Skip to main content
PLOS One logoLink to PLOS One
. 2023 Sep 15;18(9):e0286256. doi: 10.1371/journal.pone.0286256

Inducible overexpression of a FAM3C/ILEI transgene has pleiotropic effects with shortened life span, liver fibrosis and anemia in mice

Ulrike Schmidt 1, Betül Uluca 1,¤a, Iva Vokic 2, Barizah Malik 2,¤b, Thomas Kolbe 3,4, Caroline Lassnig 5, Martin Holcmann 2, Veronica Moreno-Viedma 2, Bernhard Robl 2, Carina Mühlberger 6, Dagmar Gotthardt 6, Maria Sibilia 2, Thomas Rülicke 7, Mathias Müller 5, Agnes Csiszar 2,*
Editor: Michael Klymkowsky8
PMCID: PMC10503705  PMID: 37713409

Abstract

FAM3C/ILEI is an important factor in epithelial-to-mesenchymal transition (EMT) induction, tumor progression and metastasis. Overexpressed in many cancers, elevated ILEI levels and secretion correlate with poor patient survival. Although ILEI’s causative role in invasive tumor growth and metastasis has been demonstrated in several cellular tumor models, there are no available transgenic mice to study these effects in the context of a complex organism. Here, we describe the generation and initial characterization of a Tet-ON inducible Fam3c/ILEI transgenic mouse strain. We find that ubiquitous induction of ILEI overexpression (R26-ILEIind) at weaning age leads to a shortened lifespan, reduced body weight and microcytic hypochromic anemia. The anemia was reversible at a young age within a week upon withdrawal of ILEI induction. Vav1-driven overexpression of the ILEIind transgene in all hematopoietic cells (Vav-ILEIind) did not render mice anemic or lower overall fitness, demonstrating that no intrinsic mechanisms of erythroid development were dysregulated by ILEI and that hematopoietic ILEI hyperfunction did not contribute to death. Reduced serum iron levels of R26-ILEIind mice were indicative for a malfunction in iron uptake or homeostasis. Accordingly, the liver, the main organ of iron metabolism, was severely affected in moribund ILEI overexpressing mice: increased alanine transaminase and aspartate aminotransferase levels indicated liver dysfunction, the liver was reduced in size, showed increased apoptosis, reduced cellular iron content, and had a fibrotic phenotype. These data indicate that high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis, which leads to liver dysfunction, disturbed iron metabolism and eventually to death. Overall, we show here that the novel Tet-ON inducible Fam3c/ILEI transgenic mouse strain allows tissue specific timely controlled overexpression of ILEI and thus, will serve as a versatile tool to model the effect of elevated ILEI expression in diverse tissue entities and disease conditions, including cancer.

Introduction

The FAM3 family of cytokines were first identified from a search of genomics databases for molecules predicted to be structurally similar to interleukins and other related cytokines with a four-helix-bundle [1]. The search found four genes FAM3A, FAM3B, FAM3C, and FAM3D with a hydrophobic leader sequence [1]. The protein expressed by FAM3C was later shown in the context of cancer to be involved in epithelial-to-mesenchymal transition (EMT) [2]. Based on this action and the presumed similarity to interleukins FAM3C was renamed interleukin-like EMT inducer (ILEI). However, subsequent crystal structure analysis revealed that the FAM3 family in fact constitutes a distinct class of signaling molecules [3, 4], FAM3C/ILEI being unique in the family due to its covalent dimerization properties [3, 5].

Human tumors from various cancers overexpress ILEI with altered subcellular localization, which is associated with changes in the secretion levels of the protein [6], elevated ILEI levels also linked to gene amplification [7]. EMT converts adherent epithelial tumor cells into highly invasive mesenchymal cells [8]. ILEI has been demonstrated to induce invasion in cancer cell models and metastasis from xenografts [911]. Additionally, altered ILEI localization has been linked to metastasis and survival in human breast and hepatocellular carcinomas [911], and high levels of ILEI expression have correlated with poor prognosis in colorectal cancer [12]. Despite a growing understanding of the role of elevated ILEI levels in causing invasive tumor growth and metastasis, methods available to study these effects in the context of a complex organism are limited.

ILEI has also been implicated in other diseases. Low levels of ILEI expression might be a factor in Alzheimer’s disease development [13]. While ILEI may also drive EMT in renal fibrosis [14]. FAM3C genetic variants have been associated with bone mineral density, dyslipidemia and lipid traits, and sudden sensorineural hearing loss [1517]. Therefore, mouse models have been developed to investigate non-cancer effects of ILEI. Two previous studies have described a Fam3c knockout mouse strain, with a focus on bone mineral density [18, 19]. These models demonstrated relatively minor effects on the mouse phenotype. Another study evaluated the overexpression of human FAM3C in neuronal cells under the control of the mouse prion promoter to understand the mechanism of Alzheimer’s disease [20]. However, both of these approaches lack the option to manipulate ILEI expression in time and in specific tissues of interest. The Tet-On system is a dual transgenic system involving a reverse tetracycline-controlled transactivator (i) that only recognizes the tetracycline-responsive promoter element (TRE) (ii) in the presence of doxycycline (Dox) [21, 22]. Therefore, using this system to produce transgenic mice can provide reversibly inducible gene expression in a tissue specific manner. A safe consideration for genes that are likely to have lethal or highly debilitating effects. This system has been used successfully to produce mouse models for the study of many different genes [2326].

In this study, we describe the generation and initial characterization of a Tet-On inducible Fam3c/ILEI transgenic mouse strain. This model will provide a useful tool in understanding the complex mechanisms of ILEI action in the context of a whole organism in a wide range of cancers and other diseases.

Materials and methods

Ethics statement

Mice were housed under barrier conditions in specific pathogen-free quality according to FELASA recommendations [27]. All animal experiments were discussed and approved by the Ethics and Animal Welfare Committee of the University of Veterinary Medicine Vienna, the Research Institute of Molecular Pathology, and the Medical University of Vienna and conform to the guidelines of the national authority (the Austrian Federal Ministry for Science and Research) as laid down in §8ff of the Animal Science and Experiments Act (Tierversuchsgesetz–TVG; refs BMWF-68.205/0204-C/GT/2007; BMWF-68.205/0210-II/10b/2009, MA58/001489/2008/12, BMWF-66.009/0065-II/10b/2009, BMWFW-66.009/0319-V/3b/2019) and according to the guidelines of FELASA and ARRIVE. A humane endpoint was conducted by cervical dislocation if death of the animals was to be expected during the following hours. As pale skin coloration and weight loss/retarded growth accompanied the overall phenotype, palpable hypothermia and unresponsive condition were used as the most reliable criteria to determine moribundity.

