Skip to main content
  • Correspondence
  • Open access
  • Published:

Fibrosis in PCLS: comparing TGF-β and fibrotic cocktail

Abstract

Introduction

Fibrotic cocktail (FC) is a combination of pro-fibrotic and pro-inflammatory mediators that induces early fibrotic changes in organotypic lung models. We hypothesised that transforming growth factor beta 1 (TGF-β1) alone induces a pro-fibrotic effect similar to FC. Our aim was to compare the pro-fibrotic effects of TGF-β1 with FC in human precision-cut lung slices (PCLS).

Methods

PCLS from “healthy” lung tissue of cancer patients undergoing surgery (n = 7) were incubated with TGF-β1, FC or control for 72 h. Gene expression markers for myofibroblasts differentiation, extracellular matrix (ECM), as well as TGF-β receptors were assessed (RT-qPCR). ECM proteins expression in lysates and supernatant was assessed by ELISA and immunofluorescence.

Results

We found that TGF-β1 significantly increased gene expression of ACTA2, COL1A1, CCN2, and VIM compared to control but also compared to FC. FC showed a significant increase of matrix metalloproteinase (MMP) 7 and 1 compared to control, while TGF-β receptor 2 was lower after FC compared to TGF-β1 or control. FC or TGF-β1 showed similar fibronectin protein expression in lysates and supernatants, while type I collagen protein expression in lysates was significantly greater with TGF-β1 compared to control.

Conclusions

Our findings show that TGF-β1 induces consistent pro-fibrotic changes in PCLS after 72 h. Compared to TGF-β1, FC treatment resulted in reduced gene expression of TGF-β receptor 2 and increased MMPs expression, potentially mitigating the early pro-fibrotic effects. Selecting specific pro-fibrotic stimuli may be preferable depending on the research question and time point of interest in lung fibrosis studies using PCLS.

Introduction

Fibrosing interstitial lung diseases (ILD) are a subset of ILDs that can present with a progressive phenotype leading to early mortality [1]. Idiopathic pulmonary fibrosis (IPF) is the most severe form of lung fibrosis without inflammation, resulting in the rapid decline of lung function [2]. Most ILDs are characterized by inflammation and fibrosis, making them crucial targets for therapy [3]. Increasingly, it is recognized that treatable traits may be more important in guiding treatment than the specific ILD diagnosis itself [4]. For personalised treatment strategies research models are needed that allow the investigation of specific pathomechanisms, including inflammatory and fibrotic stimuli in combination and separately.

The molecular pathomechanism of lung fibrosis involves several pro-fibrotic factors, such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF) [5], lysophosphatidic acid (LPA) [6, 7], and connective tissue growth factor (CTGF) [8], that favour an abnormal wound healing response and the development of lung fibrosis [9]. These mediators influence excessive extracellular matrix (ECM) deposition, impaired cell apoptosis or increased senescence, which contribute to sustained scarring and fibrosis progression [10].

Organotypic models including PCLS have provided valuable information about the dynamic interaction between mesenchymal cells and the ECM [11], or about the crosstalk between alveolar fibroblasts and epithelial cells mediated by the transforming growth factor beta 1 (TGF-β1) and non-canonical Wnt pathway [12]. Fibrosis induction in human PCLS has been optimised by various strategies including the addition of matrix metalloproteinase (MMP) inhibitors to TGF-β1 to improve collagen deposition [13], the combination of TGF-β1 with platelet lysate or neutrophil extracellular traps [14].

Alsafadi et al. suggested a specific fibrotic cocktail (FC), that combines TGF-β1 with two pro-fibrotic mediators, PDGF and LPA, and one pro-fibrotic but also inflammatory cytokine, tumour necrosis factor (TNF), to reproduce the physiological changes observed in the early stages of lung fibrosis [15]. The choice of stimuli resulted from current knowledge about fibrosis mediator in lung fibrosis [5, 7, 16]. Notably, the authors state, that the role of TNF in IPF remains incompletely understood [15]. Among all pro-fibrotic factors, TGF-β1 and its signalling pathway represents the major stimulus for the erroneous and permanent repair response in lung fibrogenesis [16]. TGF-β1 is equally a common mediator in fibrosis of other organs [17]. Compared with other fibrotic stimuli, such as PDGF, TGF-β1 promotes a greater ECM production as shown by pro-peptides of type I and type III collagens [18]. In PCLS, TGF-β1 increases ECM deposition and has been used to study anti-fibrotic drugs [19].

