Skip to main content
PLOS Neglected Tropical Diseases logoLink to PLOS Neglected Tropical Diseases
. 2023 Jul 25;17(7):e0011520. doi: 10.1371/journal.pntd.0011520

mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a

Shanling Cao 1,2, Dexian Wang 1, Yixuan Wu 1, Junmei Zhang 1,2, Lixia Pu 1, Xuenong Luo 1, Xueyong Zhang 1,3, Xiaolin Sun 2, Yadong Zheng 4,*, Shuai Wang 1,5,*, Xiaola Guo 1,*
Editor: Uriel Koziol6
PMCID: PMC10403128  PMID: 37490505

Abstract

Liver fibrosis is one of the histopathological characters during Echinococcus multilocularis infection. The activation of hepatic stellate cells (HSCs) is a key event in the development of liver fibrosis. However, the molecular mechanism of HSC activation in the E. multilocularis infection-induced liver fibrosis remains largely unclear. Here, we reported that mmu-miR-342-3p was most dominantly expressed in HSCs and was upregulated in the HSCs in response to E. multilocularis infection. We further showed that mmu-miR-342-3p was able to bind to the 3’ UTR of the Zbtb7a gene and regulated its expression. Moreover, mmu-miR-342-3p expression was negatively correlated with its target gene Zbtb7a in HSCs during E. multilocularis infection. Knockdown of mmu-miR-342-3p promoted the expression of Gfap in the activated HSCs in vitro. In the E. multilocularis-infected mice, knockdown of mmu-miR-342-3p suppressed the expression of α-Sma, Col1α1, and TGF-β but promoted the expression of Gfap. Therefore, mmu-miR-342-3p is a key regulator for activation of HSCs, and inhibiting mmu-miR-342-3p to suppressed Zbtb7a-mediated TGF-β signaling in activated HSCs could be a novel strategy to treat liver fibrosis induced by E. multilocularis.

Author summary

Liver fibrosis is one of the histopathological features during Echinococcus multilocularis infection. The activation of hepatic stellate cells (HSCs) is a key event in the development of liver fibrosis. However, the molecular mechanism of HSC activation in the liver fibrosis induced by E. multilocularis infection remains largely unknown. In recent years, there is increasing evidence indicate that the abnormal expression of miRNAs is closely related to the occurrence and development of liver fibrosis. The expression profile of miRNA in hepatic fibrosis induced by E. multilocularis infection has been widely characterized, but the roles of miRNAs in liver fibrosis are largely unexplored. Herein, we identified a set of differentially expressed miRNAs in the activated HSCs induced by E. multilocularis, mmu-miR-342-3p of which was dominantly expressed in HSCs. This study signifies the important role of mmu-miR-342-3p in the HSC activation, which will help us to better understand the mechanism of liver fibrosis induced by E. multilocularis infection.

Introduction

Alveolar echinococcosis (AE), a serious zoonotic parasitic disease, is characterized by continuous and infiltrative tumor-like invasive growth of Echinococcus multilocularis metacestodes [1]. Humans are occasionally infected by ingesting eggs from contaminated food or water. AE is mainly distributed in semi-agricultural and semi-pastoral areas in the northwest of China, which seriously threatens local people’s health [24].

Hepatic fibrosis is a dynamic pathological process characterized by excessive deposition of extracellular matrix (ECM) during the progression of parasitic liver disease [5]. The eggs released from Schistosoma mansoni and Schistosoma japonicum primarily deposits in the periportal zones, which generate a granulomatous reaction and causing substantial pathologic liver fibrosis [6]. Echinococcus parasites induce an imbalance of the immune responses within the hepatic tissue, leading to the hepatic architectural distortion and the development of fibrosis [7]. The resultant liver fibrosis may be associated with severe pathology in echinococcosis with granuloma formation, collagen accumulation and inflammation [8,9]. The severity degree of liver injury and fibrosis gradually aggravates with the extension of infection period [7,10]. It is well known that activation of hepatic stellate cell (HSC) is linked to the occurrence and development of liver fibrosis and has been well investigated in a number of liver diseases [1113]. Upon liver damage, the quiescent HSCs are activated and trans-differentiated into myofibroblast-like cells, which secrete a great amount of ECM. E. multilocularis infection can also induce the activation of HSCs, which express high levels of alpha-smooth muscle actin (α-SMA), Vimentin, and Collagen I (Col1α1) [14,15], however the mechanism behind remains unclear.

MicroRNAs (miRNAs) are a class of endogenous non-coding small RNAs with 20–24 nt in length [16]. MiRNAs usually bind to the 3’ untranslated region (UTR) of target genes and inhibit their translation or promote degradation. There is increasing evidence that miRNAs play an important role in many infectious liver diseases, such as viral, bacterial and parasitic hepatitis [17,18]. The abnormal expression of miRNAs is closely related to the occurrence and development of liver fibrosis. A large number of miRNAs have been proposed as predictors of fibrosis progression [19,20]. The miRNA expression profile of hepatic fibrosis induced by E. multilocularis infection have been widely characterized [21,22], but the roles of miRNAs in liver fibrosis are largely unexplored. Herein, we identified a set of differentially expressed miRNAs in the activated HSCs induced by E. multilocularis, mmu-miR-342-3p of which was dominantly expressed in HSCs. We investigated the role of mmu-miR-342-3p in the HSC activation, which will help us to better understand the mechanism of liver fibrosis induced by E. multilocularis infection.

Materials and methods

Ethics statement

Animal experiments in the study were evaluated and approved by Ethics Committee of Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences and performed in accordance with Good Animal Practice of Animal Ethics Procedures.

Parasite infection

Four-week-old BALB/c mice (n = 160) were randomly divided into two groups. One group (n = 80) was intraperitoneally injected with 600 E. multilocularis protoscoleces, which were obtained from the hydatid cysts in infected mouse in our lab as previously described [23]. The other (n = 80) was inoculated with 0.9% saline solution as an uninfected group. The high-fat diet induced mice were ordered from Wuhan Servicebio Technology Co., Ltd.

Histological assessment and immunohistochemistry staining

Liver tissue samples of uninfected (n = 3) and infected mice (n = 3) were fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin. The samples were sliced into 5 μm sections and performed with hematoxylin and eosin, Masson’s trichrome and Sirius Red staining. For Sirius Red staining, sections were incubated in Picro-Sirus Red solution (Abcam) for 60 min, followed by two quick washes in acetic acid solution and absolute alcohol. The images were taken using a light microscope (Zeiss, Germany). Images taken from 15 random 20× fields from three different liver lobes in each animal were measured. The amount of collagen deposition was measured by the method of color thresholding segmentation using ImageJ.

Immunohistochemistry staining was performed according to the following protocol. In brief, the sections were deparaffinized in xylene and rehydrated in graded alcohols. Following antigen retrieval in 0.01 M sodium citrate buffer, the tissue slides were incubated with 3% hydrogen peroxide for 5 minutes to block endogenous peroxidase activity. The sections were blocked with 5% BSA in TBST and then incubated with α-SMA antibodies (1:200, Servicebio). After wash, the sections were incubated with the anti-rabbit secondary antibodies (1:1000, SeraCare) for 1 h at 37°C. Finally, the signal was developed utilizing DAB substrate Kit (Servicebio), and the slices were stained with hematoxylin (Servicebio). The images were taken under a light microscope (Zeiss, Germany). Images taken from 15 random 20× fields from three different liver lobes in each animal were measured. The amount of α-SMA positive cells was measured using ImageJ.

