Skip to main content

MINI REVIEW article

Front. Pharmacol., 31 March 2020
Sec. Renal Pharmacology

The Therapeutic Effect of Artemisinin and Its Derivatives in Kidney Disease

Ming XiaMing XiaDi LiuDi LiuYu LiuYu LiuHong Liu*Hong Liu*
  • Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China

Artemisinin (ARS) and its derivatives (ARSs) are recommended as the first-line antimalarial drugs for the treatment of malaria. Besides antimalarial function, its potent anti-inflammatory and immunoregulatory properties, as well as the ability to regulate oxidative stress have brought them to a prominent position. As researchers around the world are continually exploring the unknown biological activities of ARS derivatives, experimental studies have shown much progress in renal therapy. This review aims to give a brief overview of the current research on ARSs applications for kidney treatment with the evaluation of therapeutic properties and potential molecular mechanisms.

Introduction

The imbalance between the molecular mechanisms that govern oxidative stress, inflammation, immunity, and cell death are important causes of acute kidney injury (AKI) and chronic kidney diseases (CKD) (Sureshbabu et al., 2015). Both AKI and CKD can lead to diminished kidney function and are associated with high mortality and morbidity. Accumulated evidence demonstrated that natural products are alternative sources for treating renal diseases on account of the conventional experience and multi-target characteristics (Chen et al., 2018).

Artemisinin (ARS) is an effective constituent with a molecular weight of 282 originally extracted from traditional Chinese medicine Artemisia annua L, which was first discovered by Chinese scientists in 1972. Its chemical structure-sesquiterpene lactone with a peroxide bridge has been demonstrated to exert an excellent antimalarial effect (White et al., 2015; Chang, 2016). In the presence of heme or free iron, the production of reactive oxygen species and carbon-centered free radicals generated by the cleavage of the endoperoxide bridge can directly poison the parasites (Vennerstrom et al., 2004). ARS selectively kills plasmodium-infected red blood cells without destroying healthy cells, making it the recommended drug for the treatment of malaria (Lalloo et al., 2016) and more clinically effective than other antimalarial drugs such as hydroxychloroquine (HCQ) and chloroquine (CQ) (Golenser et al., 2006; Efferth and Kaina, 2010). ARS has a rapid onset of action and can be rapidly absorbed by the gastrointestinal tract after oral administration, with half-live ranging from 2 to 5 h. It is mainly distributed in the liver, kidney, and bile, and approximately 80% of the drug was excreted through the urine and feces within 24 h of administration (German and Aweeka, 2008; Li, 2012). Currently, a series of ARS derivatives (ARSs) with improved pharmacological features are used in clinical treatment including artemether (ARM), artesunate (ART), β-aminoarteether maleate (SM934), and dihydroartemisinin (DHA) (chemical structures were shown in Figure 1). The half-lives of ARM (2–4 h), ART (< 1 h), DHA (∼1 h) are shorter (Krishna et al., 2004; German and Aweeka, 2008) and oral intake represents a relatively safe route in the clinic.

FIGURE 1
www.frontiersin.org

Figure 1 Chemical structures of artemisinin and its derivatives.

In addition to decades of remarkable progress against malaria, studies have demonstrated a variety of other pharmacological effects beyond antimalarial, such as anti-virus, anti-neoplastic, anti-inflammation as well as immunosuppressive effects (An J. et al., 2017). The properties of ARSs have been intensively reviewed in systemic lupus erythematosus, rheumatoid, arthritis, multiple sclerosis, etc (Shi et al., 2015; Mu and Wang, 2018), while the treatment of kidney disease has not been summarized. This review will focus on the proposed therapeutic properties and mechanisms of ARSs in kidney disease, and discuss the potential application of ARSs as novel agents for future treatment.

Search Strategy

Comprehensive literature searches for candidate studies were undertaken in two English and three Chinese biomedical databases from inception through February 2020. These databases included PubMed, Springer, Chinese National Knowledge Infrastructure, WanFang Med Online, and Chinese Biomedical Databases. Searches were limited to studies in English and Chinese. The following terms were used in the search: “artemisinin,” “artemisinins,” “artesunate,” “dihydroartemisinin,” “artemether,” “SM934,” “β-aminoarteether maleate,” “kidney,” “renal,” “nephro,” “nephritis,” nephropathy.”

Overview of Researches on ARSs in Renal Disease

The effects of ARSs were mainly studied on animal models and cells, with two clinical studies targeting lupus nephritis (LN). In a randomized, 5-year follow-up clinical trial for LN (Lu, 2002), the treatment group (ARS 0.6 g/d and cordyceps 3–4 g/d) was reported to improve 24 h urine protein, creatinine clearance rate, level of C3, and was more effective than the control group (tripterygium wilfordii polyglycosides tablets 1 mg/kg, three times a day and/or baoshenkang tablets 150 mg, three times a day). A 2-month randomized trial had similar findings (Li et al., 2011), treatment with ART (50 mg, twice a day) was reported to improve systemic symptoms and reduce the immunological activity index than either before treatment or the control group with tripterygium wilfordii polyglycosides tablets (10 mg, three times a day and/or prednisone 0.5 mg/kg/d). However, these two studies were not blinded and placebo-controlled, which may result in information bias, and observations of larger samples are still lacking.

Table 1 summarized the characteristics of animal studies with ARSs treatment, covering LN, adriamycin nephropathy (AN), subtotal nephrectomy (SNx), IgA nephropathy (IgAN), diabetic nephropathy (DN), AKI, unilateral ureteral obstruction (UUO), pristine or lipopolysaccharide (LPS)-induced nephritis, nephrotic syndrome (NS), and Heymann nephritis (HN). In vitro cell models and in vivo animal models investigations for ARSs efficacy on kidney disease involve various aspects including oxidative stress, inflammation action, and immune response, we will describe the effects of ARSs in sections below.

TABLE 1
www.frontiersin.org

Table 1 Study characteristics of animal experiments in kidney disease.

Mechanism of Arss in Kidney

Oxidative Stress Regulation of Artemisinins

Oxidative stress is an important mediator in the development and progression of CKD and AKI and its complications due to increased production of reactive oxygen species (ROS) and diminished antioxidant capacity (Ruiz et al., 2013). In the condition of a surplus of ROS, ARSs were reported to exhibit an antioxidant effect (Kim et al., 2014; Yang et al., 2018; Liu et al., 2019). In addition, according to the characteristics of ARSs, tumor cells are more vulnerable due to higher levels of iron (Robert et al., 2005) and are more susceptible to further ROS insults induced by ARSs (Hamacher-Brady et al., 2011; Efferth, 2017). Accordingly, ARSs have the potential to treat kidney cancer, and the emergence of new technologies such as ARS-based smart nanomedicine offers more possibilities (Luo et al., 2019).