FC-induced fibrosis in PCLS is used by various research groups including ours and has led to remarkable progress in lung fibrosis research resulting in numerous publications [20,21,22,23,24]. Comparison of the gene expression in FC stimulated PCLS with IPF stages, FC recapitulates 46% of the observed in vivo changes across all stages. Of these, 11% overlapped with the stage 1–specific signature in IPF, indicating that FC treated PCLS best recapitulates the molecular processes that occur in early IPF [25].

As a limitation of this model, we observe like Alsafadi et al. that some donors respond differently to FC [15]. This is independent of clinical characteristics and possibly due to tissue specific or interindividual susceptibilities [15]. To minimize factors involved in treatment response variations, we hypothesise that TGF-β1 alone may sufficiently induce some of the fibrotic changes in PCLS. This study compared TGF-β1 with FC induced fibrosis in a PCLS model of normal-appearing human lung tissue from cancer patients undergoing surgery.

Methods

PCLS generation

Human control lung tissues (n = 7) were collected from normal appearing lung areas of lung cancer resections from patients undergoing surgery at the university hospital, Inselspital, Bern (Ethical approval KEK-BE_2024 − 01841). All patients signed written consent prior sample collection. The patients’ characteristics and diagnoses are summarised in Table 1.

Table 1 Patient characteristics

Lung tissues (ca. 10 g) were processed to generate PCLS (400 μm thick, 4 mm) to ensure homogeneity and reproducibility between replicates, following established standard protocols [22]. PCLS were treated with FC (5 ng/ml TGF-β1, 10 ng/ml PDGF-AB, 10 ng/ml TNF, and 5 µM LPA) [15] or 5 ng/ml TGF-β1 only in DMEM supplemented with 0.1% FBS for 72 h. Control PCLS were treated with medium and reagents’ diluents. The medium was changed every day. PCLS (4–6 samples per donor) and supernatant were collected for protein and gene expression analysis. Supplemental Table 1 shows the analysis performed in each donor sample.

RNA isolation and gene determination

Total mRNA, cDNA synthesis reverse-transcription reactions and RT-qPCR were performed following our modified previously reported protocol [22]. The gene expression of ACTA2, FN1, COL1A1, COL3A1, CCN2, VIM, MMP7, MMP1, TGFBR1, TGFBR2, and B2M as housekeeping were quantified by real-time PCR with Fast SYBR™ Green Master Mix (#4385616, Thermo Fisher Scientific). The primer sequences are displayed in Supplemental Table 2 [22, 26].

Protein isolation

PCLS were mechanically disrupted in TissueLyser II (Qiagen) and protein lysate were obtained following previous protocols [22]. Total protein was quantified by Pierce™ BCA protein assay (#23227, Thermo Fisher Scientific) and the samples were stored at -80 °C until use.

ELISA

Human pro-collagen I alpha 1 and human fibronectin were measured in the supernatant and protein lysate from PCLS by ELISA kit (#DY6220-05 and #DY1918-05, respectively, R&D Systems), following the manufacturer’s protocol. Dilution determination for the control and treated samples was performed before total sample analysis. The total amount of type I collagen (pg/ml) and fibronectin (ng/ml) in the protein lysate (µg/ml) was assessed by dividing the respective ELISA values by the total protein content measured by BCA protein assay.

Immunofluorescence

Fixed PCLs were immunostained for human fibronectin EDA domain, with DAPI as counterstaining, following previous published protocols [22]. LSM files were processed in Fiji image processing package v1.54f (ImageJ, USA), and the Integrated density (IntDe, in arbitrary units, AU) was subsequently calculated from the positive fluorescent areas of each picture in order to estimate the fluorescence intensity, by following the formula: FN IntDen = Sample IntDen – Negative control IntDen.