Isolation, cultivation, and transfection of HSCs

Primary HSCs were isolated from uninfected (n = 6) and infected (n = 6) mouse liver by collagenase IV perfusion, followed by density gradient centrifugation as previously described [24]. Briefly, the livers were perfused with 0.09% EGTA buffer at a flow rate of 5 mL/min at 42°C for 2 min and then with Enzyme buffer containing 0.04% collagenase IV at a flow rate of 5 mL/min at 42°C for 6 min. The resultant digested livers were excised, and in vitro digestion was performed in 80 mL Enzyme buffer containing 0.08% collagenase IV and 1% DNase at 37°C for 30 min. The cells were passed through nylon filters (70 μm) and centrifuged at 50 g at 4°C for 4 min. The supernatant was centrifuged at 600 g for 10 min, and the pellet was washed by Gey’s balanced salt solution (GBSS) at 500 g at 4°C for 5min to obtain nonparenchymal cells (NPCs). The NPCs were resuspended in 5 mL GBSS and gently coated with Optiprep solutions of different concentrations, with 8 mL 11.5% Optiprep in the upper layer and 4 mL 20% Optiprep in the lower layer, and centrifuged uninterrupted at 1,400 ×g at 4°C for 17 min. The upper cells (HSCs) were transferred to tubes and washed three times with GBSS. Cells were cultured in DMEM medium containing 10% FBS for subsequent experiments.

For further validation, we first performed flow cytometry analysis using a HSC marker (Glail fibrillary acidic protein, GFAP). Next, Vitamin A lipid droplet autofluorescence in isolated primary HSCs was detected at a wavelength of 328 nm using a fluorescence microscope (Zeiss, Germany). Thirdly, the expression of the biomarkers for HSCs (α-SMA, GFAP and Col1α1), hepatocytes (albumin, Alb; Cytokeratin 18, CK18), and macrophages (EGF-like module-containing mucin-like hormone receptor-like 1, Emr1) were determined by qRT-PCR.

Primary HSCs were cultured in DMEM medium containing 10% FBS, and transfection experiments were performed (at 5 days after isolation). HSCs (5 × 105 cells per well) were plated into 12-well plates and transfected with 50 nM mmu-miR-342-3p inhibitor or 50 nM negative control (NC) (Ambion/Invitrogen) using Lipofectamine RNAiMAX Transfection Reagent (Invitrogen). Following transfection for 10 h, the medium was replaced with fresh DMEM medium supplemented with 10% FBS. Each transfection was independently repeated three times.

Identification of differentially expressed known miRNAs

The miRNA expression in HSCs after E. multilocularis infection was investigated using high-throughput sequencing [25]. Raw reads were generated subsequently, which were deposited in NCBI Sequence Read Archive (SRA) under accession number PRJNA732233. The process of raw sequencing data was performed according to the previously reported methods with some modification. In brief, after removal of the reads with low quality and adaptor sequences, the clean reads were mapped to the full annotated genome of the mouse genome (http://www.ncbi.nlm.nih.gov/genome/genomes/52). Afterward, all the mapped reads were used for identification of miRNAs (miRBase, http://www.mirbase.org/index.shtml). Relative expression levels of miRNAs in three groups were analyzed using DESeq R package (1.8.3) and differentially expressed miRNAs between the uninfected and infected groups at different time points post infection were identified.

Raw reads were generated subsequently, which were deposited in NCBI Sequence Read Archive (SRA) under accession number PRJNA732233. Differentially expressed miRNAs between the uninfected and infected groups at different time points post infection were identified.

Quantitative RT-PCR Analysis

For examining the miRNA expression, the first-strand cDNAs were synthesized using an All-in-One miRNA First-Strand cDNA Synthesis Kit (GeneCopoia) and U6 snRNA was selected as an endogenous control. For examining the mRNA expression, the first-strand cDNAs were synthesized using a RevertAid First Strand cDNA Synthesis Kit (Invitrogen) and GAPDH was selected as an endogenous reference gene. The cDNA mixture was diluted by 5-fold with nuclease-free water. Quantitative RT-PCR was performed using All-in-One qPCR Mix (GeneCopoeia) by 7500 Real Time PCR System (Applied Biosystems) under following conditions: 95°C for 10 min, followed by 40 cycles of 95°C for 10 sec, 60°C for 1min. All the primers were purchased from GeneCopoeia (Table A in S1 Text). The relative expression levels of miRNAs or mRNA were calculated using the 2-ΔΔCt formula. Statistical analysis data were taken from three independent experiments.

Plasmid construction and luciferase assay

The 3’ UTR fragment of Zinc finger and BTB domain-containing 7A (Zbtb7a) gene with restriction enzyme site was amplified using a pair of primers: 5’-GAGCTCGGTGAATTTGCGTGT-3’ and 5’-GTCGACCCTGTGTCCCTCCTA-3’ (restriction enzyme sites were underlined). The PCR product was cloned into a PmirGLO Dual-Luciferase vector (Promega, USA) and confirmed by sequencing, designated as WT-Zbtb7a. The 3’UTR of Zbtb7a with mutations in a binding site was artificially synthesized (Sangon, China), cloned into the PmirGLO Dual-Luciferase vector and confirmed by sequencing, designated as Mut-Zbtb7a.

HEK293T cells (1×105 cells per well) were plated into 24-well plates and transfected with 1μg of WT-Zbtb7a or Mut-Zbtb7a in combination with 30 pmol of mmu-miR-342-3p mimics or NC mimics using Lipofectamine 2000 (ThermoFisher Scientific). At 24 h after transfection, Renilla and firefly luciferase activity was measured by GloMax 96 Microplate Luminometer (Promega) using Dual-Glo Luciferase Assay System (Promega). Each transfection was independently repeated three times.

Western blotting

The total proteins were isolated from cells using RIPA buffer (ThermoFisher Scientific) supplemented with protease inhibitor (Sigma). The concentration of total proteins was measured using BCA protein assay Kit (ThermoFisher Scientific). A total of 50 μg proteins were separated by SDS-PAGE and transferred onto PVDF membranes (Millipore). Subsequently, the membranes were blocked with 5% BSA in TBST and then incubated with Zbtb7a antibodies (1:500, BBI) at 4°C overnight. After wash, the membranes were incubated with anti-rabbit secondary antibodies (1:1000, Abcam) for 1 h at room temperature, followed by addition of Pierce ECL Western Blotting Substrate (ThermoFisher Scientific). Light emission was recorded using a chemiluminescent detection system (G-Box, Syngene).

In vivo study

Chitosan (CS)-miRNA nanoparticles were prepared according to the method previously described with minor modifications [26]. Briefly, 95% deacetylated CS was dissolved in acetic acid solution (2.0 mg/mL, adjust to pH 5.5). A sodium tripolyphosphate solution (TPP; 2.0 mg/mL) was used as a cross-linking agent. To prepare CS-miRNA nanoparticles, 250 μL of the TPP solutions containing 3 nmol miRNA inhibitors were added drop by drop to 2.5 mL of CS solutions and the resulting solutions were maintained at room temperature for 30 min.