In Vitro

Receptor-interacting protein kinase 1 (RIP1) is verified to modulate mitochondrial ROS production via excessive generation of mitochondrial superoxide and depletion of GSH (Zhou et al., 2017). ART was reported to induce ROS production and cell death in human renal carcinoma cells, while pretreatment with RIP1 inhibitor or knockdown of RIP1 reverted ART-induced cytotoxicity (Chauhan et al., 2017).

In Vivo

Pretreated DHA or ARS could ameliorate oxidative stress in AKI mice by restoring malonyl dialdehyde (MDA), nitric oxide (NO), glutathione peroxidase (GSH), catalase (CAT), and superoxide dismutase (SOD) activity in the kidney (An Y. et al., 2017; Liu et al., 2019). In addition, ARM was shown to reduce the serum H2O2 level and elevated renal cortical PGC-1α expression, but it did not exert obvious effects on CAT and SOD expression in the renal cortex in DN (Han et al., 2019). In normal rats without oxidative stress, orally taken artemether-lumefantrine (1.14/6.86 mg/kg/d, twice a day) or artesunate-amodiaquine (2.86/8.58 mg/kg/d, twice a day) for 7 d did not apparently alter renal antioxidant status compared with the control. Although there was no significant alteration in kidney, liver, lung, and brain weights, the artesunate-amodiaquine group showed cardiotoxicity (decreased heart weight by 27.2% compared with control) (Otuechere et al., 2012).

To date, ARSs could trigger cell death by inducing oxidative stress, and could also resist oxidation to reduce cell damage. Detailed understanding of the molecular mechanisms and the events by which ARSs regulate oxidative stress to control cellular processes in different cells remain to be explored.

Anti-Inflammation Effect of ARSs

Inflammation plays a pivotal role in the pathophysiological processes of kidney diseases and associated with renal injury (Ernandez and Mayadas, 2016). The anti-inflammatory effects of ARS have been widely recognized, including repression of nuclear factor-κB (NF-κB), toll-like receptors (TLRs), signal transducer and activator of transcription (STAT), and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) activity (Aldieri et al., 2003; Ho et al., 2014; Shi et al., 2015), which are key factors mediating immune-inflammatory response and are associated with kidney disease progression (Ruiz-Andres et al., 2016).

In Vitro

ART ameliorated high glucose-induced injury by suppressing TLR4/NF-κB/nod-like receptor protein 3 (NLRP3) inflammasome pathway in rat glomerular mesangial cell (Sun et al., 2018).

In Vivo

For LN mice, it has been reported that treatment with ARSs could decrease interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) in serum, and blocked intercellular adhesion molecule-1 (ICAM-1), fractalkine, NF-κB signaling pathway in renal (Dong et al., 2003; Li et al., 2006; Wang et al., 2010; Wu X. et al., 2010; You et al., 2014; Liang et al., 2018). A similar effect was observed in the nephritis and septic AKI models (Wu P. et al., 2011; Wan and Li, 2017; Liu et al., 2019). Also, ARSs were reported to inhibit disease progression via downregulating renal monocyte chemoattractant protein 1 (MCP-1) expression in LN, IgAN, and septic AKI models (Mi et al., 2009; Jin et al., 2009; Ma et al., 2009; Liu et al., 2019). In addition, ARSs were proved to alleviate the tubule-interstitial inflammation and fibrosis by inhibiting NF‐κB and mothers against decapentaplegic homolog (Smad) signaling pathway in SNx rats, Heymann nephritis rats and UUO models (Ma et al., 2010; Li et al., 2015; Wen et al., 2019). ARSs were also reported to reduce diabetic kidney damage by inhibiting TLR4, IL-8, and the DNA-binding activity of NF−κB in renal (Zhou et al., 2014a; Nie et al., 2015).

These data indicate that ARSs act as anti-inflammatory drugs at multiple components of inflammation signals and have a potential therapeutic effect on disease activity.

Immunoregulatory Effect of ARSs

Under physiological conditions, the kidney contributes to immune homeostasis, assist in the removal of metabolic wastes and toxins, and maintain peripheral tolerance. The disruption of immune homeostasis an autoimmune response, such as the occurrence of LN and IgAN, resulting in the loss of renal function (Tecklenborg et al., 2018). T cells, B cells, and macrophages, as well as cytokines, are involved in immune regulation and are activated to varying degrees depending on disease pattern. The activated pathogenic cells are more likely to lead the breakdown of the peroxide bridge structure of ARS (Shi et al., 2015).

In Vitro

Chemokine ligand 2 (CCL2) and single immunoglobulin IL-1-related receptor (SIGIRR) are involved in the inflammatory pathogenesis of LN, DHA was reported to inhibit CCL2 secretion and increase SIGIRR expression and protect LPS-induced HK-2 inflammation (Huang et al., 2015).

In Vivo

ARS combined with prednisone was reported to increase the sensitivity of glucocorticoid compared to the group administrated glucocorticoid only in LN (Wu X.L et al., 2011), which may offer a possibility of alleviating the common side effects of existing glucocorticoids or immunosuppressants. SM934 was shown to protect LN mice by inhibiting both Th1 cells and Th17 cells responses (Hou et al., 2011) and reduce the number of activated B cells by inhibiting the expression of TLR7/9 (Gui et al., 2019). SIGIRR, as an inhibitor of TLR signal transduction, could be elevated by DHA. This might be a negative immune-modulating way for DHA to slow the progression of LN (Huang et al., 2015). In addition, ARS combined with HCQ was shown to improve IgAN rats immunity (Lin et al., 2016), possibly via inhibiting the differentiation of Th2 and Th17 cells while promoting Th1 and Treg cells differential (Bai et al., 2019).

All these studies suggest that ARS family drugs are able to perform immunosuppressive functions primarily through suppressing the activation of pathogenic immune cells and have a regulatory effect on autoimmune diseases.