Statistical analysis

All results are shown as mean ± standard deviations. Comparisons between groups were assessed by repeated measures one-way ANOVA with Tukey’s as post hoc test. Statistical analysis was performed with GraphPad Prism 10 (Insight Partners, USA). Statistical significance was assumed when p < 0.05.

Results

TGF-β1 induces gene expression for fibrotic markers in PCLS

The results revealed that TGF-β1 significantly increased the gene expression of the myofibroblast differentiation marker ACTA2 compared to control (p < 0.01) and FC (p < 0.05) (Fig. 1A). In addition, mRNA levels of ECM components such as fibronectin extra domain A (FN-EDA) (p < 0.0001), type I and III collagen (COL1A1 (p < 0.0001), COL3A1 (p < 0.05)), connective tissue growth factor (CCN2), and vimentin (VIM), were significantly upregulated in PCLS after the addition of TGF-β1 compared to control and FC.

Fig. 1
figure 1

Fibrotic marker expression in PCLS treated with a fibrotic cocktail (FC) or TGF-β1 alone. (A) Gene expression of the myofibroblast marker ACTA2, the ECM markers EDA-FN1, COL1A1, COL3A1, CCN2, and VIM, the ECM remodelling genes MMP7 and 1, and the TGF-β receptors TGFBR1 and R2 72 h after treatment with FC or TGF-β1 alone. The gene results are presented as relative gene expression (RGE) to that of the housekeeping gene B2M and were normalised to control (Ctrl) condition. (B) Concentrations of total fibronectin and type I collagen in PCLS lysate were measured by ELISA. (C) Fibronectin and type I collagen release in supernatants (SN) was measured by ELISA. (D) Immunofluorescence staining revealed that the protein content of fibronectin extra domain A (FN-EDA) was similar between FC and TGF-β1, as shown by the semi-quantification with IntDen (arbitrary units, AU). Pictures obtained at 10X magnification. (─) = 500 μm. (*) p < 0.05; (**) p > 0.01, (***) p < 0.001, (****) p < 0.0001

Compared to control (p < 0.01) and TGF-β1-treated PCLS (p < 0.05), FC-treated PCLS presented increased mRNA levels of MMP7 and MMP1 (Fig. 1A), suggesting increased ECM remodelling. FC influenced gene expression of TGF-β receptors, especially TGF-β receptor 2 (TGFBR2); FC-stimulated PCLS showed lower expression than control PCLS (p < 0.05) and those stimulated with TGF-β1 alone (p < 0.01, Fig. 1A), whereas the expression of TGF-β receptor 1 (TGFBR1) was higher in TGF-β1-treated PCLS compared to FC-treated PCLS (p < 0.05).

TGF-β1 induces protein expression of fibronectin and collagen in tissue lysates and supernatants

PCLS lysates analysed by ELISA revealed that, fibronectin in TGF-β1 treated PCLS increased similar to FC stimulation, while collagen production was significantly greater in PCLS stimulated with TGF-β1 compared to control (p < 0.05, Fig. 1B).

We observed that supernatants of FC and TGF-β1 treated PCLS showed similar significant increase of fibronectin and type I collagen compared to control (p < 0.05 and p < 0.01, respectively; Fig. 1C).

Finally, we stained PCLS for FN-EDA. By semi-quantitative analysis of fluorescence intensity, we found that compared to control, TGF-β1 significant increased fibronectin in PCLS (p < 0.05, Fig. 1D).

Discussion

The need for new anti-fibrotic drugs calls for various advanced models to better evaluate pathomechanisms and test drug candidates in preclinical studies, aiming to improve outcome in clinical trials [27]. PCLS carry the biological complexity of the lung, allowing for drug testing while considering effects on different lung cell types [28]. Modelling early fibrotic changes with this organotypic model by adding FC to human lung tissue has been an important step for fibrosis research. Our results suggests that the major pro-fibrotic cytokine TGF-β1 can induce significant fibrotic changes, including protein changes in fibronectin and collagen. In selected studies, TGF-β1 might be sufficient to investigate fibrosis for specific questions, while FC remains a more complex model for early lung fibrosis.