Cloning, gene targeting, and mice

For the establishment of ILEIind mice we used a 2nd generation Tet-On system based on site-specific recombination in embryonic stem (ES) cells [28, 29]. We targeted a construct carrying the Tet operator (tetO) followed by a FLAG-tagged Fam3c cDNA (Genbank accession number NM_138587.4) into the Col1a1 locus of KH2 ES cells by Flp/frt-mediated recombination as described earlier [30]. Briefly, Fam3c coding sequence bearing a FLAG tag at its 3’ end was re-cloned from the pcDNA3.1-ILEI-FLAG construct [6] via EcoRI (Thermo Scientific) into the vector pBS319-RGBpA directly under the control of a doxycycline-inducible promoter element [28] and designated pBS31-ILEI-FLAG. The transgenic vector and a vector encoding the Flp recombinase (pCAGGS-FLP [28]) were co-transfected into KH2 ES cells. ES cell clones with correct recombination were selected by hygromycin B, correct and single integration of the construct was verified by Southern blot analysis as described earlier [30]. Tight and inducible regulation of gene expression was confirmed by in vitro Dox induction tests. Selected ES cell clone #7 was injected into C57BL/6N blastocysts and the resulting chimeras were crossed to B6N;129Sv mice. Dual-transgenic progenies heterozygous for the M2rtTA in the Rosa26 locus and the dox-inducible ILEI-FLAG construct in the Col1a1 locus were intercrossed to obtain homozygous dual-transgenic B6N;129Sv-Gt(ROSA)26Sortm1(rtTA*M2)Jae-Col1a1tm1(tetO-ILEI-FLAG)Biat mice (referred to here as R26-ILEIind mice). In parallel, mice were backcrossed to C57BL/6J mice by speed congenic procedure [31] and crossed to Vav-rtTA3 mice [32] (referred to here as Vav-ILEIind mice). All experiments were performed using R26-ILEIind male mice in a B6N;129Sv and Vav-ILEIind male and female mice in a C57BL/6J genetic background. Genotyping of R26-ILEIind mice and cells was performed as described earlier [30]. The Vav-rtTA3 transgene was genotyped as previously described [32]. Dox was supplied to R26-ILEIind mice via drinking water (1 mg/ml, supplemented with 5% sucrose) and to Vav-ILEIind mice in the diet (1000 mg/kg, ssniff). Animals were checked daily to monitor their health status by assessing nesting performance and examining groomed fur and posture. Autopsy was performed on rare moribund mice and on all surviving animals at around day 240 of life, which was set as the end point of the experiment. During macroscopic post-mortem examinations, we did not detect any spontaneous cancer formation.

Blood tests

Whole blood was collected weekly using K3EDTA MiniCollect tubes (Greiner Bio-One) by bleeding mice on the tail vein. White blood cell count (WBC), red blood cell count (RBC), hemoglobin (HGB) and hematocrit (HCT) levels were measured using an animal blood counter (sciI Vet abc). Blood sera were collected by centrifugation, diluted 1:2–4 upon need in isotonic saline solution and used to determine aspartate aminotransferase (AST), alanine transaminase (ALT), and serum iron levels using Hitachi Cobas C111 and Hitachi Cobas C311 (Roche) devices, respectively.

Cell culture

ES and bone marrow cells were cultivated as previously described [33]. For in vitro induction experiments, cells were treated with doxycycline-hydrochloride (Sigma) dissolved in water at concentrations ranging from 0.2–5 μg/ml.

Western blot analysis

Protein lysates and western blots were performed as described [6] using FLAG (Sigma), ILEI [11], Erk1, beta-actin and vinculin (Cell Signaling Technologies) primary antibodies and horseradish peroxidase (HRP)-coupled secondary antibodies (Jackson ImmunoResearch).

Immunohistochemistry, prussian blue, and picrosirius red staining

Primary antibodies against ILEI [11], FLAG (Sigma), Ki67 (Abcam), activated caspase 3 (actCasp3, Abcam), fibronectin, E-cadherin, and S100A4 (Cell Signaling Technologies) were used for immunohistochemistry by applying standard protocols. Briefly, 4-μm sections of paraffin embedded mouse tissues were deparaffinized, antigen retrieval was performed in citrate buffer (Dako). After peroxidase treatment and blocking (2% BSA, 10% horse serum and 0.1% Tween 20 in PBS), slides were incubated overnight at 4°C with primary antibodies diluted in blocking buffer, followed by washing and then incubation with HRP-conjugated SignalStain Boost immunohistochemistry detection reagent (Cell Signaling Technology) for 30 minutes at room temperature, developed with DAB substrate (Dako) followed by hematoxylin counterstain, rehydration and mounting. Hematoxylin and eosin (H&E) staining (Sigma-Aldrich) staining was performed according to standard protocols. Prussian blue staining for ferric iron detection was performed as described earlier [34, 35]. Briefly, equal parts of 5% potassium ferrocyanide (Merck) and 5% hydrochloric acid (Sigma) were mixed and heated until vaporization in a microwave. Sections were immersed for 8 minutes in the heated solution and counterstained with nuclear fast red (Sigma) for 7 minutes, followed by washing in tap water, dehydration and embedding. Histological visualization of collagen was performed using the Picric-Sirius Red Stain Kit (Scy-Tek) according to the manufacturer’s instructions. Slides were digitized with a Pannoramic SCAN II slide scanner (3DHistech) in extended focus scanning mode using a 20X plan-apochromat objective (0.8 NA) and a 5Mpxl sCMOS camera. Quantification was performed using automized histology quantification software (Definiens Tissue studio® 4.3) and in case of Prussian blue staining by manual counting of “blue” (ferric iron-containing) hepatocytes in randomly selected areas using QuPath 0.3.2 [36].

Flow cytometry of blood and spleen

The cellular fraction of 100μl freshly isolated blood was suspended in 1x PBS and transferred into 3ml erythrocyte lysis buffer (0.15 M NH4Cl, 10 mM KHCO3, 0.1 mM Na2EDTA, pH 7.2–7.4). After lysis leukocytes were pelleted, washed, and blocked with anti-CD16/32 antibody (BioLegend), followed by staining with the following fluorophore conjugated primary antibodies for 30 min at 4°C: CD11b, Gr1, CD19 and CD3ε (Biolegend). Single-cell suspension of freshly isolated splenocytes were prepared as described earlier [37] and subsequently blocked with anti-CD16/32 antibody (BioLegend). Splenocytes were stained with following fluorophore conjugated primary antibodies for 30 min at 4°C: Ly6G, CD11b, CD11c, CD19, F4/80, MHC class II, Ly6C and CD3ε (Biolegend). After incubation, the samples were washed, filtered, and stained with SYTOXgreen viability dye (Thermo Fisher) according to the manufacturer’s recommendation to exclude dead cells. Leukocytes and splenocytes were recorded by a FACSCanto (BD Bioscience) and by a Fortessa (BD Bioscience) cytometers, respectively and analyzed by FlowJo v10 software.

Statistical analysis

Statistical analysis was done with GraphPad Prism 8.0. For comparison of one parameter between two groups we used an unpaired two-tailed student’s t-test, comparison across multiple groups was performed by one-way analysis of variance (ANOVA) with Bonferroni posthoc analysis for multiple comparisons to determine statistical significance or in specific cases with student’s t-test with pairwise comparisons. Survival analysis was performed using Kaplan-Meyer plots and log rank statistics. p-values of lower than 0.05 were considered statistically significant (*p<0.05, **p<0.01, ***p<0.001). Error bars are represented as standard error of mean (SEM) of at least three independent biological replicates.

Results

Generation of KH2-ILEI-FLAG ES cells with inducible expression of ILEI-FLAG

The first step in generating the Tet-On inducible Fam3c/ILEI transgenic mouse strain was to establish KH2 ES cells expressing Dox-inducible FLAG-tagged ILEI. As shown in Fig 1A, we used the flp-in system to produce KH2 embryonic mouse stem cells expressing the M2 reverse tetracycline controlled transactivator (M2rtTA) which recognizes the tetO sequence of the Tet response element (TRE) on the Fam3c/ILEI-FLAG transgene only in the presence of Dox. Transient transfection confirmed that the cloned tet:ILEI-FLAG construct resulted in the expression of a FLAG tagged protein with the expected 25 kDA size only upon induction with Dox (Fig 1B). Single cell clones of the flp-in genomic targeting were selected with antibiotics and further sub-selected after confirming targeted insertion of the transgene by PCR (Fig 1C) and FLAG western blot analysis to verify protein expression of correct size and tight regulation of Dox inducibility (Fig 1D). Southern blot analysis of genomic DNA with a probe specific for the engineered locus was performed to validate single copy insertion of the transgene (Fig 1E). Finally, clone #7 was selected for blastocyst injection.

Fig 1. Generation of KH2-ILEI-FLAG ES cells.

