liu, M.; Zhao, H.; Du, Z.; Xia, J. Pharmacokinetic and pharmacodynamic study of triptolide-loaded liposome hydrogel patch under microneedles on rats with collagen-induced arthritis. Acta Pharm. Sin. B 2015, 5, 569–576. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, L.; Wang, T.; Li, Q.; Huang, J.; Xu, H.; Li, J.; Wang, Y.; Liang, Q. Fabrication of novel vesicles of triptolide for antirheumatoid activity with reduced toxicity in vitro and in vivo. Int. J. Nanomed. 2016, 11, 2663–2673. [Google Scholar]
  98. Liu, M.; Dong, J.; Yang, Y.; Yang, X.; Xu, H. Anti-inflammatory effects of triptolide loaded poly(d,l-lactic acid) nanoparticles on adjuvant-induced arthritis in rats. J. Ethnopharmacol. 2005, 97, 219–225. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, M.; Dong, J.; Yang, Y.; Yang, X.; Xu, H. Effect of poly(d,l-lactic acid) nanoparticles as triptolide carrier on abating rats renal toxicity by NMR-based metabolic analysis. J. Nanosci. Nanotechnol. 2008, 8, 3493–3499. [Google Scholar] [CrossRef] [PubMed]
  100. Mei, Z.; Wu, Q.; Hu, S.; Li, X.; Yang, X. Triptolide loaded solid lipid nanoparticle hydrogel for topical application. Drug Dev. Ind. Pharm. 2005, 31, 161–168. [Google Scholar] [CrossRef] [PubMed]
  101. Mei, Z.; Chen, H.; Weng, T.; Yang, Y.; Yang, X. Solid lipid nanoparticle and microemulsion for topical delivery of triptolide. Eur. J. Pharm. Biopharm. 2003, 56, 189–196. [Google Scholar] [CrossRef]
  102. Mei, Z.; Li, X.; Wu, Q.; Hu, S.; Yang, X. The research on the anti-inflammatory activity and hepatotoxicity of triptolide-loaded solid lipid nanoparticle. Pharmacol. Res. 2005, 51, 345–351. [Google Scholar] [CrossRef] [PubMed]
  103. Xue, M.; Zhao, Y.; Li, X.-J.; Jiang, Z.-Z.; Zhang, L.; Liu, S.-H.; Li, X.-M.; Zhang, L.-Y.; Yang, S.-Y. Comparison of toxicokinetic and tissue distribution of triptolide-loaded solid lipid nanoparticles vs free triptolide in rats. Eur. J. Pharm. Sci. 2012, 47, 713–717. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, C.; Peng, F.; Liu, W.; Wan, J.; Wan, C.; Xu, H.; Lam, C.W.; Yang, X. Nanostructured lipid carriers as a novel oral delivery system for triptolide: Induced changes in pharmacokinetics profile associated with reduced toxicity in male rats. Int. J. Nanomed. 2014, 9, 1049–1063. [Google Scholar]
  105. Xu, L.; Pan, J.; Chen, Q.; Yu, Q.; Chen, H.; Xu, H.; Qiu, Y.; Yang, X. In vivo evaluation of the safety of triptolide-loaded hydrogel-thickened microemulsion. Food Chem. Toxicol. 2008, 46, 3792–3799. [Google Scholar] [CrossRef] [PubMed]
  106. Mehnert, W.; Mader, K. Solid lipid nanoparticles: Production, characterization and applications. Adv. Drug Deliv. Rev. 2001, 47, 165–196. [Google Scholar] [CrossRef]
  107. zur Muhlen, A.; Schwarz, C.; Mehnert, W. Solid lipid nanoparticles (SLN) for controlled drug delivery—Drug release and release mechanism. Eur. J. Pharm. Biopharm. 1998, 45, 149–155. [Google Scholar] [CrossRef]
  108. Kogan, A.; Garti, N. Microemulsions as transdermal drug delivery vehicles. Adv. Colloid Interface Sci. 2006, 123–126, 369–385. [Google Scholar] [CrossRef] [PubMed]
  109. Chen, H.; Chang, X.; Weng, T.; Zhao, X.; Gao, Z.; Yang, Y.; Xu, H.; Yang, X. A study of microemulsion systems for transdermal delivery of triptolide. J. Control Release 2004, 98, 427–436. [Google Scholar] [CrossRef] [PubMed]