Other Effects

Anti-Fibrosis

Myofibroblasts can be differentiated by the epithelial-mesenchymal transformation (EMT) process, and are primarily responsible for excessive extracellular matrix production. TGF-β1, smooth muscle actin (α-SMA) and connective tissue growth factor (CTGF), metalloprotease (MMP), bone morphogenetic protein (BMP) are all considered to be major regulators of EMT and renal fibrosis (Liu et al., 2018). For UUO, both in vitro and in vivo study showed anti-fibrosis effect of ARSs related to the inhibition of EMT, fibroblast proliferation, and collagen synthesis (Zhang et al., 2017). And upregulating BMP-7 and downregulating BMP antagonist-uterine sensitization-associated gene-1 (USAG-1) (Cao et al., 2016), or mitigating CTGF, α-SMA (Mi et al., 2007), or PI3K/AKT pathway (Zhang et al., 2019) are all possible mechanisms.

Anti-Proliferation

Glomerular mesangial cell proliferation is a common pathological change of glomerular disease, effective control of mesangial cell proliferation is of great clinical significance. ARSs were reported to exert an inhibitory effect on the proliferation of rat mesangial cells (MA et al., 2007a; Ma et al., 2007b), possibly by inducing apoptosis and downregulating inflammatory cytokines TNF-α and IL-6 (Wang et al., 2016) or enhance caspase-3 activity (Wu X.L. et al., 2010). Our team recently demonstrated that DHA could inhibit the proliferation of aIgA1-induced human mesangial cells through the mTOR signaling pathway in vitro (Xia et al., 2020). In addition, ARSs were found to inhibit renal carcinoma cell proliferation by inhibiting the expression of fascin (Zhang et al., 2018), meanwhile inhibiting colony formation, migration, invasion, and tumorigenesis (Yu et al., 2019).

Regulate Glomerular Filtration

Glomerular permeability is regulated by the glomerular filtration barrier (GFB), which composed of glomerular endothelium, the glomerular basement membrane, and the podocyte layer. The dysfunction of intercellular adhesion and connection will result in the loss of the structural and functional integrity of GFB and the occurrence of proteinuria (Mehta and Malik, 2006). ART was proved to reduce glomerular permeability and improve proteinuria in LN mice by inhibiting vascular endothelial growth factor (VEGF) (Jin et al., 2007). Studies also showed that DHA ameliorated the hyperpermeability of GFB by inhibiting TNF-α and maintaining occludin expression (Cheng et al., 2018) or elevation of vascular endothelial (VE)-cadherin expression in endothelial cells (Li et al., 2018). In addition, ARS was observed to attenuate podocyte effacement and fusion via nephrin and podocin regulation in adriamycin-induced nephropathy (Wu et al., 2014), and reduce the shedding of podocyte and excretion of nephrin and podocin in Heymann nephritis (Liu et al., 2017).

Anti-Virus

ARS was shown to be effective in inhibiting polyomavirus BK replication in primary human kidney cells (Sharma et al., 2014).

Renal-Protective.

ART was reported to ameliorate proteinuria and suppress the progression of NS (Razavi et al., 2007). Studies also showed that ARS could relieve renal lesions in DN rats, through inhibiting platelet-derived growth factor-B (PDGF-B) expression (Zhang et al., 2014a), metalloproteinase tissue inhibitor-2 (TIMP-2) (Zhang et al., 2014b), spatiotemporal dynamics activation of protein kinase c (PKC) (Zhang et al., 2014c) and its downstream c-fos and c-jun (Zhou et al., 2014b), and their heterodimer activator protein (AP-1) (Zhou et al., 2014c). The results from the high-throughput sequence from DN rats treated with ARS may identifying promising targets for future treatment (Xiang et al., 2019). In addition, kidney function was found to be improved in cases of malarial nephropathy after treatment with ARSs (Ezzedine et al., 2007; Calitri et al., 2014; Gleeson et al., 2019).

Interaction, Safety, and Side Effects

The toxicity of ARSs in cell culture, animals (mice, rats, rabbits, dogs, monkeys), and human clinical trials were well described (Efferth and Kaina, 2010). Large clinical studies and meta-analyses did not show serious side effects, despite mild and self-limited effects including mild nausea, vomiting, and diarrhea (Mssusa et al., 2016). Individual patients may appear transient transaminase elevated and mild rash. Non-hematological side effects include mild hepatitis, neurological, renal, cutaneous, and cardiac manifestations were uncommon (Roussel et al., 2017). Rare severe adverse events include prolongation of the QTc interval and cardiac arrhythmias (in LiverTox, 2012).

In addition, animal studies showed that artesunate can reduce glomerular filtration rate, increase renal blood flow, and has certain organ toxicity (Campos et al., 2001; Otuechere et al., 2012), while in a clinical study, liver function, kidney function, and routine blood tests remained normal in most patients treated with artesunate (von Hagens et al., 2017). A systematic review and meta-analysis showed that the use of ARS-based combination therapy in adults, children, and pregnant women in the 2nd or 3rd trimester was relatively safe (Kovacs et al., 2016). The drug interactions of ARSs are relatively unknown, more rigorous and comprehensive studies of interaction mechanisms are needed, as well as monitoring the safety of ARSs, especially concerning the genotoxicity and embryotoxicity (Amorim et al., 2013).

Conclusion and Future Directions

Much knowledge has been gained about the antimalarial drugs in recent years, and more attention has been paid to ARSs application for renal damages. Many years of laboratory applications and research proved that ARSs have excellent anti-inflammatory and immunoregulatory functions. It is also a good regulator of the balance between oxidation and oxidation resistance. The regulation of the glomerular barrier highlights a unique aspect of the use of ARSs in kidney disease.

Despite accumulating evidence on the use of ARSs, the literature on its potential as a treatment for renal disease is still insufficient due to the lack of randomized controlled clinical trials. The additive effects of ARSs in combined administration with immunosuppressants and the structure-activity relationship need to be further clarified. Investigation of the improved properties of ARSs analogs also facilitates the discovery of novel drug targets for kidney disease (Santos et al., 2015; Zuma et al., 2016; de Lange et al., 2018).

Author Contributions

MX provided direction, collected related literature, and drafted the manuscript. DL and YL made significant revisions to the manuscript and directed the review to be more focused. HL gave the final approval for the article to be published. All authors have read and approved the final manuscript.