Eight E. multilocularis-infected mice were randomly divided into two groups and injected with 100 μL of CS-NC nanoparticles (CSNP-NC) or CS-mmu-miR-342-3p inhibitor nanoparticles (CSNP- inhibitor) through the tail vein, respectively. After 40 h, mice were humanely killed and liver was collected for further use.

Statistical analysis

All statistical analyses were performed using GraphPad Prism 8 (La Jolla, USA), and differences were compared using a two-tailed unpaired t-test for two groups and ANOVA for three groups. A p < 0.05 was considered as statistically significant.

Results

Activation of HSCs and hepatic fibrosis during E. multilocularis infection

Histopathological analysis using hematoxylin and eosin, Masson’s trichrome, and Sirius Red staining revealed the excessive deposition of collagen surrounding the metacestodes in the liver of E. multilocularis-infected mice, being higher than that of control mice and high-fat diet induced mice (Figs 1A and S1). The results of α-SMA immunostaining showed that myofibroblastic HSCs (MF-HSCs) mainly distributed in the livers of E. multilocularis-infected mice around the metacestodes (Fig 2A). The proportion of MF-HSCs (α-SMA-positive cells) and macrophages (Emr1-positive cells) in the livers of E. multilocularis-infected mice were higher than those in un-infected group, whereas hepatocytes (CK18-positive cells) were significantly decreased (Figs 2B and S2). To assess the effect of E. multilocularis infection on the HSC activation, the primary HSCs were isolated and the expression of biomarkers for HSCs were examined. Flow cytometry analysis demonstrated that the isolated HSCs accounted for approximately 90% (Fig 2D) and exhibited a striking blue autofluorescence (Fig 2C). Furthermore, the isolated HSCs that highly expressed three biomarkers for HSCs (determined by α-SMA, GFAP and Col1α1) reduced the contamination with hepatocytes (determined by Alb and CK18) and macrophages (determined by Emr1) (S3 Fig). As expected, the expression of α-Sma, Col1α1, and Vimentin were significantly upregulated 30-, 60-, 90-day post infection compared with the uninfected group (Fig 2E). Increased expression of α-Sma at protein level was observed in HSCs from E. multilocularis- infected mice 30-, 60-, and 90-day post infection (Fig 2F). These results demonstrate that the hepatic fibrosis and HSC activation occur in response to E. multilocularis infection.

Fig 1. The effects of E. multilocularis infection on hepatic fibrosis in mouse.

Fig 1

(A) Representative pictures of Hematoxylin and Eosin (H&E), Masson’s Trichrome, and Picrosirius Red staining for liver fibrogenesis in control mice, high-fat diet induced mice, and E. multilocularis-infected mice. Arrows indicate cysts. (B) The birefringent collagen content was quantified in Masson’s Trichrome- or Picrosirius Red-stained sections by color thresholding-based segmentation using ImageJ.

Fig 2. The activation of HSCs induced by E. multilocularis infection.

Fig 2

(A) Representative pictures of immunofluorescence staining for α-SMA in the livers of E. multilocularis-infected and uninfected mice. Arrows indicate cysts. (B) Quantification of α-SMA-positive cells in the livers of E. multilocularis-infected and uninfected mice. (C) Vitamin A lipid droplet autofluorescence in isolated primary HSCs was detected at a wavelength of 328 nm using a fluorescence microscope. (D) Flow cytometry analysis of isolated mouse HSCs using Gfap. (E) The expression of α-sma, col1α1, and vimentin in the HSCs from E. multilocularis-infected mice 30-, 60-, and 90-day post-infection (dpi) by qRT-PCR. (F) The expression of α-SMA in the HSCs from E. multilocularis-infected mice 30-, 60-, and 90-day post-infection (dpi) by Western blotting. Data for final statistical analysis were taken from 3 independent experiments. *p <0.05, **p < 0.01, ***p < 0.001.

Dysregulation of mmu-miR-342-3p in HSCs during E. multilocularis infection

Considering the important roles of miRNAs in HSC activation, we recently characterized the global profiling of lncRNAs-miRNAs-mRNAs in hepatocytes (HCs), Kupffer cells (KCs) and HSCs of mouse liver during E. multilocularis infection [27]. We found that 49 and 67 miRNAs were differentially expressed in the HSCs 60- and 90-day post infection, respectively (Table B and C in S1 Text). Of them, 33 were commonly shared (Fig 3A). The KEGG pathway analysis indicated that target genes of these miRNAs were mainly involved in cancer pathway, Hippo signaling pathway, TGF-β signaling pathway, and PI3K-Akt signaling pathway (Fig 3B). Among these differentially expressed miRNAs, mmu-miR-342-3p was dominantly expressed in HSCs compared with that in HCs and KCs (p < 0.001, Fig 3C). Thus, we analyzed the dynamic expression of mmu-miR-342-3p in HSCs at different time points post infection. The qRT–PCR analysis showed that the expression of mmu-miR-342-3p was significantly upregulated in the HSCs from E. multilocularis-infected mice 30- and 60-day post infection, while its expression was decreased 90-day post infection (p < 0.001, Fig 3D).

Fig 3. Dysregulation of mmu-miR-342-3p in liver HSCs during E. multilocularis infection.

Fig 3

(A) A volcano plot of the differentially expressed miRNAs between two liver HSCs from E. multilocularis-infected mice at 60 and 90-day post infection (dpi), where the red and green, indicated significantly upregulated miRNAs (Up) and downregulated miRNAs (Down), respectively. (B) KEGG pathway enrichment analysis of target genes. (C) The expression of mmu-miR-342-3p in hepatocyte cells (HCs), Kupffer cells (KCs), and hepatic stellate cells (HSCs). (D) The expression of mmu-miR-342-3p in liver HSCs from E. multilocularis-infected mice at 30-, 60-, and 90-day post-infection (dpi). The uninfected mice at each sampling time point were used as controls. Data for final statistical analysis were taken from 3 independent experiments. ***p < 0.001.

Upregulation of mmu-miR-342-3p in the culture-activated HSCs

As primary HSCs are known to be activated during cultivation [28], the quiescence HSCs were isolated and cultured in plastic plates for 9 days. As expected, the expression of α-Sma and Col1α1 was significantly up-regulated at day 9 compared with that at day 0, while the expression of Gfap, a biomarker for HSC quiescence, was significantly downregulated (p < 0.01, Fig 4A). Consistently, the immunofluorescence staining showed that α-SMA protein was highly expressed in culture-activated HSCs (at day 9), suggesting that the primary HSCs are activated (Fig 4B). Next, we assessed whether the expression of mmu-miR-342-3p was changed in the culture-activated HSCs. We found that it was significantly upregulated at day 9) compared with that at day 0 (Fig 4C). The results suggest that the mmu-miR-342-3p may be involved in the HSC activation.

Fig 4. Upregulation of mmu-miR-342-3p in the culture-activated HSCs.

