State of the ART: Drug Screening Reveals Artesunate as a Promising Anti-Fibrosis Therapy

Commentary Open Access

State of the ART: Drug Screening Reveals Artesunate as a Promising Anti-Fibrosis Therapy

Author Information
1
Division of Pulmonary, Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
2
Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
3
Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
*
Authors to whom correspondence should be addressed.
Views:158
Downloads:24
Journal of Respiratory Biology and Translational Medicine 2025, 2 (1), 10023;  https://doi.org/10.70322/jrbtm.2024.10023

Received: 04 December 2024 Accepted: 13 December 2024 Published: 16 December 2024

Creative Commons

© 2024 The authors. This is an open access article under the Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/).

ABSTRACT: Fibrosis is a progressive pathological process that severely impairs normal organ function. Current treatments for fibrosis are extremely limited, with no curative approaches available. In a recent article published in Cell, Zhang and colleagues employed drug screening using ACTA2 reporter iPSC-derived cardiac fibroblasts and identified artesunate as a potent antifibrotic drug by targeting MD2/TLR4 signaling. This study provides new insights into strategies for exploiting existing drugs to treat fibrosis.
Keywords: Fibrosis; Drug screening; Artesunate; MD2/TLR4

Main Text

Fibrosis is an excessive accumulation of extracellular matrix (ECM) resulting from an aberrant wound healing response caused by repeated injury, which can lead to distortion of tissue architecture and loss of organ function. Fibrosis can occur in any solid organ and tissue, resulting in the failure of various vital organs, including the lung, heart, liver and kidney [1,2,3]. Despite its severe symptoms and poor prognosis, therapies for organ fibrosis remain limited for decades. Only two drugs, pirfenidone and nintedanib, have been approved for treating idiopathic pulmonary fibrosis (IPF), neither of which, however, can fully halt disease progression, and both have significant adverse effects [4,5]. Therefore, there is an urgent need to develop more strategies to target organ fibrosis. In recent years, phenotypic drug discovery (PDD) has attracted immense attention in drug discovery. Compared with traditional target-based drug discovery (TDD), PDD focuses on the effects of the potential drug targets on disease pathophysiology instead of on the specific molecular or chemical structures, demonstrating significantly higher efficiency in pharmaceutical industry productivity [6,7]. Meanwhile, emerging technologies such as high-throughput screening (HTS), high-content screening (HCS), and in silico simulations provide practical support for the application of PDD [8,9,10]. This strategy is highly valuable in addressing the gap for discovering more potential antifibrotic drugs. A recent study by Zhang et al. published in the journal Cell employed a drug screening system and identified artesunate (ART) as a promising anti-cardiac fibrosis compound [11]. To solve the problem that human primary cardiac fibroblasts (CFs) are insufficient for large-scale drug screening and replicates, the authors developed induced pluripotent stem cell (iPSC) derived human cardiac fibroblasts (iPSC-CFs), which remain quiescent up to P5 and serve as an ideal cell source for high-throughput screening (HTS). To enable real-time monitoring of myofibroblast activation and quantify the effectiveness of screened compounds, the authors generated an iPSC reporter cell line expressing Clover2 from the ACTA2 locus, a key myofibroblast marker gene, using CRISPR-Cas9. Then, the ACTA2Clover2 iPSC-CFs were generated in accordance with cardiac fibroblast development. Initially, the ACTA2Clover2 iPSC-CFs were stimulated with TGF-β1 and treated with approximately 5000 candidate compounds from a library of Known Bioactives and FDA Approved Drugs, followed by live cell Hoechst staining. High-content confocal microscope was then used to capture the Clover2 and Hoechst signals, generating 7-point dose-response curves and calculating half maximal effective concentration (EC50) and toxic concentration 50 (TC50) values. Compounds with high EC50 and low TC50 were excluded. The remaining candidates were further filtered using a machine learning predictor, yielding 20 compounds. After toxicity assessments using iPSC derived cardiomyocytes (iPSC-CMs) and iPSC derived endothelial cells (iPSC-ECs), the top hit was selected and used to perform in vitro and in vivo functional tests. Finally, the antifibrotic mechanism of the selected compound was investigated. Through the screening, the authors found artesunate (ART), a classic anti-malaria drug [12], emerged as a top candidate with a relatively low EC50 of 2.1 μM without toxicity to iPSC-CMs or iPSC-ECs up to 10 μM. The authors next conducted a series of in vitro and in vivo experiments to evaluate the antifibrotic effects of ART. In 2D in vitro studies, ART treatment inhibited TGF-β1 induced fibrotic gene expression, cell proliferation, migration, gel contraction, and collagen secretion from CFs. IPSC-derived 3D-engineered heart tissue models demonstrated significant improvement in contraction and relaxation velocity, alleviation of passive tissue tension, and suppression of fibrotic gene expression following ART treatment. In vivo, preventive or therapeutic TAC models, as well as an ischemia-reperfusion model, were utilized to mimic preclinical and clinical scenarios. ART was effective in attenuating fibrosis and improving cardiac function with the in vivo models. Single-nucleus RNA sequencing (snRNA-seq) revealed