While some non-significant results after FC stimulation might be due to interindividual response variability of the tissue in our hands, we speculate that these results could also suggest interference of FC components with TGF-β1 signalling and potentially influence the fibrotic response in the ex vivo model through its receptor regulation [29]. For example, TGFβR2 gene expression was decreased after FC compared to control and TGF-β1, whereas the gene encoding TGFβR1 was significantly higher in TGF-β1-treated PCLS compared to FC. Due to the positive feedback loop between TGF-β1 and its receptors, an increase compared to control would have been expected [30]. Moreover, in previous studies, TNF reduced TGFβR2 protein expression in fibroblasts from systemic sclerosis (SSc) donors at a concentration of 10 ng/ml after 48 h [31]. This might suggest a role for TNF in our finding of reduced TGFβR2 gene expression after FC, which contains TNF.

The controversial role of TNF in lung fibrosis highlights the complex relationship between inflammation and fibrosis in the fibrotic lung. While some reports have shown that TNF promotes myofibroblast differentiation of lung resident mesenchymal stem cells [32], and that TNF polymorphisms are associated with fibrotic hypersensitivity pneumonitis (HP) [33]; others have shown that TNF might have therapeutic uses in the treatment of lung fibrosis, by targeting the pro-fibrotic activity of macrophages [34]. A study showed that the overexpression of TNF in transgenic mice protected the animals from the development of bleomycin-induced lung fibrosis [35]. Moreover, unlike TGF-β1 stimulation, the treatment of alveolar fibroblasts with TNF fails to induce the expression of some markers of fibrotic phenotype (i.e., COL1A1+; collagen triple helix repeat containing 1, CTHRC1+; periostin, POSTN+) [36] and actively promotes collagen degradation by MMPs in isolated human lung fibroblasts [37]. In IPF patients, TNF serum levels were not significantly different compared to healthy controls [38], and immunostaining did not reveal differences in TNF+ cells between control lung tissue and normal-appearing lung parenchyma from IPF patients [39]. This underlines the intricate interplay between inflammatory stimuli in a pro-fibrotic environment.

The dynamic interplay TNF and TGF-β1 has been also described in the bleomycin-induced lung fibrosis model, where TNF and MMP showed their major expression during the inflammatory phase, being progressively reduced during the fibrotic phase, where TGF-β1 effects dominate [40]. In bronchial epithelial cells, TNF activates MMPs via transcription factor nuclear factor (NF)-kappa B [41]. Our results revealed increased gene expression of the MMP7 and MMP1 in FC-treated PCLS compared to TGF-β1 and controls. Overexpression of MMPs was associated with ECM restructuring and spontaneous emphysema in transgenic mice overexpressing TNF [42]. MMP-mediated ECM disassembly [43] might occur in PCLS treated with FC containing TNF. Interestingly, Khan et al. reported that the addition of MMP inhibitor to TGF-β1-treated PCLS resulted in increased collagen deposition [13].

Despite the observed increase of collagen production in the protein lysate of FC-treated PCLS, no statistically significant difference compared to control was observed. This may be due to half of the donors showing less than a 2-fold change in collagen content after FC treatment, while a consistent response to TGF-β1 was observed in most of the PCLS donors compared to control. Increased levels of MMP-1 after FC and not TGF-β1, might influence collagen at the post-transcriptional level explaining our findings [44] and suggest that some components of the FC may interfere with collagen deposition at this time point.

Overall, our protein results revealed significant changes in fibronectin in FC- and TGF-β1-treated PCLS compared to controls. Protein and gene expression results suggest that, in addition to TGF-β1, other components of the FC might enhance expression of fibronectin, as it has been described for PDGF [5] or for TNF [45]. In our hands, this did not lead to significant increases of fibrosis induction comparing FC to TGF-β1.

In fixed tissue (250 μm-thick slices) analysed with second-harmonic imaging microscopy by Khan et al. did not reveal increased deposition of fibrillar collagen in PCLS after TGF-β1 stimulation after 13 days [13]. In contrast and in parallel to our study, Roach et al. demonstrated that type I and II collagens were upregulated following 7 days of TGF-β1 treatment in an ex vivo lung model by immunostaining and transcriptomic analysis [19, 46].