Fig 4

(A) The qRT-PCR analysis of α-Sma, Col1α1, Gfap in in vitro culture-activated HSCs. (B) Immunofluorescence staining of α-SMA in in vitro culture-activated HSCs. Scale bar = 50 μm (C) The expression of mmu-miR-342-3p in in vitro culture-activated HSCs. Data for final statistical analysis were taken from 3 independent experiments. **p < 0.01, ***p < 0.001.

Inhibition of Zbtb7a by mmu-miR-342-3p via directly binding to its 3′UTR

To determine the potential role of mmu-miR-342-3p in HSC activation during E. multilocularis infection, its potential targets were identified by using TargetScan, PicTar and miRNA.org. A total of 14 commonly shared targets were screened out, which were mainly involved in cell cycle, cell differentiation, cell proliferation, and tumorigenesis (Table D in S1 Text). Among these candidates, Zbtb7a was first selected for downstream analysis, of which contains a putative binding site for mmu-miR-342-3p (Fig 5A). To validate the binding capability, the WT-Zbtb7a-3′UTR and the Mut-Zbtb7a-3′-UTR luciferase reporter systems were constructed and co-transfected with mmu-miR-342-3p mimics or NC into HEK293T cells, respectively. Luciferase reporter assay revealed that the mmu-miR-342-3p mimic significantly decreased the luciferase activity in WT-Zbtb7a-3′UTR-transfected HEK293T cells compared with that in the control (p < 0.001, Fig 5A). However, the decrease was not observed in the Mut-Zbtb7a-3′-UTR-transfected cells (p > 0.05, Fig 5A), suggesting that mmu-miR-342-3p is able to bind to the Zbtb7a-3′-UTR. Consistently, after downregulation of mmu-miR-342-3p in the HSCs from E. multilocularis-infected mice by transfecting with mmu-miR-342-3p inhibitor (p < 0.05, Fig 5B), Zbtb7a was significantly up-regulated at both mRNA and protein levels (Fig 5C). Moreover, the expression of Zbtb7a was negatively correlated with the mmu-miR-342-3p expression in the HSCs from E. multilocularis-infected mice (p = 0.0031, Fig 5D and E). These results suggest that mmu-miR-342-3p participates in the HSC activation via targeting Zbtb7a.

Fig 5. Inhibition Zbtb7a by mmu-miR-342-3p via directly binding to its 3′UTR.

Fig 5

(A) Schematic representation of a putative binding site of mmu-miR-342-3p in the 3’UTR of Zbtb7a (up) and the dual-Glo Luciferase assay for the interaction between mmu-miR-342-3p-Mimics and Zbtb7a (down). The miRNA ‘seed’ sequences are indicated in bold in black. WT: WT- Zbtb7a or Mut- Zbtb7a construct was co-transfected into 293T cells with either mimic negative control (mimic NC) or mmu-miR-342-3p mimic (miR-342-3p mimic) and luciferase activities were measured after 24 h transfection. (B) The expression of mmu-miR-342-3p in the HSCs from E. multilocularis-infected mice transfected with inhibitor negative control (NC) or mmu-miR-342-3p inhibitor (Inhibitor) by RT-qPCR. (C) The expression of Zbtb7a at mRNA and protein levels in the HSCs from E. multilocularis-infected mice transfected with mmu-miR-342-3p inhibitor (Inhibitor). (D) The expression of zbtb7a in primary HSCs from E. multilocularis-infected mice at 30-, 60-, and 90-day post-infection (dpi). (E) Correlation between the expression of Zbtb7a and mmu-miR-342-3p. (n = 9, Spearman’s correlation analysis; r, correlation coefficient) Data for final statistical analysis were taken from 3 independent experiments. **p < 0.01; ***p < 0.001.

The in vivo effect of down-regulated mmu-miR-342-3p on HSC activation during E. multilocularis infection

In order to study the effect of mmu-miR-342-3p expression on the HSC activation during E. multilocularis infection, the primary HSCs were isolated from E. multilocularis-infected mice. We found that downregulated mmu-miR-342-3p could significantly promote the expression of Gfap, while α-Sma and Vimentin were not significantly changed (Fig 6A and 6B). In order to in vivo assess the effect of mmu-miR-342-3p on hepatic fibrosis, CS-mmu-miR-342-3p nanoparticles were injected into mice through the tail vain. As expected, mmu-miR-342-3p was found to be downregulated (p < 0.01, Fig 6C) and its target gene Zbtb7a was remarkably increased at both mRNA and protein levels in the liver of mice injected with CSNP-mmu-miR-342-3p inhibitor (p < 0.01, Fig 6D and 6E). Moreover, Col1α1, Vimentin and TGF-β were significantly downregulated, while Gfap was significantly upregulated (p < 0.05, Fig 6F). These results suggest that the parasite infection induces HSC activation via the mmu-miR-342-3p-Zbtb7a axis.

Fig 6. The effect of down-regulated mmu-miR-342-3p on HSC activation during E. multilocularis infection.

Fig 6

(A) The expression of mmu-miR-342-3p in the HSCs from E. multilocularis-infected mice after transfecting with inhibitor negative control (NC) or mmu-miR-342-3p inhibitor (inhibitor). (B) Effects of downregulated mmu-miR-342-3p on the expression of HSC activation marker (α-Sma and Vimentin) and quiescent marker Gfap in the HSCs from E. multilocularis-infected mice. (C) The mmu-miR-342-3p expression was significantly downregulated in the mouse livers by CS-mmu-miR-342-3p inhibitor nanoparticles (CSNP-Inhibitor) compared with that of CS-inhibitor negative control nanoparticles (CSNP-NC). (D) The Zbtb7a expression both at mRNA and protein levels were significant upregulated in the livers of E. multilocularis-infected mice by injecting with CS-mmu-miR-342-3p inhibitor nanoparticles. (F) Effect of mmu-miR-342-3p on the expression of α-Sma, Col1α1, Vimentin, and TGF-β in the in the livers of E. multilocularis-infected mice by injecting with CS-mmu-miR-342-3p inhibitor nanoparticles. Data for final statistical analysis were taken from 3 independent experiments. ***p < 0.001.

Discussion

Liver fibrosis is a significant histological hallmark of AE and the activation of HSCs is a central link to the initiation and progression of hepatic fibrosis [15]. HSCs are mainly activated and rapidly transform into proliferative, migratory, and extracellular matrix-producing myofibroblasts [29]. In this study, we found hepatic fibrosis occurred in response to E. multilocularis infection, characterized by the excessive deposition of collagen in the liver surrounding the metacestodes. In line with this, we also found that the higher proportion of myofibroblastic HSCs as well as higher expression of Vimentin, Col1α1, and α-Sma in liver HSCs from AE mice. Our results confirmed that the HSCs were activated in response to E. multilocularis infection, thus promoted the process of liver fibrosis.

