Skip to main content
Frontiers in Pharmacology logoLink to Frontiers in Pharmacology
. 2021 Sep 16;12:744826. doi: 10.3389/fphar.2021.744826

Anlotinib Inhibits PFKFB3-Driven Glycolysis in Myofibroblasts to Reverse Pulmonary Fibrosis

Weimou Chen 1,, Jinming Zhang 1,, Wenshan Zhong 1,, Yuanyuan Liu 1, Ye Lu 1, Zhaojin Zeng 1, Haohua Huang 1, Xuan Wan 1, Xiaojing Meng 2, Fei Zou 2, Shaoxi Cai 1,*, Hangming Dong 1,*
PMCID: PMC8481786  PMID: 34603058

Abstract

Idiopathic pulmonary fibrosis (IPF) is a fatal disease in which the normal alveolar network is gradually replaced by fibrotic scars. Current evidence suggests that metabolic alterations correlate with myofibroblast activation in IPF. Anlotinib has been proposed to have antifibrotic effects, but the efficacy and mechanisms of anlotinib against lung fibrosis have not been systematically evaluated. The antifibrotic effects of anlotinib were evaluated in bleomycin-induced mouse models and transforming growth factor-beta 1 (TGF-β1)-stimulated lung fibroblasts. We measured lactate levels, 2-NBDG glucose uptake and the extracellular acidification rate (ECAR) to assess glycolysis in fibroblasts. RNA-protein coimmunoprecipitation (RIP) and polysome analyses were performed to investigate novel mechanisms of glycolytic reprogramming in pulmonary fibrosis. We found that anlotinib diminished myofibroblast activation and inhibited the augmentation of glycolysis. Moreover, we show that PCBP3 posttranscriptionally increases PFKFB3 expression by promoting its translation during myofibroblast activation, thus promoting glycolysis in myofibroblasts. Regarding mechanism, anlotinib exerts potent antifibrotic effects by downregulating PCBP3, reducing PFKFB3 translation and inhibiting glycolysis in myofibroblasts. Furthermore, we observed that anlotinib had preventative and therapeutic antifibrotic effects on bleomycin-induced pulmonary fibrosis. Therefore, we identify PCBP3 as a protein involved in the regulation of glycolysis reprogramming and lung fibrogenesis and propose it as a therapeutic target for pulmonary fibrosis. Our data suggest that anlotinib has antifibrotic effects on the lungs, and we provide a novel mechanism for this effect. Anlotinib may constitute a novel and potent candidate for the treatment of pulmonary fibrosis.

Keywords: pulmonary fibrosis, anlotinib, glycolysis, PFKFB3, PCBP3

Introduction

Fibrosis can develop in most organs and cause organ failure. The most common type of lung fibrosis is idiopathic pulmonary fibrosis (IPF), which is highly prevalent and associated with a dramatically increased disease burden worldwide (Wynn and Ramalingam, 2012; Hutchinson et al., 2015). Overall, the development of new therapeutics should be pursued. Currently, only pirfenidone and nintedanib have been approved as therapeutics for IPF (Taniguchi et al., 2010; Sato et al., 2017), and as both drugs have limited efficacy (Spagnolo and Maher, 2017), there is an urgent need to identify new potential therapeutic agents for IPF patients.

Upon chronic microinjury to the alveolar epithelium, fibroblast activation and transdifferentiation into myofibroblasts are among the first responses detectable at the site of damage (Plantier et al., 2018). Myofibroblasts are characterized by de novo expression of α-smooth muscle actin (α-SMA), the formation of stress fibers, and enhanced abilities to proliferate, migrate, and produce extracellular matrix (ECM) (Hinz, 2012; Hinz et al., 2012; Liu et al., 2021). These cells drive a wound-healing response that relies on the deposition of collagen-rich ECM and activates transforming growth factor-beta 1 (TGF-β1) signaling (Kenyon et al., 2003; Sapudom et al., 2015; Nigdelioglu et al., 2016). This transient response must be tightly controlled, otherwise it can become persistent and lead to excessive matrix accumulation and fibrosis. Understanding the molecular bases of fibroblast activation is therefore essential in identifying novel and efficient antifibrotic therapeutic targets to reduce the incidence, morbidity and mortality of people suffering from clinically refractory disorders, including IPF.

Metabolic perturbation is implicated in the pathogenesis of several kinds of tissue fibrosis (DeBerardinis and Thompson, 2012; Chen et al., 2018), including pulmonary fibrosis (Para et al., 2019; Bueno et al., 2020). To cope with the high energy demands of myofibroblasts, including increases in proliferation and matrix production, it is reasonable that activated myofibroblasts exhibit augmented aerobic glycolysis to meet additional bioenergetic and biosynthetic demands, even in oxygen-rich conditions, similar to observations in many cancer cells and other nonmalignant proliferating cells. A major driver of glycolysis is 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), which produces fructose-2,6-bisphosphate (F2,6BP), the most potent allosteric activator of the glycolytic rate-limiting enzyme phosphofructokinase-1 (PFK1) (Van Schaftingen et al., 1982; Cao et al., 2019). Previous studies have shown that augmentation of aerobic glycolysis is an essential step during myofibroblast activation (Para et al., 2019). Ramping down glycolysis is effective in diminishing myofibroblast activation, thus limiting lung fibrosis. However, metabolism-based therapeutics for treating fibrotic disorders are still lacking.

Anlotinib (AL3818) hydrochloride is a novel multitargeted tyrosine kinase inhibitor (TKI) that targets the receptor tyrosine kinases vascular endothelial growth factor receptor (VEGFR) 1 thru 3, epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR) 1 thru 4, platelet-derived growth factor receptor (PDGFR) α and β, and stem cell factor receptor (Sun et al., 2016; Xie et al., 2018). Many studies have reported the therapeutic effects of anlotinib in several diseases, such as nonsmall cell lung cancer (Liang et al., 2019), endometrial cancers (Taurin et al., 2018) and osteosarcoma (Liang et al., 2019). Interestingly, the targets of anlotinib are similar to those of nintedanib, a drug that has been approved for the treatment of IPF. A recent report suggested that intraperitoneal administration of anlotinib attenuates bleomycin-induced lung fibrosis in mice by suppressing the TGF-β signaling pathway (Ruan et al., 2020). Nevertheless, the mechanism by which anlotinib resolves lung fibrosis and whether anlotinib may be therapeutically used to improve lung function are not well understood.

In the current study, we analyzed the antifibrotic effect of anlotinib on TGF-β1-induced fibroblast transdifferentiation and bleomycin-induced pulmonary fibrosis. Our data suggest that anlotinib therapy decreases fibrotic markers in vitro and in vivo. The antifibrotic effect of anlotinib is associated with inhibition of PFKFB3-dependent glycolysis, which is posttranscriptionally regulated by the RNA binding protein PCBP3. These findings provide a theoretical basis for the clinical development and application of anlotinib for the treatment of pulmonary fibrosis.

Materials and Methods

Isolation of Primary Mouse Fibroblast Cultures

Normal mouse primary fibroblasts were generated by culturing the lungs of C57BL/6 as previously described (Meng et al., 2014). The cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM, Gibco, United States) supplemented with 15% fetal bovine serum (FBS, PAN, German). The cells were cultured at 37°C in 5% CO2 and 95% humidity. Unless specifically noted, all experiments were performed with cells at passage 3.

Cell Lines

Human lung fibroblast line IMR90 was purchased from American Type Culture Collection (Manassas, VA). IMR90 were maintained in DMEM supplemented with 10% FBS, 100 units/mL penicillin, and 100 g/ml streptomycin in 5% CO2 and 95% humidity at 37°C.

Western Blotting

Cells or dissected mouse lung tissue samples were lysed in ice-cold RIPA lysis buffer with protease inhibitors. Protein concentrations were determined using a BCA Protein Quantitative Analysis Kit (Fudebio-tech) after which protein samples were separated by 8–12% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (Millipore). The membranes were then incubated at room temperature for 1 h in TBST containing 5% BSA. After blocking, the membranes were incubated with primary antibodies for 24 h at 4°C.The following primary antibodies were used: anti-Fibronectin (Abcam, ab268020); anti-Collagen I (affinity, AF7001); anti-alpha smooth muscle (Abcam, ab5694); anti-PFKFB3 (Abcam, ab181861); anti-Beta actin (proteintech, 66009-1-Ig); anti-Hexokinase 2 (proteintech, 22029-1-AP); anti-PKM2 (Proteintech, 15822-1-AP); anti-LDHA (Proteintech, 19987-1-AP); anti-LDHB (Proteintech, 14824-1-AP); and anti-PCBP3 (Abcam, ab154252). Then, the membranes were washed three times with TBST and incubated with donkey anti-rabbit IgG H&L (Abcam, ab175772) for 1 h at room temperature. The membranes were developed using the ECL method according to the manufacturer’s instructions (Millipore) and detected on a GeneGnome XRQ chemiluminescence imaging system (Syngene). ImageJ was used to calculate the relative density of proteins.

Immunofluorescence Staining

The culture medium was washed away with PBS. The cultured cells were fixed with 4% paraformaldehyde for 30 min. Then, the samples were permeabilized with 0.5% Triton X-100 in PBS for 10 min, blocked with 1% BSA in PBS for 1 h at room temperature, and incubated with primary antibodies at 4°C overnight. The primary antibodies included anti- Fibronectin (Abcam, ab268020), anti-alpha smooth muscle (Abcam, ab5694) and anti-PCBP3 (Abcam, ab154252). Then, the cells were washed three times with PBS and incubated with goat anti-rabbit IgG/Alexa Fluor 555-conjugated secondary antibodies (Biosynthesis, bs-0296GA488 and bs-0295G-AF555) for 1 h at room temperature followed by 10 min of DAPI (4’,6-diamidino-2-phenylindole dihydrochloride) staining to visualize cell nuclei visualization as previously described (Chen et al., 2021).

Quantitative RT-PCR (qPCR)

Total RNA was isolated from primary mouse lung fibroblasts using RNA MiniPrep Kits (Zymo Research, R2050). Reverse transcription reactions were performed with a PrimeScriptTM II 1st strand cDNA synthesis Kit (Takara, 6210A/B) according to the manufacturer’s recommendations. qPCR analysis was performed using a HiScript RT- SuperMix for qPCR kit (Vazyme, R223-01) with a CFX96 Touch Real-Time PCR Detection System. The mRNA levels of target genes were normalized to the β-actin mRNA level. Primers used for qPCR are listed in (Table 1).

TABLE 1.

List of primer sequences used in this study.

Gene Species Forward primer Reverse primer
β-actin Mus musculus GGC​TGT​ATT​CCC​CTC​CAT​CG CCA​GTT​GGT​AAC​AAT​GCC​ATG​T
PFKFB3 Mus musculus CCC​AGA​GCC​GGG​TAC​AGA​A GGG​GAG​TTG​GTC​AGC​TTC​G

Wound-Healing Migration Assay

Cells were seeded in six-well plates and grown until they reach 100% confluence. A “wound” was subsequently created with a sterile 100 μL pipette tip. The cells were pretreated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for an additional 24 h. After 24 h, the cells were fixed with 4% paraformaldehyde, and images were obtained using a fluorescence microscope. Wound area can be calculated by manually tracing the cell-free area in captured images using the ImageJ public domain software (NIH, Bethesda, MD).

Cell Proliferation Assay

Cell proliferation was determined by the CCK-8 Kit (Dojindo Laboratories) according to the manufacturer’s instructions. Briefly, 10 μL of CCK-8 solution was added to cultured cells in each well, followed by incubation at 37°C for 1 h. The OD values were measured at 450 nm using a microplate reader. EdU staining was conducted using the BeyoClick™ EdU Cell Proliferation Kit with Alexa Fluor 594 (Beyotime, Cat. No: C00788L). Cells were washed with PBS. Fresh DMEM was added, and then, 10 µM EdU was added into the medium. The cells were incubated for 2 h at 37°C/5% CO2. After the incubation, the cells were washed with PBS to remove the DMEM and the free EdU probe. The cells were then fixed in 4% paraformaldehyde at room temperature for 30 min before being stained with DAPI for 3 min. After an additional wash in PBS, the cells were observed under Nikon ECLIPSE TS100 (Japan).

Glucose Uptake Assay

Primary mouse lung fibroblasts were pretreated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for an additional 24 h. Then, the four types of cells were detached and transferred to a 96-well plate in fresh growth medium at a density of 10,000 cells per well for the direct 2-NBDG glucose uptake assay. The cells were rinsed twice with PBS. Glucose uptake was initiated by the addition of 100 μM 2-NBDG to each well. After 30 min, the medium was removed. The plates were then rinsed with PBS, and the fluorescence was measured at an excitation wavelength of 485 nm and an emission wavelength of 535 nm.

Intracellular and Extracellular Lactate Analysis

To measure lactate production, cells were treated as described for the glucose uptake assay. One hundred thousand cells were then plated into a 12-well plate and incubated in DMEM containing 10% FBS for 10 h. To measure the secretion of lactate, the media were removed, and the cells were incubated in FBS-free DMEM. After incubation for 1 h, the supernatant was collected to measure lactate production (Biovision). The reaction mixture was incubated for 30 min at room temperature in the dark. The lactate levels were measured at 450 nm in a microplate reader and normalized to the protein concentrations. To measure the lactate levels in mouse lung tissue, 10 mg of lung tissues was isolated and homogenized in assay buffer (Biovision). The samples were centrifuged, and the soluble fractions were measured and normalized to the protein concentrations.

Extracellular Acidification Rate

The extracellular acidification rate (ECAR) was measured using the Agilent Seahorse XFp Extracellular Flux Analyzer (Seahorse Bioscience). Experiments were performed according to the manufacturer’s instructions. ECAR were measured using Seahorse XF Glycolysis Stress Test Kit (Agilent Technologies). Briefly, cells were transfected or infected as in glucose uptake assay. The transfected cells were harvested and the cell number was counted. After baseline measurements, glucose, the oxidative phosphorylation inhibitor oligomycin, and the glycolytic inhibitor 2-DG were sequentially injected into each well at the indicated time points. Data were analysed by Seahorse XFp Wave software. ECAR is reported in mpH/minute. The cells in each well were digested by trypsin digestion (Gibco, United States), and count cell numbers by cell counting chamber. The results were normalized to normalized to cell number in each well.