Our findings confirm the efficacy of FC to induce early fibrotic changes, while our results suggest a comparable response after TGF-β1. We suggest that depending on the experimental set up and analysis, single use of TGF-β1 can be a simplified way to induce fibrotic changes in PCLS.

Our study has limitations. Treatment for 3 days does not give information about later time points. While previous studies using FC have commonly adopted 120 h incubation times for readouts [15, 20, 21, 23], our study focuses on early fibrosis induction. Longer-term studies would be needed to determine how pro-fibrotic effects evolve over time in both models. In addition, our experimental settings including medium replacement volume and times are modified from those in the original report of FC induced fibrosis in PCLS [15]; this does not allow the direct comparisons of our results with this previous publication. Our study did not evaluate interactions between TGF-β1, PDGF, TNF or LPA to evaluate potential agonistic or antagonistic effects of FC components. While the literature supports an antagonistic relationship between TGF-β1 and TNF [34, 35, 40], future experiments in PCLS exploring the interplay between inflammation and fibrosis will be necessary. Further investigation of the interaction between individual FC components would be interesting, although beyond the scope of this short report. Future characterization of cell-type specific changes by RNA sequencing will be essential to broaden our understanding and pinpoint the key mechanisms and cells involved in the potential interactions between individual FC components.

Conclusions

In summary, our results confirm that TGF-β1 reliably triggers a fibrotic response, reducing sample variability and allowing focused investigation of this molecule. The model’s simplicity supports consistent readouts, while complex models may offer insights into broader interactions in fibrosis.

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author upon reasonable request.

Abbreviations

CTGF:

Connective tissue growth factor

CTHRC1:

Collagen triple helix repeat containing 1

ECM:

Extracellular matrix

EGR1:

Early growth response-1

FC:

Fibrotic cocktail

FGF:

Fibroblast growth factor

FN-EDA:

Fibronectin extra domain A

HP:

Hypersensitivity pneumonitis

ILD:

Interstitial lung diseases

IPF:

Idiopathic pulmonary fibrosis

LPA:

Lysophosphatidic acid

MMP:

Matrix metalloproteinase

NF-κB:

Nuclear factor-kappa B

PCLS:

Precision cut lung slices

PDGF:

Platelet derived growth factor AB

POSTN:

Periostin

SSc:

Systemic sclerosis

TGF-β1:

Transforming growth factor beta 1

TGFβR1:

Transforming growth factor beta receptor 1

TGFβR2:

Transforming growth factor beta receptor 2

TNF:

Tumour necrosis factor

References

  1. Cottin V, Wollin L, Fischer A, Quaresma M, Stowasser S, Harari S. Fibrosing interstitial lung diseases: knowns and unknowns. Eur Respir Rev 2019;28:180100.

  2. George PM, Spagnolo P, Kreuter M, Altinisik G, Bonifazi M, Martinez FJ, Molyneaux PL, Renzoni EA, Richeldi L, Tomassetti S, et al. Progressive fibrosing interstitial lung disease: clinical uncertainties, consensus recommendations, and research priorities. Lancet Respir Med. 2020;8:925–34.

    Article  PubMed  Google Scholar 

  3. Wijsenbeek M, Kreuter M, Olson A, Fischer A, Bendstrup E, Wells CD, Denton CP, Mounir B, Zouad-Lejour L, Quaresma M, Cottin V. Progressive fibrosing interstitial lung diseases: current practice in diagnosis and management. Curr Med Res Opin. 2019;35:2015–24.

    Article  PubMed  Google Scholar 

  4. Khor YH, Cottin V, Holland AE, Inoue Y, McDonald VM, Oldham J, et al. Treatable traits: a comprehensive precision medicine approach in interstitial lung disease. Eur Respir J 2023;62:2300404.

  5. Chaudhary NI, Roth GJ, Hilberg F, Müller-Quernheim J, Prasse A, Zissel G, Schnapp A, Park JE. Inhibition of PDGF, VEGF and FGF signalling attenuates fibrosis. Eur Respir J. 2007;29:976–85.