Liver fibrogenesis is regulated by multiple growth factors and cytokines. Among those regulators, the best known prosclerotic mediator is TGF-β family [11]. MiRNAs, known as small regulatory RNAs, have recently emerged as critical regulatory molecules in chronic liver diseases [30]. Many studies have shown that the abnormal expression of miRNAs is closely associated with the HSC activation during the development of liver fibrosis [18]. In this study, we identified a large number of aberrantly expressed miRNAs in the HSCs in response to E. multilocularis infection. As expected, some of them including mmu-miR-29, mmu-miR-192 and mmu-miR-378 have been reported to be associated with the activation of HSCs [31]. To understand the regulatory role of these differentially expressed miRNAs, the target genes were predicted. Among them, some were involved in the TGF-β signaling pathway and PI3K-Akt signaling pathway. For instance, miR-29 is highly expressed in HSCs and possibly inhibits the HSC activation by targeting the PI3K/AKT signaling pathway [31]. Similarly, S. japonicum miR-29b-3p prevents parasite-induced liver fibrosis by inhibiting COL1A1 and COL3A1[32]. miR-192 is highly expressed in quiescent HSCs and can suppress HSC activation by targeting TGF-β1/Smad signaling [33].

In the study, mmu-miR-342-3p was proved to be dominantly expressed in HSCs. We also observed its increased expression in the HSCs from E. multilocularis-infected mouse liver at 30- and 60-day post infection. These results suggest mmu-miR-342-3p to be a promising candidate miRNA affecting the progression of liver fibrosis induced by E. multilocularis infection. Next, we found that mmu-miR-342-3p was significantly upregulated in vitro culture-activated HSCs, suggesting its potential role in the HSC activation. Luciferase reporter assay revealed that the mmu-miR-342-3p mimic could directly bind to Zbtb7a. Moreover, we observed upregulation of Zbtb7a in activated HSCs transfected with mmu-miR-342-3p inhibitor. Consistently, the expression of Zbtb7a was negatively correlated with the mmu-miR-342-3p expression in the HSCs from E. multilocularis-infected mice. These results indicate that mmu-miR-342-3p can repress Zbtb7a expression by directly targeting its 3’-UTR. It has been shown that hypermethylation of Zbtb7a is associated with the liver fibroblast activation and fibrosis [34]. Zbtb7a can inhibit the expression of TGF-β1 through indirectly suppressing its promoter activity of TGF-β1 [34]. As the most potent fibrogenic factor, TGF-β is the primary factor in the transformation of HSCs to myofibroblasts [33,34]. TGF-β exerts its actions through binding to serine/threonine kinase transmembrane TGF-β receptor I and II, resulting in deposition of extracellular matrix (ECM) proteins, such as Col1α1,α-SMA, and fibronectin [35]. Col1α1 is the main component of the fibrotic liver and its expression is known to be induced by TGF-β [36]. We found that down-regulated mmu-miR-342-3p decreased the expression of fibrogenic genes including Col1α1 and Vimentin and up-regulated the quiescence-associated gene Gfap. It is possible that mmu-miR-342-3p may promote the HSC activation by targeting Zbtb7a-mediated TGF-β signaling during E. multilocularis infection.

Supporting information

S1 Data. Excel spreadsheet containing, in separate sheets, the underlying numerical data and statistical analysis for Figure panels 1B, 2B, 2E, 3C, 3D, 4A, 4C, 5A, 5B, 5C, 5D, 6A, 6B, 6C, 6D, 6F and S3.

(XLSX)

S1 Fig. Full-blown liver lesions of E. multilocularis-infected mice were observed.

(TIF)

S2 Fig. Immunostaining of CLEC4F (KC marker) and Cytokeratin 18 (HC marker) in livers of un-infected mice or E. multilocularis-infected mice.

(TIF)

S3 Fig. The mRNA expression levels of biomarkers in the isolated HCs (Alb and CK18), HSCs (α-SMA, Gfap, and Col1α1), and KCs (Emr1) by RT-qPCR.

(TIFF)

S1 Text

Table A. The qRT-PCR primers used in the study. Table B. Summary of the differentially expressed miRNAs in liver HSCs in E. multilocularis-infected mice at 60-day post infection. Table C. Summary of the differentially expressed miRNAs in liver HSCs in E. multilocularis-infected mice at 90-day post infection. Table D. Putative target genes of mmu-miR-342-3p.

(DOCX)