Funding

This work was supported by a research grant (81770714), (81470947), and (81570622) from the National Natural Science Foundation of China. It also funded by the postgraduate innovation project of Central South University in China (Project no.2018zzts920).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Aldieri, E., Atragene, D., Bergandi, L., Riganti, C., Costamagna, C., Bosia, A., et al. (2003). Artemisinin inhibits inducible nitric oxide synthase and nuclear factor NF-kB activation. FEBS Lett. 552 (2-3), 141–144. doi: 10.1016/S0014-5793(03)00905-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Amorim, M. H., Gil da Costa, R. M., Lopes, C., Bastos, M. M. (2013). Sesquiterpene lactones: adverse health effects and toxicity mechanisms. Crit. Rev. Toxicol. 43 (7), 559–579. doi: 10.3109/10408444.2013.813905

PubMed Abstract | CrossRef Full Text | Google Scholar

An, J., Minie, M., Sasaki, T., Woodward, J. J., Elkon, K. B. (2017). Antimalarial Drugs as Immune Modulators: New Mechanisms for Old Drugs. Annu. Rev. Med. 68, 317–330. doi: 10.1146/annurev-med-043015-123453

PubMed Abstract | CrossRef Full Text | Google Scholar

An, Y., Li, Q., Sui, C. H., Wang, C. (2017). Effects of artemisinin intragastric administration on acute kidney injury induced by cisplatin in mice. Shandong Med. J. 57 (11), 36–39. doi: 10.3969/j.issn.1002-266X.2017.11.011

CrossRef Full Text | Google Scholar

Bai, L., Li, H., Li, J., Song, J., Zhou, Y., Liu, B., et al. (2019). Immunosuppressive effect of artemisinin and hydroxychloroquine combination therapy on IgA nephropathy via regulating the differentiation of CD4+ T cell subsets in rats. Int. Immunopharmacol. 70, 313–323. doi: 10.1016/j.intimp.2019.02.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Calitri, C., Garazzino, S., Camilla, R., Peruzzi, L., Amore, A., Tovo, P. A., et al. (2014). “Collateral” effect of artemether in an atypical kidney involvement by Plasmodium falciparum malaria: a case report. J. Vector Borne Dis. 51 (2), 147–148.

PubMed Abstract | Google Scholar

Campos, S. B., Rouch, L. H., Seguro, A. C. (2001). Effects of sodium artesunate, a new antimalarial drug, on renal function. Kidney Int. 59 (3), 1044–1051. doi: 10.1046/j.1523-1755.2001.0590031044.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, J., Wang, W., Li, Y., Xia, J., Peng, Y., Zhang, Y., et al. (2016). Artesunate attenuates unilateral ureteral obstruction-induced renal fibrosis by regulating the expressions of bone morphogenetic protein-7 and uterine sensitization-associated gene-1 in rats. Int. Urol Nephrol. 48 (4), 619–629. doi: 10.1007/s11255-016-1232-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, Z. (2016). The discovery of Qinghaosu (artemisinin) as an effective anti-malaria drug: A unique China story. Sci. China Life Sci. 59 (1), 81–88. doi: 10.1007/s11427-015-4988-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chauhan, A. K., Min, K. J., Kwon, T. K. (2017). RIP1-dependent reactive oxygen species production executes artesunate-induced cell death in renal carcinoma Caki cells. Mol. Cell Biochem. 435 (1-2), 15–24. doi: 10.1007/s11010-017-3052-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, D. Q., Hu, H. H., Wang, Y. N., Feng, Y. L., Cao, G., Zhao, Y. Y., et al. (2018). Natural products for the prevention and treatment of kidney disease. Phytomedicine 50, 50–60. doi: 10.1016/j.phymed.2018.09.182

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, Z., Qi, R., Li, L., Liu, Q., Zhang, W., Zhou, X., et al. (2018). Dihydroartemisinin ameliorates sepsis-induced hyperpermeability of glomerular endothelium via up-regulation of occludin expression. BioMed. Pharmacother. 99, 313–318. doi: 10.1016/j.biopha.2018.01.078

PubMed Abstract | CrossRef Full Text | Google Scholar

de Lange, C., Coertzen, D., Smit, F. J., Wentzel, J. F., Wong, H. N., Birkholtz, L. M., et al. (2018). Synthesis, antimalarial activities and cytotoxicities of amino-artemisinin-1,2-disubstituted ferrocene hybrids. Bioorg Med. Chem. Lett. 28 (19), 3161–3163. doi: 10.1016/j.bmcl.2018.08.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, Y. J., Li, W. D., Tu, Y. Y., Zhang, H. N., Zou, W. Z., Yan, L., et al. (2003). The effects of DQHS on the pathologic changes in BXSB mice lupus nephritis and the effect mechanism. Chin. Pharmacol. Bull. 19 (10), 1125–1128. doi: 10.3321/j.issn:1001-1978.2003.10.011

CrossRef Full Text | Google Scholar

Efferth, T., Kaina, B. (2010). Toxicity of the antimalarial artemisinin and its dervatives. Crit. Rev. Toxicol. 40 (5), 405–421. doi: 10.3109/10408441003610571

PubMed Abstract | CrossRef Full Text | Google Scholar

Efferth, T. (2017). From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin. Cancer Biol. 46, 65–83. doi: 10.1016/j.semcancer.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Ernandez, T., Mayadas, T. N. (2016). The Changing Landscape of Renal Inflammation. Trends Mol. Med. 22 (2), 151–163. doi: 10.1016/j.molmed.2015.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Ezzedine, K., Pistone, T., Receveur, M. C., Cressend, T., Dieval, C., Malvy, D., et al. (2007). Haemoglobinuria in a 38-year-old French expatriate man living in Cameroon following artemisinin-based antimalarial treatment. Travel Med. Infect. Dis. 5 (4), 256–258. doi: 10.1016/j.tmaid.2007.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

German, I., Aweeka, F. T. (2008). Clinical pharmacology of artemisinin-based combination therapies. Clin. Pharmacokinet. 47 (2), 91–102. doi: 10.2165/00003088-200847020-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

Gleeson, P. J., O'Regan, J. A., McHale, T., Tuite, H., Giblin, L., Reddan, D., et al. (2019). Acute interstitial nephritis with podocyte foot-process effacement complicating Plasmodium falciparum infection. Malar J. 18 (1), 58. doi: 10.1186/s12936-019-2674-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Golenser, J., Waknine, J. H., Krugliak, M., Hunt, N. H., Grau, G. E. (2006). Current perspectives on the mechanism of action of artemisinins. Int. J. Parasitol. 36 (14), 1427–1441. doi: 10.1016/j.ijpara.2006.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Gui, J. X., Cao, L., Wang, B., Zhang, Y., Li, Y. Y., Xiong, A. L., et al. (2019). Effects of artemisinin derivatives SM934 inhibiting TLR7/9 signaling pathway on B cells in systemic lupus erythematosus mice. Chin J. Clin. Pharmacol. 35 (19), 2343–2346. doi: 10.13699/j.cnki.1001-6821.2019.19.041

CrossRef Full Text | Google Scholar

Hamacher-Brady, A., Stein, H. A., Turschner, S., Toegel, I., Mora, R., Jennewein, N., et al. (2011). Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J. Biol. Chem. 286 (8), 6587–6601. doi: 10.1074/jbc.M110.210047

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, P., Wang, Y., Zhan, H., Weng, W., Yu, X., Ge, N., et al. (2019). Artemether ameliorates type 2 diabetic kidney disease by increasing mitochondrial pyruvate carrier content in db/db mice. Am. J. Transl. Res. 11 (3), 1389–1402.