Fig 1

(A) Scheme of the flp-in targeting strategy in KH2 ES cells. Adapted from [30]. (B) FLAG Western blot analysis of KH2 ES cells after transient transfection with the Fam3c/ILEI-FLAG transgene targeting construct with or without Dox induction. Erk1 was used as loading control. (C) Validation of transgene targeted insertion by PCR on single clones of KH2 ES cells after Hygromycin selection. Clone IDs in black were selected for further analysis. (D) FLAG western blot analysis of sub-selected KH2-ILEI-FLAG clones with or without Dox induction. Erk1 was used as loading control. Clone IDs in black were selected for further analysis. (E) Southern blot analysis of sub-selected KH2-ILEI-FLAG clones with a probe specific for the engineered locus. KH2 cells were used as negative and KH2-STAT1 cells [30] as positive controls. Clone #7 in bold was selected for blastocyst injection.

The Fam3c/ILEI-FLAG transgene is expressed in a broad range of tissues and organs of R26-ILEIind mice upon doxycycline induction

Blastocyst injection gave rise to 8 male mice with 30–50% chimerism and one of them showed germ-line transmission generating one founder male mouse. This founder was crossed to C57BL/6N mice and the first offspring generation was analyzed for transgene expression (Fig 2). First, the expression of the Tet-inducible Fam3c/ILEI-FLAG transgene was analyzed in ex vivo bone marrow culture after treatment with Dox for 24- or 48-hours at different concentrations (Fig 2A). Western blots of bone marrow isolated from dual-transgenic Rosa26rtTA-ILEIind (R26-ILEIind) mice showed ILEI-FLAG protein expression upon Dox administration to the cell culture, while bone marrow derived from a control Rosa26rtTA mouse did not. Higher dose of Dox induced higher levels of protein expression (Fig 2A). To evaluate inducibility of ILEI expression in vivo, ILEI protein expression levels were analyzed in various organs from mice kept on normal or Dox-containing drinking water for three days before sacrifice (Fig 2B and 2C). Protein extracts of spleen and bone marrow, as well as blood sera were examined by western blot (Fig 2B). The results showed low levels of endogenous mouse ILEI expression in splenocytes of all mice, while samples from the mice on Dox water had obviously increased ILEI protein expression. There was no endogenous ILEI expression in the bone marrow or blood sera, but mice receiving Dox showed high levels of the ILEI protein in these samples (Fig 2B). Histology sections of the intestines, kidney, liver, lung and skin also showed that mice receiving Dox expressed elevated levels of ILEI-FLAG in these organs as indicated by increased staining intensity for ILEI, while those not receiving Dox showed little or no ILEI expression (Fig 2C, left panel). De novo transgene expression was verified by immunohistochemistry against the transgene-specific epitope tag FLAG (Fig 2C, right panel). Of note, transgene expression was not detectable in the single-layer alveolar epithelium of the lung, but in other lung structures. These results show that transgenic Fam3c/ILEI-FLAG was efficiently expressed across a wide range of tissues and organs in the R26-ILEIind mice and transgene expression was detected only after induction with Dox.

Fig 2. The Fam3c/ILEI-FLAG transgene is broadly expressed in R26-ILEIind mice upon doxycycline induction.

Fig 2

(A) Western blot analysis of bone marrow isolated from control Rosa26rtTA and dual-transgenic Rosa26rtTA-ILEIind (R26-ILEIind) mice after ex vivo culture for 24 or 48 hours in the presence of the indicated Dox concentrations. FLAG and ILEI antibodies were used for transgene detection, beta-actin was used as the loading control. (B) ILEI western blot analysis of spleen, bone marrow, and blood sera freshly isolated from R26-ILEIind mice kept on normal or switched to Dox drinking water (1 mg/ml Dox and 5% sucrose) 3 days before sacrifice. Please note, splenocytes also showed endogenous ILEI expression. (C) ILEI and FLAG immunohistochemistry on thin sections of the intestine, kidney, liver, lung, and skin of R26-ILEIind mice kept on normal or switched to Dox drinking water (1 mg/ml Dox and 5% sucrose) 3 days before sacrifice. Scale bar, 50μm; scale bar for inlets, 20μm.

ILEI overexpression induced at weaning age results in a reduced life span with reduced body weight and reversible microcytic hypochromic anemia

To analyze the effects of ubiquitous ILEI overexpression in vivo we switched mice to Dox-supplied water at weaning age and monitored phenotypic changes and their overall fitness over time. R26-ILEIind mice receiving Dox drinking water from 3 weeks of age showed a reduced life span compared to R26-ILEIind mice without Dox or mice with Dox but bearing the ILEIind transgene alone. R26-ILEIind mice receiving Dox died with 60% penetrance with a median survival of 220 days (Fig 3A). There was a significant decrease in body weight of R26-ILEIind mice on Dox water compared to R26-ILEIind mice without Dox (p<0.0001) and ILEIind mice with Dox (p<0.0001) being primarily penetrant in moribund R26-ILEIind mice on Dox (Fig 3B). On the one hand, blood analysis indicated little effects on WBC counts and major lymphoid and myeloid subpopulations of R26-ILEIind mice receiving Dox compared to age matched R26-ILEIind mice without Dox or ILEIind mice with Dox, with the exception of a reduction in the CD11b+ subpopulation (Fig 3C). This suggests that ILEI overexpression was not stimulating an immune response. On the other hand, hemoglobin concentration was significantly decreased in R26-ILEIind mice receiving Dox at 3, 2, and 1 weeks before death compared to age matched mice in the ILEIind with Dox and R26-ILEIind without Dox groups (all p<0.05) (Fig 3D). RBC counts were also significantly decreased one week prior to death compared to the control groups (both p<0.01), while HCT values were significantly decreased from 2 weeks prior to death (p<0.05) (Fig 3D). These reduced parameters were accompanied with reduced mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), and mean corpuscular hemoglobin concentration (MCHC) values, indicative of microcytic hypochromic anemia.

Fig 3. Inducible ILEI overexpression results in reduced life span, body weight, and microcytic hypochromic anemia.

Fig 3

(A) Survival plot of ILEIind and R26-ILEIind mice kept on normal or switched to Dox drinking water at 3 weeks of age. (B) Mean body weight ± SEM of R26-ILEIind mice kept on Dox drinking water measured one week before death (dark data points) or at experiment endpoint (white data points) compared to age-matched mice used as Dox treatment and genetic controls. (C) Mean number of WBC, CD11b+, Gr1+, CD11b and Gr1 double-positive, CD3+ and CD19+ cells ± SEM measured at last three weeks before death of R26-ILEIind mice kept on Dox water and compared to age-matched mice used as Dox treatment and genetic controls. (D) Mean HGB concentration (top left), RBC count (middle left) HCT percentage (bottom left), MCV (top right), MCH (middle right) and MCHC (bottom right) ± SEM measured at last three weeks before death of R26-ILEIind mice kept on Dox water and compared to age-matched mice used as Dox treatment and genetic controls. (B-D) Statistical significance was determined by one-way ANOVA and is marked with asterisks (*p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).

To investigate whether any of these effects were direct consequences of ILEI overexpression and thus, reversible upon turning off the transgene, freshly weaned mice were treated with Dox water from week 3 to 13, followed by 2 weeks of Dox withdrawal and were weekly monitored for blood parameters. As shown in Fig 4A, ILEI overexpressing mice showed lower body weight over control mice, and they did not recover from the retarded growth during the two weeks after the transgene was switched off (p<0.01). Importantly, hemoglobin significantly decreased after 3 weeks of ILEI induction, followed by a drop in RBC and HCT levels with a shift of 3 weeks accompanied with significantly reduced MCHC levels, whereas MCV reduction, a sign of microcytic anemia, was not consistent, most likely having been masked by the interfering size reduction of erythrocytes during postnatal development until adulthood [38]. All parameters recovered to similar levels of the control groups one week after dox withdrawal (Fig 4B). This suggests that the anemia was tightly linked to ILEI overexpression and was rapidly reversible.