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

The study was financially supported by grants from the National Key Research and Development Program of China (2022YFD1800200) and the National Natural Science Foundation of China (32273031) to XG, Scientific Research and Development Talent Fund of Zhejiang Agriculture and Forestry University (2021LFR038) to YZ, and State Key Laboratory for Animal Disease Control and Prevention (SKLVEB2020KFKT004) to XZ. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Casulli A, Barth TFE, Tamarozzi F. Echinococcus multilocularis. Trends Parasitol. 2019; 35:738–739. [DOI] [PubMed] [Google Scholar]
  • 2.Torgerson PR, Keller K, Magnotta M, Ragland N. The global burden of alveolar echinococcosis. PLoS Negl Trop Dis. 2010; 4:e722. doi: 10.1371/journal.pntd.0000722 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wen H, Vuitton L, Tuxun T, et al. Echinococcosis: Advances in the 21st Century. Clin Microbiol Rev. 2019; 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zhang W, Zhang Z, Wu W, et al. Epidemiology and control of echinococcosis in central Asia, with particular reference to the People’s Republic of China. Acta Trop. 2015; 141:235–243. doi: 10.1016/j.actatropica.2014.03.014 [DOI] [PubMed] [Google Scholar]
  • 5.Anthony B, Allen JT, Li YS, McManus DP. Hepatic stellate cells and parasite-induced liver fibrosis. Parasit Vectors. 2010; 3:60. doi: 10.1186/1756-3305-3-60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Liu Z, Zhang L, Liang Y, Lu L. Pathology and molecular mechanisms of Schistosoma japonicum-associated liver fibrosis. Front Cell Infect Microbiol. 2022; 12:1035765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Niu F, Chong S, Qin M, Li S, Wei R, Zhao Y. Mechanism of Fibrosis Induced by Echinococcus spp. Diseases. 2019; 7(3):51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bresson-Hadni S, Spahr L, Chappuis F. Hepatic Alveolar Echinococcosis. Semin Liver Dis. 2021; 41:393–408. doi: 10.1055/s-0041-1730925 [DOI] [PubMed] [Google Scholar]
  • 9.Labsi M, Soufli I, Amir ZC, Touil-Boukoffa C. Hepatic inflammation and liver fibrogenesis: A potential target for the treatment of cystic echinococcosis-associated hepatic injury. Acta Trop. 2022; 226:106265. doi: 10.1016/j.actatropica.2021.106265 [DOI] [PubMed] [Google Scholar]
  • 10.Tian F, Jiang T, Qi X, et al. Role of Cytokines on the Progression of Liver Fibrosis in Mice Infected with Echinococcus multilocularis. Infect Drug Resist. 2021; 14:5651–5660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Aydın MM, Akçalı KC. Liver fibrosis. Turk J Gastroenterol. 2018; 29:14–21. doi: 10.5152/tjg.2018.17330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Khatun M, Ray RB. Mechanisms Underlying Hepatitis C Virus-Associated Hepatic Fibrosis.Cells. 2019; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Carson JP, Ramm GA, Robinson MW, McManus DP, Gobert GN. Schistosome-Induced Fibrotic Disease: The Role of Hepatic Stellate Cells. Trends Parasitol. 2018; 34:524–540. doi: 10.1016/j.pt.2018.02.005 [DOI] [PubMed] [Google Scholar]
  • 14.Bi X, Yang N, Ke Y, et al. An improved experimental method for simultaneously isolating hepatocytes and hepatic stellate cells in mouse liver infected with Echinococcus multilocularis. J Clin Lab Anal. 2021; 35:e24084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gao HJ, Pang HS, Sun XD, et al. Effects of persistent Echinococcus multilocularis infections on hepatic fibrosis in mice. Chinese Journal of Schistosomiasis Control. 2021; 33:54–61. [DOI] [PubMed] [Google Scholar]
  • 16.Tafrihi M, Hasheminasab E. MiRNAs: Biology, Biogenesis, their Web-based Tools, and Databases. Microrna. 2019; 8:4–27. doi: 10.2174/2211536607666180827111633 [DOI] [PubMed] [Google Scholar]
  • 17.Otsuka M, Kishikawa T, Yoshikawa T, et al. MicroRNAs and liver disease. J Hum Genet. 2017; 62:75–80. doi: 10.1038/jhg.2016.53 [DOI] [PubMed] [Google Scholar]
  • 18.Wang X, He Y, Mackowiak B, Gao B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut. 2021; 70:784–795. doi: 10.1136/gutjnl-2020-322526 [DOI] [PubMed] [Google Scholar]
  • 19.Tadokoro T, Morishita A, Masaki T. Diagnosis and Therapeutic Management of Liver Fibrosis by MicroRNA. Int J Mol Sci. 2021; 22. doi: 10.3390/ijms22158139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Deping C, Bofan J, Yaogang Z, Mingquan P. microRNA-125b-5p is a promising novel plasma biomarker for alveolar echinococcosis in patients from the southern province of Qinghai. BMC Infect Dis. 2021; 21:246. doi: 10.1186/s12879-021-05940-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Boubaker G, Strempel S, Hemphill A, Müller N, Wang J, Gottstein B. Regulation of hepatic microRNAs in response to early stage Echinococcus multilocularis egg infection in C57BL/6 mice.PLoS Negl Trop Dis. 2020; 14:e0007640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jin X, Guo X, Zhu D, Ayaz M, Zheng Y. miRNA profiling in the mice in response to Echinococcus multilocularis infection. Acta Trop. 2017; 166:39–44. [DOI] [PubMed] [Google Scholar]
  • 23.Guo X, Zheng Y. Expression profiling of circulating miRNAs in mouse serum in response to Echinococcus multilocularis infection. Parasitology. 2017; 144:1. [DOI] [PubMed] [Google Scholar]
  • 24.Mederacke I, Dapito DH, Affò S, Uchinami H, Schwabe RF. High-yield and high-purity isolation of hepatic stellate cells from normal and fibrotic mouse livers. Nat Protoc. 2015; 10:305–315. doi: 10.1038/nprot.2015.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liu T, Wang L, Li H, et al. circRNA Expression Pattern and circRNA-miRNA-mRNA Network in HCs, HSCs, and KCs of Murine Liver After Echinococcus multilocularis Infection. Front Vet Sci. 2022; 9:825307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sun Y, Kou Y, He X, et al. Efficient delivery of Echinococcus multilocularis miRNAs using chitosan nanoparticles. Biomed Pharmacother. 2022; 150:112945. [DOI] [PubMed] [Google Scholar]
  • 27.Liu T, Li H, Li Y, et al. Integrative Analysis of RNA Expression and Regulatory Networks in Mice Liver Infected by Echinococcus multilocularis. Front Cell Dev Biol. 2022; 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Friedman SL, Roll FJ, Boyles J, Bissell DM. Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. Proc Natl Acad Sci USA. 1985; 82:8681–8685. doi: 10.1073/pnas.82.24.8681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adva Drug Deliv Rev. 2017; 121:27–42. doi: 10.1016/j.addr.2017.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhao Z, Lin CY, Cheng K. siRNA- and miRNA-based therapeutics for liver fibrosis. Transl Res. 2019; 214:17–29. doi: 10.1016/j.trsl.2019.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang J, Chu ES, Chen HY, et al. microRNA-29b prevents liver fibrosis by attenuating hepatic stellate cell activation and inducing apoptosis through targeting PI3K/AKT pathway. Oncotarget. 2015; 6:7325–7338. doi: 10.18632/oncotarget.2621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tao R, Fan XX, Yu HJ, et al. MicroRNA-29b-3p prevents Schistosoma japonicum-induced liver fibrosis by targeting COL1A1 and COL3A1. J Cell Biochem. 2018; 119:3199–3209. [DOI] [PubMed] [Google Scholar]
  • 33.Coll M, El Taghdouini A, Perea L, et al. Integrative miRNA and Gene Expression Profiling Analysis of Human Quiescent Hepatic Stellate Cells. Sci Rep. 2015; 5:11549. doi: 10.1038/srep11549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shen Y, Cao R, Liu W, et al. Negative feedback loop between ZBTB7A and TGF-β in breast cancer. Oncol Lett. 2017; 14:1403–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dewidar B, Meyer C, Dooley S, Meindl-Beinker AN. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells. 2019; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ritzenthaler JD, Goldstein RH, Fine A, Smith BD. Regulation of the alpha 1(I) collagen promoter via a transforming growth factor-beta activation element. J Biol Chem. 1993; 268:13625–13631. [PubMed] [Google Scholar]
PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r001

Decision Letter 0

Alessandra Morassutti, Eva Clark

21 Feb 2023

Dear Dr Xiaola Guo,

Thank you very much for submitting your manuscript "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a" for consideration at PLOS Neglected Tropical Diseases. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. In light of the reviews (below this email), we would like to invite the resubmission of a significantly-revised version that takes into account the reviewers' comments.

We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent to reviewers for further evaluation.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript. Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Please prepare and submit your revised manuscript within 60 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email. Please note that revised manuscripts received after the 60-day due date may require evaluation and peer review similar to newly submitted manuscripts.

Thank you again for your submission. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Alessandra Morassutti, PhD

Academic Editor

PLOS Neglected Tropical Diseases

Eva Clark

Section Editor

PLOS Neglected Tropical Diseases

***********************

Reviewer's Responses to Questions

Key Review Criteria Required for Acceptance?

As you describe the new analyses required for acceptance, please consider the following:

Methods

-Are the objectives of the study clearly articulated with a clear testable hypothesis stated?

-Is the study design appropriate to address the stated objectives?

-Is the population clearly described and appropriate for the hypothesis being tested?

-Is the sample size sufficient to ensure adequate power to address the hypothesis being tested?

-Were correct statistical analysis used to support conclusions?

-Are there concerns about ethical or regulatory requirements being met?

Reviewer #1: (No Response)

Reviewer #2: see below

Reviewer #3: the objectives of the study clearly articulated. however, the authors need to answer the following questions:

Major issues:

• A copy of ethical approval is needed.

• In line 78, it’s better to mention the total number of mice used. And how it’s grouped because here you mentioned you divided them into 2 group, then in line 98 you said that “Relative expression levels of miRNAs in three groups” ???!

• In line 83, more details about the method of isolating HSCs from mice liver is needed.