    Article  CAS  PubMed  Google Scholar 

  6. Funke M, Zhao Z, Xu Y, Chun J, Tager AM. The lysophosphatidic acid receptor LPA1 promotes epithelial cell apoptosis after lung injury. Am J Respir Cell Mol Biol. 2012;46:355–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, Polosukhin V, Wain J, Karimi-Shah BA, Kim ND, et al. The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat Med. 2008;14:45–54.

    Article  CAS  PubMed  Google Scholar 

  8. Wang Q, Usinger W, Nichols B, Gray J, Xu L, Seeley TW, Brenner M, Guo G, Zhang W, Oliver N, et al. Cooperative interaction of CTGF and TGF-β in animal models of fibrotic disease. Fibrogenesis Tissue Repair. 2011;4:4.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Froidure A, Marchal-Duval E, Homps-Legrand M, Ghanem M, Justet A, Crestani B, et al. Chaotic activation of developmental signalling pathways drives idiopathic pulmonary fibrosis. Eur Respir Rev 2020;29:190140.

  10. Wu B, Tang L, Kapoor M. Fibroblasts and their responses to chronic injury in pulmonary fibrosis. Semin Arthritis Rheum. 2021;51:310–7.

    Article  CAS  PubMed  Google Scholar 

  11. Burgstaller G, Vierkotten S, Lindner M, Königshoff M, Eickelberg O. Multidimensional immunolabeling and 4D time-lapse imaging of vital ex vivo lung tissue. Am J Physiol Lung Cell Mol Physiol. 2015;309:L323–332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cohen ML, Brumwell AN, Ho TC, Garakani K, Montas G, Leong D, et al. A fibroblast-dependent TGF-β1/sFRP2 noncanonical wnt signaling axis promotes epithelial metaplasia in idiopathic pulmonary fibrosis. J Clin Invest 2024;134:e174598.

  13. Khan MM, Poeckel D, Halavatyi A, Zukowska-Kasprzyk J, Stein F, Vappiani J, et al. An integrated multiomic and quantitative label-free microscopy-based approach to study pro-fibrotic signalling in ex vivo human precision-cut lung slices. Eur Respir J 2021;58:2000221.

  14. Bozzini S, Bozza E, Bagnera C, Morbini P, Lettieri S, Della Zoppa M, et al. Assessment of Imatinib Anti-remodeling activity on a Human Precision cut lung slices Model. Int J Mol Sci 2024;25:8186.

  15. Alsafadi HN, Staab-Weijnitz CA, Lehmann M, Lindner M, Peschel B, Königshoff M, Wagner DE. An ex vivo model to induce early fibrosis-like changes in human precision-cut lung slices. Am J Physiol Lung Cell Mol Physiol. 2017;312:L896–902.

    Article  PubMed  Google Scholar 

  16. Massagué J, Sheppard D. TGF-β signaling in health and disease. Cell. 2023;186:4007–37.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020;217:e20190103.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Madsen SF, Sand JMB, Juhl P, Karsdal M, Thudium CS, Siebuhr AS, Bay-Jensen AC. Fibroblasts are not just fibroblasts: clear differences between dermal and pulmonary fibroblasts’ response to fibrotic growth factors. Sci Rep. 2023;13:9411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Roach KM, Sutcliffe A, Matthews L, Elliott G, Newby C, Amrani Y, Bradding P. A model of human lung fibrogenesis for the assessment of anti-fibrotic strategies in idiopathic pulmonary fibrosis. Sci Rep. 2018;8:342.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Ahangari F, Becker C, Foster DG, Chioccioli M, Nelson M, Beke K, Wang X, Justet A, Adams T, Readhead B, et al. Saracatinib, a selective src kinase inhibitor, blocks fibrotic responses in Preclinical models of Pulmonary Fibrosis. Am J Respir Crit Care Med. 2022;206:1463–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chioccioli M, Roy S, Newell R, Pestano L, Dickinson B, Rigby K, Herazo-Maya J, Jenkins G, Ian S, Saini G, et al. A lung targeted miR-29 mimic as a therapy for pulmonary fibrosis. EBioMedicine. 2022;85:104304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Guler SA, Machahua C, Geiser TK, Kocher G, Marti TM, Tan B, Trappetti V, Ryerson CJ, Funke-Chambour M. Dehydroepiandrosterone in fibrotic interstitial lung disease: a translational study. Respir Res. 2022;23:149.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lehmann M, Buhl L, Alsafadi HN, Klee S, Hermann S, Mutze K, Ota C, Lindner M, Behr J, Hilgendorff A, et al. Differential effects of Nintedanib and Pirfenidone on lung alveolar epithelial cell function in ex vivo murine and human lung tissue cultures of pulmonary fibrosis. Respir Res. 2018;19:175.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Marimoutou M, Patel V, Kim JH, Schaible N, Alvarez J, Hughes J, et al. The Fibrotic phenotype of Human Precision-Cut lung slices is maintained after Cryopreservation. Toxics 2024;12:637.