PubMed Abstract | Google Scholar

Ho, W. E., Peh, H. Y., Chan, T. K., Wong, W. S. (2014). Artemisinins: pharmacological actions beyond anti-malarial. Pharmacol. Ther. 142 (1), 126–139. doi: 10.1016/j.pharmthera.2013.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, L. F., He, S. J., Li, X., Yang, Y., He, P. L., Zhou, Y., et al. (2011). Oral administration of artemisinin analog SM934 ameliorates lupus syndromes in MRL/lpr mice by inhibiting Th1 and Th17 cell responses. Arthritis Rheum 63 (8), 2445–2455. doi: 10.1002/art.30392

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, M., Jin, X. K., Cai, Q. C., Li, M., Lin, Z. B., Li, W. D., et al. (2015). Therapeutic effect of DHA on lupus nephritis and its relationship with SIGIRR inducing immune negative regulation. Chin. J. Immunol. 31 (12), 1637–1647. doi: 10.3969/j.issn.1000-484X.2015.12.012

CrossRef Full Text | Google Scholar

in LiverTox (2012). Clinical and Research Information on Drug-Induced Liver Injury. (Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases).

Google Scholar

Jin, O., Zhang, H., Xu, T., Zhao, S., Zhou, K., Sun, L., et al. (2007). Pathological Change and Mechanism Of Artesunate Treatment for Lupus Nephritis in MRL/lpr Mice. J. Clin. Med. Pract. 11 (4), 5–9. doi: 10.3969/j.issn.1672-2353.2007.07.003

CrossRef Full Text | Google Scholar

Jin, O., Zhang, H., Gu, Z., Zhao, S., Xu, T., Zhou, K., et al. (2009). A pilot study of the therapeutic efficacy and mechanism of artesunate in the MRL/lpr murine model of systemic lupus erythematosus. Cell Mol. Immunol. 6 (6), 461–467. doi: 10.1038/cmi.2009.58

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, M. H., Seo, J. Y., Liu, K. H., Kim, J. S. (2014). Protective effect of Artemisia annua L. extract against galactose-induced oxidative stress in mice. PloS One 9 (7), e101486. doi: 10.1371/journal.pone.0101486

PubMed Abstract | CrossRef Full Text | Google Scholar

Kovacs, S. D., van Eijk, A. M., Sevene, E., Dellicour, S., Weiss, N. S., Emerson, S., et al. (2016). The Safety of Artemisinin Derivatives for the Treatment of Malaria in the 2nd or 3rd Trimester of Pregnancy: A Systematic Review and Meta-Analysis. PloS One 11 (11), e0164963. doi: 10.1371/journal.pone.0164963

PubMed Abstract | CrossRef Full Text | Google Scholar

Krishna, S., Uhlemann, A. C., Haynes, R. K. (2004). Artemisinins: mechanisms of action and potential for resistance. Drug Resist. Update 7 (4-5), 233–244. doi: 10.1016/j.drup.2004.07.001

CrossRef Full Text | Google Scholar

Lalloo, D. G., Shingadia, D., Bell, D. J., Beeching, N. J., Whitty, C. J. M., Chiodini, P. L., et al. (2016). UK malaria treatment guidelines 2016. J. Infect. 72 (6), 635–649. doi: 10.1016/j.jinf.2016.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W. D., Dong, Y. J., Tu, Y. Y., Lin, Z. B. (2006). Dihydroarteannuin ameliorates lupus symptom of BXSB mice by inhibiting production of TNF-alpha and blocking the signaling pathway NF-kappa B translocation. Int. Immunopharmacol. 6 (8), 1243–1250. doi: 10.1016/j.intimp.2006.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Wei, Y., Zhang, P. L., Deng, Z. Y. (2011). Clinical study on immune function regulation of artesunate in patients with lupus nephritis. Lishizhen Med. Materia Med. Res. 22 (7), 1673–1674. doi: 10.3969/j.issn.1008-0805.2011.07.057

CrossRef Full Text | Google Scholar

Li, T. T., Zhang, X. H., Jing, J. F., Li, X., Yang, X. Q., Zhu, F. H., et al. (2015). Artemisinin analogue SM934 ameliorates the proteinuria and renal fibrosis in rat experimental membranous nephropathy. Acta Pharmacol. Sin. 36 (2), 188–199. doi: 10.1038/aps.2014.134

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, L., Chen, X., Dong, F., Liu, Q., Zhang, C., Xu, D., et al. (2018). Dihydroartemisinin up-regulates VE-cadherin expression in human renal glomerular endothelial cells. J. Cell Mol. Med. 22 (3), 2028–2032. doi: 10.1111/jcmm.13448

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. (2012). Qinghaosu (artemisinin): chemistry and pharmacology. Acta Pharmacol. Sin. 33 (9), 1141–1146. doi: 10.1038/aps.2012.104

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, N., Zhong, Y., Zhou, J., Liu, B., Lu, R., Guan, Y., et al. (2018). Immunosuppressive effects of hydroxychloroquine and artemisinin combination therapy via the nuclear factor-kappaB signaling pathway in lupus nephritis mice. Exp. Ther. Med. 15 (3), 2436–2442. doi:10.3892/etm.2018.5708

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, X. Q., Yuan, J., Song, J. P., Guan, Y. Z., Fu, L., Zhou, J. Y., et al. (2016). Experimental Study on Artemisinin Combined with Hydroxychloroquine in Treating Rats with IgA Nephropathy. Traditional Chin. Drug Res. & Clin. Pharmacol. 27 (5), 608–613. doi: 10.19378/j.issn.1003-9783.2016.05.003