Fig 4. Anemic phenotype is reversible upon withdrawal of temporary ILEI overexpression.

Fig 4

(A, B) Mean (A) body weight and (B) HGB concentration (top left), RBC count (middle left), HCT percentage (bottom left), MCV (top right), MCH (middle right) and MCHC (bottom right) ± SEM of R26-ILEIind mice kept temporarily on Dox water from week 3 to 13 of age and measured weekly until week 15 of age compared to littermates used as Dox treatment and genetic controls. (A, B) Statistical significance was determined by one-way ANOVA and is marked with asterisks (*p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).

ILEI overexpression in hematopoietic cells does not influence overall fitness and red blood cell parameters

In order to investigate if anemia was caused by a malfunction in the erythroid lineage due to ILEI overexpression, we next examined the effects of Vav1-driven overexpression of the Fam3c/ILEI transgene on overall fitness and blood parameters. Vav1 is exclusively expressed in all hematopoietic cells, so this provides tissue specific expression to contrast with the ubiquitous expression driven by Rosa26. Western blot analysis demonstrated that ILEI-FLAG was expressed in the bone marrow of Vav-ILEIind mice upon Dox administration via the drinking water or diet and appeared dose dependent (Fig 5A). Importantly, overexpression of ILEI-FLAG by Dox induction in the hematopoietic compartment did not alter the body weight of the mice even after over 200 days of Dox in their diet (Fig 5B). Flow cytometry analysis of the spleen with focus on myeloid leukocyte populations revealed that ILEI overexpression led to reduced number of eosinophils (Fig 5C), indicating that the reduced number of CD11b+ myeloid cells observed in the R26-ILEIind ubiquitous overexpression model might be a consequence of intrinsic ILEI-linked mechanisms in the hematopoietic system. This had, however, no influence on parameters related to anemia, the hemoglobin levels, RBC count, and HCT percentage of Vav-ILEIind mice kept temporarily on Dox diet were similar compared to littermates used as Dox treatment and genetic controls (Fig 5D). These data demonstrate that no intrinsic mechanisms of erythroid development were dysregulated by ILEI and that hematopoietic ILEI hyperfunction despite of an effect on myeloid lineage composition did not contribute to the morbidity and mortality of the mice.

Fig 5. ILEI overexpression in hematopoietic cells does not influence overall fitness and red blood cell parameters.

Fig 5

(A) Western blot analysis of freshly isolated bone marrow from dual-transgenic Vav-ILEIind mice kept on Dox supplemented either in the drinking water or in the food at different concentrations for 3 days. FLAG and ILEI antibodies were used for transgene detection, vinculin as loading control. (B) Mean body weight ± SEM of ILEIind and Vav-ILEIind mice kept on normal or Dox diet. (C) Mean percentage of live cells ± SEM of listed myeloid subpopulations in the spleen of ILEIind and Vav-ILEIind mice kept on normal or Dox diet. (D) Mean HGB concentration, RBC count and HCT percentage ± SEM of Vav-ILEIind mice kept temporarily on Dox diet from week 3 to 10 of age and measured weekly until week 12 of age compared to littermates used as Dox treatment and genetic controls. Statistical significance was determined by student’s t-test and is marked with asterisks (*p<0.05; **p<0.01; ***p<0.001).

Ubiquitous ILEI overexpression leads to reduced iron levels in the serum and in hepatocytes, and to liver damage

Next, to further understand the anemic effects of ubiquitous ILEI overexpression we analyzed the iron levels of the transgenic mice. The serum iron levels were significantly reduced in R26-ILEIind mice on Dox compared to control R26-ILEIind mice without Dox (p<0.01) and ILEIind mice on Dox (p<0.05) (Fig 6A). As the liver is the main organ involved in iron metabolism, we also undertook analysis for serum markers of liver function: ALT and AST. ALT (Fig 6B) was significantly increased in R26-ILEIind mice on Dox drinking water relative to the mice without Dox (p<0.05) and genetic control mice (ILEIind, p<0.01), and AST levels were also increased (Fig 6C) in R26-ILEIind mice on Dox relative to the mice without Dox (p<0.05). Though mice overexpressing ILEI already had a lower body weight, the liver-to-body weight ratio (Fig 6D) was additionally significantly lower in these animals compared to Dox treatment and genetic controls (p<0.001 and p<0.05, respectively), also represented on macroscopic images in Fig 6E. Furthermore, all these observations had an exaggerated penetrance in moribund ILEI overexpressing mice compared to mice of the same group that survived to experimental endpoint (dark vs. white data points in Figs 3B and 6B–6D), indicating that beside an overall retarded growth and lower fitness Dox-induced R26-ILEIind mice had specific problems in liver function and size maintenance that were linked to death. There were also some evident differences in histological analysis of liver sections. H&E staining suggested that hepatocytes were smaller with small nuclei and the liver lost its typical hexagonal lobular structure with intermitting central vein and portal venule arrangements in mice expressing ILEI-FLAG compared to the two control groups (Fig 6F). This was alongside significantly decreased numbers of cells positive for Ki67, a proliferation marker (p<0.05 vs. Dox control, Fig 6G and 6H), and increased rates of activated caspase 3 positive cells, indicative of apoptosis, in R26-ILEIind mice on Dox (p<0.001 vs. Dox control and p<0.0001 vs. genetic control, Fig 6I and 6J). Prussian blue staining showed lower number of hepatocytes with ferric iron (Fe3+) storage in the liver tissue of ILEI-FLAG expressing mice compared to the controls (p< 0.05, Fig 6K and 6L). Therefore, these results suggest that the reduced serum iron levels observed upon ILEI overexpression were linked with reduced iron content in the liver. In addition, the liver showed malfunction and reduced hepatoprotection, which in combination with the reduced iron levels may be indicative of reduced iron uptake and heme production by hepatocytes, contributing to anemia.

Fig 6. Ubiquitous ILEI overexpression leads to reduced iron levels in the serum and in hepatocytes and to liver dysfunction.

Fig 6

(A) Mean serum iron levels ± SEM of R26-ILEIind mice kept on Dox drinking water measured one week before death compared to age-matched mice used as Dox treatment and genetic controls. (B-C) Mean (B) ALT and (C) AST levels ± SEM of R26-ILEIind mice kept on Dox drinking water measured one week before death (dark data points) or at experiment endpoint (white data points) compared to age-matched mice used as Dox treatment and genetic controls. (D) Mean liver-to-body weight ratio ± SEM of R26-ILEIind mice kept on Dox drinking water measured one week before death (dark data points) or at experiment endpoint (white data points) compared to age-matched mice used as Dox treatment and genetic controls. (E-F) Representative (E) macroscopic images and (F) H&E stained thin sections of the liver of moribund R26-ILEIind mice kept on Dox drinking water compared to age-matched mice used as Dox treatment and genetic controls. (G-L) Mean percentage of (G) Ki67 and (I) activated Caspase 3 (actCasp3) positive cells ± SEM with representative images of corresponding (H) Ki67 and (J) actCasp3 immunohistochemistry of the liver of moribund R26-ILEIind mice kept on Dox drinking water compared to age-matched mice used as Dox treatment and genetic controls. (K,L) Iron content of the liver shown as (K) Fe3+ positive hepatocytes per mm2 ± SEM and (L) representative images of moribund R26-ILEIind mice kept on Dox drinking water compared to age-matched mice used as Dox treatment and genetic controls. Scale bars, (E) 1 cm, (F,H,J,L) 100μm. Statistical significance was determined by one-way ANOVA (A,B,C,D,G,I) or student’s t-tests (K) and marked with asterisks (*p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).