  110. He, B.; Lu, C.; Zheng, G.; He, X.; Wang, M.; Chen, G.; Zhang, G.; Lu, A. Combination therapeutics in complex diseases. J. Cell. Mol. Med. 2016, 20, 2231–2240. [Google Scholar] [CrossRef] [PubMed]
  111. Ye, X.; Li, W.; Yan, Y.; Mao, C.; Cai, R.; Xu, H.; Yang, X. Effects of cytochrome P4503A inducer dexamethasone on the metabolism and toxicity of triptolide in rat. Toxicol. Lett. 2010, 192, 212–220. [Google Scholar] [CrossRef] [PubMed]
  112. Cao, L.; Ding, W.; Jia, R.; Du, J.; Wang, T.; Zhang, C.; Gu, Z.; Yin, G. Anti-inflammatory and hepatoprotective effects of glycyrrhetinic acid on CCl4-induced damage in precision-cut liver slices from Jian carp (Cyprinus carpio var. jian) through inhibition of the NF-κB pathway. Fish Shellfish Immunol. 2017, 64, 234–242. [Google Scholar] [CrossRef] [PubMed]
  113. Qiu, D.; Kao, P.N. Immunosuppressive and anti-inflammatory mechanisms of triptolide, the principal active diterpenoid from the Chinese medicinal herb Tripterygium wilfordii Hook. f. Drugs R D 2003, 4, 1–18. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, B.J. Triptolide, a novel immunosuppressive and anti-inflammatory agent purified from a Chinese herb Tripterygium wilfordii Hook F. Leuk. Lymphoma 2001, 42, 253–265. [Google Scholar] [CrossRef] [PubMed]
  115. Feng, X.; Ding, L.; Qiu, F. Potential drug interactions associated with glycyrrhizin and glycyrrhetinic acid. Drug Metab. Rev. 2015, 47, 229–238. [Google Scholar] [CrossRef] [PubMed]
  116. Shao, F.; Wang, G.; Xie, H.; Zhu, X.; Sun, J.; A, J. Pharmacokinetic study of triptolide, a constituent of immunosuppressive chinese herb medicine, in rats. Biol. Pharm. Bull. 2007, 30, 702–707. [Google Scholar] [CrossRef] [PubMed]
  117. Kong, L.-L.; zhuang, X.-M.; Yang, H.-Y.; Yuan, M.; Xu, L.; Li, H. Inhibition of P-glycoprotein Gene Expression and Function Enhances Triptolide-induced Hepatotoxicity in Mice. Sci. Rep. 2015, 5, 11747. [Google Scholar] [CrossRef] [PubMed]
  118. Han, F.M.; Peng, Z.H.; Wang, J.J.; Chen, Y. In vivo effect of triptolide combined with glycyrrhetinic acid on rat cytochrome P450 enzymes. Yao Xue Xue Bao 2013, 48, 1136–1141. [Google Scholar] [PubMed]
  119. Li, Z.; Yan, M.; Cao, L.; Fang, P.; Guo, Z.; Hou, Z.; Zhang, B. Glycyrrhetinic Acid Accelerates the Clearance of Triptolide through P-gp In Vitro. Phytother. Res. 2017, 31, 1090–1096. [Google Scholar] [CrossRef] [PubMed]
  120. Arafa, H.M. Uroprotective effects of curcumin in cyclophosphamide-induced haemorrhagic cystitis paradigm. Basic Clin. Pharmacol. Toxicol. 2009, 104, 393–399. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, L.; Huang, Q.H.; Li, Y.X.; Huang, Y.F.; Xie, J.H.; Xu, L.Q.; Dou, Y.X.; Su, Z.R.; Zeng, H.F.; Chen, J.N. Protective effects of silymarin on triptolide-induced acute hepatotoxicity in rats. Mol. Med. Rep. 2018, 17, 789–800. [Google Scholar] [CrossRef] [PubMed]
  122. Yuan, Z.X.; Jia, L.; Lim, L.Y.; Lin, J.C.; Shu, G.; Zhao, L.; Ye, G.; Liang, X.X.; Ji, H.; Fu, H.L. Renal-targeted delivery of triptolide by entrapment in pegylated TRX-20-modified liposomes. Int. J. Nanomed. 2017, 12, 5673–5686. [Google Scholar] [CrossRef] [PubMed]