CrossRef Full Text | Google Scholar

Liu, L. Z., Zhang, M. X., Wang, S. (2017). Effects of artemisinin on excretion of nephrin, podocin mRNA and urinary podocyte cells in Heymann nephritis rats. Chin. Traditional Patent Med. 39 (10), 2176–2178. doi: 10.3969/j.issn.1001-1528.2017.10.041

CrossRef Full Text | Google Scholar

Liu, J. H., He, L., Zou, Z. M., Ding, Z. C., Zhang, X., Wang, H., et al. (2018). A Novel Inhibitor of Homodimerization Targeting MyD88 Ameliorates Renal Interstitial Fibrosis by Counteracting TGF-beta1-Induced EMT in Vivo and in Vitro. Kidney Blood Press Res. 43 (5), 1677–1687. doi: 10.1159/000494745

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Lu, J., Liao, Y., Liu, S., Chen, Y., He, R., et al. (2019). Dihydroartemisinin attenuates lipopolysaccharide-induced acute kidney injury by inhibiting inflammation and oxidative stress. BioMed. Pharmacother. 117, 109070. doi: 10.1016/j.biopha.2019.109070

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, L. (2002). Study on effect of Cordyceps sinensis and artemisinin in preventing recurrence of lupus nephritis. Chin. J. Integrated Traditional Western Med. 22 (3), 169–171. doi: 10.3321/j.issn:1003-5370.2002.03.003

CrossRef Full Text | Google Scholar

Luo, Y., et al. (2019). Artemisinin-Based Smart Nanomedicines with Self-Supply of Ferrous Ion to Enhance Oxidative Stress for Specific and Efficient Cancer Treatment. ACS Appl. Mater Interfaces 11 (33), 29490–29497. doi: 10.1021/acsami.9b07390

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, X. Y., Yang, M., Mi, X. H., Wu, W. H., Zeng, Y., Ma, Y. X., et al. (2007a). Effects of artesunate on the proliferation of rats’ glomerular mesangial cells. Modern Preventive Med. 34 (23), 4415–4416, 4439. doi: 10.3969/j.issn.1003-8507.2007.23.007

CrossRef Full Text | Google Scholar

MA, X. Y., Yang, M., Wu, W. H., Zeng, Y., Mi, X. H. (2007b). Effects of artesunate on the cell cycle of rat glomerular mesangial cells. Sichuan Med. J. 28 (12), 1308. doi: 10.3969/j.issn.1004-0501.2007.12.001

CrossRef Full Text | Google Scholar

Ma, Y. X., Luan, S., Ma, X. Y., Zeng, Y., Wu, W. H., Mi, X. H., et al. (2009). The effect of artemisia on IL-2 and IL-6 in IgA nephropathy in rat. Med. J. West China 21 (5), 716–718. doi: 10.3969/j.issn.1672-3511.2009.05.005

CrossRef Full Text | Google Scholar

Ma, X. Y., Wu, W. H., Zeng, Y., Ma, Y. X., Mi, X. H. (2010). The anti-inflammatory effect and its mechanism of artemisia on the renal of UUO rats. Chongqing Med. 39 (12), 1514–1516. doi: 10.3969/j.issn.1671-8348.2010.12.013

CrossRef Full Text | Google Scholar

Mehta, D., Malik, A. B. (2006). Signaling mechanisms regulating endothelial permeability. Physiol. Rev. 86 (1), 279–367. doi: 10.1152/physrev.00012.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

Mi, X. H., Luan, S., Zeng, Y., Shu, B., Wu, W. H., Ma, Y. X., et al. (2007). Effects of Artesunate on the expression of CTGF and α-SMA in renal tissue of unilateral ureteral obstructed rat model. Med. J. West China 19 (3), 332–338. doi: 10.3969/j.issn.1672-3511.2007.03.002

CrossRef Full Text | Google Scholar

Mi, X. H., Luan, S., Ma, X. Y., Ma, Y. X., Zeng, Y., Wu, W. H., et al. (2009). Effects of Artesunate on MCP-1 and MCP-1 mRNA expression of renal tissue in the rat IgA nephropathy model. J. Sichuan Univ (Med Sci. Edi) 40 (5), 821–825. doi: 10.3969/j.issn.1672-173X.2009.05.013

CrossRef Full Text | Google Scholar

Mssusa, A. K., Fimbo, A. M., Nkayamba, A. F., Irunde, H. F., Sillo, H. B., Shewiyo, D. H., et al. (2016). Safety Profile of Artemether-Lumefantrine: A Cohort Event Monitoring Study in Public Health Facilities in Tanzania. Clin. Drug Invest. 36 (5), 401–411. doi: 10.1007/s40261-016-0385-z

CrossRef Full Text | Google Scholar

Mu, X., Wang, C. (2018). Artemisinins-a Promising New Treatment for Systemic Lupus Erythematosus: a Descriptive Review. Curr. Rheumatol Rep. 20 (9), 55. doi: 10.1007/s11926-018-0764-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Nie, H., Su, K., Long, Y., Jang, S. S., Sun, Q. (2015). Effect of artesunate on expressions of Toll-like receptor 4 and interleukin-8 in renaI tissues of diabetic nephropathy rat. Tianjin Med. J. 43 (4), 356–359. doi: 10.11958/j.issn.0253-9896.2015.04.006

CrossRef Full Text | Google Scholar

Otuechere, C. A., Edewor, G., Kale, O. E., Ekor, M. (2012). Subacute therapeutic dosing of artemether-lumefantrine and artesunate-amodiaquine combination preserves plasma cholesterol, renal antioxidant status, and organ weights in rats. Malar Res. Treat 2012, 257986. doi: 10.1155/2012/257986

PubMed Abstract | CrossRef Full Text | Google Scholar

Razavi, A., Nouri, H. R., Mehrabian, F., Mirshafiey, A. (2007). Treatment of experimental nephrotic syndrome with artesunate. Int. J. Toxicol. 26 (4), 373–380. doi: 10.1080/10915810701493293

PubMed Abstract | CrossRef Full Text | Google Scholar

Robert, A., Benoit-Vical, F., Claparols, C., Meunier, B. (2005). The antimalarial drug artemisinin alkylates heme in infected mice. Proc. Natl. Acad. Sci. U S A 102 (38), 13676–13680. doi: 10.1073/pnas.0500972102

PubMed Abstract | CrossRef Full Text | Google Scholar

Roussel, C., Caumes, E., Thellier, M., Ndour, P. A., Buffet, P. A., Jaureguiberry, S., et al. (2017). Artesunate to treat severe malaria in travellers: review of efficacy and safety and practical implications. J. Travel Med. 24 (2), 1–9. doi: 10.1093/jtm/taw093