  25. Lang NJ, Gote-Schniering J, Porras-Gonzalez D, Yang L, De Sadeleer LJ, Jentzsch RC, Shitov VA, Zhou S, Ansari M, Agami A, et al. Ex vivo tissue perturbations coupled to single-cell RNA-seq reveal multilineage cell circuit dynamics in human lung fibrogenesis. Sci Transl Med. 2023;15:eadh0908.

    Article  CAS  PubMed  Google Scholar 

  26. Machahua C, Vicens-Zygmunt V, Ríos-Martín J, Llatjós R, Escobar-Campuzano I, Molina-Molina M, Montes-Worboys A. Collagen 3D matrices as a model for the study of cell behavior in pulmonary fibrosis. Exp Lung Res. 2022;48:126–36.

    Article  CAS  PubMed  Google Scholar 

  27. Kolanko E, Cargnoni A, Papait A, Silini AR, Czekaj P, Parolini O. The evolution of in vitro models of lung fibrosis: promising prospects for drug discovery. Eur Respir Rev 2024;33:230127.

  28. Koziol-White C, Gebski E, Cao G, Panettieri RA Jr. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res. 2024;25:231.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin. 2024;45:1337–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Duan D, Derynck R. Transforming growth factor-β (TGF-β)-induced up-regulation of TGF-β receptors at the cell surface amplifies the TGF-β response. J Biol Chem. 2019;294:8490–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yamane K, Ihn H, Asano Y, Jinnin M, Tamaki K. Antagonistic effects of TNF-alpha on TGF-beta signaling through down-regulation of TGF-beta receptor type II in human dermal fibroblasts. J Immunol. 2003;171:3855–62.

    Article  CAS  PubMed  Google Scholar 

  32. Hou J, Ma T, Cao H, Chen Y, Wang C, Chen X, Xiang Z, Han X. TNF-α-induced NF-κB activation promotes myofibroblast differentiation of LR-MSCs and exacerbates bleomycin-induced pulmonary fibrosis. J Cell Physiol. 2018;233:2409–19.

    Article  CAS  PubMed  Google Scholar 

  33. Freitas C, Lima B, Melo N, Mota P, Novais-Bastos H, Alves H, Sokhatska O, Delgado L, Morais A. Distinct TNF-alpha and HLA polymorphisms associate with fibrotic and non-fibrotic subtypes of hypersensitivity pneumonitis. Pulmonology. 2023;29(Suppl 4):S63–9.

    Article  PubMed  Google Scholar 

  34. Redente EF, Keith RC, Janssen W, Henson PM, Ortiz LA, Downey GP, Bratton DL, Riches DW. Tumor necrosis factor-α accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages. Am J Respir Cell Mol Biol. 2014;50:825–37.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Fujita M, Shannon JM, Morikawa O, Gauldie J, Hara N, Mason RJ. Overexpression of tumor necrosis factor-alpha diminishes pulmonary fibrosis induced by bleomycin or transforming growth factor-beta. Am J Respir Cell Mol Biol. 2003;29:669–76.