• In line 133, the authors had mentioned that the liver was fixed in 4% paraformaldehyde, is the paraformaldehyde is prepared in Phosphate buffer saline or what, and is this protocol for immunochemistry does not require antigen retrieval?

Minor Issues:

Clarification of abbreviations is needed to provide better understanding especially for those who are not very familiar with the field of the study.

--------------------

Results

-Does the analysis presented match the analysis plan?

-Are the results clearly and completely presented?

-Are the figures (Tables, Images) of sufficient quality for clarity?

Reviewer #1: (No Response)

Reviewer #2: see below

Reviewer #3: the results was clearly presented.

--------------------

Conclusions

-Are the conclusions supported by the data presented?

-Are the limitations of analysis clearly described?

-Do the authors discuss how these data can be helpful to advance our understanding of the topic under study?

-Is public health relevance addressed?

Reviewer #1: (No Response)

Reviewer #2: see below

Reviewer #3: the conclusions is supported by the data. however the author should mention the limitations of the study clearly.

--------------------

Editorial and Data Presentation Modifications?

Use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity. If the only modifications needed are minor and/or editorial, you may wish to recommend “Minor Revision” or “Accept”.

Reviewer #1: (No Response)

Reviewer #2: see below

Reviewer #3: “Minor Revision”

--------------------

Summary and General Comments

Use this section to provide overall comments, discuss strengths/weaknesses of the study, novelty, significance, general execution and scholarship. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. If requesting major revision, please articulate the new experiments that are needed.

Reviewer #1: The manuscript entitled "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a" has been reviewed. Generally, the manuscript clearly indicated the role of miRNA-342-3p in liver fibrosis associated with Echinococcus multilocularis infection.

The reviewer has several suggestions and comments to improve the quality of current manuscript.

1. Liver fibrosis could be caused by other flatworm infection such as Schistosoma. The authors should include related studies in section of introduction and discussion to enrich the informative manuscript for understanding the mechanism of liver fibrosis.

2. The authors globally characterized the profiles of lncRNAs-miRNAs-mRNAs in hepatocytes (HCs), Kupffer cells (KCs) and HSCs of mouse liver during E. multilocularis infection. Did they note that the Zbtb7a has different expression in different time of different type cells? The author could include the related results in the discussion.

3. In Fig 1A, the data indicated that the expression of Alpha-Sma has not different at 60dpi between infection and uninfection. How to explain the results?

4. In Fig 5E, the manuscript is lack of the legend of fig 5E. What mean 1,2,3 in fig 5E?

Reviewer #2: The MS by Cao et al. entitled: “mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a” is interesting in basics but suffers from major flaws.

The authors state that infection induces fibrosis in liver of mice infected by EM. All of the message of the MS is based on this histological finding. However, this fundamental part of the analysis is badly documented:

1. In the materials and methods section there is no description how the fibrosis was evaluated nor by whom neither by which method.

2. Infection mice with EM is deleterious. In the few pics shown the authors do not show any metacestodes but just some portal field with fibrosis. Fibrosis develops around the metacestode and not in the portal fields. Authors have to show full blown liver lesions, measure the differences of inflammatory infiltrates, and then measure the fibrotic rim (or computer-based and with values that have been used to generate the box-plots: Combining Computed Tomography and Histology Leads to an Evolutionary Concept of Hepatic Alveolar Echinococcosis - PubMed (nih.gov).

3. Definition of stellate cells is poor. Are HSC increased during infection in situ? What is about the KCs and/or hepatocytes?

4. The authors must demonstrate that the effect is really based on this specific kind of infection (e.g. control groups with fat induced liver disease, or hepatitis are missing).

5. In the paper the number of experiments und number of cell cultures and repetition of experiments are not clearly given. This not acceptable.

Minor: It remains a long time unclear whether these experiments have been performed in mice or humans. Authors should consider to shift the experiments in the human system. This is the major health problem, not mice. Cultures of Human stellate cells are available.

If all this major issue are taken in account I am willing to re-review the MS.

Reviewer #3: I think this paper miRNA-342-3p promotes HSC activation induced by E. multilocularis infection is very important and novel.

Major issues:

• A copy of ethical approval is needed.

• In line 78, it’s better to mention the total number of mice used. And how it’s grouped because here you mentioned you divided them into 2 group, then in line 98 you said that “Relative expression levels of miRNAs in three groups” ???!

• In line 83, more details about the method of isolating HSCs from mice liver is needed.

• In line 133, the authors had mentioned that the liver was fixed in 4% paraformaldehyde, is the paraformaldehyde is prepared in Phosphate buffer saline or what, and is this protocol for immunochemistry does not require antigen retrieval?

Minor Issues:

Clarification of abbreviations is needed to provide better understanding especially for those who are not very familiar with the field of the study.

--------------------

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r003

Decision Letter 1

Francesca Tamarozzi

25 Apr 2023

Dear Dr Guo,

Thank you very much for submitting your manuscript "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a" for consideration at PLOS Neglected Tropical Diseases. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. In light of the reviews (below this email), we would like to invite the resubmission of a significantly-revised version that takes into account the reviewers' comments.

We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent to reviewers for further evaluation.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript. Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Please prepare and submit your revised manuscript within 60 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email. Please note that revised manuscripts received after the 60-day due date may require evaluation and peer review similar to newly submitted manuscripts.

Thank you again for your submission. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Francesca Tamarozzi

Section Editor

PLOS Neglected Tropical Diseases

Eva Clark

Section Editor

PLOS Neglected Tropical Diseases

***********************

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r005

Decision Letter 2

Francesca Tamarozzi

17 May 2023

Dear Dr Guo,

Thank you very much for submitting your manuscript "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a" for consideration at PLOS Neglected Tropical Diseases. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. The reviewers appreciated the attention to an important topic. Based on the reviews, we are likely to accept this manuscript for publication, providing that you modify the manuscript according to the review recommendations.

It is of particular importance the last point raised by Reviewer #2 is addressed.

Please prepare and submit your revised manuscript within 30 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to all review comments, and a description of the changes you have made in the manuscript.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Thank you again for your submission to our journal. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Francesca Tamarozzi

Section Editor

PLOS Neglected Tropical Diseases

Eva Clark

Section Editor

PLOS Neglected Tropical Diseases

***********************

It is of particular importance the last point raised by Reviewer #2 is addressed.

Reviewer's Responses to Questions

Key Review Criteria Required for Acceptance?

As you describe the new analyses required for acceptance, please consider the following:

Methods

-Are the objectives of the study clearly articulated with a clear testable hypothesis stated?

-Is the study design appropriate to address the stated objectives?

-Is the population clearly described and appropriate for the hypothesis being tested?

-Is the sample size sufficient to ensure adequate power to address the hypothesis being tested?

-Were correct statistical analysis used to support conclusions?

-Are there concerns about ethical or regulatory requirements being met?

Reviewer #1: (No Response)

Reviewer #2: Improved.

--------------------

Results

-Does the analysis presented match the analysis plan?

-Are the results clearly and completely presented?

-Are the figures (Tables, Images) of sufficient quality for clarity?