Mice with induced ubiquitous ILEI overexpression develop liver fibrosis

The apparent liver dysfunction showed signs indicative of liver fibrosis, thus, fibrosis markers were analyzed histologically as shown in Fig 7. Immunohistochemistry with anti-fibronectin antibodies showed that R26-ILEIind mice on Dox had significantly higher rates of stained areas compared to R26-ILEIind mice without Dox and ILEIind mice who received Dox (both p<0.0001, Fig 7A and 7C). Similar results were found with S100A4 immunohistochemistry (both p<0.05, Fig 7B and 7D). Furthermore, R26-ILEIind mice on Dox were highly enriched for areas of disordered and fibrous collagen deposition compared to the two control groups that had an overall weak collagen signal with the exception of accumulations around blood vessels as visualized by Sirius Picric Red staining (Fig 7E). Similarly, R26-ILEIind mice on Dox showed a more dense appearance of bile ducts and an upregulation of E-cadherin expression in these structures that was not evident in the two control groups of mice by immunohistochemistry with anti-E-cadherin antibodies (Fig 7F). These data support the view that liver fibrosis results from overexpression of ILEI in the liver. This could in turn lead to severe liver dysfunction and eventually to death.

Fig 7. Mice with induced ubiquitous ILEI overexpression develop liver fibrosis.

Fig 7

(A-D) Representative images of (A) Fibronectin and (B) S100A4 immunohistochemistry and their quantifications shown as mean percentage of (C) Fibronectin and (D) S100A4 positive area ± SEM of the liver of moribund R26-ILEIind mice kept on Dox drinking water compared to age-matched mice used as Dox treatment and genetic controls. (E-F) Increased collagen deposition and bile duct density shown by (E) Sirius Picric Red staining and (F) E-cadherin immunohistochemistry. Scale bar, 200μm; scale bar for inlets, 50μm. (B,C) Statistical significance was determined by one-way ANOVA and marked with asterisks (*p<0.05; ****p<0.0001).

Discussion

In this study we present the development of a mouse model to study the effects of elevated ILEI expression in vivo. We demonstrate the successful generation and establishment of a transgenic mouse line with inducible ubiquitous ILEI overexpression (R26-ILEIind mice). The R26-ILEIind mice have the M2rtTA inserted into the Rosa26 locus and the Dox-dependent tetO:ILEI-FLAG construct in the 3’UTR region of the Col1a1 locus, resulting in ubiquitous inducible, Rosa26 promoter-driven expression of M2rtTA which recognizes the Tet-operon (tetO) only in the presence of Dox to control the expression of the Fam3c/ILEI-FLAG transgene. The Dox induction showed dose dependency. Mice with the Dox-dependent ILEI-FLAG construct at the Col1a1 locus without the M2rtTA inserted at the Rosa26 locus were used as genetic control and showed no measurable expression of ILEI-FLAG upon Dox administration. The Tet-On system used in this model also allows tissue specific expression. In this case we used the rtTA under the control of the Vav1 promoter, which is exclusively active in hematopoietic cells [39], to limit the overexpression of ILEI to those cell types. Therefore, this novel Tet-On inducible Fam3c/ILEI transgenic mouse strain will provide a useful model for tissue specific and timely controlled overexpression of ILEI and by this represents a versatile tool to model the effect of elevated ILEI expression in diverse disease conditions, including cancer.

As ILEI is vital in the EMT process, overexpression of ILEI early in development could not be excluded from interfering with mouse viability in a drastic if not lethal manner. Therefore, we started Dox induction at weaning age. ILEI-FLAG was expressed at high levels in most tissues examined, suggesting that the transgenic mice were ubiquitously expressing the fusion protein. Characterization of the mouse phenotype on induction of ILEI overexpression showed a shorter lifespan, reduced body weight, microcytic hypochromic anemia, and liver dysfunction and fibrosis compared to control mice. However, the anemia was quickly reversible at a young age upon withdrawal of ILEI induction. While this is the first study to develop an inducible ubiquitous overexpression of ILEI, previous mouse models have been established that involved full knock-out of ILEI expression or neuron specific overexpression of ILEI. Those studies were investigating specific effects of ILEI in influencing bone mineral density or Alzheimer’s disease. They found very small effects on the whole organism [1820]). Interestingly, one of the Fam3c knock-out studies, alongside their main interest in bone morphology, found male mice showing hematological changes not evident in females [19]. As our study was not intended to investigate a role of ILEI in bone we did not undertake a detailed analysis of the bones of the transgenic mice. This will obviously be an interesting study in the future alongside a detailed examination of other important organs such as the heart, skeletal muscles, and brain. However, looking at the hematological changes of the previous study in detail reveals that male mice with Fam3c knock-out had elevated numbers of polymorphonuclear neutrophils and reduced amounts of lymphocytes, RBC counts were higher with slightly smaller corpuscular volume of the cells, total hemoglobin did not differ significantly [19]. This reasonably supports the hematological changes and the role of ILEI in anemia found in our study and suggests that in future it might be relevant to examine gender differences.

To understand the cause of the anemia in mice ubiquitously expressing ILEI we utilized the ability of the Tet-On system to be switched to tissue specific expression. Hematopoietic cells undergo proliferation and differentiation events to produce mature blood cells throughout the lifespan of the individual [40]. Overexpression of the Fam3c/ILEI transgene in hematopoietic cells alone did not render mice anemic or lower overall fitness. These results suggest that ILEI overexpression was not dysregulating intrinsic mechanisms of erythroid development and that overdriving of hematopoietic ILEI function did not contribute to the mortality of the mice. Ubiquitous induction of ILEI overexpression reduced serum iron levels, so this was indicative of a malfunction in iron absorption, cellular uptake, or heme production. Iron deficiency anemia normally presents as microcytic hypochromic anemia where RBCs have a smaller mean corpuscular volume (MCV) than normal. In accordance with this, mice in this study also had reduced MCV, indicative of microcytic hypochromic anemia caused by iron deficiency.

As the liver plays the major role in controlling the systemic iron balance within the body [41], we next examined the effect of ILEI overexpression in the liver. Our results show that the liver was severely affected with increased ALT and AST levels, reduced liver size, increased apoptosis, and reduced ferric iron content of hepatocytes. This dysfunction was easily visualized in histological sections having a fibrotic appearance with increased areas of fibronectin and S100A4 compared to control sections, and obvious collagen bundles. Meanwhile detection of E-cadherin showed areas of intense staining around bile ducts that was not seen in the control samples. This is reminiscent of the enhanced hepatic E-cadherin production in mouse bile duct ligation models of cholestatic fibrosis [42, 43]. These data indicate that high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis, which leads to liver dysfunction, disturbed iron metabolism and eventually to death. This result needs careful consideration by researchers interested in the role ILEI plays in metabolic diseases. In particular, diabetes and non-alcoholic liver disease models suggest overexpression of ILEI as potential therapy to overcome glucose intolerance, insulin resistance and liver steatosis linked to reduced ILEI expression in the liver [44, 45]. However, our data suggest that therapeutic approaches involving overexpression of ILEI would have to be very carefully controlled because they might result in liver fibrosis. Thus, our model can serve as a suitable tool to investigate long term effects of the described short-term and in vitro observations, and our data indicate that ILEI overexpression might be linked to unexpected risks in liver function.