  123. Yuan, Z.X.; Wu, X.J.; Mo, J.; Wang, Y.L.; Xu, C.Q.; Lim, L.Y. Renal targeted delivery of triptolide by conjugation to the fragment peptide of human serum albumin. Eur. J. Pharm. Biopharm. 2015, 94, 363–371. [Google Scholar] [CrossRef] [PubMed]
  124. Fu, Y.; Lin, Q.; Gong, T.; Sun, X.; Zhang, Z.R. Renal-targeting triptolide-glucosamine conjugate exhibits lower toxicity and superior efficacy in attenuation of ischemia/reperfusion renal injury in rats. Acta Pharmacol. Sin. 2016, 37, 1467–1480. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, Z.; Zheng, Q.; Han, J.; Gao, G.; Liu, J.; Gong, T.; Gu, Z.; Huang, Y.; Sun, X.; He, Q. The targeting of 14-succinate triptolide-lysozyme conjugate to proximal renal tubular epithelial cells. Biomaterials 2009, 30, 1372–1381. [Google Scholar] [CrossRef] [PubMed]
  126. Lin, C.; Wong, B.C.K.; Chen, H.; Bian, Z.; Zhang, G.; Zhang, X.; Kashif Riaz, M.; Tyagi, D.; Lin, G.; Zhang, Y.; et al. Pulmonary delivery of triptolide-loaded liposomes decorated with anti-carbonic anhydrase IX antibody for lung cancer therapy. Sci. Rep. 2017, 7, 1097. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, C.; Liu, B.; Xu, X.; Zhuang, B.; Li, H.; Yin, J.; Cong, M.; Xu, W.; Lu, A. Toward targeted therapy in chemotherapy-resistant pancreatic cancer with a smart triptolide nanomedicine. Oncotarget 2016, 7, 8360–8372. [Google Scholar] [CrossRef] [PubMed]
  128. Ling, D.; Xia, H.; Park, W.; Hackett, M.J.; Song, C.; Na, K.; Hui, K.M.; Hyeon, T. pH-sensitive nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular carcinoma. ACS Nano 2014, 8, 8027–8039. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of (Triptolide)TP.
Figure 1. Chemical structure of (Triptolide)TP.
Ijms 19 00376 g001
Figure 2. Schematic illustration of TP properties in the treatment of rheumatoid arthritis (RA). The anti-RA properties of TP have been attributed to its immunosupressive and antiproliferative effect. MIP: macrophage inflammatory protein; MCP: monocyte chemoattractant protein; RANTES: regulated upon activation normal T cell expressed and secreted; IP: interferon-induced protein; IL: interleukin; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; Ang: angiopoietin; TNF: tumor necrosis factor; CCR: C-C chemokine receptor; MMP: matrix metalloproteinase; COX: cyclooxygenase; PG: prostaglandin; NO: nitric oxide; TREM: triggering receptors expressed on myeloid cells-1; TLR: toll-like receptor; BMD: bone mineral density; RANK: receptor activator of nuclear factor-κB; RNAKL: receptor activator of nuclear factor-κB ligand; OPG: osteoprotegerin; TGF: transforming growth factor.
Figure 2. Schematic illustration of TP properties in the treatment of rheumatoid arthritis (RA). The anti-RA properties of TP have been attributed to its immunosupressive and antiproliferative effect. MIP: macrophage inflammatory protein; MCP: monocyte chemoattractant protein; RANTES: regulated upon activation normal T cell expressed and secreted; IP: interferon-induced protein; IL: interleukin; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; Ang: angiopoietin; TNF: tumor necrosis factor; CCR: C-C chemokine receptor; MMP: matrix metalloproteinase; COX: cyclooxygenase; PG: prostaglandin; NO: nitric oxide; TREM: triggering receptors expressed on myeloid cells-1; TLR: toll-like receptor; BMD: bone mineral density; RANK: receptor activator of nuclear factor-κB; RNAKL: receptor activator of nuclear factor-κB ligand; OPG: osteoprotegerin; TGF: transforming growth factor.