CrossRef Full Text | Google Scholar

Ruiz, S., Pergola, P. E., Zager, R. A., Vaziri, N. D. (2013). Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int. 83 (6), 1029–1041. doi: 10.1038/ki.2012.439

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruiz-Andres, O., Sanchez-Niño, M. D., Moreno, J. A., Ruiz-Ortega, M., Ramos, A. M., Sanz, A. B., et al. (2016). Downregulation of kidney protective factors by inflammation: role of transcription factors and epigenetic mechanisms. Am. J. Physiology-Renal Physiol. 311 (6), F1329–F1340. doi: 10.1152/ajprenal.00487.2016

CrossRef Full Text | Google Scholar

Santos, C. B., Lobato, C. C., Braga, F. S., Costa Jda, S., Favacho, H. A., Carvalho, J. C., et al. (2015). Rational Design of Antimalarial Drugs Using Molecular Modeling and Statistical Analysis. Curr. Pharm. Des. 21 (28), 4112–4127. doi: 10.2174/1381612821666150528121423

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, B. N., Marschall, M., Henriksen, S., Rinaldo, C. H. (2014). Antiviral effects of artesunate on polyomavirus BK replication in primary human kidney cells. Antimicrob Agents Chemother. 58 (1), 279–289. doi: 10.1128/AAC.01800-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, C., Li, H., Yang, Y., Hou, L. (2015). Anti-inflammatory and immunoregulatory functions of artemisinin and its derivatives. Mediators Inflammation 2015, 435713. doi: 10.1155/2015/435713

CrossRef Full Text | Google Scholar

Sun, Z., Ma, Y., Chen, F., Wang, S., Chen, B., Shi, J., et al. (2018). Artesunate ameliorates high glucose-induced rat glomerular mesangial cell injury by suppressing the TLR4/NF-kappaB/NLRP3 inflammasome pathway. Chem. Biol. Interact. 293, 11–19. doi: 10.1016/j.cbi.2018.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Sureshbabu, A., Ryter, S. W., Choi, M. E. (2015). Oxidative stress and autophagy: crucial modulators of kidney injury. Redox Biol. 4, 208–214. doi: 10.1016/j.redox.2015.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Tecklenborg, J., Clayton, D., Siebert, S., Coley, S. M. (2018). The role of the immune system in kidney disease. Clin. Exp. Immunol. 192 (2), 142–150. doi: 10.1111/cei.13119

PubMed Abstract | CrossRef Full Text | Google Scholar

Vennerstrom, J. L., Arbe-Barnes, S., Brun, R., Charman, S. A., Chiu, F. C., Chollet, J., et al. (2004). Identification of an antimalarial synthetic trioxolane drug development candidate. Nature 430 (7002), 900–904. doi: 10.1038/nature02779

PubMed Abstract | CrossRef Full Text | Google Scholar

von Hagens, C., Walter-Sack, I., Goeckenjan, M., Osburg, J., Storch-Hagenlocher, B., Sertel, S., et al. (2017). Prospective open uncontrolled phase I study to define a well-tolerated dose of oral artesunate as add-on therapy in patients with metastatic breast cancer (ARTIC M33/2). Breast Cancer Res. Treat 164 (2), 359–369. doi: 10.1007/s10549-017-4261-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wan, R. J., Li, Y. H. (2017). Effects of Artesunate prevent nephritis via the Tolllike receptor 4/nuclear factorkappaB signaling pathway in rats. Mol. Med. Rep. 16 (5), 6389–6395. doi: 10.3892/mmr.2017.7362

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Wang, H., Jiang, B., Zhang, H. Y., Liu, B. J., Sun, L. Y., et al. (2010). Artesunate relieves lupus nephritis by inhibiting the expression of ICAM-1. J. Clin. Med. Pract. 14 (17), 1–3. doi: 10.3969/j.issn.1672-2353.2010.17.001

CrossRef Full Text | Google Scholar

Wang, H. X., Su, K., Xun, J. Q., Huang, X. J., Wang, J. G., et al. (2016). Effects of artesunate on proliferation, apoptosis and expression of TNF-α and IL-6 in rat mesangial cells. China Health Care Nutr. 26, 208.

Google Scholar

Wen, Y., Pan, M. M., Lv, L. L., Tang, T. T., Zhou, L. T., Wang, B., et al. (2019). Artemisinin attenuates tubulointerstitial inflammation and fibrosis via the NF-kappaB/NLRP3 pathway in rats with 5/6 subtotal nephrectomy. J. Cell Biochem. 120 (3), 4291–4300. doi: 10.1002/jcb.27714

PubMed Abstract | CrossRef Full Text | Google Scholar

White, N. J., Hien, T. T., Nosten, F. H. (2015). A Brief History of Qinghaosu. Trends Parasitol. 31 (12), 607–610. doi: 10.1016/j.pt.2015.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Wang, X., Zhang, W., Shi, X., An, P., Sun, W., Wang, Z., et al. (2010). Therapeutic effect of artemisinin on lupus nephritis mice and its mechanisms. Acta Biochim. Biophys. Sin. (Shanghai) 42 (12), 916–923. doi: 10.1093/abbs/gmq101

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. L., Zhang, W. G., Shi, X. M., An, P., Sun, W. S., Qiao, C. L., et al. (2010). Effects of Artemisinin on Proliferation, Apoptosis and Caspase-3 Expression of Rat Mesangial Cell. J. Chin. Med. Mater. 33 (3), 407–410.

Google Scholar

Wu, P., Li, Q. J., Xia, Z. L., Zhang, F. Q., Xia, Q. J. (2011). Establishment of Mice Nephritis Models and Observation of the Effects of Dihydroartemisinin on the Release of Inflammatory Cytokines. West China Med. J. 26 (7), 1028–1031.