The livers of mice overexpressing ILEI have many similar characteristics to liver cirrhosis in human disease, with evidence of liver dysfunction alongside increased levels of fibrosis markers and fibrous and disordered collagen. Cirrhosis is an advanced stage of liver fibrosis accompanied by distortion of the hepatic vasculature, which results in compromised liver function [46]. Late-stage cirrhosis often results in smaller liver volume, as seen in this study, and this may be related to poor prognosis of patients with hepatocellular disease [47]. On the other hand, cirrhosis usually develops after a sustained period of inflammation [46]. In this study there was no evidence of a period of inflammation or hepatitis from ILEI overexpression. Similarly, while cirrhosis increases the risk of liver cancer [46], we did not observe any sign of malignant transformation in the liver. Overall, ILEI overexpression did not cause spontaneous tumor formation in any organs as analyzed by autopsy of moribund and endpoint mice.

This study raises the issue of low levels of iron in the liver and liver dysfunction. However, most investigations of iron and liver damage highlight the role of high levels of iron being a cause of oxidative damage resulting in liver fibrosis [4850]. In this situation with low iron levels in the liver, blood analysis indicated few effects on WBC counts or major lymphoid and myeloid subpopulations suggesting that ILEI overexpression was not stimulating an immune response. It seems more likely from our results that overexpression of ILEI in the liver results in liver dysfunction and as a consequence iron metabolism is disturbed. This is supported by the results from specific overexpression of ILEI in hematopoietic cells. However, it is also possible that low iron levels are primary and iron deficiency is initially not tightly linked to liver dysfunction. Detailed analysis of this likely complicated mechanism is beyond the initial characterization of the mouse model presented here. Therefore, the mechanisms behind iron malabsorption indicative in this mouse model need to be investigated in more detail in the future. Those studies will involve uncoupling the effects of overexpression of ILEI in the liver from other systemic effects on iron levels and will require detailed analysis of various tissue specific ILEI expression profiles, including the intestine as main place of iron absorption. It is also important to fully understand the microcytic hypochromic anemia discovered in the mice, including analysis of the factors involved, such as transferrin and ferritin, to give a more precise understanding of its nature.

Conclusion

The study describes the generation and initial characterization of a transgenic mouse with inducible expression of ILEI. Ubiquitous induction of ILEI overexpression at weaning age resulted in mice with a shortened lifespan, reduced body weight, and microcytic hypochromic anemia. These characteristics seemed to be related to high levels of ILEI expression in the liver, which showed dysfunction and fibrosis. Therefore, high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis. This could in turn lead to liver dysfunction, disturbed iron metabolism, and eventually to death.

Supporting information

S1 File

(ZIP)

Acknowledgments

We thank Nicole Leitner and Simone Müller for their involvement in the generation of the transgenic mouse line and in the speed congenic procedure, Gabriella Litos for assisting at genotyping, Boris Kovacic for experimental guidance in Flow Cytometry and Gergely Szakacs, Robert Eferl and Lukas Kenner for constructive discussions and critical reading of the manuscript. The mouse strain VavrtTA was kindly provided by Michael Bader and Ross Dickins. CRediT (Contributor Roles Taxonomy) was used to define contributor roles of the authors. Melanie Colegrave from Molecular Cell Research provided writing assistance.