    Article  CAS  PubMed  Google Scholar 

  36. Tsukui T, Sun KH, Wetter JB, Wilson-Kanamori JR, Hazelwood LA, Henderson NC, Adams TS, Schupp JC, Poli SD, Rosas IO et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat Commun 2020;11:1920.

  37. Fang Q, Liu X, Al-Mugotir M, Kobayashi T, Abe S, Kohyama T, Rennard SI. Thrombin and TNF-alpha/IL-1beta synergistically induce fibroblast-mediated collagen gel degradation. Am J Respir Cell Mol Biol. 2006;35:714–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Alhamad EH, Cal JG, Shakoor Z, Almogren A, AlBoukai AA. Cytokine gene polymorphisms and serum cytokine levels in patients with idiopathic pulmonary fibrosis. BMC Med Genet. 2013;14:66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nuovo GJ, Hagood JS, Magro CM, Chin N, Kapil R, Davis L, Marsh CB, Folcik VA. The distribution of immunomodulatory cells in the lungs of patients with idiopathic pulmonary fibrosis. Mod Pathol. 2012;25:416–33.

    Article  CAS  PubMed  Google Scholar 

  40. Ye H, Cai PC, Zhou Q, Ma WL. Transforming growth factor-β1 suppresses the up-regulation of matrix metalloproteinase-2 by lung fibroblasts in response to tumor necrosis factor-α. Wound Repair Regen. 2011;19:392–9.

    Article  PubMed  Google Scholar 

  41. Hozumi A, Nishimura Y, Nishiuma T, Kotani Y, Yokoyama M. Induction of MMP-9 in normal human bronchial epithelial cells by TNF-alpha via NF-kappa B-mediated pathway. Am J Physiol Lung Cell Mol Physiol. 2001;281:L1444–1452.

    Article  CAS  PubMed  Google Scholar 

  42. Thomson EM, Williams A, Yauk CL, Vincent R. Overexpression of tumor necrosis factor-α in the lungs alters immune response, matrix remodeling, and repair and maintenance pathways. Am J Pathol. 2012;180:1413–30.

    Article  CAS  PubMed  Google Scholar 

  43. Chen K, Xu M, Lu F, He Y. Development of Matrix metalloproteinases-mediated extracellular matrix remodeling in Regenerative Medicine: a Mini Review. Tissue Eng Regen Med. 2023;20:661–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Devos H, Zoidakis J, Roubelakis MG, Latosinska A, Vlahou A. Reviewing the regulators of COL1A1. Int J Mol Sci 2023;24:10004.

  45. Hetzel M, Bachem M, Anders D, Trischler G, Faehling M. Different effects of growth factors on proliferation and matrix production of normal and fibrotic human lung fibroblasts. Lung. 2005;183:225–37.

    Article  CAS  PubMed  Google Scholar 

  46. Roach KM, Castells E, Dixon K, Mason S, Elliott G, Marshall H, Poblocka MA, Macip S, Richardson M, Khalfaoui L, Bradding P. Evaluation of Pirfenidone and Nintedanib in a human lung model of Fibrogenesis. Front Pharmacol. 2021;12:679388.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Lungenliga Bern.

Author information

Authors and Affiliations

Authors

Contributions

MFC and CM designed the experimental study; TMM, and PD contributed to tissue and clinical data acquisition; CM and MFC contributed to analysis, interpretation of the data and drafting of the manuscript. All the authors critically revised and approved the final manuscript.

Corresponding author

Correspondence to Carlos Machahua.

Ethics declarations

Ethics approval and consent to participate

The study and experimentation in human control lung tissue were approved by the local Swiss Ethical Committee, Bern, Switzerland (approval number: KEK-BE_2024 − 01841). Human samples were collected and handle in accordance with the Declaration of Helsinki, and donors were informed prior to recruitment, and signed a written consent form.

Consent for publication

NA.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Machahua, C., Marti, T.M., Dorn, P. et al. Fibrosis in PCLS: comparing TGF-β and fibrotic cocktail. Respir Res 26, 44 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12931-025-03110-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12931-025-03110-2

Keywords