Reviewer #1: (No Response)

Reviewer #2: Improved. However, as I recommeded in my first review I ask the authors again to insert a histological figure of a full blown liver lesion of a mouse after infection as a proof of principle. Up to now, the authors show histology of mice with non characteristic periportal fibrosis; to insert a histological figure of the charcteristic liver lesion after infection is an item that should be easily done since infection rates are high (as have stated the authors); to solve this point an additional supplemental figure is enough. The authors have not answered to this point raised in my previous review.

--------------------

Conclusions

-Are the conclusions supported by the data presented?

-Are the limitations of analysis clearly described?

-Do the authors discuss how these data can be helpful to advance our understanding of the topic under study?

-Is public health relevance addressed?

Reviewer #1: (No Response)

Reviewer #2: Improved

--------------------

Editorial and Data Presentation Modifications?

Use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity. If the only modifications needed are minor and/or editorial, you may wish to recommend “Minor Revision” or “Accept”.

Reviewer #1: (No Response)

Reviewer #2: Improved.

--------------------

Summary and General Comments

Use this section to provide overall comments, discuss strengths/weaknesses of the study, novelty, significance, general execution and scholarship. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. If requesting major revision, please articulate the new experiments that are needed.

Reviewer #1: (No Response)

Reviewer #2: Improved.

--------------------

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

References

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article's retracted status in the References list and also include a citation and full reference for the retraction notice.

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r007

Decision Letter 3

Uriel Koziol

18 Jun 2023

Dear Dr Guo,

Thank you very much for submitting your manuscript "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a" for consideration at PLOS Neglected Tropical Diseases. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. The reviewers appreciated the attention to an important topic. Based on the reviews, we are likely to accept this manuscript for publication, providing that you modify the manuscript according to the review recommendations.

Due to the previous editor being currently unavailable, I have been assigned as the editor for the revised version of your manuscript.

While reviewing the revised manuscript, I have concluded that all comments and criticisms presented by the reviewers have been correctly addressed.

However, I have also noticed that in Fig. 5A, the graph shows that the miRNA mimic increased luciferase activity, while the text indicates that luciferase activity was decreased by the RNA mimic (as would be expected, given the rest of the results shown in Figs. 5 and 6). Please check if there was an error during the preparation of Fig. 5A, or revise the text and conclusions if necessary.

Additionally, in Fig. 5A the inset reads "Mimcs" instead of "mimic", and the NC mimic is not described in the text or figure legend.

Finally, please check l. 258, "hepatocytes (Alb-positive cells) were significantly decreased (Fig 2B and S2 Fig).", as the marker for hepatocytes used in Fig. S2 was cytokeratin 18, and not albumin.

Please prepare and submit your revised manuscript within 30 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to all review comments, and a description of the changes you have made in the manuscript.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Thank you again for your submission to our journal. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Uriel Koziol

Section Editor

PLOS Neglected Tropical Diseases

Eva Clark

Section Editor

PLOS Neglected Tropical Diseases

***********************

Due to the previous editor being currently unavailable, I have been assigned as the editor for the revised version of your manuscript.

While reviewing the revised manuscript, I have concluded that all comments and criticisms presented by the reviewers have been correctly addressed.

However, I have also noticed that in Fig. 5A, the graph shows that the miRNA mimic increased luciferase activity, while the text indicates that luciferase activity was decreased by the RNA mimic (as would be expected, given the rest of the results shown in Figs. 5 and 6). Please check if there was an error during the preparation of Fig. 5A, or revise the text and conclusions if necessary.

Additionally, in Fig. 5A the inset reads "Mimcs" instead of "mimic", and the NC mimic is not described in the text or figure legend.

Finally, please check l. 258, "hepatocytes (Alb-positive cells) were significantly decreased (Fig 2B and S2 Fig).", as the marker for hepatocytes used in Fig. S2 was cytokeratin 18, and not albumin.

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

References

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article's retracted status in the References list and also include a citation and full reference for the retraction notice.

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r009

Decision Letter 4

Uriel Koziol

8 Jul 2023

Dear Dr Guo,

We are pleased to inform you that your manuscript 'mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a' has been provisionally accepted for publication in PLOS Neglected Tropical Diseases.

Before your manuscript can be formally accepted you will need to complete some formatting changes, which you will receive in a follow up email. A member of our team will be in touch with a set of requests.

Please note that your manuscript will not be scheduled for publication until you have made the required changes, so a swift response is appreciated.

IMPORTANT: The editorial review process is now complete. PLOS will only permit corrections to spelling, formatting or significant scientific errors from this point onwards. Requests for major changes, or any which affect the scientific understanding of your work, will cause delays to the publication date of your manuscript.

Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us now if you or your institution is planning to press release the article. All press must be co-ordinated with PLOS.

Thank you again for supporting Open Access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Uriel Koziol

Section Editor

PLOS Neglected Tropical Diseases

Eva Clark

Section Editor

PLOS Neglected Tropical Diseases

***********************************************************

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0011520.r010

Acceptance letter

Uriel Koziol

20 Jul 2023

Dear Dr Guo,

We are delighted to inform you that your manuscript, "mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a," has been formally accepted for publication in PLOS Neglected Tropical Diseases.

We have now passed your article onto the PLOS Production Department who will complete the rest of the publication process. All authors will receive a confirmation email upon publication.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any scientific or type-setting errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript. Note: Proofs for Front Matter articles (Editorial, Viewpoint, Symposium, Review, etc...) are generated on a different schedule and may not be made available as quickly.

Soon after your final files are uploaded, the early version of your manuscript will be published online unless you opted out of this process. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Shaden Kamhawi

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

Paul Brindley

co-Editor-in-Chief

PLOS Neglected Tropical Diseases

Associated Data

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

    Supplementary Materials

    S1 Data. Excel spreadsheet containing, in separate sheets, the underlying numerical data and statistical analysis for Figure panels 1B, 2B, 2E, 3C, 3D, 4A, 4C, 5A, 5B, 5C, 5D, 6A, 6B, 6C, 6D, 6F and S3.

    (XLSX)

    S1 Fig. Full-blown liver lesions of E. multilocularis-infected mice were observed.

    (TIF)

    S2 Fig. Immunostaining of CLEC4F (KC marker) and Cytokeratin 18 (HC marker) in livers of un-infected mice or E. multilocularis-infected mice.

    (TIF)

    S3 Fig. The mRNA expression levels of biomarkers in the isolated HCs (Alb and CK18), HSCs (α-SMA, Gfap, and Col1α1), and KCs (Emr1) by RT-qPCR.

    (TIFF)

    S1 Text

    Table A. The qRT-PCR primers used in the study. Table B. Summary of the differentially expressed miRNAs in liver HSCs in E. multilocularis-infected mice at 60-day post infection. Table C. Summary of the differentially expressed miRNAs in liver HSCs in E. multilocularis-infected mice at 90-day post infection. Table D. Putative target genes of mmu-miR-342-3p.

    (DOCX)

    Attachment

    Submitted filename: response to reviewers final.docx

    Attachment

    Submitted filename: response to reviewers final.docx

    Attachment

    Submitted filename: Responses to Reviewer.docx

    Attachment

    Submitted filename: Responses to Reviewer.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLOS Neglected Tropical Diseases are provided here courtesy of PLOS

    RESOURCES