Data Availability

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

Funding Statement

The study was supported by a grant of the Austrian Science Fund (FWF) ZK-81B (DG and CM), the Overseas Scholarship of the University of the Punjab, Pakistan (BM) and the Fellinger Krebsforschung (AC). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Zhu Y, Xu G, Patel A, McLaughlin MM, Silverman C, Knecht K, et al. Cloning, expression, and initial characterization of a novel cytokine-like gene family. Genomics. 2002;80(2):144–50. doi: 10.1006/geno.2002.6816 [DOI] [PubMed] [Google Scholar]
  • 2.Jechlinger M, Grunert S, Tamir IH, Janda E, Ludemann S, Waerner T, et al. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003;22(46):7155–69. doi: 10.1038/sj.onc.1206887 [DOI] [PubMed] [Google Scholar]
  • 3.Jansson AM, Csiszar A, Maier J, Nystrom AC, Ax E, Johansson P, et al. The interleukin-like epithelial-mesenchymal transition inducer ILEI exhibits a non-interleukin-like fold and is active as a domain-swapped dimer. The Journal of biological chemistry. 2017;292(37):15501–11. doi: 10.1074/jbc.M117.782904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Johansson P, Bernstrom J, Gorman T, Oster L, Backstrom S, Schweikart F, et al. FAM3B PANDER and FAM3C ILEI represent a distinct class of signaling molecules with a non-cytokine-like fold. Structure. 2013;21(2):306–13. doi: 10.1016/j.str.2012.12.009 [DOI] [PubMed] [Google Scholar]
  • 5.Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, et al. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. The FEBS journal. 2017;284(20):3484–505. doi: 10.1111/febs.14207 [DOI] [PubMed] [Google Scholar]
  • 6.Csiszar A, Kutay B, Wirth S, Schmidt U, Macho-Maschler S, Schreiber M, et al. Interleukin-like epithelial-to-mesenchymal transition inducer activity is controlled by proteolytic processing and plasminogen inverted question mark urokinase plasminogen activator receptor system-regulated secretion during breast cancer progression. Breast Cancer Res. 2014;16(5):433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schmidt U, Heller G, Timelthaler G, Heffeter P, Somodi Z, Schweifer N, et al. The FAM3C locus that encodes interleukin-like EMT inducer (ILEI) is frequently co-amplified in MET-amplified cancers and contributes to invasiveness. Journal of experimental & clinical cancer research: CR. 2021;40(1):69. doi: 10.1186/s13046-021-01862-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Liao TT, Yang MH. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11(7):792–804. doi: 10.1002/1878-0261.12096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lahsnig C, Mikula M, Petz M, Zulehner G, Schneller D, van Zijl F, et al. ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression. Oncogene. 2009;28(5):638–50. doi: 10.1038/onc.2008.418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Song Q, Sheng W, Zhang X, Jiao S, Li F. ILEI drives epithelial to mesenchymal transition and metastatic progression in the lung cancer cell line A549. Tumour Biol. 2014;35(2):1377–82. doi: 10.1007/s13277-013-1188-y [DOI] [PubMed] [Google Scholar]
  • 11.Waerner T, Alacakaptan M, Tamir I, Oberauer R, Gal A, Brabletz T, et al. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006;10(3):227–39. doi: 10.1016/j.ccr.2006.07.020 [DOI] [PubMed] [Google Scholar]
  • 12.Gao ZH, Lu C, Wang ZN, Song YX, Zhu JL, Gao P, et al. ILEI: a novel marker for epithelial-mesenchymal transition and poor prognosis in colorectal cancer. Histopathology. 2014;65(4):527–38. doi: 10.1111/his.12435 [DOI] [PubMed] [Google Scholar]
  • 13.Watanabe N, Nakano M, Mitsuishi Y, Hara N, Mano T, Iwata A, et al. Transcriptional downregulation of FAM3C/ILEI in the Alzheimer’s brain. Hum Mol Genet. 2021;31(1):122–32. doi: 10.1093/hmg/ddab226 [DOI] [PubMed] [Google Scholar]
  • 14.Zhou J, Jiang H, Jiang H, Fan Y, Zhang J, Ma X, et al. The ILEI/LIFR complex induces EMT via the Akt and ERK pathways in renal interstitial fibrosis. J Transl Med. 2022;20(1):54. doi: 10.1186/s12967-022-03265-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cho YS, Go MJ, Kim YJ, Heo JY, Oh JH, Ban HJ, et al. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009;41(5):527–34. doi: 10.1038/ng.357 [DOI] [PubMed] [Google Scholar]
  • 16.Song Q, Song J, Li C, Wang Y, Qi L, Wang H. Genetic variants in the FAM3C gene are associated with lipid traits in Chinese children. Pediatr Res. 2021;89(3):673–8. doi: 10.1038/s41390-020-0897-3 [DOI] [PubMed] [Google Scholar]
  • 17.Yang A, Liu S, Yang X, Guo Z, Li J, Li X, et al. Uncovering Novel Prognostic Factors of Sudden Sensorineural Hearing Loss by Whole-Genome Sequencing of Cell-Free DNA. J Int Adv Otol. 2022;18(6):459–64. doi: 10.5152/iao.2022.21493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zheng HF, Tobias JH, Duncan E, Evans DM, Eriksson J, Paternoster L, et al. WNT16 influences bone mineral density, cortical bone thickness, bone strength, and osteoporotic fracture risk. PLoS Genet. 2012;8(7):e1002745. doi: 10.1371/journal.pgen.1002745 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Määttä JA, Bendre A, Laanti M, Büki KG, Rantakari P, Tervola P, et al. Fam3c modulates osteogenic cell differentiation and affects bone volume and cortical bone mineral density. Bonekey Rep. 2016;5:787. doi: 10.1038/bonekey.2016.14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hasegawa H, Liu L, Tooyama I, Murayama S, Nishimura M. The FAM3 superfamily member ILEI ameliorates Alzheimer’s disease-like pathology by destabilizing the penultimate amyloid-beta precursor. Nat Commun. 2014;5:3917. [DOI] [PubMed] [Google Scholar]
  • 21.Baron U, Gossen M, Bujard H. Tetracycline-controlled transcription in eukaryotes: novel transactivators with graded transactivation potential. Nucleic acids research. 1997;25(14):2723–9. doi: 10.1093/nar/25.14.2723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Das AT, Tenenbaum L, Berkhout B. Tet-On Systems For Doxycycline-inducible Gene Expression. Curr Gene Ther. 2016;16(3):156–67. doi: 10.2174/1566523216666160524144041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Giménez E, Lavado A, Giraldo P, Cozar P, Jeffery G, Montoliu L. A transgenic mouse model with inducible Tyrosinase gene expression using the tetracycline (Tet-on) system allows regulated rescue of abnormal chiasmatic projections found in albinism. Pigment Cell Res. 2004;17(4):363–70. doi: 10.1111/j.1600-0749.2004.00158.x [DOI] [PubMed] [Google Scholar]
  • 24.Song X, Guo Y, Duo S, Che J, Wu C, Ochiya T, et al. A mouse model of inducible liver injury caused by tet-on regulated urokinase for studies of hepatocyte transplantation. Am J Pathol. 2009;175(5):1975–83. doi: 10.2353/ajpath.2009.090349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lottmann H, Vanselow J, Hessabi B, Walther R. The Tet-On system in transgenic mice: inhibition of the mouse pdx-1 gene activity by antisense RNA expression in pancreatic beta-cells. J Mol Med (Berl). 2001;79(5–6):321–8. doi: 10.1007/s001090100229 [DOI] [PubMed] [Google Scholar]
  • 26.Yao F, Walker PD, MacKenzie RG. A Tet-on system for DRD1-expressing cells. PLoS One. 2013;8(8):e72681. doi: 10.1371/journal.pone.0072681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.rodents Fwgorogfhmo, rabbits, Mahler Convenor M, Berard M, Feinstein R, Gallagher A, et al. FELASA recommendations for the health monitoring of mouse, rat, hamster, guinea pig and rabbit colonies in breeding and experimental units. Laboratory animals. 2014;48(3):178–92. doi: 10.1177/0023677213516312 [DOI] [PubMed] [Google Scholar]
  • 28.Beard C, Hochedlinger K, Plath K, Wutz A, Jaenisch R. Efficient method to generate single-copy transgenic mice by site-specific integration in embryonic stem cells. Genesis. 2006;44(1):23–8. doi: 10.1002/gene.20180 [DOI] [PubMed] [Google Scholar]
  • 29.Hochedlinger K, Yamada Y, Beard C, Jaenisch R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell. 2005;121(3):465–77. doi: 10.1016/j.cell.2005.02.018 [DOI] [PubMed] [Google Scholar]
  • 30.Leitner NR, Lassnig C, Rom R, Heider S, Bago-Horvath Z, Eferl R, et al. Inducible, dose-adjustable and time-restricted reconstitution of STAT1 deficiency in vivo. PLoS One. 2014;9(1):e86608. doi: 10.1371/journal.pone.0086608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Teppner I, Aigner B, Schreiner E, Müller M, Windisch M. Polymorphic microsatellite markers in the outbred CFW and ICR stocks for the generation of speed congenic mice on C57BL/6 background. Laboratory animals. 2004;38(4):406–12. doi: 10.1258/0023677041958882 [DOI] [PubMed] [Google Scholar]
  • 32.Takiguchi M, Dow LE, Prier JE, Carmichael CL, Kile BT, Turner SJ, et al. Variability of inducible expression across the hematopoietic system of tetracycline transactivator transgenic mice. PLoS One. 2013;8(1):e54009. doi: 10.1371/journal.pone.0054009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Leitner NR, Strobl B, Bokor M, Painz R, Kolbe T, Rülicke T, et al. A time- and dose-dependent STAT1 expression system. BMC Biotechnol. 2006;6:48. doi: 10.1186/1472-6750-6-48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Luna L. Manual of Histologic Staining Methods of the AFIP. 3rd Edition ed: McGraw-Hill, NY; 1968. [Google Scholar]
  • 35.Sheehan D, Hrapchak B. Theory and practice of Histotechnology. 2nd Edition ed: Battelle Press, Ohio; 1980. [Google Scholar]
  • 36.Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: Open source software for digital pathology image analysis. Scientific reports. 2017;7(1):16878. doi: 10.1038/s41598-017-17204-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Novoszel P, Drobits B, Holcmann M, Fernandes CS, Tschismarov R, Derdak S, et al. The AP-1 transcription factors c-Jun and JunB are essential for CD8alpha conventional dendritic cell identity. Cell death and differentiation. 2021;28(8):2404–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chen L, Wang J, Liu J, Wang H, Hillyer CD, Blanc L, et al. Dynamic changes in murine erythropoiesis from birth to adulthood: implications for the study of murine models of anemia. Blood Adv. 2021;5(1):16–25. doi: 10.1182/bloodadvances.2020003632 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ilan L, Katzav S. Human Vav1 expression in hematopoietic and cancer cell lines is regulated by c-Myb and by CpG methylation. PLoS One. 2012;7(1):e29939. doi: 10.1371/journal.pone.0029939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tavian M, Biasch K, Sinka L, Vallet J, Péault B. Embryonic origin of human hematopoiesis. Int J Dev Biol. 2010;54(6–7):1061–5. doi: 10.1387/ijdb.103097mt [DOI] [PubMed] [Google Scholar]
  • 41.Meynard D, Babitt JL, Lin HY. The liver: conductor of systemic iron balance. Blood. 2014;123(2):168–76. doi: 10.1182/blood-2013-06-427757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Van Campenhout R, Crespo Yanguas S, Cooreman A, Gijbels E, Leroy K, Vilas-Boas V, et al. Increased Expression of Adherens Junction Components in Mouse Liver following Bile Duct Ligation. Biomolecules. 2019;9(10). doi: 10.3390/biom9100636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jirouskova M, Nepomucka K, Oyman-Eyrilmez G, Kalendova A, Havelkova H, Sarnova L, et al. Plectin controls biliary tree architecture and stability in cholestasis. J Hepatol. 2018;68(5):1006–17. doi: 10.1016/j.jhep.2017.12.011 [DOI] [PubMed] [Google Scholar]
  • 44.Chen Z, Ding L, Yang W, Wang J, Chen L, Chang Y, et al. Hepatic Activation of the FAM3C-HSF1-CaM Pathway Attenuates Hyperglycemia of Obese Diabetic Mice. Diabetes. 2017;66(5):1185–97. doi: 10.2337/db16-0993 [DOI] [PubMed] [Google Scholar]
  • 45.Zhang X, Yang W, Wang J, Meng Y, Guan Y, Yang J. FAM3 gene family: A promising therapeutical target for NAFLD and type 2 diabetes. Metabolism. 2018;81:71–82. doi: 10.1016/j.metabol.2017.12.001 [DOI] [PubMed] [Google Scholar]
  • 46.Ginès P, Krag A, Abraldes JG, Solà E, Fabrellas N, Kamath PS. Liver cirrhosis. Lancet. 2021;398(10308):1359–1376. doi: 10.1016/S0140-6736(21)01374-X [DOI] [PubMed] [Google Scholar]
  • 47.Hagan MT, Sayuk GS, Lisker-Melman M, Korenblat K M, Kerr TA, Chapman WC, et al. Liver volume in the cirrhotic patient: does size matter?. Dig Dis Sci. 2014;59(4):886–891. doi: 10.1007/s10620-014-3038-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fujikura Y, Krijt J, Povýšil C, Mělková Z, Přikryl P, Vokurka M, et al. Iron Overload Causes Alterations of E-Cadherin in the Liver. Folia Biol (Praha). 2016;62(3):95–102. [DOI] [PubMed] [Google Scholar]
  • 49.Britton LJ, Subramaniam VN, Crawford DH. Iron and non-alcoholic fatty liver disease. World J Gastroenterol. 2016;22(36):8112–22. doi: 10.3748/wjg.v22.i36.8112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mehta KJ, Farnaud SJ, Sharp PA. Iron and liver fibrosis: Mechanistic and clinical aspects. World J Gastroenterol. 2019;25(5):521–38. doi: 10.3748/wjg.v25.i5.521 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Michael Klymkowsky

16 Feb 2023

PONE-D-23-00773Inducible overexpression of a FAM3C/ILEI transgene has pleiotropic effects with shortened life span, liver fibrosis and anemia in micePLOS ONE

Dear Dr. csiszar,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

 Please consider and address, point by point, reviewer #2's objections and recommendations.  While major new studies (recommended by the reviwer) may not be necessary, clearly indicate why that is so. 

Please submit your revised manuscript by Apr 02 2023 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Michael Klymkowsky, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Thank you for stating the following financial disclosure: 

"The study was supported by a grant of the Austrian Science Fund (FWF) ZK-81B (DG and CM), the Overseas Scholarship of the University of the Punjab, Pakistan (BM) and the Fellinger Krebsforschung (AC)."

Please state what role the funders took in the study.  If the funders had no role, please state: "The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript." If this statement is not correct you must amend it as needed. 

Please include this amended Role of Funder statement in your cover letter; we will change the online submission form on your behalf.

3. Thank you for stating the following in the Acknowledgments Section of your manuscript: 

"The study was supported by a grant of the Austrian Science Fund (FWF) ZK-81B (DG and CM), the Overseas Scholarship of the University of the Punjab, Pakistan (BM) and the Fellinger Krebsforschung (AC)."

We note that you have provided funding information that is not currently declared in your Funding Statement. However, funding information should not appear in the Acknowledgments section or other areas of your manuscript. We will only publish funding information present in the Funding Statement section of the online submission form. 

Please remove any funding-related text from the manuscript and let us know how you would like to update your Funding Statement. Currently, your Funding Statement reads as follows: 

"The study was supported by a grant of the Austrian Science Fund (FWF) ZK-81B (DG and CM), the Overseas Scholarship of the University of the Punjab, Pakistan (BM) and the Fellinger Krebsforschung (AC)."

Please include your amended statements within your cover letter; we will change the online submission form on your behalf.

4. In your Data Availability statement, you have not specified where the minimal data set underlying the results described in your manuscript can be found. PLOS defines a study's minimal data set as the underlying data used to reach the conclusions drawn in the manuscript and any additional data required to replicate the reported study findings in their entirety. All PLOS journals require that the minimal data set be made fully available. For more information about our data policy, please see http://journals.plos.org/plosone/s/data-availability.

Upon re-submitting your revised manuscript, please upload your study’s minimal underlying data set as either Supporting Information files or to a stable, public repository and include the relevant URLs, DOIs, or accession numbers within your revised cover letter. For a list of acceptable repositories, please see http://journals.plos.org/plosone/s/data-availability#loc-recommended-repositories. Any potentially identifying patient information must be fully anonymized.

Important: If there are ethical or legal restrictions to sharing your data publicly, please explain these restrictions in detail. Please see our guidelines for more information on what we consider unacceptable restrictions to publicly sharing data: http://journals.plos.org/plosone/s/data-availability#loc-unacceptable-data-access-restrictions. Note that it is not acceptable for the authors to be the sole named individuals responsible for ensuring data access.

We will update your Data Availability statement to reflect the information you provide in your cover letter.

5. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels. 

  

In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions.

6. 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.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The manuscript PONE_D_23-00773 by Schmidt et al. is very interesting indicating tissue-specific effects for ILEI overexpression. The manuscript is well written and the data presented support the conclusions.

Reviewer #2: In this study, the authors generated and characterized a Tet-ON inducible ILEI-transgenic mouse strain. They found that mice with induced ubiquitous ILEI-FLAG overexpression at weaning age exhibited a shortened lifespan, reduced body weight, iron-deficiency anemia, and liver fibrosis with dysfunction. Reduced iron levels were observed in the serum and in hepatocytes.

This study is an initial and incomplete characterization of Tet-ON inducible ILEI-transgenic mice. This reviewer has the following comments on this manuscript.

1) Using quantitative methods such as immunoblotting and ELISA, the authors should show expression levels of ILEI in major organs including the brain, heart, lung, liver, spleen, kidney, skeletal muscles, and bone marrow of induced ILEI-Tg mice used in this study as comparing with those of control and wild-type mice.

2) Previously, the author's group reported that homodimers of ILEI are exclusively active (Krai M, 2017; Jansson AM, 2017). They should examine whether overexpressed ILEI-FLAG in their mice is dimerized.

3) Iron deficiency anemia is usually microcytic hypochromic but not normocytic hypochromic. The authors should show MCV and MCH (or MCHC) values. In addition, effectiveness to iron administration can indicate whether anemia of ILEI-overexpressed mice is caused by iron deficiency.

4) Transferrin plays a critical role in transfer and metabolism of iron. The authors should measure serum transferrin levels of mice with induced ILEI overexpression.

5) Livers of induced ILEI Tg mice were reduced in size, showed increased apoptosis, reduced cellular iron content, and had a fibrotic phenotype. Which finding appeared firstly? Were these findings reversible upon withdrawal of ILEI induction?

6) Livers of mice with induced ILEI overexpression were somewhat reminiscent of liver cirrhosis in human. The authors should discuss about similarity and difference between these conditions.

7) Previous studies suggested that ILEI overexpression caused carcinogenesis. The authors should evaluate frequency of spontaneous cancer development after induction of ILEI overexpression.

8) In line 370 on page 17; "Figure F" ?

9) The authors refer to the same mouse line as "Fam3c transgenic" and "ILEI transgenic".

**********

6. 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

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment 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. Registration is free. 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 PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

Decision Letter 1

Michael Klymkowsky

12 May 2023

Inducible overexpression of a FAM3C/ILEI transgene has pleiotropic effects with shortened life span, liver fibrosis and anemia in mice

PONE-D-23-00773R1

Dear Dr. csiszar,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Michael Klymkowsky, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: (No Response)

**********

7. 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 #2: No

**********

Acceptance letter

Michael Klymkowsky

16 May 2023

PONE-D-23-00773R1

Inducible overexpression of a FAM3C/ILEI transgene has pleiotropic effects with shortened life span, liver fibrosis and anemia in mice

Dear Dr. Csiszar:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Michael Klymkowsky

Academic Editor

PLOS ONE


Articles from PLOS ONE are provided here courtesy of PLOS

RESOURCES