Google Scholar

Wu, X. L., Shi, X. M., An, P., Sun, W. S., Qiao, C. L., et al. (2011). Effect of artemisinin combined with glucocorticoid on the expressions of glucocorticoid receptor alpha mRNA, glucocorticoid receptor beta mRNA and P300/CBP protein in lupus nephritis mice. Chin J. Integr. Med. 17 (4), 277–282. doi: 10.1007/s11655-011-0693-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., An, P., Ye, B., Shi, X., Dang, H., Fu, R., et al. (2014). Artemisinin ameliorated proteinuria in rats with adriamycin-induced nephropathy through regulating nephrin and podocin expressions. J. Tradit. Chin Med. 34 (1), 63–68. doi: 10.1016/S0254-6272(14)60056-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Xia, M., Liu, D., Tang, X., Liu, Y., Liu, H., Liu, Y., et al. (2020). Dihydroartemisinin inhibits the proliferation of IgAN mesangial cells through the mTOR signaling pathway. Int. Immunopharmacol. 80, 106125. doi: 10.1016/j.intimp.2019.106125

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, M., Chen, Z., He, L., Xiong, G., Lu, J. (2019). Transcription profiling of artemisinin-treated diabetic nephropathy rats using high-throughput sequencing. Life Sci. 219, 353–363. doi: 10.1016/j.lfs.2019.01.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, D. X., Qiu, J., Zhou, H. H., Yu, Y., Zhou, D. L., Xu, Y., et al. (2018). Dihydroartemisinin alleviates oxidative stress in bleomycin-induced pulmonary fibrosis. Life Sci. 205, 176–183. doi: 10.1016/j.lfs.2018.05.022

PubMed Abstract | CrossRef Full Text | Google Scholar

You, Y. W., Liao, P. H., Yang, F. F., Lin, X. (2014). Regulating effect of dihydroartemisinin on fractalkine expression in renal cortex of lupus-prone MRL/Ipr mice. Immunological J. 30 (7), 617–622. doi: 10.13431/j.cnki.immunol.j.20140135

CrossRef Full Text | Google Scholar

Yu, C., Sun, P., Zhou, Y., Shen, B., Zhou, M., Wu, L., et al. (2019). Inhibition of AKT enhances the anti-cancer effects of Artemisinin in clear cell renal cell carcinoma. BioMed. Pharmacother. 118, 109383. doi: 10.1016/j.biopha.2019.109383

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L. K., Su, Y. J., Zhou, F. J., Zhang, J. Y., An, J. X. (2014a). Effect of Artemisinin on the upregulation of PDGF-B protein expression in the kidney of experimental diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (13), 1392–1396. doi: 10.3969/j.issn.1008-8849.2014.13.009

CrossRef Full Text | Google Scholar

Zhang, L. K., Zhang, J. Y., Zhou, F. J., Su, Y. J., Zhang, J. Y, An, J. X. (2014b). Study on the mechanism of renoprotective effects of artemisinin in diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (26), 2862–2864. doi: 10.3969/j.issn.1008-8849.2014.26.005

CrossRef Full Text | Google Scholar

Zhang, L. K., Zhang, J. Y., Zhou, F. J., Su, Y. J., An, J. X., et al. (2014c). Inhibitory effect of artemisinin on the spatiotemporal dynamics activation of protein kinase C in the kidney of experimental diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (18), 1964–1966.

Google Scholar

Zhang, Y., Li, H., Zhu, J., Wei, T., Peng, Y., Li, R., et al. (2017). Role of artesunate in TGF-β1-induced renal tubular epithelial-mesenchymal transdifferentiation in NRK-52E cells. Mol. Med. Rep. 16 (6), 8891–8899. doi: 10.3892/mmr.2017.7728

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Cha, W. Q., Chen, X. H., Jiang, L., Jiang, Y. X. (2018). Effect of Artemisinin on Cell Proliferation and Fascin Expression of Human Renal Cell Carcinoma Cell Line 786-0. Med. Innovation China 15 (24), 29–31. doi: 10.3969/j.issn.1674-4985.2018.24.008

CrossRef Full Text | Google Scholar

Zhang, B., Liu, P., Zhou, Y., Chen, Z., He, Y., Mo, M., et al. (2019). Dihydroartemisinin attenuates renal fibrosis through regulation of fibroblast proliferation and differentiation. Life Sci. 223, 29–37. doi: 10.1016/j.lfs.2019.03.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, F. J., Su, Y. J., Zhang, L. K., Zhang, J. Y., An, J. X. (2014a). Inhibitory effect of artemisinin on the upregulation of DNA-binding activity of NF-κB in kidney tissue of diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (19), 2075–2079. doi: 10.3969/j.issn.1008-8849.2014.19.009

CrossRef Full Text | Google Scholar

Zhou, F. J., Zhang, L. K., Su, Y. J., Zhang, J. Y., An, J. X. (2014b). Inhibitory effect of artemisinin on the upregulation of c-fos and c-jun gene expression in kidney tissue of diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (21), 2294–2298. doi: 10.3969/j.issn.1008-8849.2014.21.005

CrossRef Full Text | Google Scholar

Zhou, F. J., Zhang, J. Y., Zhang, L. K., Su, Y. J., An, J. X. (2014c). Inhibitory effect of artemisinin on the upregulation of the DNA-binding activity of AP-1 in the kidney tissue of experimental diabetic rats. Modern J. Integrated Traditional Chin. Western Med. 23 (24), 2648–2652. doi: 10.3969/j.issn.1008-8849.2014.24.008

CrossRef Full Text | Google Scholar

Zhou, Z., Lu, B., Wang, C., Wang, Z., Luo, T., Piao, M., et al. (2017). RIP1 and RIP3 contribute to shikonin-induced DNA double-strand breaks in glioma cells via increase of intracellular reactive oxygen species. Cancer Lett. 390, 77–90. doi: 10.1016/j.canlet.2017.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Zuma, N. H., Smit, F. J., de Kock, C., Combrinck, J., Smith, P. J., N'Da, D. D., et al. (2016). Synthesis and biological evaluation of a series of non-hemiacetal ester derivatives of artemisinin. Eur. J. Med. Chem. 122, 635–646. doi: 10.1016/j.ejmech.2016.07.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: artemisinin derivatives (artemisinins), kidney disease, inflammation, immunity, oxidative stress

Citation: Xia M, Liu D, Liu Y and Liu H (2020) The Therapeutic Effect of Artemisinin and Its Derivatives in Kidney Disease. Front. Pharmacol. 11:380. doi: 10.3389/fphar.2020.00380

Received: 19 September 2019; Accepted: 12 March 2020;
Published: 31 March 2020.

Edited by:

Norberto Perico, Istituto Di Ricerche Farmacologiche Mario Negri, Italy

Reviewed by:

Ilse Sofia Daehn, Icahn School of Medicine at Mount Sinai, United States
Bente Jespersen, Aarhus University Hospital, Denmark

Copyright © 2020 Xia, Liu, Liu and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hong Liu, liuhong618@csu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.