that ART inhibited TAC-induced dynamic transformation in CFs. ART is well known for its role in malaria therapy by binding heme [13]. However, heme-related genes were absent in cardiac fibroblasts. The authors then investigated the role of ART in inhibiting MD2/TLR4 signaling. Surface plasmon resonance (SPR) assay confirmed the binding between ART and MD2. In silico simulations suggested that ART reduced the flexibility of the TLR4 binding domain in MD2, thus antagonizing MD2-TLR4 interaction. MD2 conformational change was further validated by intrinsic tryptophan fluorescence at Tryptophan 23, which was transitioned from a buried hydrophobic state to a solvent-exposed position upon ART binding. Additionally, the ability of ART to interfere with LPS-MD2 binding was also evaluated, showing a stronger competitive binding to LPS compared with L6H21, which is another MD2-TLR4 inhibitor that interferes with the interaction between LPS and MD2. Proximity ligation assay (PLA) and co-immunoprecipitation (coIP) further verified that MD2-TLR4 interaction was inhibited by ART. ERK signaling, one of the major downstream pathways of MD2/TLR4, was also inhibited by ART. Transposase-accessible chromatin (ATAC)-seq indicated AP-1, especially c-FOS, was the top downregulated transcription factor by ART upon TGF-β1 stimulation. In summary, this study demonstrated that ART inhibited MD2-TLR4 interaction by targeting MD2 molecules, thereby suppressing ERK and AP-1 signaling to reduce fibrotic gene expression in cardiac fibroblasts (Figure 1). This study employs several state-of-the-art approaches to discover ART as an anti-fibrotic agent. Cell availability poses a significant challenge in anti-fibrosis drug screening due to the limited quantity and unsustainability of primary cells. The authors developed iPSC derived cells as the cell model, addressing the issue of cell quantity while preserving the features of primary cells. ACTA2Clover2 reporter cells were further generated to screen out potent antifibrotic compounds. This approach significantly increased the sensitivity and efficiency of drug screening, as well as the convenience of successive functional tests. The candidate compounds used in the screening were from libraries of Known Bioactives and FDA Approved Drugs [14], which are typically clinically established drugs repurposed for new applications rather than newly developed drugs. This approach of repurposing clinically established drugs for new applications significantly reduced time and resource costs. For functional tests, the authors utilized a range of in vitro and in vivo models, including a 2D cell model, a 3D engineered heart tissue model, and three in vivo models that mimic cardiac hypertrophy and myocardial infarction in clinical settings. The results strongly supported the therapeutic potential of ART in treating cardiac fibrosis. The in silico simulations and sequencing data provided insights into the potential mechanism of ART in treating fibrosis, which involves ART binding to MD2 to inhibit TLR4 signaling and its cascades. We previously reported that TLR4 interacted with hyaluronan to participate in the regulation of lung inflammation and injury [15], while also playing a critical role in the renewal of type 2 alveolar epithelial cells and limiting lung fibrosis [16]. TLR4 was also reported to promote fibrogenesis by activating quiescent hepatic stellate cells (HSCs) [17]. These studies collectively emphasized the crucial role of TLR4 in the pathogenesis of fibrosis. As the authors cited in the article, among all TLR family members, only TLR4 requires MD2 as a co-receptor [18], highlighting the link between specific inhibition of MD2/TLR4 signaling by ART and its therapeutic potential in organ fibrosis. Additionally, ERK, a critical factor in myofibroblast activation and lung fibrosis [19], was investigated to be suppressed by ART, further implicating its effectiveness in anti-fibrosis. This study also underscores the endless new applications of artemisinins [20,21]. While the study was comprehensive and well-detailed, some points remain elusive. The mechanistic link between ART-MD2 and TGF-β signaling was unclear. Furthermore, other pathways beyond TGF-β signaling may also be involved in ART’s antifibrotic effects. In addition, while LPS is the most well-known activator of the TLR4 pathway [22,23], it is not a common inducer in fibrosis, especially in non-infectious cardiac injury. Additional damage-associated molecular patterns (DAMPs), such as hyaluronan [15,16] and the nuclear protein high-mobility group box 1 (HMGB1) [24,25], which are more relevant to non-infectious tissue fibrosis, should be considered for molecular interaction studies. Moreover, the long-term effects and safety of ART, and comprehensive investigations into its potential in other organ fibrosis, remain further to be evaluated.
Figure 1. A schematic diagram illustrating the study workflow and the functional pathway of ART in CFs. ACTA2<sup>Clover2</sup> iPSC-CFs were differentiated from ACTA2<sup>Clover2</sup> iPSCs, which were generated using CRISPR-Cas9. Drug screening was conducted following TGF-β1 stimulation and treatment with candidate compounds, leading to the identification of artesunate as a potent antifibrotic drug. Artesunate inhibited MD2/TLR4 signaling activated by DAMPs, thereby suppressing ERK activation—a noncanonical TGF-β pathway—and the subsequent AP-1 pathway, ultimately reducing fibrotic genes transcription and preventing myofibroblast activation. The diagram was generated with BioRender (https://www.biorender.com/).

Acknowledgments

We thank lab members for their insightful and helpful discussions.

Author Contributions

D.J. conceived the study. Y.Q. performed a literature search and drafted the paper. Y.Q., J.L. and D.J. wrote the paper.

Ethics Statement

Not applicable.

Informed Consent Statement

Not applicable.

Funding

The work was supported in part by NIH R01-AG078655 (J.L.) and R01-HL172990 (D.J.).

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships.

References

1.
Henderson NC, Rieder F, Wynn TA. Fibrosis: From mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar]
2.
Jun JI, Lau LF. Resolution of organ fibrosis. J. Clin. Investig. 2018, 128, 97–107. [Google Scholar]
3.
Rockey DC, Bell PD, Hill JA. Fibrosisa common pathway to organ injury and failure. N. Engl. J. Med. 2015, 372, 1138–1149. [Google Scholar]
4.
King TE, Jr., Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al.  A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis.  N. Engl. J. Med. 2014, 370, 2083–2092. [Google Scholar]
5.
Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2071–2082. [Google Scholar]
6.
Moffat JG, Vincent F, Lee JA, Eder J, Prunotto M. Opportunities and challenges in phenotypic drug discovery: An industry perspective. Nat. Rev. Drug Discov. 2017, 16, 531–543. [Google Scholar]
7.
Swinney DC, Anthony J. How were new medicines discovered? Nat. Rev. Drug Discov. 2011, 10, 507–519. [Google Scholar]
8.
Gerckens M, Schorpp K, Pelizza F, Wograth M, Reichau K, Ma H, et al. Phenotypic drug screening in a human fibrosis model identified a novel class of antifibrotic therapeutics. Sci. Adv. 2021, 7, eabb3673. [Google Scholar]
9.
Moshkov N, Becker T, Yang K, Horvath P, Dancik V, Wagner BK, et al. Predicting compound activity from phenotypic profiles and chemical structures.  Nat. Commun. 2023, 14, 1967. [Google Scholar]
10.
Nelson AR, Christiansen SL, Naegle KM, Saucerman JJ. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts. Proc. Natl. Acad. Sci. USA 2024, 121, e2303513121. [Google Scholar]
11.
Zhang H, Thai PN, Shivnaraine RV, Ren L, Wu X, Siepe DH, et al. Multiscale drug screening for cardiac fibrosis identifies MD2 as a therapeutic target. Cell 2024, 187, 7143–7163. [Google Scholar]
12.
Talman AM, Clain J, Duval R, Menard R, Ariey F.  Artemisinin Bioactivity and Resistance in Malaria Parasites. Trends Parasitol. 2019, 35, 953–963. [Google Scholar]
13.
Adebayo JO, Tijjani H, Adegunloye AP, Ishola AA, Balogun EA, Malomo SO. Enhancing the antimalarial activity of artesunate. Parasitol. Res. 2020, 119, 2749–2764. [Google Scholar]
14.
Stanford Medicine High-Throughput Screening Knowledge Center (HTSKC). Avalaible online: http://med.stanford.edu/htbc.html (accessed on 13 December 2024).
15.
Jiang D, Liang J, Fan J, Yu S, Chen S, Luo Y, et al.  Regulation of lung injury and repair by Toll-like receptors and hyaluronan.  Nat. Med. 2005, 11, 1173–1179. [Google Scholar]
16.
Liang J, Zhang Y, Xie T, Liu N, Chen H, Geng Y, et al. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice.  Nat. Med. 2016, 22, 1285–1293. [Google Scholar]
17.
Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis.  Nat. Med. 2007, 13, 1324–1332. [Google Scholar]
18.
Botos I, Segal DM, Davies DR. The structural biology of Toll-like receptors. Structure 2011, 19, 447–459. [Google Scholar]
19.
Ding Q, Gladson CL, Wu H, Hayasaka H, Olman MA. Focal adhesion kinase (FAK)-related non-kinase inhibits myofibroblast differentiation through differential MAPK activation in a FAK-dependent manner.  J. Biol. Chem. 2008, 283, 26839–26849. [Google Scholar]
20.
Krishna S, Bustamante L, Haynes RK, Staines HM. Artemisinins: Their growing importance in medicine. Trends Pharmacol. Sci. 2008, 29, 520–527. [Google Scholar]
21.
Li J, Casteels T, Frogne T, Ingvorsen C, Honore C, Courtney M, et al. Artemisinins Target GABA(A) Receptor Signaling and Impair alpha Cell Identity.  Cell 2017, 168, 86–100.e15. [Google Scholar]
22.
Medzhitov R, Janeway CA, Jr. Decoding the patterns of self and nonself by the innate immune system.  Science 2002, 296, 298–300. [Google Scholar]
23.
Akira S, Takeda K. Toll-like receptor signalling. Nat. Rev. Immunol. 2004, 4, 499–511. [Google Scholar]
24.
Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, et al. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J. Biol. Chem. 2004, 279, 7370–7377. [Google Scholar]
25.
Tsung A, Klune JR, Zhang X, Jeyabalan G, Cao Z, Peng X, et al. HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. J. Exp. Med. 2007, 204, 2913–2923. [Google Scholar]
TOP