RNA Immunoprecipitation (RIP)

Cells were rinsed twice with ice-cold PBS and lysed with an equal pellet volume of RIPA-2 buffer. Protein-A Dynabeads (Invitrogen) were incubated with either mouse IgG or FLAG antibody (Abcam, ab205606). Beads coated in antibody were resuspended in NT2 buffer. Thawed and clarified lysates were added and the bead/antibody/lysate mixture was incubated at 4°C overnight rotating end-over-end. Beads were washed with cold NT2 buffer five times. Proteinase K treatment released RNAs from bound proteins and input and bound RNA was isolated with TRIzol (Invitrogen) and reverse transcribed as described above.

Polysome Analysis

Cells were transfected with empty vector or Flag-PCBP3 and incubated with 100 g/ml cycloheximide for 10 min and lysed with polysome extraction buffer containing 20 mM Tris–HCl, pH 7.5, 100 mM KCl, 5 mM MgCl2 and 0.5% NP-40 as previously described (Kim et al., 2015). Cytoplasmic lysates were fractionated by ultracentrifugation through 10–50% linear sucrose gradients and divided into 12 fractions. The total RNA in each fraction was extracted and analyzed by quantitative RT-PCR analysis.

Overexpression Experiments and RNA Interference

The Plasmid vector encoding PCBP3 and the empty vector were purchased from Hanbio (Shanghai, China). Primary mouse lung fibroblasts were cultured in six well plates (105 cells/well) and added with 2.5 μg of target plasmid per well. After 12 h, the transfection medium was changed to normal medium. Effects of overexpression on mRNA and protein levels were examined 36 h later. The siRNA targeting mouse PFKFB3 (PFKFB3 siRNA: 5′- CCU​CUU​GAC​CCU​GAU​AAA​UTT-3′) were synthesized by Genepharma Co. (Shanghai, China). Primary mouse lung fibroblasts were cultured in six well plates (105 cells/well) and transfected using Lipofectamine 3,000 (Invitrogen, CA) with PFKFB3 siRNA or negative control siRNA (NC siRNA) for 48 h following the manufacturer’s instructions.

Animal Experiments

All experiments were conducted in accordance with protocols approved by the Southern Medical University Institutional Animal Care and Use Committee. Female mice (C57BL/6), 6–8 weeks of age, were purchased from Southern Medical University. The mice were kept on a 12 h light-dark cycle with free access to food and water. For bleomycin administration, the mice were anesthetized with 2, 2, 2-tribromoethanol (Sigma-Aldrich), followed by intratracheal instillation of BLM (5 U/kg, i. t.) in 50 μL phosphate-buffered saline (PBS) or equally volume PBS for 21 days. The mice were administered dimethyl sulfoxide (DMSO) (control group) or anlotinib (1 mg/kg, i. p.) once daily for 21 consecutive days. Further experiments were designed to measure the effects of delayed anlotinib administration. Anlotinib treatment was initiated 1 week after exposure to bleomycin, and the mice were administered with anlotinib (1 or 2 mg/kg/day) for 2 weeks, and the mice were sacrificed at day 21. The lungs were harvested for further analyses.

Pulmonary Function Test

At endpoint, at least 5 mice from each group were anesthetized with 2,2,2-tribromoethanol in saline, tracheotomized below the larynx, and intubated with a tracheal cannula. After the surgery, the mice were placed inside the plethysmographic chamber and the cannula was connected to the machine. Pulmonary function was measured by pulmonary function test system (BUXCO, United States). The system’s software automatically records and displays the pulmonary function parameters.

Hydroxyproline Assay

Lung collagen content was measured with a hydroxyproline (HYP) kit (Nanjing Jian Cheng Institute, Nanjing, China). The lung tissues were prepared for hydrolysis, adjusting the PH value to 6.0–6.8. Subsequently, the developing solution was added to the tissues that were incubated at 37°C for 5 min. Absorbance was read at 550 nm using a microplate reader. Data were expressed as micrograms (µg) of HYP per mg of wet lung tissue.

Materials

TGF-β1 were purchased from R&D Systems, Inc. (Minneapolis, MN, United States). Anlotinib dihydrochloride (AL3818, S8726) were purchased from Selleck (Houston, TX, United States).

Statistical Analysis

The results are expressed as the means ± standard deviation (SD). Multigroup comparisons were performed using one-way ANOVA. Student’s t-test was used for comparisons between two groups. A p value of less than 0.05 was considered significant. Replicates consisted of at least three independent experiments. Analyses were performed on SPSS version 25.0 (IBM) for Windows and GraphPad Prism version 6.0 (GraphPad Software, CA).

Results

Anlotinib Represses Myofibroblast Activation and the Profibrogenic Phenotype in vitro

Given that TGF-β1 is the predominant cytokine that stimulates the differentiation of lung fibroblasts into myofibroblasts and induces ECM production (Sapudom et al., 2015; Huang et al., 2020), we examined the effect of anlotinib (the chemical structure is shown in Supplementary Figure S1) on TGF-β1-induced activation of primary mouse lung fibroblasts (MLFs). The CCK-8 assay results showed that anlotinib did not cause significant cytotoxicity at doses of 1 µM (Figure 1A). To mimic the inhibitory effect of anlotinib on the progression of lung fibrosis, primary MLFs were pretreated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for an additional 24 h. Our results demonstrated that TGF-β1 induced the expression of fibronectin, collagen I, and α-SMA, but anlotinib reversed the expression of these fibrotic markers (Figures 1B,C). Immunofluorescence analysis of α-SMA and fibronectin showed similar results (Figures 1D,E). We also examined whether anlotinib affected the proliferation and migration of fibroblasts, which have been shown to significantly contribute to many fibrotic pathologies (Jarman et al., 2014; Huang et al., 2020). As shown by the EdU (Figures 1F,G) and CCK-8 results (Figure 1H), anlotinib treatment prevented the TGF-β1-induced proliferation of primary MLFs. Moreover, anlotinib inhibited the TGF-β1-induced migration of fibroblasts (Figures 1I,J). These results were confirmed in the human IMR90 cell line (Figures 1K, L and Supplementary Figures S2A–D). These data indicate that anlotinib can repress myofibroblast activation and the profibrogenic phenotype in vitro.

FIGURE 1.

FIGURE 1

Anlotinib represses myofibroblast activation and the profibrogenic phenotype in vitro.(A) Dose-dependent cytotoxicity of anlotinib in primary mouse lung fibroblast isolated from healthy mice by CCK-8. (B) Western blots analysis of Fibronectin, Collagen I, α-SMA and β-actin in mouse lung fibroblasts treated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for 3  h, followed by TGF-β1 for an additional 24 h. (C) Quantification for the indicated protein (mean ± SD, n = 3). Immunofluorescence for α-SMA (green) (D) and Fibronectin (green) (E). DAPI-stained nuclei (blue). Scale bar, 25 µm. (F) An EdU assay was used to observe the proliferative cells. Scale bar, 100 µm. (G) The number of EdU-positive cells was recorded (mean ± SD, n = 3). (H) A cck8 assay was used to observe the proliferative cells. (I) Representative images to show scratch-wound assay. Scale bars, 100 µm. Experiments were performed as in B. Images were taken 0, and 24 h after assay (white lines indicate wound edge). (J) Quantitative analysis of migration distance (mean ± SD, n = 3). (K) Western blots analysis of Fibronectin, Collagen I, α-SMA and β-actin in IMR90 cells treated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for an additional 24 h. (L) Quantification for the indicated proteins in IMR90 cells (mean ± SD, n = 3). *p < 0.05, **p < 0.01, ***p < 0.001 VS TGF-β1-treated group (ANOVA).

Anlotinib Inhibits PFKFB3-Driven Glycolysis in Lung Myofibroblasts

To investigate the potential antifibrotic mechanisms of anlotinib, we estimated the most likely macromolecular targets of anlotinib and obtained 100 potential targets through SwissTargetPrediction (Table 2) (Gfeller et al., 2014). A total of 7,360 lung fibrosis-related targets were obtained from the GeneCards database (Table 3) (Safran et al., 2002). To clarify the interaction between potential anlotinib targets and lung fibrosis-related targets, the intersection of the targets was mapped by drawing a Venn diagram and constructing a target network (Figure 2A). Seventy-four common targets were shared between the potential anlotinib targets and lung fibrosis-related targets (Table 4). STRING (version 11.0) was used for protein-protein interaction (PPI) analysis (Figure 2B) (Szklarczyk et al., 2019). Bioinformatics analysis data identified mitogen-activated protein kinase (MAPK) signaling pathway-related genes as the top hits among the 74 genes (Figure 2C). Given that the MAPK pathway is well recognized as a metabolic regulator and that many of these genes control metabolic processes (Figure 2D) (Ho et al., 2004; Papa et al., 2019; Hu et al., 2020; Wang F. et al., 2020), we first confirmed the presence of glycolytic alterations in lung myofibroblasts. We directly measured the levels of lactate and found that both intracellular and extracellular lactate levels in lung myofibroblasts treated with TGF-β1 were significantly increased (Figures 2E,F). Consistent with the augmented glycolysis in lung myofibroblasts, these cells also demonstrated increased glucose consumption (Figure 2G). However, anlotinib treatment decreased the production and secretion of lactate and reduced the consumption of glucose (Figures 2E–G). Accordingly, extracellular acidification rate (ECAR) analysis indicated that treatment with TGF-β1 increased glycolysis and glycolytic activity in primary MLFs, both of which were also reduced by anlotinib (Figures 2H,I). To delineate the mechanisms by which anlotinib inhibits the augmented glycolysis observed in lung myofibroblasts, we assessed the expression of key glycolytic enzymes in these cells. We found that PFKFB3 was induced by TGF-β1 in lung fibroblasts and that anlotinib significantly decreased its expression at the protein level (Figures 2J,K). PFKFB3 is not a rate-limiting glycolytic enzyme; instead, PFKFB3 catalyzes the conversion of fructose-6-phosphate to fructose-2,6-bisphosphate, which is an allosteric activator of PFK1 and a potent stimulator of glycolysis (Atsumi et al., 2002; De Bock et al., 2013). Taken together, these data suggest that anlotinib can abrogate the PFKFB3-driven increase in glycolysis, participating in myofibroblast activation.

TABLE 2.

Targets of anlotinib obtained through SwissTargetPrediction.