Ijms 19 00376 g002
Table 1. Main derivatives of TP.
Table 1. Main derivatives of TP.
No.Compound NameChemical StructureModification SitesImproved Characteristics Compared with TPReferences
1(5R)-5-hydroxytriptolide (LLDT-8) Ijms 19 00376 i001C-5 sitemuch lower toxicity[88]
2LLDT-67 Ijms 19 00376 i002C-14 sitelow toxicity[86]
3LLDT-288 Ijms 19 00376 i003C-14 sitelow toxicity[87]
4PG490-88 Ijms 19 00376 i004C-14-hydroxyl siteWater soluble[85]
5Minnelide Ijms 19 00376 i005C-14-hydroxyl siteWater soluble[92]
6MRx102————low toxicity[93]
Note: “——“means that there are no corresponding chemical structure and modification site in the literature we cited.
Table 2. Innovative delivery system studies of TP.
Table 2. Innovative delivery system studies of TP.
Drug CarrierIn Vivo/In VitroAdvantagesReferences
liposome hydrogel patchCIA ratsimproves bioavailability of TP; bypasses hepatic first-pass metabolism, and reduces the incidence or severity of gastrointestinal reactions[96]
nanodrug carrier system
(γ-PGA-l-PAE-TP (PPT))
normal C57/B6 mice/RAW264.7 cell linesreduces free TP toxicity in vitro and in vivo[97]
poly(d,l-lactic acid) (PLA) nanoparticlesAIA ratsimprove bioavailability of TP[98]
poly(d,l-lactic acid) (PLA) nanoparticlesnormal SD ratsabate the renal toxicity caused by TP[99]
solid lipid nanoparticle hydrogelcarrageenan-induced ratsimproves safety and minimizes the toxicity induced by TP[100]
solid lipid nanoparticle/microemulsionscarrageenan-induced rats and AIA ratsincrease therapeutic index[101]
solid lipid nanoparticlescarrageenan-induced ratsenhance the anti-inflammatory activity of TP have a protective effect against TP-induced hepatotoxicity[102]
solid lipid nanoparticlesnormal SD ratsreduce gastric irritation[78]
solid lipid nanoparticlesnormal SD ratsenhance efficacy, decrease reproductive toxicity[103]
nanostructured lipid carriersnormal SD ratsreduce subacute toxicity in male rats[104]
hydrogel-thickened microemulsionnormal rabbits, mice, beagle dogs, guinea pigsno obvious toxicities[105]
Note: CIA: collagen-induced arthritis; AIA: adjuvant-induced arthritis; SD: Sprague Dawley; TP: triptolide.

Share and Cite

MDPI and ACS Style

Fan, D.; Guo, Q.; Shen, J.; Zheng, K.; Lu, C.; Zhang, G.; Lu, A.; He, X. The Effect of Triptolide in Rheumatoid Arthritis: From Basic Research towards Clinical Translation. Int. J. Mol. Sci. 2018, 19, 376. https://doi.org/10.3390/ijms19020376

AMA Style

Fan D, Guo Q, Shen J, Zheng K, Lu C, Zhang G, Lu A, He X. The Effect of Triptolide in Rheumatoid Arthritis: From Basic Research towards Clinical Translation. International Journal of Molecular Sciences. 2018; 19(2):376. https://doi.org/10.3390/ijms19020376

Chicago/Turabian Style

Fan, Danping, Qingqing Guo, Jiawen Shen, Kang Zheng, Cheng Lu, Ge Zhang, Aiping Lu, and Xiaojuan He. 2018. "The Effect of Triptolide in Rheumatoid Arthritis: From Basic Research towards Clinical Translation" International Journal of Molecular Sciences 19, no. 2: 376. https://doi.org/10.3390/ijms19020376

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop