Skip to main content
Erschienen in: Respiratory Research 1/2023

Open Access 01.12.2023 | Research

TGFβ1-RCN3-TGFBR1 loop facilitates pulmonary fibrosis by orchestrating fibroblast activation

verfasst von: Mingting Wu, Zhenyan Wang, Xiaoqian Shi, Danni Zan, Hong Chen, Shuqiao Yang, Fangping Ding, Liu Yang, Pingping Tan, Runlin Z. Ma, Jing Wang, Lishuang Ma, Yingmin Ma, Jiawei Jin

Erschienen in: Respiratory Research | Ausgabe 1/2023

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Idiopathic pulmonary fibrosis (IPF) bears high mortality due to unclear pathogenesis and limited therapeutic options. Therefore, identifying novel regulators is required to develop alternative therapeutic strategies.

Methods

The lung fibroblasts from IPF patients and Reticulocalbin 3 (RCN3) fibroblast-selective knockdown mouse model were used to determine the importance of Rcn3 in IPF; the epigenetic analysis and protein interaction assays, including BioID, were used for mechanistic studies.

Results

Reticulocalbin 3 (RCN3) upregulation is associated with the fibrotic activation of lung fibroblasts from IPF patients and Rcn3 overexpression blunts the antifibrotic effects of pirfenidone and nintedanib. Moreover, repressing Rcn3 expression in mouse fibroblasts ameliorates bleomycin-induced lung fibrosis and pulmonary dysfunction in vivo. Mechanistically, RCN3 promotes fibroblast activation by maintaining persistent activation of TGFβ1 signalling via the TGFβ1-RCN3-TGFBR1 positive feedback loop, in which RCN3 upregulated by TGFβ1 exposure detains EZH2 (an epigenetic methyltransferase) in the cytoplasm through RCN3-EZH2 interaction, leading to the release of the EZH2-H3K27me3 epigenetic repression of TGFBR1 and the persistent expression of TGFBR1.

Conclusions

These findings introduce a novel regulating mechanism of TGFβ1 signalling in fibroblasts and uncover a critical role of the RCN3-mediated loop in lung fibrosis. RCN3 upregulation may cause resistance to IPF treatment and targeting RCN3 could be a novel approach to ameliorate pulmonary fibrosis.

Graphical Abstract

Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12931-023-02533-z.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
IPF
Idiopathic pulmonary fibrosis
ILD
Intestinal lung diseases
Rcn3
Reticulocalbin 3
TGF β
Transforming growth factor β
FGF
Fibroblast growth factor
PDGF
Platelet-derived growth factor
TGFBR1/2
TGF β1 type I/II receptor
ER
Endoplasmic reticulum
UPR
Unfolded protein response
AEC II
Type II alveolar epithelial cells
ALI
Acute lung injury
DHLF
Disease human lung fibroblast
NHLF
Normal human lung fibroblast
MLF
Mouse lung fibroblast
H3K27me3
Tri-methylation of histone H3 at Lys27
qPCR
Quantitative real-time PCR
CoIP
Co-immunoprecipitation
ChIP
Chromatin immunoprecipitation
EDU
5-Ethynyl-2-deoxyuridine
FVC
Forced vital capacity
Re
Expiratory resistance
Ri
Inspiratory resistance
Cdyn
Dynamic compliance
CCND1
Cyclin D1
PCNA
Proliferating cell nuclear antigen
4-PBA
4-Phenylbutyrate
CCK8
Cell Counting Kit-8
Col1a1/2
Collagen I type 1/II
α-SMA
Actin alpha 2
PRC2
Poly comb repressive complex 2
EZH2
Epigenetic methyltransferase in poly comb repressive complex 2
rhEZH2
Recombinant human EZH2 protein

Background

Idiopathic pulmonary fibrosis (IPF) is the most aggressive fibrotic intestinal lung disease (ILD) with irreversible destruction of lung architecture, and it bears high mortality due to unclear pathogenesis and limited therapeutic options [1, 2]. The inevitable progression of IPF is linked to its complex pathogenesis, which involves with different cell types, signalling pathways, and structural changes [3]. Despite past extensive studies, the mortality of IPF is barely improved [1, 2]. IPF is believed to arise from aberrant fibrotic remodelling caused by excessive fibroblast activation, which is associated with the deregulation of signalling initiated by fibrotic factors, such as transforming growth factor-β (TGF-β), fibroblast growth factor (FGF) and platelet-derived growth factor (PDGF) [1]. Recently, two approved antifibrotic drugs (pirfenidone and nintedanib) targeting these signalling pathways can slow the progression and relieve symptoms in IPF patients. However, they still exhibit inconsistent efficacy and fail to reduce mortality [36]. Therefore, identifying novel regulators is required to develop alternative therapeutic strategies.
Repeated injuries to the alveolar epithelium are primarily cause of IPF, resulting in the release of profibrotic mediators. The most predominant mediator is TGFβ1 which induces fibroblast activation through both canonical and non-canonical pathways, including smad, AKT, ERK and STAT3 pathways [79]. TGFβ1 binds to the type II receptor (TGFBR2) and then recruits the type I receptor (TGFBR1) to the heterotetrametric complex, activating TGFBR1 which then phosphorylates and activates downstream effectors [10]. The regulation of TGFBRs through transcription, protein stability and trafficking are essential for maintaining TGF-β signalling balance [11, 12]. Increasing evidence indicates that the epigenetic regulation of TGFBRs also plays a critical role in the dysregulation of TGF-β signalling in cancers [1316]. However, the regulatory mechanism in the upstream of TGFβ1 signalling during pulmonary fibrosis remains unclear.
Reticulocalbin 3 (RCN3), as an ER chaperone protein localised to the secretory pathway, contains a signal sequence at its N-terminus, six EF-hands, and an HDEL ER-retention signal at its C-terminus [1719]. Our earlier study showed that Rcn3 deletion caused a failure of alveolar epithelial cells (AECII) maturation with the impaired secretion of surfactant proteins, leading to disrupted perinatal lung development [19]. In addition, our recent studies indicated the critical roles of Rcn3 in regulating AEC apoptosis and inflammatory response, involving in lung fibrosis and acute lung injury (ALI) [20, 21]. Interestingly, we found a manifest upregulation of Rcn3 in the fibrotic area of the lung from the bleomycin-induced fibrosis mouse model, suggesting a potential role of Rcn3 in fibroblast during pulmonary fibrosis [21]. Increasing studies also showed that Rcn3 was associated with collagen production and collagen fibrillogenesis in cardiac fibrosis and postnatal tendon development [22, 23]. Given these findings, we hypothesised that RCN3 could be a critical regulator in modulating fibroblast activation during pulmonary fibrosis.
Herein, we found that RCN3 was remarkably upregulated in fibroblasts from both patient and mouse fibrotic lungs, whereas RCN3 knockdown diminished the activation of disease human lung fibroblasts from IPF patients (DHLF-IPF). On the other hand, Rcn3 overexpression induced fibroblast activation and significantly blunted the anti-fibrotic effects of both pirfenidone and nintedanib. Furthermore, fibroblast-selective Rcn3 in vivo knockdown ameliorated bleomycin-induced lung fibrosis and pulmonary dysfunction. The mechanistic study revealed that RCN3 promoted fibrogenesis by enhancing TGFβ1 signalling via the TGFβ1-RCN3-TGFBR1 positive feedback loop. This loop epigenetically maintains TGFBR1 transcription by releasing the EZH2-H3K27me3 epigenetic repression via the Rcn3-EZH2 interaction, leading to persistent activation of TGFβ1 signalling. These findings suggest a determinate role of the Rcn3-mediated loop in lung fibrosis and introduce a novel regulating mechanism for TGFβ1 signalling. Elevated RCN3 level may contribute to resistance to IPF treatment, so targeting Rcn3 could be a novel therapeutic approach for pulmonary fibrosis.

Methods

Mouse models

All animal procedures were approved by the Animal Care and Ethics Committee of the Institute of Genetics and Developmental Biology, Chinese Academy of Sciences and were performed in accordance with the Guide for the Care and Use of Laboratory Animals of the Chinese Academy of Sciences. By crossing the fibroblast-specific protein (FSP)1-Cre (Jackson Lab) and Rcn3flox/flox mice (generated in our lab [21]), the fibroblast-selective Rcn3 deletion mice (CKO, FSP1-Cre/Rcn3f/f) were generated and the littermates (Rcn3flox/flox) served as controls. The intratracheal administration of bleomycin (Sigma-Aldrich) was performed in 8-week-old mice at the dose of 0.08 U/kg in 50 μl saline (25 μl × 2), and sterile saline was instilled at the same procedure as a control [21].

Human tissues

Human fibrotic lung samples were obtained by diagnostic surgical lung biopsies from patients diagnosed as IPF at Beijing Chao-Yang Hospital. Normal human lung samples were collected from surgical lung resections from patients with lung cancer at the same hospital. All studies involving human lung tissues were approved by the Ethics Committee of Beijing Chao-Yang Hospital (2021-KE-295) and signed informed consents for research use of samples were obtained from all subjects. All methods were carried out in accordance with relevant guidelines and regulations.

Measurement of airway resistance and compliance

The airway resistance and compliance were measured using AniRes 2005 system (Beijing Biolab, Beijing, China) as manufacturer’s manual and previously described [24]. Briefly, mice were anesthetised by intraperitoneal injection of avertin (250 mg/kg) followed by tracheal exposure and cannulation with a Y-type cannula (2.5 mm inner diameter). After the mice were then placed inside a plethysmographic chamber at the supine position, one branch of the tracheal Y-type cannula was connected to a ventilator at outside of the chamber and the other was connected to the pressure detection channel. The mice were mechanically ventilated at a breath rate of 90 breaths/minute at a tidal volume equal to 10 ml/kg with maintaining peak respiratory pressure at 10–16 cm of water. The display pulmonary function parameters were calculated, including forced vital capacity (FVC), dynamic compliance (Cdyn), inspiratory resistance (Ri), and expiratory resistance (Re).

Histological and immunohistochemical staining

The lung tissues fixed with 4% paraformaldehyde (PFA) were embedded in paraffin and sliced into 5 µm thickness for Haematoxylin & eosin (HE) and Masson’s Trichrome staining (ScyTek Laboratories). The fibrotic score for mouse fibrotic lung was scored as semi-quantitative (0–4 score), as previously described [21]. Through evaluating 10 sequential and nonoverlapping fields on Masson’s trichrome-stained sections for each mouse, the fibrotic condition was scored: 0, normal lung architecture; 1, increased thickness of some (≤ 50%) of interalveolar septa; 2, thickening of > 50% of interalveolar septa without formation of fibrotic patches; 3, thickening of the interalveolar septa with formation of isolated fibrotic patches; 4, formation of multiple fibrotic patches with total or subtotal distortion of parenchymal architecture.
For immunohistochemical staining, paraffin-embedded tissues were performed through standard methods and incubated with primary antibodies: anti-αSMA (1:2000, Abcam: ab124964) and anti-Rcn3 (1:4000, sigma: HPA050402).

The isolation of mouse alveolar epithelial cells (AECs) and lung fibroblasts

Mouse alveolar epithelial cells were prepared as described in our previous published study [21]. Briefly, the trachea in exsanguinated mice were cannulated and 2 ml of 5 U/ml dispase II (Sigma, D4693) in DPBS was rapidly instilled into the lung, followed by a slow infusion of 0.5 ml agarose (45 °C 1% w/v, low melting). The lung was taken out and incubated in dispase for 45 min (25 °C), and then lung tissue was gently teased from the bronchi in HEPES-buffered DMEM containing 100 U/ml DNase I. The cell suspension was filtered through progressively smaller cell strainers (100 and 40 μm) and metal gauze (25 μm), and cells were placed on CD 45/32 coated culture dished for 1 h. Afterwards, fibroblasts were enriched by adherence for 2 h on cell culture dishes loosely. The adhered fibroblast and unadhered AECII cells were collected to check Rcn3 expression.
The mouse lung fibroblasts were prepared as below. Mouse lungs perfused by DPBS were harvested, minced and digested in HBSS buffer containing 0.1% type I collagenase, 2.5 U/ml dispase II and 100 U/ml DNase I for 1 h at 37 °C water bath. The digestion was mixed with DMEM (10% FBS), followed by pipetting to separate cells from tissues. The cells were plated into cell culture dishes and incubated at 37 °C for 24 h, followed by washing and cultured for further experiments.

Human lung fibroblasts (HLF)

Human lung fibroblasts were purchased from merchandise. Normal primary human lung fibroblasts (NHLF) and Disease human lung fibroblasts derived from IPF patients (DHLF-IPF) were purchased from Lonza (American, CC-2512 and CC-7231). Cells were cultured in Fibroblast Medium (CM-H011, Procell) and DMEM (sigma, D6429).
Human lung fibroblasts were prepared from human lung tissues. Fresh lung tissue was cut into small pieces and washed with DPBS three or more times till clean blood. The lung tissues were further cut into smaller fragments (~ 1 mm3), pooled in DMEM (10% FBS) and plated into cell culture dishes. After fibroblasts proliferated from these tissues, the tissue pieces were washed out. The fibroblast finally grew to 80% confluence and serially passed in the same medium.

Cell fraction isolation

Isolated different cell protein components (cytoplasmic, nuclear and membrane proteins) using a protein extraction kit (KeyGen BioTEC, KGBSP002) following the product manufactures instruction. The Histone protein was extracted using EpiQuik™ Total Histone Extraction Kit (EPIGENTEC, OP-0006) as manufacturer’s manual. Briefly, resuspend cells in diluted pre-lysis buffer on ice for 10 min with gentle shaking. After the supernatant was removed by centrifugation at 12000×g, the pellet was mixed with lysis buffer for 30 min on ice. The supernatant containing histone was collected by spinning at 12,000 rpm and mixed with 0.3 volume of balance-DTT buffer immediately for the following experiments.

RCN3 knockdown and over-expression

NHLF were planted about 2 × 105 cells per well in six wells plates with 10% FBS DMEM overnight, and cells were transfected by 50 uM of siRNA-RCN3 (targeting sequence shown below) or siRNA-scramble through lipo3000 (Invitrogen, L3000015) in Opti-MEM (Gibco, 51985034). 36 h post transfection, cells were cultured in FBS free medium overnight and then treated with or without TGFβ1 (sigma, GF346) at 5 ng/ml for 24 h (for qPCR) or 48 h (for Western blot). As for Rcn3 overexpression, the lentiviral particles containing human Rcn3 (NM_020650.3) or empty vector were produced in 293T cells using lentiviral shuttle vector FUGW. 24 h after infection, cells were subjected to further experiments. The siRNA sequence for human Rcn3:
  • RNA oligo sequences 21nt guide UUCAGCAAUCACGAUGUCCCG;
  • 21nt passenger GGACAUCGUGAUUGCUGAAAC.

TGFβ1 stimulation and inhibition in fibroblasts

HLF and MLF were cultured in serum-free medium and treated with TGFβ1 (sigma, GF346) at 5 ng/ml or other concentrations as indicated. For inhibition, the cells were co-treated by inhibitors 4-PBA (MCE, HY-A0281) or LY2109761 (sigma, HY-12075-TMP) with TGFβ1, followed by downstream experiments. For assaying signalling activation in response to TGFβ1 stimulation, after 5 ng/ml of TGFβ1 treatment, cell lysates were collected at 30 min for p-smad3 and at 12 h for p-AKT and p-stat3.

Bioi-ID assay and protein–protein interactions

The BioID-Rcn3 fusion protein was constructed by inserting Rcn3 (without signal peptide) into the C-terminal of BioID2 and the signal peptide fragment of Rcn3 into the N-terminal of BioID2; BioID-only was constructed by inserting Rcn3 signal peptide segment in the N-terminal of BioID2. The overexpression of BioID-Rcn3 and BioID-only by inserting into lentiviral shuttle vector FUGW and the lentiviral were produced to infect normal human lung fibroblast (NHLF). After the expressions of these proteins were validated, the BioID assay was performed according to the previous detailed protocol [25]. Briefly, cells reached approximately 70–80% confluency in 100 mm dishes and were infected by lentivirus for 24 h, followed by exposure to TGFβ1 (5 ng/ml) and biotin (50 µM, sigma, B4639) for another 24 h. The cell lysate was triturated on ice by sonication twice and subjected into streptavidin-affinity purification process to enrich biotinylated proteins proximal to Rcn3. The enriched proteins were identified by LC–MS/MS. By eliminating the common proteins in BioID-Rcn3, BioID-only and Biotin controls, specific potential interaction proteins with Rcn3 were identified.

Quantitative real-time PCR (qPCR)

RNA was isolated from cells or tissues with Trizol (Invitrogen, 15596018) according to the standard protocols, and the cDNA was synthesised using PrimeSTAR® Max DNA Polymerase (Takara, R045A). qPCR assays on these cDNA were performed using TB Green® Premix Ex Taq™ II (Takara, RR820) and data were normalised to house-keeping gene (RPL19 content for mouse samples and GAPDH for human samples) and analysed by the 2−△△Ct method relative to saline/vehicle treated control groups. The data are derived from at least three independent experiments performed in triplicate and the primers used in qPCR see Additional file 2: Table S1.

Biolayer interferometry (BLI)

Rcn3–EZH2 direct interaction determined by biolayer interferometry technology by OctetRED system (ForteBio) using human recombinant proteins (hrRcn3: Abcam, ab276552; EZH2: Origene, TP302054), following manufacturer’s protocol. In brief, rhEZH2 was biotinylated by Biotinylation Kit (Thermo Scientific, 21329) followed by desalting using desalt columns (thermos scientific, 89889). rhRcn3 with his-tag (5 µg/ml) were immobilised with NTA biosensors (ForteBio, 18-5001) followed by washing with binding buffer three times. The biotinylated rhEZH2 was serially diluted in binding buffer (262.3, 131.1, 65.56 and 32.78 µg/ml) and the Rcn3-EZH2 association and dissociation were detected by OctetRED system for 10 min at 25 °C. The baseline signal drift was controlled by monitoring immobilized Rcn3 without EZH2. OctetRED analysis software was used to analyse the data.

Immunoblotting assay

30–50 µg of extracted protein containing protease inhibitor and phosphatase inhibitor was boiled in loading buffer and was separated by 8–12% SDS-PAGE, flowed by blotting onto 0.2 µm PVDF membrane. The membrane was then blocked with 5% skim-milk in Tris-buffered saline, 0.1% Tween 20 (TBS-T) for 90 min at room temperature and in turn incubated with diluent primary antibody as recommended in the instructions overnight at 4 °C. After washing with TBS-T, the membrane was incubated with horseradish peroxidase (HRP)-conjugated secondary antibody at room temperature and signals were visualised by enhanced chemiluminescence (GE Healthcare). The band-densitometry data were measured by using the Image J software. The antibodies were listed in Additional file 2: Table S2.

Hydroxyproline assay

The lung tissue samples (right middle lobes) were transferred to 6 M HCl to final 100 mg/ml, followed by incubation for 20 h at 95 °C. After centrifugation at 13000×g, the supernatant was diluted with water to 4 M HCL at the ratio of 1 volume sample to 0.5 volume water and then 35 µl of the sample was used to quantitate collagen content by using a collagen assay kit (QuickZyme, QZBtotcol1-TMP) according to manufacturer’s manual.

Cell proliferation assay

CCK8 assay

After transferring siRNA for 24 h, cells were seeded in a 96-well cell culture plate at a density of 5 × 103 for 24 h followed by 12 h of serum starvation. Cells were then exposed to TGFβ1 (5 ng/ml), FGF (25 ng/ml, R&D Systems, 3718-FB-010) or 10% FBS. After 24 h post treatment, cell proliferation was detected by adding 10 µl of CCK8 solution for each well and incubating for 2 h in 37 °C (CCK8 assay kit, LABLEAD, CK001). The final absorbance at 450 nm was used to calculate cellular proliferation rate.

EDU proliferation assay

Cell proliferation was measured by EdU Cell Proliferation Kit with Alexa Fluor 488 (Beyotime, C0071S) according to the instruction. Briefly, after cells were treated with TGFβ1 (5 ng/ml) for 24 h, pre-heat EDU working solution (10 μM) was added into each well followed by 2 h of incubation at 37 °C. Cells were fixed in 4% PFA and permeabilizated with 0.3% Triton X-100 in DPBS. Cells were incubated with click addictive solution and stained with Hoechst 33342. The images of EDU were photographed by Leica fluorescence microscopy and calculated by Image J software.

Cell migration assay

Transwell assay

siRNA transfected cells were reseeded with serum free medium at the density of 1 × 104 into the upper chamber, while the lower chamber was equipped with DMEM containing 10% FBS. After 24 h, cells in the upper chamber were fixed by 4% PFA and stained by 0.1% Crystal Violet Ammonium Oxalate Solution (solarbio, G1063). Each chamber was photographed for 9 views under the inverted microscope and calculated the number of invaded cells through Image J software.

Scratch assay

Until the seeded transfected cells reached 90% on the 6-well plate, scrapped two straight lines on each well and washed with DPBS to remove detached cells. Cells were incubated with or without TGFβ1 (5 ng/ml) in 1% FBS DMEM for 48 h photographed and recorded the images at 0 and 48 h. Compared each image and obtained the distance of each scratch closure by Image J software.

High-throughput RNA sequencing and data analyses

RNA-Sequencing analyses of the total RNA from NHLF cells with Rcn3-siRNA (Si-T) and Ctl-siRNA (SiC-T) after TGFβ1 exposure (5 ng/ml) for 24 h (n = 4 per group) by OE Biotech (Shanghai, China). In brief, total RNA was extracted using the mirVana miRNA Isolation Kit (Ambion) and The RNA Integrity Number (RIN) ≥ 7 were subjected to libraries construction using TruSeq Stranded mRNA LTSample Prep Kit (Illumina, San Diego, CA, USA). RNA sequencing was performed using the Illumina sequencing platform (HiSeqTM 2500 or Illumina HiSeq X Ten) and 125 bp/150 bp paired-end reads were generated. Quantification of mRNA transcript abundance was performed by normalised expression values as fragments per kb per million reads using cufflinks, and the read counts of each gene were obtained by htseq-count. A total of 180 differentially-expressed genes (DEGs) with the foldchange (FC) > 1.2 and FDR < 0.05 were identified between Si-T versus SiC-T by using DEseq. Hierarchical clustering presented the gene expression profiles separated based on Rcn3 knockdown. The Gene Ontology analysis on these DEGs prioritised by strength (Gene-ratio: the ratio of number of deferentially expressed genes between the number of genes associated the GO term) and FDR p values corrected by BH procedure (< 0.05); The most affected processes were presented. BP: Biological process, CC: Cellular component, MF: Molecular function.

Protein stability assay

Cycloheximide pulse chase assay was used for examining TGFBR1 protein stability. Control and Rcn3-knockdown NHLF were treated with 100 μg/ml cycloheximide (MedChemExpress, HY-12320) to inhibit protein synthesis. The immunoblotting was used to check TGFBR1 protein levels. The ratio of (TGFBR1/β-tubulin) was determined at different time points and expressed as percentages relative to the ratio at time 0. β-tubulin was used as a control protein.

Co-immunoprecipitation (Co-IP)

NHLF cells were used for endogenous Co-IP experiments and cells at 48 h after infection by lentivirus containing overexpression constructors as indicated for exogenous Co-IP. Cells were lysed in triton lysis Buffer (TLB) by sonication on ice and the protein lysis was collected by centrifugation at 12000×g, 30 min, 4 °C. Following, 1 mg of protein lysis was mixed with 30 µl protein A/G and incubated 1.5 h on a rotator at 4 °C to eliminate nonspecific binding proteins. After centrifugation at 1000×g, the supernatant was incubated with 1 µg anti-flag antibody, anti-EZH2 (Santa Cruz, sc-13725s) and mouse IgG (negative control, (Beyotime, A7016) on the rotator at 4 °C overnight. Following 50 µl protein A/G was used to pull down antibody-conjugated proteins, then washing beads with TLB for more than three times. Finally, beads were resuspended in equal volume of 2 × loading and boiled at 95 °C for 10 min to separate the protein and beads. Samples were then analysed by immunoblotting.

Chromatin immunoprecipitation (ChIP) assay

ChIP assay was conducted by using the ChIP assay kit (Beyotime, P2078) according to the manufacture-instruction. Briefly, cells grown to 80–90% conference in 100 mm dishes were washed in cold DPBS twice and cross-linked by 1% formaldehyde for 10 min at 37 °C. The cross-linking fixation was stopped by adding 1/10 volume of glycine solution (10×) and incubated for 5 min at room temperature. Cells were washed with pre-cold DPBS containing 1 mM PMSF three times and lysed in SDS lysis buffer on ice for 10 min followed by sonication. The supernatant was collected for immunoprecipitation with anti-H3K27me3 antibody (CST, 9733s) and IgG (Beyotime, A7028) control. The cross-linked DNA was purified with DNA Purification Kit (Beyotime, D0033) and examined by qPCR using specific primers which were listed in Additional file 2: Table S3.

Cellular immunofluorescent staining (IF)

The NHLF cells were seeded and grown on glass coverslips coated with poly-l-lysine and fixed in 4% paraformaldehyde prior to being incubated with antibodies specific to Rcn3 (1:200, sigma, HPA050402) and α-actinin EZH2 (1:100, Santa Cruz, sc-13725s). The secondary anti-rabbit antibody conjugated to Alexa Fluor 488 (1:1000, ZSGB-BIO, ZF0511) and Alexa Fluor 594 (1:1000, ZSGB-BIO, ZF0513) was applied respectively to detect the immune signals, respectively. Fluorescence images were viewed with a Zeiss LSM980 confocal microscope.

Statistical analysis

All data are presented as the mean ± SD. To compare continuous variables, the Kolmogorov–Smirnov test was used to test the normality of the data. Statistical comparisons between two groups were performed by the two-tailed Student’s t-test for normally distributed continuous variables or the Mann–Whitney U test for non-normally distributed continuous variables. Statistical comparisons of three or more groups were performed by using the one-way ANOVA (Bonferroni post hoc test) for normally distributed variables, one-way Kruskal–Wallis tests (Dunn’s post hoc test) for non-normally distributed variables, or two-way ANOVA (Tukey post hoc test). The statical significance was set at p < 0.05. All analyses were performed using GraphPad Prism 8.0 software.

Results

The upregulation of RCN3 in lung fibroblasts is critical for pulmonary fibrosis

First, we examined RCN3 expression in lung fibroblast (LF) from IPF patients. Immunohistochemical (IHC) on the continues sections from patient lungs with intestinal fibrosis showed prominent RCN3 induction in α-SMA positive area, whereas Rcn3 positive staining in normal lung tissues was primarily in the corner of alveoli (suggestive of AECIIs) in line with our previous publication [21] (Fig. 1A). Additionally, we explored the published single-cell RNA-seq on IPF patients (Sci. Adv. 6, eaba1983, 2020) and found a remarkable upregulation of Rcn3 in myofibroblasts rather than other cell types [26] (Additional file 1: Fig. S1A). Moreover, RCN3, along with α-SMA and collagen-I, was upregulated in disease lung fibroblasts derived from an IPF patient (DHLF-IPF, from merchandise Lonza) versus normal human lung fibroblast (NHLF, from merchandise Lonza), which was consistent with observations in fibroblasts isolated from patient lungs (Fig. 1B). Furthermore, RCN3 knockdown remarkably diminished the expressions of α-SMA and collagen-I in DHLF-IPF (Fig. 1C).
Next, we examined Rcn3 expression in mouse fibrotic lung fibroblasts. Intratracheal bleomycin instillation caused prominent pulmonary intestinal fibrosis (with isolated fibrotic foci) as indicated by Masson’s trichrome and IHC staining of α-SMA (Fig. 1D, E, Additional file 1: Fig. S1B). In the primer lung fibroblasts from these mice, Rcn3 was remarkably upregulated along with α-SMA (Fig. 1D). IHC staining on the continuous lung sections further confirmed that the striking increase of Rcn3 was localised in α-SMA positive area (Fig. 1E and Additional file 1: Fig. S1B). These findings on the patient and the mouse model suggested a critical role of Rcn3 upregulation in lung fibroblast during IPF.

The selective repression of Rcn3 expression in fibroblast ameliorates bleomycin-induced lung fibrosis

The role of Rcn3 in LF during lung fibrosis was further investigated by using the mouse model with fibroblast-selective Rcn3 repression (CKO), which were generated by crossing the fibroblast-specific protein (FSP)1-Cre and Rcn3flox/flox mice. The CKO mice (FSP1-Cre/Rcn3f/f) developed normally and their littermates (Rcn3f/f) served as controls (Additional file 1: Fig. S2). The immunoblotting assay showed that Rcn3 expression was remarkably repressed in LFs, but not in ACEs, isolated from the same CKO lung (Fig. 2A, Additional file 1: Fig. S2C). The ICH of Rcn3 further confirmed the declined Rcn3 staining inside the lung fibrotic foci from intratracheal bleomycin-treated CKO mice, whereases the Rcn3 staining in corner of alveoli (ACEs) and around the blood vessels (endothelial cells) was intact in these mice (Fig. 2B), which was consistent with a previous study that Fsp-1 is relatively fibroblast specific in the lung [27]. Since the previous study reports that Fsp1 may also express in immunocytes and hematopoietic cells [28], we checked the inflammatory condition in the lung to evaluate the potential nonspecific effects of FSP1-Cre mediated Rcn3 deletion. The CKO mice exhibit normal gross morphologies including body weight, motor activity and responses to painful stimuli, normal red blood cell (RBC) count, and comparable levels of IL-1β, TNFα and MCP1 mRNAs in the lung (Additional file 1: Fig. S2A, B).
In response to bleomycin instillation, CKO mice showed attenuated bleomycin-lung fibrosis versus controls, as indicated by constricted fibrotic foci formation and parenchymal distortion, which was in line with decreases in the semiquantitative fibrotic score, hydroxyproline content and the expressions of α-SAM, collagen I and Cyclin D1 (Fig. 2C–F, Additional file 1: Fig. S2D). Consistently, the bleomycin-induced decline of pulmonary function was significantly ameliorated in CKO mice, evidenced by limited changes of forced vital capacity (FVC), expiratory resistance (Re), inspiratory resistance (Ri) and dynamic compliance (Cdyn) (Fig. 2G). Additionally, although there have been reports of the non-specific effects of FSP1-Cre in immunocytes [28], CKO and control lungs showed comparable levels of IL-1β, TNFα and MCP1 at acute/fibrotic phases (3, 7 and 14 days) after bleomycin exposure, suggesting attenuated fibrosis in CKO lung was not due to decreased inflammation (Additional file 1: Fig. S3). Furthermore, the isolated LFs from CKO lungs exhibited significantly decreased Rcn3, α-SAM, collagen-I and Cyclin-D1 expressions, but not TGFβ1 (Fig. 2H, Additional file 1: Fig. S5). These observations suggested that Rcn3 induction in LFs was critical for bleomycin-induced fibrosis.

RCN3 induction by TGFβ1 in an ER stress-dependent manner is essential for LF functional activation and Rcn3 upregulation blunted antifibrotic effects of pirfenidone and nintedanib

To investigate whether RCN3 induction during lung fibrosis was associated with TGFβ1 signalling, NHLF and mouse lung fibroblast (MLF) were treated with TGFβ1 at different concentrations. TGFβ1-exposure at 2 and 5 ng/ml doses enhanced the expressions of RCN3, α-SAM, collagen-I and ER stress marker GRP78 (Fig. 3A, B). Interestingly, ER stress inhibitor 4-phenylbutyrate (4-PBA) strikingly attenuated the inductions of GRP78 and RCN3 in response to TGFβ1-exposure, suggesting an ER stress-dependent RCN3 induction (Fig. 3C and Additional file 1: Fig. S4A). Furthermore, RCN3 knockdown by siRNA suppressed the activation of fibroblast induced by TGFβ1, but not FGF, as demonstrated by diminished expressions of α-SAM, collagen-I and Cyclin-D1 (Fig. 3D, E, Additional file 1: Fig. S4B). CCK8 and EdU incorporation assays consistently showed that RCN3 deficiency significantly blunted NHLF proliferation in response to TGFβ1 and FBS (10%) rather than FGF (Fig. 4A, B). RCN3 deficiency also caused a notable depression of TGFβ1-induced migration, as indicated by transwell and scratch assays (Fig. 4C, D). These results indicated the importance of Rcn3 upregulation for activating LF upon TGFβ1-exposure.
Next, we investigated whether RCN3 would exhibit the difference in the anti-profibrotic effects of Nintedanib and Pirfenidone, because Nintedanib inhibits multiple receptor tyrosine kinases (FGFR and VEGFR) and Pirfenidone primarily inhibits the TGFβ1 signalling [29]. TGFβ1-induced upregulations of RCN3 were blunted by pirfenidone rather than nintedanib, although both of two drugs significantly inhibited TGFβ1-induced upregulations of α-SMA, Col1a1 and Col1a2 (Fig. 4E and Additional file 1: Fig. S5). Interestingly, Rcn3 overexpression-caused NHLF activation was significantly bunted by both drugs. Furthermore, it is worth nothing that RCN3 overexpression significantly dampened the anti-fibrotic effects of both drugs against TGFβ1-induced LF activation, suggesting that Rcn3 upregulation could restrain their therapeutic effects for IPF (Fig. 4F).

RCN3 was involved in the activations of both canonical and non-canonical TGFβ1 signalling in lung fibroblasts

To further dissect the regulatory mechanism of RCN3 in TGFβ1 signalling, TGFβ1-treated NHLF bearing RCN3-siRNA (si-T) and control-siRNA (siC-T) were subjected to RNA-Sequencing analysis. A total of 181 differentially-expressed genes (51 upregulation, 129 downregulation genes) were identified (foldchange (FC) > 1.2, FDR < 0.05), including genes involved in fibrosis and cell cycle, suggesting that Rcn3 deficiency could affect both canonical and non-canonical TGFβ1 signalling (Fig. 5A). Most RCN3 deficiency-affected processes enriched by Gene Ontology (GO) analysis were primarily associated with fibrosis including ECM organisation, wound healing and ECM structure constitutes (Fig. 5B). Of note, TGFBR1 was found in the down-regulation panel, suggesting a potential role of RCN3 in regulating the initiation of TGFβ1 signalling. This hypothesis was further supported by the observation that RCN3 knockdown markedly blunted both canonical and non-canonical TGFβ1 downstream signalling pathways (Fig. 5C). Moreover, the TGFBRs inhibitor, LY2109761, completely suppressed the activation of smad3 caused by RCN3 overexpression (Fig. 5D).

RCN3 facilitates TGFβ1 signalling by the transcriptional increase of TGFBR1, establishing a positive feedback loop

We further investigated the regulatory mechanism of RCN3 in moderating TGFβ1-TGFBR1 signalling. We found that primary lung fibroblasts from CKO mice had significantly reduced expression of TGFBR1, but not TGFβ1, along with diminished Rcn3 expression (Additional file 1: Fig. S2C). The significantly altered mRNA level of TGFBR1, but not TGFBR2, was also observed in NHLF with either RCN3 knockdown or overexpression (Fig. 6A). Consistently, RCN3 knockdown significantly diminished the protein level of TGFBR1 in both NHLF and DHLF-IPF. NHLF bearing RCN3 knockdown also were unable to maintain the level of TGFBR1 upon TGFβ1 stimulation (Fig. 6B, C, Additional file 1: Fig. S6). The reduction of TGFBR1 protein was observed in both cytoplasm and membrane fractions (Fig. 6D), which was in line with blunted TGFβ1 signalling. Additionally, RCN3 overexpression significantly upregulated TGFBR1 expression, accompanied with the inductions of fibrotic markers (Fig. 6E). Furthermore, protein stability by cycloheximide chase assay showed an unchanged protein degradation rate of TGFBR1 in RCN3 deficiency NHLF (Fig. 6F), suggesting a transcriptional regulation of TGFBR1 by RCN3. Additionally, RCN3 deficiency failed to change TGFBR2 transcription, validated by using three sets of qPCR primers targeting different regions in either Rcn3-siRNA or shRNA-targeted NHLFs (Additional file 1: Fig. S6).
Taken together, the TGFβ1-RCN3-TGFBR1 positive-feedback loop was proposed based on the evidences: TGFβ1-exposure upregulated RCN3 and TGFBR1; RCN3 deficiency diminished TGFβ1-induced TGFBR1 upregulation; RCN3 overexpression upregulated TGFBR1 leading to TGFβ1 signalling activation, which was repressed by TGFBR inhibitor LY2109761.

The TGFβ1 exposure promoted RCN3-EZH2 interaction which restrained nuclear EZH2 level and decreased EZH2/H3K27me3 enrichment at the TGFBR1 promoter region

To further determine the regulatory mechanism of Rcn3 in TGFBR1 transcription, the BioID labelling was used to screen physiological interaction proteins of RCN3 in living NHLF cells in response to TGFβ1-exposure. After eliminating the common proteins in BioID-RCN3, BioID-only and Biotin controls, a total of 12 proteins were identified as specific interaction proteins of Rcn3 upon TGFβ1 stimulation (Fig. 7A). One of these proteins is Enhancer of zeste homolog 2 (EZH2) which is an epigenetic methyltransferase in polycomb repressive complex 2 (PRC2) and has been shown to suppress TGFBR1 transcription by catalysing tri-methylation of histone H3 at Lys 27 (H3K27me3) at the promoter region [16]. The RCN3-EZH2 interaction was confirmed by co-immunoprecipitation (Co-IP) by using exogenously expressed Flag-RCN3 and Myc-EZH2 (Fig. 7B). The quantitative assessment of the binding affinity by the Bio-Layer Interferometry (BLI, Octet-RED system) further confirmed their direct interaction as indicated by a concentration-dependent interaction of Rcn3 with EZH2 with an estimated dissociation constant (Kd) of 1.54 µM (Fig. 7C). Furthermore, we gained insight into RCN3-EZH2 complex formation from protein–protein docking analysis (Cluspro 2.0), exhibiting several high-confidence structural models of RCN3-EZH2 complex as rigid bodies with acceptable interaction weighted scores. The top potential models suggested that a segment of RCN3 (121–191 residues) seemed located in a pock-like structure of EZH2 (Additional file 1: Fig. S7).
EZH2 functions as a transcriptional suppressor through H3K27me3 within the nucleus, while immunofluorescence assay on Hela cells showed some co-localization of EZH2 and RCN3 within the cytoplasm (Additional file 1: Fig. S8). Furthermore, RCN3 knockdown significantly increased the level of nuclear EZH2 in HLF, while RCN3 overexpression decreased the level of nuclear EZH2 (Fig. 7D). Immunofluorescence on HLF showed more cytoplasmatic EZH2 than that in Hela cells; either TGFβ1 exposure or Rcn3 overexpression promoted the colocalization of Rcn3-EZH2 in the cytoplasm (Fig. 7E). In line with the change of nuclear-EZH2 level, RCN3 knockdown and overexpression markedly enhanced and diminished H3K27me3 level, respectively (Fig. 7F). Furthermore, the ChIP assay consistently indicated that RCN3 deficiency caused dramatically increased EZH2/H3K27me3 enrichment at the TGFBR1 promoter region (Fig. 7G, H). Therefore, RCN3-EZH2 interaction in cytoplasm likely plays a vital role in releasing EZH2/H3K27me3 epigenetic repression of TGFBR1 to maintain the TGFBR1 level during the receptor turnover upon TGFβ1 stimulation. The TGFβ1-RCN3/EZH2/H3K27me3-TGFBR1 is a kind of dynamic turnover modification for TGFβ1 signalling.

Discussion

IPF is the most aggressive fibrotic ILD and lacks effective therapy due to unclear pathogenesis, so the discovery of novel regulators could shed light on developing alternative therapeutic strategies [30, 31]. Herein, we discovered a critical role of RCN3 in orchestrating fibroblast function during pulmonary fibrosis, as evidenced by key findings: (1) RCN3 was notably upregulated in fibroblasts from both patient and mouse fibrotic lungs, whereas RCN3 knockdown diminished activation of lung fibroblast from IPF patients. (2) The repression of Rcn3 in fibroblast ameliorated bleomycin-induced pulmonary fibrosis and dysfunction with suppressed LF activation. (3) the in vitro TGFβ1-exposure significantly upregulated RCN3 in an ER stress-dependent manner, while RCN3 knockdown diminished TGFβ1-induced fibrotic activation. Consistently, RCN3 overexpression promoted fibroblast activation and suppressed the effects of both pirfenidone and nintedanib against TGFβ1-induced fibroblast activation. (4) RCN3 facilitated fibroblasts activation by enhancing TGFβ1 signalling through transcriptional upregulation of TGFBR1, which is associated with reduced EZH2/H3K27me3 enrichment at TGFBR1 promoter. (5) The physiological RCN3-EZH2 interaction released EZH2/H3K27me3 epigenetic repression of TGFBR1 upon TGFβ1 stimulation, suggesting a dynamic turnover modification. Taken together, these results depict a positive-feedback loop TGFβ1-RCN3-TGFBR1 as a critical mechanism in the pathogenesis of pulmonary fibrosis (shown as a schematic illustration in Fig. 8): upon injury-repair activation, TGFβ1 stimulation enhances RCN3 expression in interstitial lung fibroblast and such induction of RCN3 releases the EZH2/H3K27me3-dependent repression of TGFBR1 via RCN3-EZH2 interaction, leading to enhanced TGFBR1 expression and then persistent activation of TGFβ1 signalling.

The RCN3-mediated positive feedback loop of TGFβ1 in lung fibroblast is likely a determinant for the imbalance of the pulmonary injury-repair process

The predominant cause of IPF is repeated alveolar epithelium injuries accompanied by the release of proinflammatory and profibrotic mediators, leading to an aberrant injury-repair process represented as progressive and irreversible interstitial fibrosis [2]. Our previous study using Rcn3 AECII-selective deletion mice indicates that Rcn3 in AECII is antifibrotic against bleomycin-induced pulmonary fibrosis via alleviating ER stress-induced AEC apoptosis [21]. Herein, observations on mice with fibroblast-selective Rcn3 repression indicate a profibrotic function of Rcn3 in lung fibroblast. Since the injury-repair process includes alveolar epithelial damage, repair and interstitial remodelling, Rcn3 likely facilitates the whole process by functioning in different roles in epithelium and fibroblasts. However, the aberrant fibrotic remodelling in IPF is largely due to the excessive fibroblast activation, so Rcn3 upregulation in fibroblast could be an essential trigger of shifting to imbalanced interstitial remodelling. This hypothesis was confirmed by the evidences: a striking induction of RCN3 was observed in fibroblasts from both patient and mouse fibrotic lungs; RCN3 overexpression promoted LF activation; Rcn3 deficiency markedly blunted DHLF-IPF activation and bleomycin-induced fibrosis.
The abundant evidence from human and animal studies demonstrates a central role of TGFβ1 in IPF pathogenesis [32]. Upon alveolar epithelial injury, TGFβ1 is the predominant profibrotic factor to orchestrate fibroblast functions, including differentiation, proliferation and ECM synthesis through smad and non-smad signalling pathways [33]. Herein, TGFβ1 exposure strikingly upregulates RCN3 in both human and mouse fibroblasts, while the suppression of RCN3 induction dampened both smad and non-smad signalling with depressed fibroblast activation. Interestingly, the RCN3 overexpression enhanced TGFβ1 signalling even without extra TGFβ1 exposure, which is associated with increased TRFBR1 expression rather than TGFβ1. Consistently, RCN3 deficiency transcriptionally diminished TGFBR1 expression upon TGFβ1 stimulation, suggesting that RCN3 is indispensable for maintaining persistent activation of TGFβ1 signalling via preserving membrane TGFBR1 level. These findings, for the first time, uncover a determinate role of the positive-feedback loop of TGFβ1-RCN3-TGFBR1 in lung fibrosis, as well as introduce a novel regulating mechanism of TGFβ1 signalling.
Since IPF is associated with a variety of pathways, including TGFβ1 and other tyrosine kinases pathways (PDGF, FGF and VEGF), The treatments for IPF require a multiple-targeting strategy, in which the TGFβ1 signalling is an essential pharmacological target. However, TGFβ1 acts in different functions in a variety of cell types, including epithelial, mesenchymal and inflammatory cells, so the direct inhibition of TGFβ1/TGFBR may be of inconsistent efficacy as well as cause side-effects such as increased inflammation [34]. The novel positive feedback TGFβ1-Rcn3-TGFBR1 loop plays a crucial role in the persistent activation of TGFβ1 signalling in fibroblasts. Therefore, targeting RCN3 could be an effective approach for treating IPF.

The regulation of TGFBR1 rather than TGFBR2 is involved in the Rcn3-mediated positive feedback loop

A dynamic regulation of membrane TGFBR1/2 orchestrates TGFβ1 signalling through ligand-independent and ligand-dependent recycling [35, 36]. The dynamic regulation of TGFBR1 (the executor of TGFβ1 action) is essential to modulate the amplitude and duration of the response, and an imbalanced regulation can cause excessive signalling activation. Our results indicate an Rcn3-dependent transcriptional upregulation of TGFBR1 upon TGFβ1-exposure without altering the turnover and recycling. Therefore, during the injury-repair process, Rcn3 deregulation may cause persistent TGFBR1 expression, triggering balance toward hyperactivation of TGFβ1 signalling. Consistently, recent studies have highlighted the importance of increased TGFBR1 in fibroblast activation in IPF [12, 37]. Our results also showed that Rcn3 knockdown and overexpression altered TGFBR1 expression but not TGFBR2. Therefore, the Rcn3-mediated feedback loop is not involved with TGFBR2, which is consistent with previous studies that the expression and basolateral membranes trafficking of TGFBR1 and TGFBR2 are independently regulated [12, 35, 36].
In this study, we identified the Rcn3-EZH2 interaction in repose to TGFβ1 stimulation and uncovered a novel EZH2/H3K27me3-mediated epigenetic regulation of TGFBR1 by Rcn3-EZH2 interaction. Consistently, a previous study showed that DNMT3A suppressed TGFBR1 transcription by recruiting EZH2 to TGFBR1 promoter [16]. However, cancer studies showed that EZH2 regulated TGFBR2 via the H3K27me3-dependent mechanism, by which hypoxia and YAP/TAZ signal attenuated TGFBR2 expression in prostate and lung cancer, respectively [14, 38]. Inconsistently, we did not find that Rcn3-EZH2 could alter TGFBR2 transcription, which might be due to distinct regulatory mechanisms in cancer cells and fibroblasts.
Additionally, increasing evidence shows that EZH2 can function as both H3K27me3-dependent transcriptional suppressor and H3K27me3-independent co-activator [3943]. Several regulatory mechanisms have been identified in different cellular contexts. For example, the phosphorylation of EZH2 at K307 by TAK1 in injured respiratory epithelium or at T487 by CDK in cancer triggered the switch from H3K27me3-dependent to -independent function [41, 43]. Conversely, the methylation of EZH2 at K307 enhanced the H3K27me3-dependent repression of tumour-suppressor genes, promoting breast cancer cell proliferation and invasion [40]. These various regulatory mechanisms occurring in distinct cellular contexts could explain why the Rcn3-EZH2 axis failed to regulate TGFBR2 transcription, as well as suggest the specificity of the Rcn3-mediated loop in regulating fibroblast function.

The enhanced RCN3 expression in IPF patients could lead to the resistance to pirfenidone and nintedanib treatments

Pirfenidone and nintedanib are recently approved anti-profibrotic drugs for IPF and fibrotic ILD treatments [5, 6, 29, 44]. The anti-fibrotic effect of pirfenidone is mainly attributed to the inhibitions of both TGFβ1 production and signalling activation, while nintedanib primarily functions as the receptor tyrosine kinases inhibitor for profibrotic mediators, FGFR, PDGF and VEGFR [6, 29]. In line with different phonological mechanisms, pirfenidone, rather than nintedanib, inhibited TGFβ1-induced upregulation of Rcn3, suggesting pirfenidone would be more effective against the Rcn3-mediate loop. However, Rcn3 overexpression significantly repressed their effects against TGFβ1-induced fibroblast activation, suggesting the deregulation of Rcn3 expression may restrain the therapeutic effect of both pirfenidone and nintedanib. Therefore, the level of Rcn3 in IPF patients could be a valuable clinical prognostic marker or an index guiding IPF therapeutic strategies. Future prospective clinical studies are required to evaluate the clinical significance of Rcn3.

Conclusions

Taken together, we uncover a determinate role of the TGFβ1-Rcn3-TGFBR1 loop in the pathogenesis of pulmonary fibrosis, as well as introduce a novel regulating mechanism of TGFβ1 signalling. These findings suggest that Rcn3 upregulation may cause resistance to IPF treatment and targeting Rcn3 could be a novel approach for pulmonary fibrosis treatment.

Acknowledgements

We would like to acknowledge the equipment and animal facility assistances of Beijing Institute of Respiratory Medicine and Institute of Genetics and Developmental Biology, Chinese Academy of Sciences; the clinical sample assistance of Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University; experimental material and regent assistance of Beijing Youan Hospital, Capital Medical University.

Declarations

For human samples: all studies involving human lung tissues were approved by the Ethics Committee of Beijing Chao-Yang Hospital (2021-KE-295) and signed informed consent for research use of samples were obtained from all subjects. All methods were carried out in accordance with relevant guidelines and regulations including the Code of Ethics of the World Medical Association (Declaration of Helsinki). For mouse study: all animal procedures were approved by the Animal Care and Ethics Committee of the Institute of Genetics and Developmental Biology, Chinese Academy of Sciences and were performed in accordance with the Guide for the Care and Use of Laboratory Animals of the Chinese Academy of Sciences.
Not applicable.

Competing interests

The authors have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
2.
3.
Zurück zum Zitat Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov. 2017;16:755–72.PubMedCrossRef Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov. 2017;16:755–72.PubMedCrossRef
4.
Zurück zum Zitat Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, Cottin V, Flaherty KR, Hansell DM, Inoue Y, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2071–82.PubMedCrossRef Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, Cottin V, Flaherty KR, Hansell DM, Inoue Y, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2071–82.PubMedCrossRef
5.
Zurück zum Zitat King TE, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, Gorina E, Hopkins PM, Kardatzke D, Lancaster L, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083–92.PubMedCrossRef King TE, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, Gorina E, Hopkins PM, Kardatzke D, Lancaster L, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083–92.PubMedCrossRef
6.
Zurück zum Zitat Ruwanpura SM, Thomas BJ, Bardin PG. Pirfenidone: molecular mechanisms and potential clinical applications in lung disease. Am J Respir Cell Mol Biol. 2020;62:413–22.PubMedCrossRef Ruwanpura SM, Thomas BJ, Bardin PG. Pirfenidone: molecular mechanisms and potential clinical applications in lung disease. Am J Respir Cell Mol Biol. 2020;62:413–22.PubMedCrossRef
7.
Zurück zum Zitat Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, Swigris JJ, Taniguchi H, Wells AU. Idiopathic pulmonary fibrosis. Nat Rev Dis Prim. 2017;3:17074.PubMedCrossRef Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, Swigris JJ, Taniguchi H, Wells AU. Idiopathic pulmonary fibrosis. Nat Rev Dis Prim. 2017;3:17074.PubMedCrossRef
8.
9.
Zurück zum Zitat Selman M, Pardo A. Revealing the pathogenic and aging-related mechanisms of the enigmatic idiopathic pulmonary fibrosis. An integral model. Am J Respir Crit Care Med. 2014;189:1161–72.PubMedCrossRef Selman M, Pardo A. Revealing the pathogenic and aging-related mechanisms of the enigmatic idiopathic pulmonary fibrosis. An integral model. Am J Respir Crit Care Med. 2014;189:1161–72.PubMedCrossRef
10.
Zurück zum Zitat Budi EH, Duan D, Derynck R. Transforming growth factor-β receptors and Smads: regulatory complexity and functional versatility. Trends Cell Biol. 2017;27:658–72.PubMedCrossRef Budi EH, Duan D, Derynck R. Transforming growth factor-β receptors and Smads: regulatory complexity and functional versatility. Trends Cell Biol. 2017;27:658–72.PubMedCrossRef
11.
Zurück zum Zitat Miller DSJ, Bloxham RD, Jiang M, Gori I, Saunders RE, Das D, Chakravarty P, Howell M, Hill CS. The dynamics of TGF-β signaling are dictated by receptor trafficking via the ESCRT machinery. Cell Rep. 2018;25:1841-1855.e1845.PubMedPubMedCentralCrossRef Miller DSJ, Bloxham RD, Jiang M, Gori I, Saunders RE, Das D, Chakravarty P, Howell M, Hill CS. The dynamics of TGF-β signaling are dictated by receptor trafficking via the ESCRT machinery. Cell Rep. 2018;25:1841-1855.e1845.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Lee TH, Yeh CF, Lee YT, Shih YC, Chen YT, Hung CT, You MY, Wu PC, Shentu TP, Huang RT, et al. Fibroblast-enriched endoplasmic reticulum protein TXNDC5 promotes pulmonary fibrosis by augmenting TGFbeta signaling through TGFBR1 stabilization. Nat Commun. 2020;11:4254.PubMedPubMedCentralCrossRef Lee TH, Yeh CF, Lee YT, Shih YC, Chen YT, Hung CT, You MY, Wu PC, Shentu TP, Huang RT, et al. Fibroblast-enriched endoplasmic reticulum protein TXNDC5 promotes pulmonary fibrosis by augmenting TGFbeta signaling through TGFBR1 stabilization. Nat Commun. 2020;11:4254.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Wanna-Udom S, Terashima M, Suphakhong K, Ishimura A, Takino T, Suzuki T. KDM2B is involved in the epigenetic regulation of TGF-beta-induced epithelial-mesenchymal transition in lung and pancreatic cancer cell lines. J Biol Chem. 2021;296: 100213.PubMedCrossRef Wanna-Udom S, Terashima M, Suphakhong K, Ishimura A, Takino T, Suzuki T. KDM2B is involved in the epigenetic regulation of TGF-beta-induced epithelial-mesenchymal transition in lung and pancreatic cancer cell lines. J Biol Chem. 2021;296: 100213.PubMedCrossRef
14.
Zurück zum Zitat Lo Sardo F, Pulito C, Sacconi A, Korita E, Sudol M, Strano S, Blandino G. YAP/TAZ and EZH2 synergize to impair tumor suppressor activity of TGFBR2 in non-small cell lung cancer. Cancer Lett. 2021;500:51–63.PubMedCrossRef Lo Sardo F, Pulito C, Sacconi A, Korita E, Sudol M, Strano S, Blandino G. YAP/TAZ and EZH2 synergize to impair tumor suppressor activity of TGFBR2 in non-small cell lung cancer. Cancer Lett. 2021;500:51–63.PubMedCrossRef
15.
Zurück zum Zitat Xu Y, Sun X, Zhang R, Cao T, Cai SY, Boyer JL, Zhang X, Li D, Huang Y. A positive feedback loop of TET3 and TGF-beta1 promotes liver fibrosis. Cell Rep. 2020;30:1310-1318.e1315.PubMedPubMedCentralCrossRef Xu Y, Sun X, Zhang R, Cao T, Cai SY, Boyer JL, Zhang X, Li D, Huang Y. A positive feedback loop of TET3 and TGF-beta1 promotes liver fibrosis. Cell Rep. 2020;30:1310-1318.e1315.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Jia Y, Xie H, Zhang J, Ying H. Induction of TGF-beta receptor I expression in a DNA methylation-independent manner mediated by DNMT3A downregulation is involved in early-onset severe preeclampsia. FASEB J. 2020;34:13224–38.PubMedCrossRef Jia Y, Xie H, Zhang J, Ying H. Induction of TGF-beta receptor I expression in a DNA methylation-independent manner mediated by DNMT3A downregulation is involved in early-onset severe preeclampsia. FASEB J. 2020;34:13224–38.PubMedCrossRef
17.
Zurück zum Zitat Honore B. The rapidly expanding CREC protein family: members, localization, function, and role in disease. BioEssays. 2009;31:262–77.PubMedCrossRef Honore B. The rapidly expanding CREC protein family: members, localization, function, and role in disease. BioEssays. 2009;31:262–77.PubMedCrossRef
18.
Zurück zum Zitat Tsuji A, Kikuchi Y, Sato Y, Koide S, Yuasa K, Nagahama M, Matsuda Y. A proteomic approach reveals transient association of reticulocalbin-3, a novel member of the CREC family, with the precursor of subtilisin-like proprotein convertase, PACE4. Biochem J. 2006;396:51–9.PubMedPubMedCentralCrossRef Tsuji A, Kikuchi Y, Sato Y, Koide S, Yuasa K, Nagahama M, Matsuda Y. A proteomic approach reveals transient association of reticulocalbin-3, a novel member of the CREC family, with the precursor of subtilisin-like proprotein convertase, PACE4. Biochem J. 2006;396:51–9.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Jin J, Li Y, Ren J, Man Lam S, Zhang Y, Hou Y, Zhang X, Xu R, Shui G, Ma RZ. Neonatal respiratory failure with retarded perinatal lung maturation in mice caused by reticulocalbin 3 disruption. Am J Respir Cell Mol Biol. 2016;54:410–23.PubMedCrossRef Jin J, Li Y, Ren J, Man Lam S, Zhang Y, Hou Y, Zhang X, Xu R, Shui G, Ma RZ. Neonatal respiratory failure with retarded perinatal lung maturation in mice caused by reticulocalbin 3 disruption. Am J Respir Cell Mol Biol. 2016;54:410–23.PubMedCrossRef
20.
Zurück zum Zitat Shi X, An X, Yang L, Wu Z, Zan D, Li Z, Pang B, Chen Y, Li J, Tan P, et al. Reticulocalbin 3 deficiency in alveolar epithelium attenuated LPS-induced ALI via NF-kappaB signaling. Am J Physiol Lung Cell Mol Physiol. 2021;320:L627–39.PubMedCrossRef Shi X, An X, Yang L, Wu Z, Zan D, Li Z, Pang B, Chen Y, Li J, Tan P, et al. Reticulocalbin 3 deficiency in alveolar epithelium attenuated LPS-induced ALI via NF-kappaB signaling. Am J Physiol Lung Cell Mol Physiol. 2021;320:L627–39.PubMedCrossRef
21.
Zurück zum Zitat Jin J, Shi X, Li Y, Zhang Q, Guo Y, Li C, Tan P, Fang Q, Ma Y, Ma RZ. Reticulocalbin 3 deficiency in alveolar epithelium exacerbated bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2018;59:320–33.PubMedCrossRef Jin J, Shi X, Li Y, Zhang Q, Guo Y, Li C, Tan P, Fang Q, Ma Y, Ma RZ. Reticulocalbin 3 deficiency in alveolar epithelium exacerbated bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2018;59:320–33.PubMedCrossRef
22.
Zurück zum Zitat Park NR, Shetye SS, Bogush I, Keene DR, Tufa S, Hudson DM, Archer M, Qin L, Soslowsky LJ, Dyment NA, Joeng KS. Reticulocalbin 3 is involved in postnatal tendon development by regulating collagen fibrillogenesis and cellular maturation. Sci Rep. 2021;11:10868.PubMedPubMedCentralCrossRef Park NR, Shetye SS, Bogush I, Keene DR, Tufa S, Hudson DM, Archer M, Qin L, Soslowsky LJ, Dyment NA, Joeng KS. Reticulocalbin 3 is involved in postnatal tendon development by regulating collagen fibrillogenesis and cellular maturation. Sci Rep. 2021;11:10868.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Martinez-Martinez E, Ibarrola J, Fernandez-Celis A, Santamaria E, Fernandez-Irigoyen J, Rossignol P, Jaisser F, Lopez-Andres N. Differential proteomics identifies reticulocalbin-3 as a novel negative mediator of collagen production in human cardiac fibroblasts. Sci Rep. 2017;7:12192.PubMedPubMedCentralCrossRef Martinez-Martinez E, Ibarrola J, Fernandez-Celis A, Santamaria E, Fernandez-Irigoyen J, Rossignol P, Jaisser F, Lopez-Andres N. Differential proteomics identifies reticulocalbin-3 as a novel negative mediator of collagen production in human cardiac fibroblasts. Sci Rep. 2017;7:12192.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Ruan H, Luan J, Gao S, Li S, Jiang Q, Liu R, Liang Q, Zhang R, Zhang F, Li X, et al. Fedratinib attenuates bleomycin-induced pulmonary fibrosis via the JAK2/STAT3 and TGF-β1 signaling pathway. Molecules. 2021;26:4491.PubMedPubMedCentralCrossRef Ruan H, Luan J, Gao S, Li S, Jiang Q, Liu R, Liang Q, Zhang R, Zhang F, Li X, et al. Fedratinib attenuates bleomycin-induced pulmonary fibrosis via the JAK2/STAT3 and TGF-β1 signaling pathway. Molecules. 2021;26:4491.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Roux KJ, Kim DI, Burke B. BioID: a screen for protein-protein interactions. Curr Protoc Protein Sci. 2013;74:19–23.CrossRef Roux KJ, Kim DI, Burke B. BioID: a screen for protein-protein interactions. Curr Protoc Protein Sci. 2013;74:19–23.CrossRef
26.
Zurück zum Zitat Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N, Ahangari F, Chu SG, Raby BA, DeIuliis G, Januszyk M, et al. Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci Adv. 2020;6: eaba1983.PubMedPubMedCentralCrossRef Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N, Ahangari F, Chu SG, Raby BA, DeIuliis G, Januszyk M, et al. Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci Adv. 2020;6: eaba1983.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Lawson WE, Polosukhin VV, Zoia O, Stathopoulos GT, Han W, Plieth D, Loyd JE, Neilson EG, Blackwell TS. Characterization of fibroblast-specific protein 1 in pulmonary fibrosis. Am J Respir Crit Care Med. 2005;171:899–907.PubMedCrossRef Lawson WE, Polosukhin VV, Zoia O, Stathopoulos GT, Han W, Plieth D, Loyd JE, Neilson EG, Blackwell TS. Characterization of fibroblast-specific protein 1 in pulmonary fibrosis. Am J Respir Crit Care Med. 2005;171:899–907.PubMedCrossRef
28.
Zurück zum Zitat Österreicher CH, Penz-Österreicher M, Grivennikov SI, Guma M, Koltsova EK, Datz C, Sasik R, Hardiman G, Karin M, Brenner DA. Fibroblast-specific protein 1 identifies an inflammatory subpopulation of macrophages in the liver. Proc Natl Acad Sci USA. 2011;108:308–13.PubMedCrossRef Österreicher CH, Penz-Österreicher M, Grivennikov SI, Guma M, Koltsova EK, Datz C, Sasik R, Hardiman G, Karin M, Brenner DA. Fibroblast-specific protein 1 identifies an inflammatory subpopulation of macrophages in the liver. Proc Natl Acad Sci USA. 2011;108:308–13.PubMedCrossRef
29.
Zurück zum Zitat Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, Kolb M. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45:1434–45.PubMedPubMedCentralCrossRef Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, Kolb M. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45:1434–45.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Spagnolo P, Kropski JA, Jones MG, Lee JS, Rossi G, Karampitsakos T, Maher TM, Tzouvelekis A, Ryerson CJ. Idiopathic pulmonary fibrosis: disease mechanisms and drug development. Pharmacol Ther. 2021;222: 107798.PubMedCrossRef Spagnolo P, Kropski JA, Jones MG, Lee JS, Rossi G, Karampitsakos T, Maher TM, Tzouvelekis A, Ryerson CJ. Idiopathic pulmonary fibrosis: disease mechanisms and drug development. Pharmacol Ther. 2021;222: 107798.PubMedCrossRef
31.
Zurück zum Zitat Khor YH, Ng Y, Barnes H, Goh NSL, McDonald CF, Holland AE. Prognosis of idiopathic pulmonary fibrosis without anti-fibrotic therapy: a systematic review. Eur Respir Rev. 2020;29: 190158.PubMedPubMedCentralCrossRef Khor YH, Ng Y, Barnes H, Goh NSL, McDonald CF, Holland AE. Prognosis of idiopathic pulmonary fibrosis without anti-fibrotic therapy: a systematic review. Eur Respir Rev. 2020;29: 190158.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Hewlett JC, Kropski JA, Blackwell TS. Idiopathic pulmonary fibrosis: epithelial–mesenchymal interactions and emerging therapeutic targets. Matrix Biol. 2018;71–72:112–27.PubMedPubMedCentralCrossRef Hewlett JC, Kropski JA, Blackwell TS. Idiopathic pulmonary fibrosis: epithelial–mesenchymal interactions and emerging therapeutic targets. Matrix Biol. 2018;71–72:112–27.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Fernandez IE, Eickelberg O. The impact of TGF-β on lung fibrosis: from targeting to biomarkers. Proc Am Thorac Soc. 2012;9:111–6.PubMedCrossRef Fernandez IE, Eickelberg O. The impact of TGF-β on lung fibrosis: from targeting to biomarkers. Proc Am Thorac Soc. 2012;9:111–6.PubMedCrossRef
35.
Zurück zum Zitat Yin X, Murphy SJ, Wilkes MC, Ji Y, Leof EB. Retromer maintains basolateral distribution of the type II TGF-beta receptor via the recycling endosome. Mol Biol Cell. 2013;24:2285–98.PubMedPubMedCentralCrossRef Yin X, Murphy SJ, Wilkes MC, Ji Y, Leof EB. Retromer maintains basolateral distribution of the type II TGF-beta receptor via the recycling endosome. Mol Biol Cell. 2013;24:2285–98.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Yin X, Kang J-H, Andrianifahanana M, Wang Y, Jung M-Y, Hernandez DM, Leof EB. Basolateral delivery of the type I transforming growth factor beta receptor is mediated by a dominant-acting cytoplasmic motif. Mol Biol Cell. 2017;28:2701–11.PubMedPubMedCentralCrossRef Yin X, Kang J-H, Andrianifahanana M, Wang Y, Jung M-Y, Hernandez DM, Leof EB. Basolateral delivery of the type I transforming growth factor beta receptor is mediated by a dominant-acting cytoplasmic motif. Mol Biol Cell. 2017;28:2701–11.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Wang Y, Zhang L, Huang T, Wu GR, Zhou Q, Wang FX, Chen LM, Sun F, Lv Y, Xiong F, et al. The methyl-CpG-binding domain 2 facilitates pulmonary fibrosis by orchestrating fibroblast to myofibroblast differentiation. Eur Respir J. 2022;60:2003697.PubMedPubMedCentralCrossRef Wang Y, Zhang L, Huang T, Wu GR, Zhou Q, Wang FX, Chen LM, Sun F, Lv Y, Xiong F, et al. The methyl-CpG-binding domain 2 facilitates pulmonary fibrosis by orchestrating fibroblast to myofibroblast differentiation. Eur Respir J. 2022;60:2003697.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Zhou H, Wu G, Ma X, Xiao J, Yu G, Yang C, Xu N, Zhang B, Zhou J, Ye Z, Wang Z. Attenuation of TGFBR2 expression and tumour progression in prostate cancer involve diverse hypoxia-regulated pathways. J Exp Clin Cancer Res. 2018;37:89.PubMedPubMedCentralCrossRef Zhou H, Wu G, Ma X, Xiao J, Yu G, Yang C, Xu N, Zhang B, Zhou J, Ye Z, Wang Z. Attenuation of TGFBR2 expression and tumour progression in prostate cancer involve diverse hypoxia-regulated pathways. J Exp Clin Cancer Res. 2018;37:89.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Zhang L, Qu J, Qi Y, Duan Y, Huang YW, Zhou Z, Li P, Yao J, Huang B, Zhang S, Yu D. EZH2 engages TGFbeta signaling to promote breast cancer bone metastasis via integrin beta1-FAK activation. Nat Commun. 2022;13:2543.PubMedPubMedCentralCrossRef Zhang L, Qu J, Qi Y, Duan Y, Huang YW, Zhou Z, Li P, Yao J, Huang B, Zhang S, Yu D. EZH2 engages TGFbeta signaling to promote breast cancer bone metastasis via integrin beta1-FAK activation. Nat Commun. 2022;13:2543.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Zeng Y, Qiu R, Yang Y, Gao T, Zheng Y, Huang W, Gao J, Zhang K, Liu R, Wang S, et al. Regulation of EZH2 by SMYD2-mediated lysine methylation is implicated in tumorigenesis. Cell Rep. 2019;29:1482-1498.e1484.PubMedCrossRef Zeng Y, Qiu R, Yang Y, Gao T, Zheng Y, Huang W, Gao J, Zhang K, Liu R, Wang S, et al. Regulation of EZH2 by SMYD2-mediated lysine methylation is implicated in tumorigenesis. Cell Rep. 2019;29:1482-1498.e1484.PubMedCrossRef
41.
Zurück zum Zitat Tang B, Sun R, Wang D, Sheng H, Wei T, Wang L, Zhang J, Ho TH, Yang L, Wei Q, Huang H. ZMYND8 preferentially binds phosphorylated EZH2 to promote a PRC2-dependent to -independent function switch in hypoxia-inducible factor-activated cancer. Proc Natl Acad Sci USA. 2021;118: e2019052118.PubMedPubMedCentralCrossRef Tang B, Sun R, Wang D, Sheng H, Wei T, Wang L, Zhang J, Ho TH, Yang L, Wei Q, Huang H. ZMYND8 preferentially binds phosphorylated EZH2 to promote a PRC2-dependent to -independent function switch in hypoxia-inducible factor-activated cancer. Proc Natl Acad Sci USA. 2021;118: e2019052118.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Kim J, Lee Y, Lu X, Song B, Fong KW, Cao Q, Licht JD, Zhao JC, Yu J. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Rep. 2018;25:2808-2820.e2804.PubMedPubMedCentralCrossRef Kim J, Lee Y, Lu X, Song B, Fong KW, Cao Q, Licht JD, Zhao JC, Yu J. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Rep. 2018;25:2808-2820.e2804.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Le HQ, Hill MA, Kollak I, Keck M, Schroeder V, Wirth J, Skronska-Wasek W, Schruf E, Strobel B, Stahl H, et al. An EZH2-dependent transcriptional complex promotes aberrant epithelial remodelling after injury. EMBO Rep. 2021;22: e52785.PubMedPubMedCentralCrossRef Le HQ, Hill MA, Kollak I, Keck M, Schroeder V, Wirth J, Skronska-Wasek W, Schruf E, Strobel B, Stahl H, et al. An EZH2-dependent transcriptional complex promotes aberrant epithelial remodelling after injury. EMBO Rep. 2021;22: e52785.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Rangarajan S, Kurundkar A, Kurundkar D, Bernard K, Sanders YY, Ding Q, Antony VB, Zhang J, Zmijewski J, Thannickal VJ. Novel mechanisms for the antifibrotic action of nintedanib. Am J Respir Cell Mol Biol. 2016;54:51–9.PubMedPubMedCentralCrossRef Rangarajan S, Kurundkar A, Kurundkar D, Bernard K, Sanders YY, Ding Q, Antony VB, Zhang J, Zmijewski J, Thannickal VJ. Novel mechanisms for the antifibrotic action of nintedanib. Am J Respir Cell Mol Biol. 2016;54:51–9.PubMedPubMedCentralCrossRef
Metadaten
Titel
TGFβ1-RCN3-TGFBR1 loop facilitates pulmonary fibrosis by orchestrating fibroblast activation
verfasst von
Mingting Wu
Zhenyan Wang
Xiaoqian Shi
Danni Zan
Hong Chen
Shuqiao Yang
Fangping Ding
Liu Yang
Pingping Tan
Runlin Z. Ma
Jing Wang
Lishuang Ma
Yingmin Ma
Jiawei Jin
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2023
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-023-02533-z

Weitere Artikel der Ausgabe 1/2023

Respiratory Research 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Eingreifen von Umstehenden rettet vor Erstickungstod!

15.05.2024 Fremdkörperaspiration Nachrichten

Wer sich an einem Essensrest verschluckt und um Luft ringt, benötigt vor allem rasche Hilfe. Dass Umstehende nur in jedem zweiten Erstickungsnotfall bereit waren, diese zu leisten, ist das ernüchternde Ergebnis einer Beobachtungsstudie aus Japan. Doch es gibt auch eine gute Nachricht.

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.