Target Common name Uniprot ID ChEMBL ID Target class Probability* Known actives (3D/2D)
Tyrosine-protein kinase receptor UFO AXL P30530 CHEMBL4895 Kinase 0.106165761464 72/8
Tyrosine-protein kinase receptor TYRO3 TYRO3 Q06418 CHEMBL5314 Kinase 0.106165761464 53/3
Proto-oncogene tyrosine-protein kinase MER MERTK Q12866 CHEMBL5331 Kinase 0.106165761464 46/3
Receptor protein-tyrosine kinase erbB-2 ERBB2 P04626 CHEMBL1824 Kinase 0.106165761464 178/5
Tyrosine-protein kinase BRK PTK6 Q13882 CHEMBL4601 Kinase 0.106165761464 22/2
Serine/threonine-protein kinase Aurora-B AURKB Q96GD4 CHEMBL2185 Kinase 0.106165761464 143/14
Vascular endothelial growth factor receptor 1 FLT1 P17948 CHEMBL1868 Kinase 0.106165761464 79/18
Epidermal growth factor receptor erbB1 EGFR P00533 CHEMBL203 Kinase 0.106165761464 597/31
Vascular endothelial growth factor receptor 2 KDR P35968 CHEMBL279 Kinase 0.106165761464 485/95
Tyrosine-protein kinase SRC SRC P12931 CHEMBL267 Kinase 0.106165761464 448/42
Hepatocyte growth factor receptor MET P08581 CHEMBL3717 Kinase 0.106165761464 314/108
Serine/threonine-protein kinase GAK GAK O14976 CHEMBL4355 Kinase 0.106165761464 16/2
Kinesin-1 heavy chain/Tyrosine-protein kinase receptor RET RET P07949 CHEMBL2041 Kinase 0.106165761464 72/11
ALK tyrosine kinase receptor ALK Q9UM73 CHEMBL4247 Kinase 0.106165761464 207/3
Tyrosine-protein kinase ABL ABL1 P00519 CHEMBL1862 Kinase 0.106165761464 127/11
Stem cell growth factor receptor KIT P10721 CHEMBL1936 Kinase 0.106165761464 106/10
Activin receptor type-1 ACVR1 Q04771 CHEMBL5903 Kinase 0.106165761464 29/1
Vascular endothelial growth factor receptor 3 FLT4 P35916 CHEMBL1955 Kinase 0.106165761464 37/8
Tyrosine-protein kinase receptor FLT3 FLT3 P36888 CHEMBL1974 Kinase 0.106165761464 156/14
Platelet-derived growth factor receptor alpha PDGFRA P16234 CHEMBL2007 Kinase 0.106165761464 57/14
Fibroblast growth factor receptor 1 FGFR1 P11362 CHEMBL3650 Kinase 0.106165761464 189/9
TGF-beta receptor type I TGFBR1 P36897 CHEMBL4439 Kinase 0.106165761464 78/4
Tyrosine-protein kinase LCK LCK P06239 CHEMBL258 Kinase 0.106165761464 167/19
Tyrosine-protein kinase BTK BTK Q06187 CHEMBL5251 Kinase 0.106165761464 67/5
Tyrosine-protein kinase Lyn LYN P07948 CHEMBL3905 Kinase 0.106165761464 76/6
Fibroblast growth factor receptor 3 FGFR3 P22607 CHEMBL2742 Kinase 0.106165761464 37/3
Platelet-derived growth factor receptor beta PDGFRB P09619 CHEMBL1913 Kinase 0.106165761464 114/8
Tyrosine-protein kinase YES YES1 P07947 CHEMBL2073 Kinase 0.106165761464 34/3
Fibroblast growth factor receptor 2 FGFR2 P21802 CHEMBL4142 Kinase 0.106165761464 35/27
Tyrosine-protein kinase FGR FGR P09769 CHEMBL4454 Kinase 0.106165761464 17/4
Macrophage colony stimulating factor receptor CSF1R P07333 CHEMBL1844 Kinase 0.106165761464 149/7
Tyrosine-protein kinase BLK BLK P51451 CHEMBL2250 Kinase 0.106165761464 20/4
Serine/threonine-protein kinase PLK4 PLK4 O00444 CHEMBL3788 Kinase 0.106165761464 14/2
Ephrin receptor EPHB4 P54760 CHEMBL5147 Kinase 0.106165761464 30/3
Tyrosine-protein kinase FYN FYN P06241 CHEMBL1841 Kinase 0.106165761464 36/3
Dual specificity mitogen-activated protein kinase kinase 2 MAP2K2 P36507 CHEMBL2964 Kinase 0.106165761464 11/3
Tyrosine-protein kinase HCK HCK P08631 CHEMBL3234 Kinase 0.106165761464 30/4
Serine/threonine-protein kinase 10 STK10 O94804 CHEMBL3981 Kinase 0.106165761464 13/4
Tyrosine-protein kinase ABL2 ABL2 P42684 CHEMBL4014 Kinase 0.106165761464 13/4
Tyrosine-protein kinase TIE-2 TEK Q02763 CHEMBL4128 Kinase 0.106165761464 24/16
Ephrin type-A receptor 8 EPHA8 P29322 CHEMBL4134 Kinase 0.106165761464 9/3
Serine/threonine-protein kinase 2 SLK Q9H2G2 CHEMBL4202 Kinase 0.106165761464 17/4
Tyrosine-protein kinase FRK FRK P42685 CHEMBL4223 Kinase 0.106165761464 15/3
Ephrin type-A receptor 6 EPHA6 Q9UF33 CHEMBL4526 Kinase 0.106165761464 10/4
TRAF2- and NCK-interacting kinase TNIK Q9UKE5 CHEMBL4527 Kinase 0.106165761464 20/4
Serine/threonine-protein kinase MST1 STK4 Q13043 CHEMBL4598 Kinase 0.106165761464 11/3
Mitogen-activated protein kinase kinase kinase kinase 5 MAP4K5 Q9Y4K4 CHEMBL4852 Kinase 0.106165761464 12/4
Casein kinase I epsilon CSNK1E P49674 CHEMBL4937 Kinase 0.106165761464 12/3
Ephrin type-A receptor 3 EPHA3 P29320 CHEMBL4954 Kinase 0.106165761464 11/3
Tyrosine-protein kinase receptor Tie-1 TIE1 P35590 CHEMBL5274 Kinase 0.106165761464 11/4
Mitogen-activated protein kinase kinase kinase kinase 3 MAP4K3 Q8IVH8 CHEMBL5432 Kinase 0.106165761464 14/3
Serine/threonine-protein kinase SIK2 SIK2 Q9H0K1 CHEMBL5699 Kinase 0.106165761464 12/3
Mitogen-activated protein kinase kinase kinase kinase 1 MAP4K1 Q92918 CHEMBL5749 Kinase 0.106165761464 14/3
Serine/threonine-protein kinase 33 STK33 Q9BYT3 CHEMBL6005 Kinase 0.106165761464 16/4
Mitogen-activated protein kinase kinase kinase kinase 4 MAP4K4 O95819 CHEMBL6166 Kinase 0.106165761464 17/4
Serine/threonine-protein kinase TAO2 TAOK2 Q9UL54 CHEMBL1075195 Kinase 0.106165761464 7/3
Serine/threonine-protein kinase TAO3 TAOK3 Q9H2K8 CHEMBL5701 Kinase 0.106165761464 12/2
Voltage-gated calcium channel alpha2/delta subunit 1 CACNA2D1 P54289 CHEMBL1919 Calcium channel auxiliary subunit alpha2delta family 0.106165761464 15/0
Voltage-gated calcium channel alpha2/delta subunit 2 CACNA2D2 Q9NY47 CHEMBL3896 Calcium channel auxiliary subunit alpha2delta family 0.106165761464 4/0
Receptor protein-tyrosine kinase erbB-4 ERBB4 Q15303 CHEMBL3009 Kinase 0.106165761464 29/3
Mitogen-activated protein kinase kinase kinase 8 MAP3K8 P41279 CHEMBL4899 Kinase 0.106165761464 32/0
Serine/threonine-protein kinase/endoribonuclease IRE1 ERN1 O75460 CHEMBL1163101 Enzyme 0.106165761464 18/1
Macrophage-stimulating protein receptor MST1R Q04912 CHEMBL2689 Kinase 0.106165761464 8/11
Serine/threonine-protein kinase Aurora-C AURKC Q9UQB9 CHEMBL3935 Kinase 0.106165761464 15/2
Fibroblast growth factor receptor 4 FGFR4 P22455 CHEMBL3973 Kinase 0.106165761464 15/2
Ephrin type-A receptor 7 EPHA7 Q15375 CHEMBL4602 Kinase 0.106165761464 7/3
Dual specificity mitogen-activated protein kinase kinase 5 MAP2K5 Q13163 CHEMBL4948 Kinase 0.106165761464 11/4
Serine/threonine-protein kinase RIPK2 RIPK2 O43353 CHEMBL5014 Kinase 0.106165761464 6/4
Discoidin domain-containing receptor 2 DDR2 Q16832 CHEMBL5122 Kinase 0.106165761464 13/3
Activin receptor type-1B ACVR1B P36896 CHEMBL5310 Kinase 0.106165761464 6/1
Epithelial discoidin domain-containing receptor 1 DDR1 Q08345 CHEMBL5319 Kinase 0.106165761464 14/4
Mitogen-activated protein kinase kinase kinase kinase 2 MAP4K2 Q12851 CHEMBL5330 Kinase 0.106165761464 15/3
Misshapen-like kinase 1 MINK1 Q8N4C8 CHEMBL5518 Kinase 0.106165761464 14/4
Leukocyte tyrosine kinase receptor LTK P29376 CHEMBL5627 Kinase 0.106165761464 10/3
Serine/threonine-protein kinase 35 STK35 Q8TDR2 CHEMBL5651 Kinase 0.106165761464 9/4
Ephrin type-A receptor 1 EPHA1 P21709 CHEMBL5810 Kinase 0.106165761464 9/3
Ephrin receptor EPHB6 O15197 CHEMBL5836 Unclassified protein 0.106165761464 11/4
Receptor tyrosine-protein kinase erbB-3 ERBB3 P21860 CHEMBL5838 Kinase 0.106165761464 4/3
Serine/threonine-protein kinase MST4 STK26 Q9P289 CHEMBL5941 Kinase 0.106165761464 12/2
Eukaryotic translation initiation factor 2-alpha kinase 1 EIF2AK1 Q9BQI3 CHEMBL6029 Kinase 0.106165761464 3/3
SPS1/STE20-related protein kinase YSK4 MAP3K19 Q56UN5 CHEMBL6191 Kinase 0.106165761464 15/4
Serine/threonine-protein kinase AKT2 AKT2 P31751 CHEMBL2431 Kinase 0.106165761464 318/0
Protein kinase C gamma PRKCG P05129 CHEMBL2938 Kinase 0.106165761464 38/0
Serine/threonine-protein kinase AKT AKT3 Q9Y243 CHEMBL4816 Kinase 0.106165761464 73/0
Serine/threonine-protein kinase PIM1 PIM1 P11309 CHEMBL2147 Kinase 0.106165761464 638/0
Serine/threonine-protein kinase PIM2 PIM2 Q9P1W9 CHEMBL4523 Kinase 0.106165761464 448/0
Serine/threonine-protein kinase PIM3 PIM3 Q86V86 CHEMBL5407 Kinase 0.106165761464 331/0
Cyclin-dependent kinase 2/cyclin E1 CCNE1 CDK2 P24864 P24941 CHEMBL1907605 Kinase 0.106165761464 74/0
Tyrosine-protein kinase JAK1 JAK1 P23458 CHEMBL2835 Kinase 0.106165761464 137/0
Dipeptidyl peptidase VIII DPP8 Q6V1X1 CHEMBL4657 Protease 0.106165761464 346/0
Dipeptidyl peptidase IX DPP9 Q86TI2 CHEMBL4793 Protease 0.106165761464 239/0
Phosphodiesterase 4B PDE4B Q07343 CHEMBL275 Phosphodiesterase 0.106165761464 43/0
Protein kinase C iota PRKCI P41743 CHEMBL2598 Kinase 0.106165761464 287/0
Cyclin-dependent kinase 2 CDK2 P24941 CHEMBL301 Kinase 0.106165761464 170/0
Cyclin-dependent kinase 1 CDK1 P06493 CHEMBL308 Kinase 0.106165761464 146/0
Cyclin T1 CCNT1 O60563 CHEMBL2108 Other cytosolic protein 0.106165761464 111/0
Telomerase reverse transcriptase TERT O14746 CHEMBL2916 Enzyme 0.106165761464 79/0
Sodium/calcium exchanger 1 SLC8A1 P32418 CHEMBL4076 Electrochemical transporter 0.106165761464 44/0
Gonadotropin-releasing hormone receptor GNRHR P30968 CHEMBL1855 Family A G protein-coupled receptor 0.106165761464 431/0
Amine oxidase, copper containing AOC3 Q16853 CHEMBL3437 Enzyme 0.106165761464 19/0

TABLE 3.

Lung fibrosis-related targets obtained from the GeneCards database.

Number Gene Number Gene Number Gene Number Gene
1 CFTR 335 PDGFA 669 CPLANE1 1,003 HSPH1
2 TGFB1 336 MIR197 670 MKI67 1,004 MT-ATP6
3 TERT 337 MIRLET7B 671 TBX1 1,005 MIR212
4 TP53 338 MIR195 672 PI3 1,006 CASP10
5 TNF 339 MIR96 673 BMP4 1,007 PDE5A
6 SFTPC 340 CHI3L1 674 PMS2 1,008 SH2D1A
7 EGFR 341 MIRLET7E 675 SLC7A7 1,009 SCN5A
8 IL6 342 MIR16-1 676 E2F1 1,010 RSPH4A
9 MUC5B 343 MIR199A1 677 S100A1 1,011 PIK3R2
10 RTEL1 344 MIR210 678 TKT 1,012 DNAH9
11 IL10 345 MIR20A 679 MYRF 1,013 PSMA7
12 SFTPB 346 MIR378A 680 LAMA5 1,014 KIF3A
13 SFTPA1 347 THBD 681 COL2A1 1,015 CYP3A5
14 ELN 348 DMBT1 682 DEFB1 1,016 LOC111674466
15 ABCA3 349 MIR185 683 RPL5 1,017 ADM
16 CAV1 350 IL9 684 SOD1 1,018 PSMC3
17 IFNG 351 CFM1 685 PLK1 1,019 WDPCP
18 MUC1 352 CD79A 686 SCGB3A2 1,020 MIR101-1
19 SFTPA2 353 DYNC2LI1 687 SLC9A3R1 1,021 PRKCB
20 SERPINA1 354 NEK1 688 IREB2 1,022 IL13RA2
21 STAT3 355 IQCB1 689 LOC113664106 1,023 MYH11
22 TERC 356 MIR204 690 FADD 1,024 PRKG2
23 CCN2 357 HLA-A 691 CYP3A4 1,025 SUFU
24 IL13 358 IGF2 692 HSPB1 1,026 PIK3C2A
25 TLR4 359 FLNA 693 MAPK14 1,027 PSMC5
26 PARN 360 BCL2 694 CTSB 1,028 KCNQ1OT1
27 CTNNB1 361 GSN 695 IGF2R 1,029 MIR133A1
28 CCR6 362 MAP2K2 696 BAP1 1,030 GOPC
29 AKT1 363 BPIFA1 697 GATA2 1,031 TRAF2
30 MIR21 364 MIR15A 698 MMP8 1,032 TNFRSF6B
31 NKX2-1 365 EPHX1 699 ITGAV 1,033 MIR139
32 MMP1 366 HPS5 700 PTX3 1,034 LOC111674470
33 SMAD4 367 TRAF3IP1 701 MSLN 1,035 IGHMBP2
34 ACE 368 CFTR-AS1 702 AFF4 1,036 PSMA3
35 SPP1 369 AP3B1 703 CTCF 1,037 PSMC6
36 CXCL8 370 GAPDH 704 ADA 1,038 GADD45B
37 EGF 371 KRT19 705 KITLG 1,039 FKRP
38 FASLG 372 MIR107 706 ZNF423 1,040 DNAAF2
39 CDH1 373 CP 707 IL12A 1,041 ERCC5
40 KRAS 374 HLA-B 708 PRKCA 1,042 E2F3
41 SERPINE1 375 MMP12 709 EGR1 1,043 PIK3R3
42 BRAF 376 AGER 710 DLL4 1,044 PRF1
43 SMAD3 377 DDR1 711 IFNA1 1,045 IFNAR1
44 PRTN3 378 TNNT2 712 EZR 1,046 PF4
45 PIK3CA 379 CCL18 713 LGALS3 1,047 MIR27B
46 IL1RN 380 MIR148B 714 AFP 1,048 VTN
47 HMOX1 381 MIR141 715 TUBB2B 1,049 DNAJC5
48 IL1B 382 INS 716 EPCAM 1,050 BGLAP
49 HLA-DRB1 383 GLI3 717 APOB 1,051 MIR455
50 FAM13A 384 JAK2 718 SPPL2C 1,052 LOC111674471
51 FAS 385 EVC 719 CDK2 1,053 CSF3R
52 PTEN 386 HPS3 720 GDF2 1,054 VCL
53 SFTPD 387 IL33 721 EPO 1,055 NAGLU
54 ITGAM 388 NOS2 722 VCP 1,056 GUSB
55 DSP 389 SLPI 723 HSPA8 1,057 NEAT1
56 STN1 390 MIR133B 724 ATP4A 1,058 TBX4
57 TGFBR1 391 TCTN3 725 HBB 1,059 PSMD1
58 SRC 392 TLR5 726 SELE 1,060 PSMD12
59 CDKN2A 393 MUC4 727 NAT2 1,061 GAST
60 ERBB2 394 MIR93 728 FGF10 1,062 MIR99A
61 CTLA4 395 MIR18A 729 BMP7 1,063 CREB1
62 NFE2L2 396 SLC11A1 730 ETS1 1,064 TRPC6
63 MMP9 397 PSMA6 731 DEFB4A 1,065 EPAS1
64 MTOR 398 RNASE3 732 PRDM10 1,066 MIR125B1
65 HRAS 399 SMAD2 733 GJA1 1,067 IFNGR1
66 TINF2 400 ICAM1 734 RPS6KB1 1,068 PGR
67 FGFR1 401 TGFA 735 JAK3 1,069 TLR1
68 COL1A1 402 RPGR 736 TP63 1,070 CD40
69 NPHP3 403 ANXA5 737 HNF1B 1,071 DLK1
70 F2 404 CXCR1 738 BBS10 1,072 FOS
71 DPP9 405 IL2 739 ITGA2 1,073 DNAL1
72 TGFB2 406 GREM1 740 HOTAIR 1,074 MAD1L1
73 ATP11A 407 CD274 741 SGK1 1,075 PSMD11
74 HFE 408 MIR142 742 DNAAF1 1,076 HBA2
75 PDGFRB 409 GSTP1 743 DNMT1 1,077 RAB11B
76 ITGA3 410 PHOX2A 744 DAB2 1,078 CCKBR
77 MIRLET7D 411 SOS2 745 CDH2 1,079 CLCN3
78 MBL2 412 LMNA 746 HJV 1,080 PPARA
79 NOTCH1 413 ENPP2 747 PRKAA1 1,081 MB
80 MET 414 TOP1 748 HSPA5 1,082 GAS5-AS1
81 TIMP1 415 APEX1 749 CHUK 1,083 LOC111674474
82 SERPINH1 416 CXCL9 750 SKP2 1,084 KCNJ1
83 SCNN1A 417 PLAU 751 ILK 1,085 KIAA0319L
84 MMP2 418 MIRLET7C 752 ADORA2B 1,086 PSMA2
85 ELANE 419 MIR30E 753 SST 1,087 PSMD3
86 VEGFA 420 MIR214 754 CFLAR 1,088 LOC111674473
87 PLG 421 MIR486-1 755 CCNB1 1,089 ABCF2
88 MIR130A 422 KRT7 756 ADCY10 1,090 PSMD8
89 ADRB2 423 TNFRSF13B 757 HDAC9 1,091 MIR590
90 KIT 424 MIR106B 758 ARG1 1,092 PSMD7
91 CC2D2A 425 MIR181A1 759 BAK1 1,093 RNF5
92 TGFBR2 426 MTHFR 760 DICER1 1,094 GLIS3
93 PDGFRA 427 BTNL2 761 IKBKG 1,095 CHAT
94 IL4 428 PTPN11 762 MYH6 1,096 LOC111674476
95 NHP2 429 KDR 763 COL4A3 1,097 MIR23B
96 STK11 430 ELMOD2 764 FGA 1,098 MIR130B
97 NOP10 431 STING1 765 ABL1 1,099 MIR151A
98 ALOX5 432 THY1 766 GJA5 1,100 LOC111674478
99 TMEM67 433 MIR140 767 ARHGAP31 1,101 CD82
100 CCL2 434 VCAM1 768 FCGR3B 1,102 E2F2
101 MIR34C 435 CCR2 769 SLC26A9 1,103 RNF185
102 STAT1 436 CDKN1A 770 LRBA 1,104 MIR423
103 ALB 437 STAT4 771 PMM2 1,105 DEFB103B
104 HGF 438 CEP164 772 ACD 1,106 NFKB2
105 RPGRIP1L 439 MIR128-2 773 CHIT1 1,107 MIR186
106 NPHP1 440 S100A9 774 CDK6 1,108 AFAP1-AS1
107 TSC2 441 TLR3 775 CSF1 1,109 PSMC2
108 MDM2 442 SERPINA3 776 ABCG2 1,110 ERLIN2
109 TTC21B 443 MIR25 777 CASP1 1,111 GADD45G
110 EDN1 444 CD44 778 P2RX7 1,112 CS
111 WDR19 445 MALAT1 779 ASCL1 1,113 CD22
112 CRP 446 COL3A1 780 VWF 1,114 PSMB1
113 CCL11 447 CD28 781 PROM1 1,115 LOC111674479
114 AGT 448 MIR23A 782 PDPN 1,116 LOC111674467
115 CCND1 449 MIR19A 783 PRODH 1,117 SDHB
116 COL1A2 450 HAMP 784 ALG9 1,118 LOC111674465
117 COPA 451 MIR429 785 HOXD13 1,119 PSMD14
118 CASP8 452 MIR335 786 TNFRSF10A 1,120 IFNB1
119 DKC1 453 TTR 787 IL12B 1,121 PLAT
120 ACTC1 454 C2CD3 788 BBS9 1,122 PSMA5
121 FGFR2 455 NR1H4 789 NR3C1 1,123 NEB
122 IL17A 456 G6PD 790 MIR122 1,124 CHGA
123 SCNN1B 457 MIR127 791 ITGB3 1,125 MIR187
124 ATM 458 TNNI3 792 ERF 1,126 LOC113523647
125 MIR155 459 CXCL1 793 CCR5 1,127 HSD3B7
126 SHH 460 MIR424 794 ADIPOQ 1,128 LOC108491823
127 RB1 461 IFT52 795 TF 1,129 IL4R
128 CSF2 462 ZCCHC8 796 RXRA 1,130 PSMB4
129 CEP290 463 AGTR2 797 FAT4 1,131 DRC1
130 FCGR2A 464 NSMCE3 798 PSMD4 1,132 MIR361
131 PKHD1 465 TYR 799 UMOD 1,133 COL11A2
132 NPHP4 466 ABCB4 800 TMEM107 1,134 MIR497
133 MIR200B 467 NOS3 801 BLOC1S6 1,135 SBDS
134 MKS1 468 LPAR1 802 MMP14 1,136 TLR6
135 SCNN1G 469 CXCR2 803 PTK2B 1,137 MIR339
136 FAM111B 470 ZIC3 804 PIEZO2 1,138 RECQL4
137 FN1 471 LOC111674475 805 CDK1 1,139 DERL2
138 INVS 472 CCR7 806 SIRT3 1,140 SH2B3
139 STAT6 473 ANKS6 807 IL15 1,141 GUCA2A
140 WRAP53 474 CALR 808 TAP1 1,142 CCDC103
141 FGFR3 475 TCTN1 809 GATA6 1,143 PCNA
142 RET 476 MIR224 810 EPOR 1,144 PSME2
143 NRAS 477 IGF1R 811 PVT1 1,145 NCOR1
144 DDR2 478 XRCC1 812 MUSK 1,146 TTC37
145 FBN1 479 CD34 813 ESR2 1,147 PSMB5
146 OFD1 480 EVC2 814 MAP1B 1,148 PRKG1
147 MAP2K1 481 CEACAM5 815 MT-CYB 1,149 LEPR
148 ENG 482 MIR338 816 TRAF6 1,150 TNC
149 NEK8 483 GATA4 817 PKD1L1 1,151 PTGS1
150 FGF2 484 MIRLET7G 818 RBPJ 1,152 IFNA2
151 MUC5AC 485 SOD2 819 DMD 1,153 DZIP1L
152 NFKB1 486 MIR191 820 FABP4 1,154 MIR296
153 RARB 487 CCR3 821 CD80 1,155 CD86
154 TNFRSF1A 488 CXCL5 822 PSMA4 1,156 MYCL
155 IL5 489 MYLK 823 IGHE 1,157 VPS45
156 APC 490 H2AC18 824 EGFR-AS1 1,158 GBE1
157 SMPD1 491 GPC3 825 DNMT3B 1,159 IL7R
158 MPO 492 TMEM138 826 MIR26A1 1,160 CYP27A1
159 ACTA2 493 MIR377 827 FLT4 1,161 PLA2G7
160 MIR126 494 CCL22 828 CCNA2 1,162 NOTCH2
161 NOD2 495 SLC34A2 829 DNAI2 1,163 CEACAM1
162 MIR17 496 PTPN22 830 TUG1 1,164 MIR485
163 FGF7 497 PKD2 831 IFIH1 1,165 LOC113633876
164 IFT140 498 DNAH5 832 KATNIP 1,166 REG3A
165 KRT18 499 S100A4 833 MMP3 1,167 EPRS1
166 REN 500 BAX 834 KLF6 1,168 LOC113633875
167 PDCD1 501 THPO 835 BLOC1S3 1,169 LOC113604967
168 VEGFC 502 MIR137 836 SERPINB1 1,170 ZEB1
169 IFT80 503 FOXJ1 837 GLA 1,171 FOXM1
170 SCGB1A1 504 BCL2L1 838 LPO 1,172 CASR
171 BRCA2 505 TRPV4 839 F2R 1,173 TALDO1
172 AHI1 506 CCR4 840 BTK 1,174 MIR219A1
173 TCTN2 507 CADM1 841 SOCS3 1,175 CCL26
174 BMPR2 508 TMEM237 842 COL4A2 1,176 LOC113664107
175 RAF1 509 LOC111674463 843 NBAS 1,177 CLCN4
176 CXCR4 510 GBA 844 CEP104 1,178 SNHG1
177 KIAA0586 511 EZH2 845 MAGEA4 1,179 LAMA2
178 HIF1A 512 SOX9 846 DERL1 1,180 TCF7
179 FARSB 513 MIR24-2 847 AKT3 1,181 TFRC
180 TLR2 514 SPARC 848 NPPA 1,182 PLCZ1
181 AGTR1 515 S100A8 849 BCL2L11 1,183 POMC
182 BMP6 516 LTA 850 LAMA1 1,184 ABCC3
183 CDK4 517 MKKS 851 IL3 1,185 CRYAB
184 TMEM216 518 HYDIN 852 ITGA5 1,186 LOC110806263
185 RELA 519 GLI1 853 CYP2D6 1,187 MCM4
186 CSF3 520 BIRC5 854 DOCK6 1,188 TNFSF13B
187 CCL5 521 MAPK3 855 CANX 1,189 MIR149
188 ESR1 522 PSMB8 856 AXIN2 1,190 PLA2G6
189 MIR223 523 AP3D1 857 SCARB2 1,191 KNG1
190 FOXP3 524 MIRLET7A3 858 CDX2 1,192 HOTTIP
191 TTN 525 IDH1 859 SLC9A3 1,193 AR
192 MARS1 526 CYP2E1 860 ICOS 1,194 PSMB2
193 MAPK8 527 TNFSF10 861 PTGER4 1,195 LSM1
194 MEG3 528 PRKCD 862 SLC17A5 1,196 NIPBL
195 CCL3 529 MIR181C 863 CXCR5 1,197 PSMA8
196 HPS4 530 CYCS 864 TCTEX1D2 1,198 IL16
197 BRCA1 531 IFT27 865 MXRA5 1,199 CCAT1
198 IL2RA 532 TUBB3 866 FOXP1 1,200 SPRY2
199 HPS1 533 HP 867 MIR138-1 1,201 RIPK1
200 MIR34A 534 ALMS1 868 CLDN1 1,202 PHKG2
201 MIR200C 535 NOX4 869 MIR154 1,203 MIR503
202 B9D1 536 RYR1 870 INSR 1,204 GSTT1
203 Dnase1 537 TSLP 871 UCHL1 1,205 PRKCI
204 TSC1 538 VDAC1 872 DCN 1,206 AGL
205 WDR35 539 U2AF1 873 XPNPEP3 1,207 RAG2
206 WT1 540 CAT 874 KRT13 1,208 HNF1A-AS1
207 INPP5E 541 CALCA 875 COMT 1,209 CCAT2
208 MPL 542 DNAH11 876 EDNRA 1,210 ERLIN1
209 MIR125A 543 NF1 877 CA4 1,211 MIR125B2
210 CSPP1 544 PTK2 878 HLA-DQA1 1,212 MIR15B
211 GRP 545 SP110 879 BBS12 1,213 CREBBP
212 MIR144 546 IL12RB1 880 KRT8 1,214 CTSL
213 RTEL1-TNFRSF6B 547 NQO1 881 CMA1 1,215 NR5A1
214 SERPINC1 548 GAA 882 AKR1B10 1,216 MSR1
215 CD4 549 MIR409 883 LOC113633877 1,217 UTP4
216 DYNC2H1 550 VIP 884 CEP55 1,218 MIR301A
217 MIR145 551 GPT 885 MAP2K4 1,219 MUC7
218 FLT1 552 MIR708 886 CDK5 1,220 CASC2
219 TNFRSF1B 553 NFKBIA 887 P2RY2 1,221 ZNRD1ASP
220 MIR29A 554 KEAP1 888 NLRP3 1,222 PLCG2
221 HLA-DQB1 555 LOC111674477 889 PSMC4 1,223 MIR124-1
222 FHIT 556 CD36 890 HSPA4 1,224 MIR382
223 H19 557 IGFBP3 891 AVPR2 1,225 LAMP1
224 PKD1 558 NKX2-5 892 RPS27A 1,226 CD69
225 SOS1 559 CEP83 893 MIR375 1,227 DANCR
226 SPINK1 560 BDNF 894 MIR33A 1,228 TP53COR1
227 MIR31 561 ABCB11 895 ATF6 1,229 MYL3
228 IFT172 562 FGFR4 896 RXRB 1,230 CYSLTR2
229 MMP7 563 CTSG 897 UBC 1,231 SELL
230 CD8A 564 ITGB1 898 NPC2 1,232 LINC-ROR
231 PDGFB 565 CSF2RA 899 CLCN2 1,233 NOS1
232 SLC2A1 566 DNAI1 900 FOXE1 1,234 IFT20
233 JUN 567 ARL3 901 MIR132 1,235 MIR10B
234 BMP2 568 MECP2 902 HDGF 1,236 CD81
235 HMGB1 569 CDKN1B 903 WNT7B 1,237 ANTXR1
236 ERBB3 570 DNMT3A 904 SLC9A3R2 1,238 PRSS2
237 MIR150 571 VHL 905 H2AX 1,239 SNHG20
238 PRSS1 572 THBS1 906 PRKAG2 1,240 MIR95
239 MIR29C 573 YAP1 907 SERPINB3 1,241 GALC
240 MIR146B 574 TIMP2 908 LRP1B 1,242 DGCR5
241 ABCC1 575 TOLLIP 909 DNAAF3 1,243 HNMT
242 BBS2 576 KCNK3 910 BCL10 1,244 SLCO2A1
243 ABCB1 577 NOTCH3 911 RAG1 1,245 MLH1
244 CXCL10 578 RASSF1 912 LAMA4 1,246 PLA2G2A
245 XIAP 579 SP1 913 HLA-G 1,247 MME
246 KIF21A 580 RHOA 914 PSMB3 1,248 TYMS
247 PIK3R1 581 ABCC2 915 SIRT1 1,249 MIR198
248 BBS1 582 IKBKB 916 MIRLET7A1 1,250 JAK1
249 IRF1 583 GZMB 917 PLA2G1B 1,251 PDE4D
250 IL1A 584 LEP 918 KDM4C 1,252 LAMC2
251 PTPRC 585 BBS5 919 MUC6 1,253 AHR
252 HLA-DPB1 586 APOE 920 TUBB1 1,254 TPM1
253 ERCC6 587 NHLRC2 921 SKIV2L 1,255 MT-CO2
254 STX1A 588 NPPB 922 TPM2 1,256 ASCC1
255 SETD2 589 SYP 923 CCDC40 1,257 EOGT
256 CCL17 590 ACTB 924 TET2 1,258 CCL7
257 IDH2 591 PSMA1 925 TRIP11 1,259 TRPM4
258 CXCL12 592 PTCH1 926 ITGA2B 1,260 GPSM2
259 MIR27A 593 LOXL2 927 ENO2 1,261 TARS1
260 GSTM1 594 CEP41 928 EIF2AK3 1,262 TPM3
261 DCTN4 595 MYPN 929 RSPH9 1,263 CD14
262 PRKN 596 CR1 930 MIR193A 1,264 MIR216A
263 CXCR3 597 ERCC1 931 PXN 1,265 CTAG1B
264 MIR200A 598 IGFBP5 932 DNAAF5 1,266 MIR22HG
265 ARL13B 599 MGMT 933 LAT 1,267 MIR10A
266 PTGS2 600 VIM 934 RSPH1 1,268 PCAT1
267 MIR148A 601 ENO1 935 CDKN2B-AS1 1,269 SNHG15
268 MYC 602 NLRC4 936 ADORA1 1,270 BANCR
269 IGF1 603 PIK3CG 937 RAD51 1,271 PSAP
270 IL18 604 MYH7 938 DIABLO 1,272 IL11
271 CYP1A1 605 F13A1 939 TFR2 1,273 MIR362
272 NEK9 606 WNT4 940 RSPO2 1,274 UCA1
273 LOX 607 NME1 941 C4A 1,275 UBE2L3
274 MIR30D 608 CD63 942 IFI27 1,276 FBLN5
275 MIR146A 609 RPGRIP1 943 MIR29B1 1,277 GC
276 IFT122 610 ATP8B1 944 HSPD1 1,278 TP73-AS1
277 B9D2 611 GNAS 945 CCDC39 1,279 PPBP
278 KCNQ1 612 TUBB 946 PDE4A 1,280 LINC00473
279 CYP2A6 613 IFRD1 947 TRIM21 1,281 SOX2-OT
280 MIR483 614 CASP9 948 PLCG1 1,282 MIR181B1
281 MIR183 615 LAMP2 949 IL1RL1 1,283 XIST
282 IL1R1 616 HDAC2 950 BBIP1 1,284 MIR129-1
283 SDCCAG8 617 ANXA1 951 MIR196A1 1,285 PRL
284 PTRH2 618 IFT74 952 BAD 1,286 MIR193B
285 MIR182 619 ROS1 953 MYBPC3 1,287 NR3C2
286 CDKN3 620 CLEC7A 954 ARAF 1,288 MYL1
287 GUCY2C 621 AKT2 955 VDR 1,289 SDC1
288 MIR222 622 WRN 956 MAGEA1 1,290 MIR24-1
289 LOC111674472 623 DTNBP1 957 TAC1 1,291 PHB
290 KIF7 624 CD19 958 IDUA 1,292 MYL2
291 FLNC 625 SMARCA4 959 EPX 1,293 APOA1
292 PPARG 626 ANGPT2 960 ASXL1 1,294 SNHG12
293 XRCC3 627 PARP1 961 MAGEA3 1,295 CEP57
294 SNAI1 628 BBS7 962 RAC1 1,296 IL6R
295 POSTN 629 MIF 963 GATA1 1,297 SERPINF2
296 MIR192 630 SELP 964 GAS5 1,298 CALB2
297 JAG1 631 HSPG2 965 MIR9-1 1,299 MIR152
298 CD40LG 632 ATP12A 966 RYR2 1,300 ADK
299 DYNC2I1 633 HSP90AA1 967 JPH2 1,301 NRG1
300 ACVRL1 634 CHRM3 968 RIOX2 1,302 MIR501
301 MIR221 635 MVP 969 LOC111674464 1,303 GPRC5A
302 IFT43 636 DCDC2 970 TNFAIP3 1,304 LZTR1
303 DES 637 ERCC2 971 PIK3CB 1,305 TLR7
304 MIR22 638 PPP2R1B 972 DHCR7 1,306 POT1
305 TNFRSF10B 639 TYMP 973 C1S 1,307 EP300
306 IRF5 640 CLCA1 974 SNAI2 1,308 FBL
307 ALK 641 OGG1 975 ERBB4 1,309 PLOD2
308 CEP120 642 MCL1 976 ACVR1 1,310 DLL1
309 MIR451A 643 CCN4 977 NPC1 1,311 TOP2A
310 DYNC2I2 644 F3 978 LNX1 1,312 MAGEC2
311 CASP3 645 STAT5B 979 GRB2 1,313 MIR499A
312 CCL4 646 FUZ 980 TFAP2B 1,314 ENSG00000266919
313 HPS6 647 LBR 981 SLC6A4 1,315 TNFRSF11B
314 MIR143 648 CXCL2 982 IFT88 1,316 HOXA11-AS
315 ICOSLG 649 PLAUR 983 NPHS1 1,317 RASGRP1
316 CLCA4 650 ASAH1 984 MAP2K7 1,318 SPRY4-IT1
317 HLA-DPA1 651 BPI 985 PSMD2 1,319 FIP1L1
318 FOXF1 652 NPM1 986 DNAH8 1,320 GSR
319 BIRC3 653 ACTA1 987 B2M 1,321 CST3
320 SMAD7 654 ANGPT1 988 INTU 1,322 LTBP4
321 MIR203A 655 GGT1 989 ENTPD1 1,323 ZFAS1
322 MIR30A 656 CAMP 990 PRSS8 1,324 F5
323 MIR324 657 SOD3 991 CCNE1 1,325 STMN1
324 MIR199B 658 F2RL3 992 LRRC56 1,326 AIRE
325 ACP5 659 TEK 993 MAP3K8 1,327 RETN
326 MIR205 660 LTF 994 SHC1 1,328 NTS
327 TP73 661 MT-CO1 995 CCDC114 1,329 KRT5
328 TLR9 662 LIPA 996 ACHE 1,330 F2RL1
329 MAPK1 663 WNT3 997 TBX20 1,331 TNFSF11
330 CLCN5 664 GDF1 998 AURKB 1,332 COL4A5
331 TMEM231 665 SLC40A1 999 ASL 1,333 PIK3CD
332 BBS4 666 GLIS2 1,000 MIR30C1 1,334 FGF9
333 RMRP 667 AREG 1,001 IL2RB 1,335 KRT20
334 TGFB3 668 CDKN2B 1,002 NCF2

FIGURE 2.

FIGURE 2

Anlotinib inhibits PFKFB3-driven glycolysis in lung myofibroblasts. (A) Venn diagram to show the overlaps between anlotinib targets and lung fibrosis-related targets. (B) Protein-protein interaction (PPI) network of common targets between anlotinib and IPF. (C) The KEGG enrichment analysis of 74 targets of common targets. (D)The GO enrichment for each section listed. The mouse lung fibroblasts were pretreated with anlotinib (1 µM) for 3 h and then exposed to TGF-β1 (10 ng/ml) for an additional 24 h, and then the cells were lysed and lactate contents in the cellular lysates (E) and culture media (F) were determined. The data are presented as fold change relative to the levels of the untreated control group (mean ± SD, n = 3). (G) Glucose uptake detected with 2-NBDG were determined. The data are presented as fold change relative to the levels of the untreated control group (mean ± SD, n = 3). (H) Extracellular acidification rate (ECAR) was assessed. (I) Glycolysis and glycolysis capacity were quantified and shown as histograms (mean ± SD, n = 3). (J) Western blot analysis of HK2、PKM2、PFKFB3、LDHA and LDHB, β-actin was used as a loading control. (K) Quantification of HK2、PKM2、PFKFB3、LDHA and LDHB protein levels relative to β-Actin is shown (mean ± SD, n = 3). I, K, *p < 0.05, **p < 0.01, ***p < 0.001 VS TGF-β1-treated group by ANOVA.

TABLE 4.

Common targets shared between the potential anlotinib targets and lung fibrosis-related targets.

Number Gene Number Gene
1 AXL 38 EPHA3
2 MERTK 39 TIE1
3 ERBB2 40 CACNA2D1
4 AURKB 41 ERBB4
5 FLT1 42 MAP3K8
6 EGFR 43 ERN1
7 KDR 44 MST1R
8 SRC 45 FGFR4
9 MET 46 EPHA7
10 RET 47 MAP2K5
11 ALK 48 RIPK2
12 ABL1 49 DDR2
13 KIT 50 ACVR1B
14 ACVR1 51 DDR1
15 FLT4 52 MAP4K2
16 FLT3 53 EPHA1
17 PDGFRA 54 EPHB6
18 FGFR1 55 ERBB3
19 TGFBR1 56 EIF2AK1
20 BTK 57 MAP3K19
21 LYN 58 AKT2
22 FGFR3 59 PRKCG
23 PDGFRB 60 AKT3
24 YES1 61 PIM1
25 FGFR2 62 PIM2
26 FGR 63 JAK1
27 CSF1R 64 DPP8
28 BLK 65 DPP9
29 PLK4 66 PDE4B
30 FYN 67 PRKCI
31 MAP2K2 68 CDK2
32 HCK 69 CDK1
33 STK10 70 CCNT1
34 ABL2 71 TERT
35 TEK 72 SLC8A1
36 SLK 73 GNRHR
37 STK4 74 AOC3

PCBP3 Posttranscriptionally Increases PFKFB3 Expression by Promoting Its Translation During Myofibroblast Activation

Interestingly, the progressive upregulation of PFKFB3 during myofibroblast activation induced by TGF-β1 that was observed at the protein level was not confirmed at the mRNA level, as measured by RT-PCR (Supplementary Figures S3A, B). These results indicate that TGF-β1-induced overexpression does not require de novo transcription of PFKFB3. To further verify these findings, primary MLFs were incubated with cycloheximide to block new protein synthesis, and immunoblotting was used to measure PFKFB3 levels (Figure 3A). The half-life of PFKFB3 was not significantly altered, indicating that TGF-β1 does not influence PFKFB3 protein stability. Therefore, we postulated that PFKFB3 upregulation is modulated through posttranscriptional mechanisms in this context. To verify this hypothesis, we used the online tool catRAPID to screen for potential proteins that may interact with PFKFB3 mRNA and identified that PCBP3 (Table 5) (Agostini et al., 2013; Livi et al., 2016), a member of the PCBP family, has a high probability of directly interacting with PFKFB3 mRNA (Figure 3B) (Choi et al., 2007; Kang et al., 2012; Leidgens et al., 2013; Wang J. et al., 2020). We comparatively analyzed the expression of PCBP3 after treatment with different doses of TGF-β1 by immunoblot analysis and found that PCBP3 protein expression was increased in primary MLFs after TGF-β1 treatment (Figures 3C,D), which correlated with PFKFB3 overexpression. To better define the connection between PCBP3 function and PFKFB3, we performed RNA-protein coimmunoprecipitation (RIP) studies in primary MLFs transfected with FLAG-tagged PCBP3 (FLAG-PCBP3). An antibody targeting the FLAG protein was used to immunoprecipitate FLAG-PCBP3 and any interacting molecules from the cell lysates. Reverse transcription followed by PCR was then used to identify individual PFKFB3 mRNAs isolated with FLAG-PCBP3. We found that PFKFB3 transcripts were enriched by PCBP3 coimmunoprecipitation compared to control IgG coimmunoprecipitation (Figure 3E), demonstrating that PFKFB3 mRNA is indeed a direct target of PCBP3 in MLFs. To test the possibility that PCBP3 may influence PFKFB3 translation, we performed polysome analysis in cells transfected with FLAG-PCBP3. Cytoplasmic lysates were fractionated through sucrose gradients to separate ribosomal subunits (40S and 60S), monosomes (80S) and progressively larger polysomes. RNA was extracted from each of the 12 fractions, and the levels of PFKFB3 and β-actin mRNA were quantified by quantitative RT-PCR. While PFKFB3 mRNA levels peaked in fraction 7 in control cells, the distribution of PFKFB3 mRNA shifted rightward when PCBP3 was overexpressed, peaking in fraction 9, indicating that PFKFB3 mRNA formed, on average, larger polysomes after PCBP3 overexpression (Figure 3F). The distribution of β-actin mRNA was not affected by PCBP3 overexpression. These results indicated that overexpression of PCBP3 increases the translation of PFKFB3. Overall, these results suggest that PCBP3 improves PFKFB3 expression levels by increasing its translation rather than by influencing its protein stability.

FIGURE 3.

FIGURE 3

PCBP3 posttranscriptionally increases PFKFB3 expression by promoting its translation during myofibroblast activation. (A) PFKFB3 degradation in mouse lung fibroblasts when protein synthesis was inhibited by 50 μM cycloheximide (mean ± SD, n = 3). (B) Prediction of RNA–protein interaction of PFKFB3 mRNA with PCBP3 protein using the catRAPID algorithm. Red represents interaction strength. (C) Western blot analysis of PFKFB3 and PCBP3 protein levels in mouse lung fibroblasts stimulated with TGF-β1 for the indicated concentrations. (D) Quantification of PFKFB3 and PCBP3 protein levels relative to β-actin is shown (mean ± SD, n = 3, **p < 0.01, ***p < 0.001 compared with 0 by one-way ANOVA). (E) Quantitative RT-PCR (qPCR) to show the effect of PCBP3 overexpressing on PFKFB3 RNA immunoprecipitation (RIP) in mouse lung fibroblasts. Values were plotted as mean ± SD from three independent experiments. p value was calculated by Student t test. ***p < 0.001. (F) Mouse lung fibroblasts expressing Flag-PCBP3 were fractionated into cytoplasmic extracts through sucrose gradients. The arrow indicates the direction of sedimentation. The distribution of PFKFB3 and β-actin mRNAs was quantified by RT-PCR analysis of RNA isolated from 12 gradient fractions. Statistical analyses were performed using Student t test. ***p < 0.001. (G) Mouse lung fibroblasts were transfected with Flag-PCBP3, and then transfected with PFKFB3-siRNAs or NC-siRNA. The levels of Fibronectin、Collagen I、PFKFB3、α-SMA、PCBP3 and β-actin assessed by western blot. (H) Graphical representation of the relative levels of indicated proteins (mean ± SD, n = 3, *p < 0.05, **p < 0.01, ***p < 0.001 by ANOVA).

TABLE 5.

Potential proteins may interact with PFKFB3 mRNA through the catRAPID algorithm.

# Protein D RNA ID Z score? Discriminative power (%)? Interaction strength (%)? Domain? Motif? Ranking?
1 ELAV1_MOUSE_247-308 NC_000068.7:c11_1_5924-6036 −0.13 50 98 yes yes HHH
2 ELAV1_MOUSE_247-308 NC_000068.7:c11_1_26816-27008 −0.08 67 99 yes yes HHH
3 ELAV1_MOUSE_247-308 NC_000068.7:c11_1_15806-15983 −0.23 40 90 yes yes HHH
4 PCBP3_MOUSE_301-351 NC_000068.7:c11_1_41382-41564 −0.69 14 13 yes yes HHH
5 ELAV1_MOUSE_247-308 NC_000068.7:c11_1_77223-77340 −0.47 22 74 yes yes HHH
6 ELAV1_MOUSE_247-308 NC_000068.7:c11_1_10399-10532 −0.50 20 64 yes yes HHH
7 ELAV1_MOUSE_109-176 NC_000068.7:c11_1_5924-6036 −0.14 50 98 yes yes HHH
8 ELAV1_MOUSE_109-176 NC_000068.7:c11_1_45706-45815 −0.39 26 85 yes yes HHH
9 ELAV1_MOUSE_109-176 NC_000068.7:c11_1_26816-27008 −0.04 63 99 yes yes HHH
10 ELAV1_MOUSE_109-176 NC_000068.7:c11_1_15806-15983 −0.24 40 90 yes yes HHH
11 ELAV1_MOUSE_109-172 NC_000068.7:c11_1_5924-6036 −0.15 47 97 yes yes HHH
12 ELAV1_MOUSE_109-172 NC_000068.7:c11_1_45706-45815 −0.40 26 85 yes yes HHH
13 ELAV1_MOUSE_109-172 NC_000068.7:c11_1_15806-15983 −0.26 37 87 yes yes HHH
14 PCBP3_MOUSE_301-351 NC_000068.7:c11_1_77461-77636 −0.71 14 9 yes yes HHH
15 PCBP3_MOUSE_301-351 NC_000068.7:c11_1_66050-66214 −0.66 14 17 yes yes HHH

To determine the functional impact of PCBP3-mediated regulation of PFKFB3 expression in lung fibrosis, we transfected lung fibroblasts with FLAG-PCBP3. Expression of PFKFB3 was significantly increased by PCBP3 overexpression compared to that of the empty vector control. Reliable markers of the phenotypic transformation of fibroblasts into myofibroblasts, fibronectin, collagen I and α-SMA, were markedly increased in FLAG-PCBP3-treated cells at the protein level (Figures 3G,H) compared with vector-treated cells. In turn, using small interfering RNA (siRNA) to silence PFKFB3, the FLAG-PCBP3-induced overexpression of fibronectin, collagen I and α-SMA was abolished (Figures 3G,H). These findings suggest that PCBP3 protein upregulation is an early and sustained event during fibroblast activation and that the profibrogenic effects of PCBP3 are mediated by PFKFB3 expression. Taken together, these data suggest that PCBP3 posttranscriptionally increases PFKFB3 expression by promoting its translation during myofibroblast activation.

Anlotinib Represses PCBP3 Expression Levels During Myofibroblast Activation

To confirm the regulation of PCBP3 by anlotinib in vitro, we evaluated the protein expression of PCBP3 in MLFs and IMR90 cells. We found that TGF-β1 induced the expression of PCBP3 in MLFs and that anlotinib prevented PCBP3 expression by immunofluorescence analysis (Figure 4A). Western blot analysis of PCBP3 showed a similar result (Figures 4B,C) in MLFs, and these results were confirmed in the human IMR90 cell line (Figures 4D,E). Taken together, these data suggest that anlotinib can repress PCBP3 expression levels during myofibroblast activation in vitro.

FIGURE 4.

FIGURE 4

Anlotinib represses PCBP3 expression levels during myofibroblast activation. (A) Immunofluorescence for PCBP3 (green) in mouse lung fibroblasts treated with anlotinib for 3  h, followed by TGF-β1 for an additional 24 h. DAPI-stained nuclei (blue). Scale bar, 25 µm. (B) Western blots analysis of PCBP3 and β-actin in primary mouse lung fibroblasts. (C) Quantification for the indicated proteins (mean ± SD, n = 3). (D) The Western blots analysis of PCBP3 and β-actin in IMR90 cells treated with anlotinib for 3  h, followed by TGF-β1 for an additional 24 h. (E) Quantification for the indicated proteins (mean ± SD, n = 3). *p < 0.05, **p < 0.01, ***p < 0.001 VS TGF-β1-treated group by ANOVA.

Anlotinib Attenuates Bleomycin-Induced Pulmonary Fibrosis

To investigate the biological effects of anlotinib on pulmonary fibrosis in vivo, we established a bleomycin (BLM)-induced mouse model of pulmonary fibrosis. The mice were intraperitoneally injected with 1 mg/kg anlotinib daily after BLM administration (Figure 5A). From the first week after bleomycin instillation, the bleomycin-treated mice showed a certain reduction in activity, accompanied by slight shortness of breath. 21 days after bleomycin administration, bleomycin-treated mice showed obvious hyperventilation, accompanied by reduced activity and weight loss, but no similar symptoms were observed in the control group. A single dose of BLM (5 mg/kg) administered by intratracheal instillation successfully induced pulmonary fibrosis in C57BL/6 mice, as evidenced by a decline in pulmonary function, decreased tidal volume (TV, Figure 5B) and dynamic compliance (Cdyn, Figure 5C), and increased lung resistance (RI, Figure 5D). However, treatment with anlotinib significantly reversed bleomycin-induced pulmonary dysfunction. Moreover, we evaluated collagen deposition in the lung tissues by analyzing the hydroxyproline (HYP) content and found that anlotinib treatment reduced the amount of collagen in the lungs of bleomycin-treated mice (Figure 5E). Hematoxylin and eosin (H&E) staining indicated that anlotinib-treated mice had decreased lung inflammation and reduced lung architectural damage (Figure 5F). Accordingly, Masson’s trichrome staining showed decreased collagen deposition in anlotinib-treated mice compared with vehicle-treated mice (Figure 5F). Furthermore, attenuated fibrosis was supported by decreased protein levels of fibronectin and α-SMA by immunohistochemical (IHC) staining (Figure 5G). We also found that anlotinib treatment reduced fibronectin, collagen I and α-SMA expression by western blotting (Figures 5H,I). Taken together, these data show that anlotinib attenuates bleomycin-induced pulmonary fibrosis in vivo.

FIGURE 5.

FIGURE 5

Anlotinib attenuates bleomycin-induced pulmonary fibrosis. (A) Intervention dosing regimen of anlotinib in experimental mouse model of fibrosis. C57BL/6 mice were intraperitonealy injuected with 1 mg/kg of anlotinib or vehicle (n = 5-6 per group) daily after bleomycin instillation. Lungs were harvested at 21 days for the following analyses. Analysis of tidal volume (TV) (B), dynamic compliance (Cdyn) (C), and lung resistance (RI) (D) (mean ± SD, n = 5). (E) Hydroxyproline (HYP) contents in lung tissues from mice (mean ± SD, n = 5). Representative images show haematoxylin and eosin (H&E), Masson’s trichrome (F), α-SMA and Fibronectin staining (G) of lung sections from the indicated groups of mice. Scale bars, 100 µm. (H) Western blot analysis of Fibronectin、Collagen I and α-SMA, β-actin was used as a loading control. (I) Quantification of Fibronectin、Collagen I and α-SMA protein levels relative to β-actin is shown (mean ± SD, n = 6). *p < 0.05, **p < 0.01, ***p < 0.001 VS BLM-treated group by one-way ANOVA.

Anlotinib Decreases PCBP3 Expression and Inhibits PFKFB3-Driven Glycolysis in Fibrotic Rodent Lungs

We next examined whether the levels of PCBP3 were regulated by anlotinib in vivo. We evaluated the expression of PCBP3 in lung tissues and found that the protein levels of PCBP3 were markedly increased after bleomycin instillation, while anlotinib treatment decreased PCBP3 expression (Figures 6A,B). Accordingly, IHC staining showed decreased PCBP3 protein levels in anlotinib-treated mice compared with vehicle-treated mice (Figure 6C). In addition, to confirm the regulation of PFKFB3-driven glycolysis by anlotinib in vivo, we measured the levels of lactate and the expression of PFKFB3 in the lungs of mice. We found that there were significantly higher levels of lactate in the lungs of bleomycin-treated mice than in the lungs of control mice, and anlotinib decreased lactate levels (Figure 6D). Western blot and IHC staining studies revealed that bleomycin-induced PFKFB3 expression in the lungs of mice was prevented by anlotinib (Figures 6E–G). Overall, these results suggest that anlotinib decreases PCBP3 expression and inhibits PFKFB3-driven glycolysis in fibrotic rodent lungs.

FIGURE 6.

FIGURE 6

Anlotinib decreases PCBP3 expression and inhibit the PFKFB3-driven glycolysis in fibrotic rodent lungs. (A) Intervention dosing regimen of anlotinib in experimental mouse model of fibrosis. C57BL/6 mice were intraperitonealy injuected with 1 mg/kg of anlotinib or vehicle (n = 5-6 per group) daily after bleomycin instillation. Lungs were harvested at 21 days. Western blot analysis of PCBP3, β-actin was used as a loading control (n = 6). (B) Quantification of PCBP3 protein levels relative to β-actin is shown (mean ± SD, n = 6). (C) Representative images show PCBP3 staining of lung sections from the indicated groups of mice. Scale bars, 100 µm. (D) lactate contents in lung tissues from mice (mean ± SD, n = 5). (E) Western blot analysis of PFKFB3, β-actin was used as a loading control (n = 6). (F) Quantification of PFKFB3 protein levels relative to β-actin is shown (mean ± SD, n = 6). (G) Representative images show PFKFB3 staining of lung sections from the indicated groups of mice. Scale bars, 100 µm. **p < 0.01, ***p < 0.001 VS BLM-treated group by one-way ANOVA.

Anlotinib Accelerates the Resolution of Bleomycin-Induced Lung Fibrosis

We demonstrated that anlotinib treatment could attenuate bleomycin-induced pulmonary fibrosis. In that in vivo experiment, anlotinib was administered at approximately the same time as bleomycin instillation. We further examined whether anlotinib could postpone the progression of established fibrosis. Therefore, we performed another in vivo experiment in which anlotinib was intraperitoneally injected 7 days after bleomycin instillation (Figure 7A). As interventions beginning 7 days post bleomycin were classified as therapeutic (Izbicki et al., 2002; Moeller et al., 2008), we initially treated mice with anlotinib (1 mg/kg/day or 2 mg/kg/day) beginning on day 7 after bleomycin instillation. Pulmonary function tests showed that anlotinib treatment reversed the bleomycin-induced decline in pulmonary function, with increases in TV (Figure 7B) and Cdyn (Figure 7C) and a decrease in RI (Figure 7D). HYP measurements showed that the collagen content was significantly decreased in anlotinib-treated mice compared with vehicle-treated mice (Figure 7E). H&E staining and Masson’s trichrome staining of lungs collected at day 21 showed enhanced recovery from fibrosis upon anlotinib treatment (Figure 7F). Correspondingly, IHC staining showed that anlotinib treatment reduced fibronectin and α-SMA expression in the lungs (Figure 7G). Western blot analysis also showed that anlotinib decreased the protein levels of fibronectin, collagen I and α-SMA in the lungs (Figures 7H,I). Collectively, these data clearly demonstrate that anlotinib accelerates fibrosis resolution in vivo even after the establishment of fibrosis.

FIGURE 7.

FIGURE 7

Anlotinib accelerates resolution of bleomycin-induced pulmonary fibrosis. (A) Intervention dosing regimen of anlotinib in established pulmonary fibrosis. Bleomycin instillation was used to induce fibrosis and no treatment was given during the first 7 d. Then, mice were intraperitonealy injuected with 1 or 2 mg/kg of anlotinib or vehicle (n = 5-6 per group) daily. Lungs were harvested at 21 days. Tidal volume (TV) (B), dynamic compliance (Cdyn) (C), and lung resistance (RI) (D) of mice were measured (mean ± SD, n = 5). (E) HYP contents in lung tissues from mice (mean ± SD, n = 5). Representative images show H&E, Masson’s trichrome (F), α-SMA and Fibronectin staining (G) of lung sections from the indicated groups of mice. Scale bars, 100 µm. (H) Western blot analysis of Fibronectin、Collagen I and α-SMA, β-actin was used as a loading control (n = 4). (I) Quantification of Fibronectin、Collagen I and α-SMA protein levels relative to β-actin is shown (mean ± SD, n = 5). *p < 0.05, **p < 0.01, ***p < 0.001 VS BLM-treated group by one-way ANOVA.

Discussion

Despite recent advances in our understanding of IPF pathology, there is still no curative treatment for this disease; indeed, the currently available antifibrotic treatment modalities slow but do not completely stop the progression of the disease (Spagnolo and Maher, 2017). In this study, we demonstrate that anlotinib strongly inhibits fibroblast-to-myofibroblast transdifferentiation and reduces extracellular matrix production in primary MLFs and in the human IMR90 cell line. Accordingly, preventative and therapeutic administration of anlotinib to bleomycin-administered mice resulted in accelerated resolution of fibrosis. No adverse, systemic side effects were observed. Here, we demonstrate a novel mechanism by which anlotinib exerts antifibrotic effects by downregulating PCBP3, reducing PFKFB3 translation and inhibiting glycolysis in myofibroblasts (Figure 8).

FIGURE 8.

FIGURE 8

Schematic representation of PCBP3-PFKFB3-dependent glycolysis and the inhibitory effect of anlotinib on this pathway. Lung injury induces PCBP3 expression, which results in an increase in PFKFB3 expression by promoting its translation, resulting in the augmentation of glycolysis in lung fibroblasts. Glycolytic reprogramming participates in myofibroblast activation and furthers lung fibrosis. The tyrosine kinase inhibitor anlotinib inhibits PFKFB3-driven glycolysis by decreasing the expression of PCBP3, thereby suppressing myofibroblast activation and inhibiting the exacerbation of lung fibrosis.

A previous study revealed that male and female C57BL/6 mice did not differ in terms of their lung fibrotic responses, including cellular infiltration, collagen deposition, and quantifiable morphological changes in the lung architecture, but that the bleomycin-induced decrease in static compliance was significantly greater in males than in females (Voltz et al., 2008). This adverse effect on lung function was found to be due to male sex hormones. So sex differences should be carefully considered when interpreting experimental models of pulmonary fibrosis in mice (Blaauboer et al., 2014). In our study, we used only female mice to avoid the sex differences. We found that anlotinib exerted the preventative effects on bleomycin model of pulmonary fibrosis. Furthermore, anlotinib can also accelerate fibrosis resolution after the establishment of fibrosis.

A recent report showed that anlotinib inhibits the profibrotic effect of TGF-β1 in lung fibroblasts by attenuating inflammation and oxidative stress (Ruan et al., 2020). Our data are in line with that report and reveal an additional mechanism by which anlotinib acts on lung fibroblasts to attenuate fibrosis. There is emerging evidence about the association between metabolic disorders and IPF (Yin et al., 2019; Cho et al., 2020; Hu et al., 2020). Similar to highly proliferative cancer cells, myofibroblasts are highly dependent on glycolysis in vitro (Bueno et al., 2020). Furthermore, glycolysis is necessary not only for fibroblast growth and migration but also for the acquisition and maintenance of a myofibroblastic phenotype (Xu et al., 2017; Para et al., 2019). A previous study revealed that inhibition of glycolysis by the PFKFB3 inhibitor 3PO or by genomic disruption of the PFKFB3 gene blunted the differentiation of lung fibroblasts into myofibroblasts and attenuated profibrotic phenotypes in myofibroblasts (Xie et al., 2015). Another study revealed that lung fibroblasts displayed augmented aerobic glycolysis through activation of the PI3K-Akt-mTOR/PFKFB3 pathway in LPS-induced pulmonary fibrosis (Hu et al., 2020). Our data, along with previous studies, demonstrated that glycolytic reprogramming was critical to lung myofibroblast activation and pulmonary fibrosis. Furthermore, we demonstrated that anlotinib could strongly inhibit glycolytic reprogramming in vitro and in vivo.

The results presented herein provide new insights into the molecular mechanisms of lung fibrogenesis. This work unveils a previously unrecognized posttranscriptional regulation in activated lung fibroblasts composed of the RNA binding protein PCBP3 and the critical glycolytic enzyme PFKFB3, which maintains fibroblasts with higher glycolytic activity in fibrotic lungs compared to normal lung fibroblasts in healthy lungs. PCBP family members perform multiple functions by binding to the poly(C) sequence in both DNA and RNA to modulate mRNA stabilization, translation silencing, or translation enhancement (Blyn et al., 1997; Andino et al., 1999; Ostareck et al., 2001). Our present findings showing that PCBP3 plays an important role in myofibroblast activation and fibrogenesis and significantly extends our previous understanding by identifying an additional node of interaction between PCBP3-mediated posttranscriptional dysregulation and lung disease. We found that PFKFB3 protein overexpression was not accompanied by PFKFB3 mRNA upregulation, indicating that this increase was not transcriptionally derived. Instead, we observed that high PFKFB3 protein levels were maintained during fibroblast transdifferentiation, owing to PCBP3-mediated translational activation. Thus, the PCBP3 protein is upregulated during myofibroblast activation and binds directly to PFKFB3 during transcription. This binding activates PFKFB3 mRNA translation and generates high levels of the glycolysis activator PFKFB3. This mechanism does not exclude additional pathways of regulating PFKFB3 expression. Hence, it is not unusual for key proteins to be regulated at multiple levels, including through transcription, translation, and posttranslational modifications.

Our study is the first to report that anlotinib inhibits PFKFB3-mediated glycolysis in myofibroblasts. Moreover, anlotinib attenuates glycolysis in myofibroblasts by repressing PCBP3 expression levels rather than directly regulating the expression of PFKFB3, as anlotinib treatment does not decrease the mRNA levels of PFKFB3. Our work contributes novel mechanistic insight into the action of anlotinib. However, one of the limitations of this study is that we didn’t knock out PCBP3/PFKFB3 in mice to verify their effects in lung fibrosis, which may be explored in the further research. This future direction may be important to better understand how PCBP3 regulates PFKFB3-mediated glycolysis in pulmonary fibrosis. The other one is that this study only used bleomycin mice model for the research. Although the bleomycin model is the most widely used and best-characterized mouse model, the fibrosis of the bleomycin model is self-resolving, which contrasts with the progressive chronic fibrosis typical of human IPF (Liu et al., 2017). Therefore, whether anlotinib could attenuate fibrosis in human IPF still requires ex vivo models of pulmonary fibrosis.

In conclusion, our study demonstrated a clear antifibrotic role for anlotinib in the lungs. Its antifibrotic activity is mediated by its ability to decrease PCBP3 expression and attenuate PFKFB3-driven glycolysis, thereby inhibiting myofibroblast activation. Anlotinib might be considered as a potential therapeutic option for IPF patients.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material, further inquiries can be directed to the corresponding authors.

Ethics Statement

The animal study was reviewed and approved by Southern Medical University Institutional Animal Care and Use Committee.

Author Contributions

WC, JZ, and WZ performed study concept and design; YL, YL, and ZZ performed development of methodology and writing; HH and XW provided acquisition, analysis and interpretation of data, and statistical analysis; XM and FZ provided technical and material support; SC and HD performed review and revision of the paper.

Funding

This work was supported by National Natural Science Foundation of China (No. 81870058, 81600648, 81970032, and 81870058), National Natural Science Foundation of Guangdong Province (NO. 2017A030313849).

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.

Publisher’s Note

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.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphar.2021.744826/full#supplementary-material

Abbreviations

IPF, idiopathic pulmonary fibrosis; BLM, bleomycin; RBP, RNA binding protein; α-SMA, α-smooth muscle actin; ECM, extracellular matrix; PFKFB3, 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase-3; F2, 6BP, fructose-2, 6-bisphosphate; PFK1, phosphofructokinase-1; TKI, tyrosine kinase inhibitor; VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; TGF-β1, transforming growth factor-beta 1; TV, tidal volume; Cdyn, dynamic compliance; RI, resistance; HYP, hydroxyproline; IHC, immunohistochemical; H&E, Hematoxylin and eosin; MLF, mouse lung fibroblasts; PPI, protein-protein interaction; MAPK, mitogen-activated protein kinase; ECAR, extracellular acidification rate; siRNA, small interfering RNA; DMSO, dimethylsulfoxide; PBS, phosphate buffered saline; DMEM, Dulbecco’s Modified Eagle Medium; FBS, fetal bovine serum; DAPI, 4′,6-diamidino-2-phenylindole.

References

  1. Agostini F., Zanzoni A., Klus P., Marchese D., Cirillo D., Tartaglia G. G. (2013). catRAPID Omics: a Web Server for Large-Scale Prediction of Protein-RNA Interactions. Bioinformatics 29 (22), 2928–2930. 10.1093/bioinformatics/btt495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Andino R., Böddeker N., Silvera D., Gamarnik A. V. (1999). Intracellular Determinants of Picornavirus Replication. Trends Microbiol. 7 (2), 76–82. 10.1016/s0966-842x(98)01446-2 [DOI] [PubMed] [Google Scholar]
  3. Atsumi T., Chesney J., Metz C., Leng L., Donnelly S., Makita Z., et al. (2002). High Expression of Inducible 6-Phosphofructo-2-Kinase/fructose-2,6-Bisphosphatase (iPFK-2; PFKFB3) in Human Cancers. Cancer Res. 62 (20), 5881–5887. 10.2337/diabetes.54.12.3349 [DOI] [PubMed] [Google Scholar]
  4. Blaauboer M. E., Boeijen F. R., Emson C. L., Turner S. M., Zandieh-Doulabi B., Hanemaaijer R., et al. (2014). Extracellular Matrix Proteins: a Positive Feedback Loop in Lung Fibrosis? Matrix Biol. 34, 170–178. 10.1016/j.matbio.2013.11.002 [DOI] [PubMed] [Google Scholar]
  5. Blyn L. B., Towner J. S., Semler B. L., Ehrenfeld E. (1997). Requirement of poly(rC) Binding Protein 2 for Translation of Poliovirus RNA. J. Virol. 71 (8), 6243–6246. 10.1128/jvi.71.8.6243-6246.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bueno M., Calyeca J., Rojas M., Mora A. L. (2020). Mitochondria Dysfunction and Metabolic Reprogramming as Drivers of Idiopathic Pulmonary Fibrosis. Redox Biol. 33, 101509. 10.1016/j.redox.2020.101509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cao Y., Zhang X., Wang L., Yang Q., Ma Q., Xu J., et al. (2019). PFKFB3-mediated Endothelial Glycolysis Promotes Pulmonary Hypertension. Proc. Natl. Acad. Sci. U S A. 116 (27), 13394–13403. 10.1073/pnas.1821401116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Chen Y., Xu T., Li M., Li C., Ma Y., Chen G., et al. (2021). Inhibition of SENP2-Mediated Akt deSUMOylation Promotes Cardiac Regeneration via Activating Akt Pathway. Clin. Sci. (Lond) 135 (6), 811–828. 10.1042/cs20201408 [DOI] [PubMed] [Google Scholar]
  9. Chen Z., Liu M., Li L., Chen L. (2018). Involvement of the Warburg Effect in Non-tumor Diseases Processes. J. Cel Physiol. 233 (4), 2839–2849. 10.1002/jcp.25998 [DOI] [PubMed] [Google Scholar]
  10. Cho S. J., Moon J. S., Nikahira K., Yun H. S., Harris R., Hong K. S., et al. (2020). GLUT1-dependent Glycolysis Regulates Exacerbation of Fibrosis via AIM2 Inflammasome Activation. Thorax 75 (3), 227–236. 10.1136/thoraxjnl-2019-213571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Choi H. S., Kim C. S., Hwang C. K., Song K. Y., Law P. Y., Wei L. N., et al. (2007). Novel Function of the Poly(C)-binding Protein Alpha CP3 as a Transcriptional Repressor of the Mu Opioid Receptor Gene. FASEB J. 21 (14), 3963–3973. 10.1096/fj.07-8561com [DOI] [PubMed] [Google Scholar]
  12. De Bock K., Georgiadou M., Schoors S., Kuchnio A., Wong B. W., Cantelmo A. R., et al. (2013). Role of PFKFB3-Driven Glycolysis in Vessel Sprouting. Cell 154 (3), 651–663. 10.1016/j.cell.2013.06.037 [DOI] [PubMed] [Google Scholar]
  13. DeBerardinis R. J., Thompson C. B. (2012). Cellular Metabolism and Disease: what Do Metabolic Outliers Teach Us? Cell 148 (6), 1132–1144. 10.1016/j.cell.2012.02.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Gfeller D., Grosdidier A., Wirth M., Daina A., Michielin O., Zoete V. (2014). SwissTargetPrediction: a Web Server for Target Prediction of Bioactive Small Molecules. Nucleic Acids Res. 42, W32–W38. 10.1093/nar/gku293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Hinz B. (2012). Mechanical Aspects of Lung Fibrosis: a Spotlight on the Myofibroblast. Proc. Am. Thorac. Soc. 9 (3), 137–147. 10.1513/pats.201202-017AW [DOI] [PubMed] [Google Scholar]
  16. Hinz B., Phan S. H., Thannickal V. J., Prunotto M., Desmoulière A., Varga J., et al. (2012). Recent Developments in Myofibroblast Biology: Paradigms for Connective Tissue Remodeling. Am. J. Pathol. 180 (4), 1340–1355. 10.1016/j.ajpath.2012.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Ho R. C., Alcazar O., Fujii N., Hirshman M. F., Goodyear L. J. (2004). p38gamma MAPK Regulation of Glucose Transporter Expression and Glucose Uptake in L6 Myotubes and Mouse Skeletal Muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 286 (2), R342–R349. 10.1152/ajpregu.00563.2003 [DOI] [PubMed] [Google Scholar]
  18. Hu X., Xu Q., Wan H., Hu Y., Xing S., Yang H., et al. (2020). PI3K-Akt-mTOR/PFKFB3 Pathway Mediated Lung Fibroblast Aerobic Glycolysis and Collagen Synthesis in Lipopolysaccharide-Induced Pulmonary Fibrosis. Lab. Invest. 100 (6), 801–811. 10.1038/s41374-020-0404-9 [DOI] [PubMed] [Google Scholar]
  19. Huang Y., Wang Y., Wang X., Lin L., Wang P., Sun J., et al. (2020). The Effects of the Transforming Growth Factor-Β1 (TGF-Β1) Signaling Pathway on Cell Proliferation and Cell Migration Are Mediated by Ubiquitin Specific Protease 4 (USP4) in Hypertrophic Scar Tissue and Primary Fibroblast Cultures. Med. Sci. Monit. 26, e920736. 10.12659/msm.920736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Hutchinson J., Fogarty A., Hubbard R., McKeever T. (2015). Global Incidence and Mortality of Idiopathic Pulmonary Fibrosis: a Systematic Review. Eur. Respir. J. 46 (3), 795–806. 10.1183/09031936.00185114 [DOI] [PubMed] [Google Scholar]
  21. Izbicki G., Segel M. J., Christensen T. G., Conner M. W., Breuer R. (2002). Time Course of Bleomycin-Induced Lung Fibrosis. Int. J. Exp. Pathol. 83 (3), 111–119. 10.1046/j.1365-2613.2002.00220.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Jarman E. R., Khambata V. S., Cope C., Jones P., Roger J., Ye L. Y., et al. (2014). An Inhibitor of NADPH Oxidase-4 Attenuates Established Pulmonary Fibrosis in a Rodent Disease Model. Am. J. Respir. Cel. Mol. Biol. 50 (1), 158–169. 10.1165/rcmb.2013-0174OC [DOI] [PubMed] [Google Scholar]
  23. Kang D. H., Song K. Y., Choi H. S., Law P. Y., Wei L. N., Loh H. H. (2012). Novel Dual-Binding Function of a Poly (C)-binding Protein 3, Transcriptional Factor Which Binds the Double-Strand and Single-Stranded DNA Sequence. Gene 501 (1), 33–38. 10.1016/j.gene.2012.04.001 [DOI] [PubMed] [Google Scholar]
  24. Kenyon N. J., Ward R. W., McGrew G., Last J. A. (2003). TGF-beta1 Causes Airway Fibrosis and Increased Collagen I and III mRNA in Mice. Thorax 58 (9), 772–777. 10.1136/thorax.58.9.772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Kim Y., Noren Hooten N., Dluzen D. F., Martindale J. L., Gorospe M., Evans M. K. (2015). Posttranscriptional Regulation of the Inflammatory Marker C-Reactive Protein by the RNA-Binding Protein HuR and MicroRNA 637. Mol. Cel. Biol. 35 (24), 4212–4221. 10.1128/mcb.00645-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Leidgens S., Bullough K. Z., Shi H., Li F., Shakoury-Elizeh M., Yabe T., et al. (2013). Each Member of the poly-R(C)-binding Protein 1 (PCBP) Family Exhibits Iron Chaperone Activity toward Ferritin. J. Biol. Chem. 288 (24), 17791–17802. 10.1074/jbc.M113.460253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Liang L., Hui K., Hu C., Wen Y., Yang S., Zhu P., et al. (2019). Autophagy Inhibition Potentiates the Anti-angiogenic Property of Multikinase Inhibitor Anlotinib through JAK2/STAT3/VEGFA Signaling in Non-small Cell Lung Cancer Cells. J. Exp. Clin. Cancer Res. 38 (1), 71. 10.1186/s13046-019-1093-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Liu G., Philp A. M., Corte T., Travis M. A., Schilter H., Hansbro N. G., et al. (2021). Therapeutic Targets in Lung Tissue Remodelling and Fibrosis. Pharmacol. Ther. 225, 107839. 10.1016/j.pharmthera.2021.107839 [DOI] [PubMed] [Google Scholar]
  29. Liu T., De Los Santos F. G., Phan S. H. (2017). The Bleomycin Model of Pulmonary Fibrosis. Methods Mol. Biol. 1627, 27–42. 10.1007/978-1-4939-7113-8_2 [DOI] [PubMed] [Google Scholar]
  30. Livi C. M., Klus P., Delli Ponti R., Tartaglia G. G. (2016). catRAPID Signature: Identification of Ribonucleoproteins and RNA-Binding Regions. Bioinformatics 32 (5), 773–775. 10.1093/bioinformatics/btv629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Meng Y., Yu C. H., Li W., Li T., Luo W., Huang S., et al. (2014). Angiotensin-converting Enzyme 2/angiotensin-(1-7)/Mas axis Protects against Lung Fibrosis by Inhibiting the MAPK/NF-κB Pathway. Am. J. Respir. Cel. Mol. Biol. 50 (4), 723–736. 10.1165/rcmb.2012-0451OC [DOI] [PubMed] [Google Scholar]
  32. Moeller A., Ask K., Warburton D., Gauldie J., Kolb M. (2008). The Bleomycin Animal Model: a Useful Tool to Investigate Treatment Options for Idiopathic Pulmonary Fibrosis? Int. J. Biochem. Cel. Biol. 40 (3), 362–382. 10.1016/j.biocel.2007.08.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Nigdelioglu R., Hamanaka R. B., Meliton A. Y., O'Leary E., Witt L. J., Cho T., et al. (2016). Transforming Growth Factor (TGF)-β Promotes De Novo Serine Synthesis for Collagen Production. J. Biol. Chem. 291 (53), 27239–27251. 10.1074/jbc.M116.756247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Ostareck D. H., Ostareck-Lederer A., Shatsky I. N., Hentze M. W. (2001). Lipoxygenase mRNA Silencing in Erythroid Differentiation: The 3'UTR Regulatory Complex Controls 60S Ribosomal Subunit Joining. Cell 104 (2), 281–290. 10.1016/s0092-8674(01)00212-4 [DOI] [PubMed] [Google Scholar]
  35. Papa S., Choy P. M., Bubici C. (2019). The ERK and JNK Pathways in the Regulation of Metabolic Reprogramming. Oncogene 38 (13), 2223–2240. 10.1038/s41388-018-0582-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Para R., Romero F., George G., Summer R. (2019). Metabolic Reprogramming as a Driver of Fibroblast Activation in PulmonaryFibrosis. Am. J. Med. Sci. 357 (5), 394–398. 10.1016/j.amjms.2019.02.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Plantier L., Cazes A., Dinh-Xuan A. T., Bancal C., Marchand-Adam S., Crestani B. (2018). Physiology of the Lung in Idiopathic Pulmonary Fibrosis. Eur. Respir. Rev. 27 (147), 170062. 10.1183/16000617.0062-2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Ruan H., Lv Z., Liu S., Zhang L., Huang K., Gao S., et al. (2020). Anlotinib Attenuated Bleomycin-Induced Pulmonary Fibrosis via the TGF-Β1 Signalling Pathway. J. Pharm. Pharmacol. 72 (1), 44–55. 10.1111/jphp.13183 [DOI] [PubMed] [Google Scholar]
  39. Safran M., Solomon I., Shmueli O., Lapidot M., Shen-Orr S., Adato A., et al. (2002). GeneCards 2002: towards a Complete, Object-Oriented, Human Gene Compendium. Bioinformatics 18 (11), 1542–1543. 10.1093/bioinformatics/18.11.1542 [DOI] [PubMed] [Google Scholar]
  40. Sapudom J., Rubner S., Martin S., Thoenes S., Anderegg U., Pompe T. (2015). The Interplay of Fibronectin Functionalization and TGF-Β1 Presence on Fibroblast Proliferation, Differentiation and Migration in 3D Matrices. Biomater. Sci. 3 (9), 1291–1301. 10.1039/c5bm00140d [DOI] [PubMed] [Google Scholar]
  41. Sato S., Shinohara S., Hayashi S., Morizumi S., Abe S., Okazaki H., et al. (2017). Anti-fibrotic Efficacy of Nintedanib in Pulmonary Fibrosis via the Inhibition of Fibrocyte Activity. Respir. Res. 18 (1), 172. 10.1186/s12931-017-0654-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Spagnolo P., Maher T. M. (2017). Clinical Trial Research in Focus: Why Do So many Clinical Trials Fail in IPF? Lancet Respir. Med. 5 (5), 372–374. 10.1016/s2213-2600(17)30122-4 [DOI] [PubMed] [Google Scholar]
  43. Sun Y., Niu W., Du F., Du C., Li S., Wang J., et al. (2016). Safety, Pharmacokinetics, and Antitumor Properties of Anlotinib, an Oral Multi-Target Tyrosine Kinase Inhibitor, in Patients with Advanced Refractory Solid Tumors. J. Hematol. Oncol. 9 (1), 105. 10.1186/s13045-016-0332-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Szklarczyk D., Gable A. L., Lyon D., Junge A., Wyder S., Huerta-Cepas J., et al. (2019). STRING V11: Protein-Protein Association Networks with Increased Coverage, Supporting Functional Discovery in Genome-wide Experimental Datasets. Nucleic Acids Res. 47, D607–D613. 10.1093/nar/gky1131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Taniguchi H., Ebina M., Kondoh Y., Ogura T., Azuma A., Suga M., et al. (2010). Pirfenidone in Idiopathic Pulmonary Fibrosis. Eur. Respir. J. 35 (4), 821–829. 10.1183/09031936.00005209 [DOI] [PubMed] [Google Scholar]
  46. Taurin S., Yang C. H., Reyes M., Cho S., Coombs D. M., Jarboe E. A., et al. (2018). Endometrial Cancers Harboring Mutated Fibroblast Growth Factor Receptor 2 Protein Are Successfully Treated with a New Small Tyrosine Kinase Inhibitor in an Orthotopic Mouse Model. Int. J. Gynecol. Cancer 28 (1), 152–160. 10.1097/igc.0000000000001129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Van Schaftingen E., Lederer B., Bartrons R., Hers H. G. (1982). A Kinetic Study of Pyrophosphate: Fructose-6-Phosphate Phosphotransferase from Potato Tubers. Application to a Microassay of Fructose 2, 6-bisphosphate. Eur. J. Biochem. 129 (1), 191–195. 10.1111/j.1432-1033.1982.tb07039.x [DOI] [PubMed] [Google Scholar]
  48. Voltz J. W., Card J. W., Carey M. A., Degraff L. M., Ferguson C. D., Flake G. P., et al. (2008). Male Sex Hormones Exacerbate Lung Function Impairment after Bleomycin-Induced Pulmonary Fibrosis. Am. J. Respir. Cel. Mol. Biol. 39 (1), 45–52. 10.1165/rcmb.2007-0340OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Wang F., Qi X. M., Wertz R., Mortensen M., Hagen C., Evans J., et al. (2020a). p38γ MAPK Is Essential for Aerobic Glycolysis and Pancreatic Tumorigenesis. Cancer Res. 80 (16), 3251–3264. 10.1158/0008-5472.CAN-19-3281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Wang J., Guo X., Wang D., Yang S. (2020b). Expression Patterns of Pcbp Gene Family Members during Zebrafish Embryogenesis. Gene Expr. Patterns 35, 119097. 10.1016/j.gep.2020.119097 [DOI] [PubMed] [Google Scholar]
  51. Wynn T. A., Ramalingam T. R. (2012). Mechanisms of Fibrosis: Therapeutic Translation for Fibrotic Disease. Nat. Med. 18 (7), 1028–1040. 10.1038/nm.2807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Xie C., Wan X., Quan H., Zheng M., Fu L., Li Y., et al. (2018). Preclinical Characterization of Anlotinib, a Highly Potent and Selective Vascular Endothelial Growth Factor Receptor-2 Inhibitor. Cancer Sci. 109 (4), 1207–1219. 10.1111/cas.13536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Xie N., Tan Z., Banerjee S., Cui H., Ge J., Liu R. M., et al. (2015). Glycolytic Reprogramming in Myofibroblast Differentiation and Lung Fibrosis. Am. J. Respir. Crit. Care Med. 192 (12), 1462–1474. 10.1164/rccm.201504-0780OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Xu J., Li J., Yu Z., Rao H., Wang S., Lan H. (2017). HMGB1 Promotes HLF-1 Proliferation and ECM Production through Activating HIF1-α-Regulated Aerobic Glycolysis. Pulm. Pharmacol. Ther. 45, 136–141. 10.1016/j.pupt.2017.05.015 [DOI] [PubMed] [Google Scholar]
  55. Yin X., Choudhury M., Kang J. H., Schaefbauer K. J., Jung M. Y., Andrianifahanana M., et al. (2019). Hexokinase 2 Couples Glycolysis with the Profibrotic Actions of TGF-β. Sci. Signal. 12 (612), eaax4067. 10.1126/scisignal.aax4067 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material, further inquiries can be directed to the corresponding authors.


Articles from Frontiers in Pharmacology are provided here courtesy of Frontiers Media SA

RESOURCES