Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (158)

Search Parameters:
Keywords = bleomycin-induced pulmonary fibrosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 9750 KiB  
Article
SIK2 Drives Pulmonary Fibrosis by Enhancing Fibroblast Glycolysis and Activation
by Jianhan He, Ruihan Dong, Huihui Yue, Fengqin Zhang, Xinran Dou, Xuan Li, Hui Li and Huilan Zhang
Biomedicines 2025, 13(8), 1919; https://doi.org/10.3390/biomedicines13081919 - 6 Aug 2025
Abstract
Background: Pulmonary fibrosis (PF), the end-stage manifestation of interstitial lung disease, is defined by excessive extracellular matrix deposition and alveolar destruction. Activated fibroblasts, the primary matrix producers, rely heavily on dysregulated glucose metabolism for their activation. While Salt Inducible Kinase 2 (SIK2) regulates [...] Read more.
Background: Pulmonary fibrosis (PF), the end-stage manifestation of interstitial lung disease, is defined by excessive extracellular matrix deposition and alveolar destruction. Activated fibroblasts, the primary matrix producers, rely heavily on dysregulated glucose metabolism for their activation. While Salt Inducible Kinase 2 (SIK2) regulates glycolytic pathways in oncogenesis, its specific contributions to fibroblast activation and therapeutic potential in PF pathogenesis remain undefined. This study elucidates the functional role of SIK2 in PF and assesses its viability as a therapeutic target. Methods: SIK2 expression/localization in fibrosis was assessed by Western blot and immunofluorescence. Fibroblast-specific Sik2 KO mice evaluated effects on bleomycin-induced fibrosis. SIK2’s role in fibroblast activation and glucose metabolism impact (enzyme expression, metabolism assays, metabolites) were tested. SIK2 inhibitors were screened and evaluated therapeutically in fibrosis models. Results: It demonstrated significant SIK2 upregulation, specifically within activated fibroblasts of fibrotic lungs from both PF patients and murine models. Functional assays demonstrated that SIK2 is crucial for fibroblast activation, proliferation, and migration. Mechanistically, SIK2 enhances fibroblast glucose metabolism by increasing the expression of glycolysis-related enzymes. Additionally, this study demonstrated that the SIK2 inhibitor YKL06-061 effectively inhibited PF in both bleomycin and FITC-induced PF mouse models with the preliminary safety profile. Furthermore, we identified a novel therapeutic application for the clinically approved drug fostamatinib, demonstrating it inhibits fibroblast activation via SIK2 targeting and alleviates PF in mice. Conclusions: Our findings highlight SIK2 as a promising therapeutic target and provide compelling preclinical evidence for two distinct anti-fibrotic strategies with significant potential for future PF treatment. Full article
(This article belongs to the Special Issue New Insights in Respiratory Diseases)
Show Figures

Figure 1

20 pages, 6034 KiB  
Article
Pexidartinib and Nintedanib Combination Therapy Targets Macrophage Polarization to Reverse Pulmonary Fibrosis: A Preclinical Study
by Ji-Hee Kim, Jae-Kyung Nam, Min-Sik Park, Seungyoul Seo, Hyung Chul Ryu, Hae-June Lee, Jeeyong Lee and Yoon-Jin Lee
Int. J. Mol. Sci. 2025, 26(15), 7570; https://doi.org/10.3390/ijms26157570 - 5 Aug 2025
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease with limited therapeutic options and increasing global incidence, with a median survival of only 2–5 years. The clinical utility of macrophage polarization to regulate the progression of pulmonary fibrosis remains understudied. This [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease with limited therapeutic options and increasing global incidence, with a median survival of only 2–5 years. The clinical utility of macrophage polarization to regulate the progression of pulmonary fibrosis remains understudied. This study determined the efficacy of nintedanib and pexidartinib (PLX3397) combination therapy for treating IPF. Combination treatment effectively inhibited the progression of radiation-induced pulmonary fibrosis (RIPF) and prolonged survival in bleomycin-treated mice. Micro-CT analysis revealed a significant tissue repair efficacy. The therapy significantly normalized the abnormal vascular structure observed during RIPF and bleomycin-induced pulmonary fibrosis progression and was accompanied by a decrease in the M2 population. Polarized M1 macrophages enhanced normalized tube formation of irradiated endothelial cells (ECs) in vitro; M2 macrophages increased adhesion in irradiated ECs and abnormal tube formation. Single-cell RNA sequencing data from patients with IPF further supports colony stimulating factor (CSF) 1 upregulation in macrophages and downregulation of capillary EC markers. This study highlights a promising combination strategy to overcome the therapeutic limitations of monotherapy with nintedanib for the treatment of IPF. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

23 pages, 40218 KiB  
Article
ACSL4 Drives C5a/C5aR1–Calcium-Induced Fibroblast-to-Myofibroblast Transition in a Bleomycin-Induced Mouse Model of Pulmonary Fibrosis
by Tingting Ren, Jia Shi, Lili Zhuang, Ruiting Su, Yimei Lai and Niansheng Yang
Biomolecules 2025, 15(8), 1106; https://doi.org/10.3390/biom15081106 - 31 Jul 2025
Viewed by 307
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by excessive extracellular matrix (ECM) deposition driven by aberrant fibroblast-to-myofibroblast transition (FMT). However, the upstream regulators and downstream effectors of this process remain incompletely understood. Here, we identify acyl-CoA synthetase long-chain family member 4 (ACSL4), a lipid [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is characterized by excessive extracellular matrix (ECM) deposition driven by aberrant fibroblast-to-myofibroblast transition (FMT). However, the upstream regulators and downstream effectors of this process remain incompletely understood. Here, we identify acyl-CoA synthetase long-chain family member 4 (ACSL4), a lipid metabolic enzyme, as a critical mediator linking complement component 5a (C5a)/C5a receptor 1 (C5aR1) signaling to FMT via calcium signaling. In bleomycin (BLM)-induced pulmonary fibrosis of C57BL/6JGpt mice, and in C5a-stimulated primary lung fibroblasts, the expression of ACSL4 was markedly upregulated. Pharmacological inhibition of ACSL4 (PRGL493) or C5aR1 (PMX53) attenuated the deposition of ECM and suppressed the expression of fibrotic markers in vivo and in vitro. Mechanistically, the activation of C5a/C5aR1 signaling increased intracellular calcium levels and promoted the expression of ACSL4, while inhibition of calcium signaling (FK506) reversed the upregulation of ACSL4 and FMT-related changes, including the expression of α-smooth muscle actin (αSMA) and the migration of fibroblasts. Notably, inhibition of ACSL4 did not affect the proliferation of fibroblasts, suggesting its specific role in phenotypic transition. These findings demonstrate that ACSL4 functions downstream of C5a/C5aR1-induced calcium signaling to promote FMT and the progression of pulmonary fibrosis. Targeting ACSL4 may therefore offer a novel therapeutic strategy for IPF. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

20 pages, 2643 KiB  
Article
Modulation of Pulmonary Fibrosis by Pulmonary Surfactant-Associated Phosphatidylethanolamine In Vitro and In Vivo
by Beatriz Tlatelpa-Romero, Luis G. Vázquez-de-Lara Cisneros, Olga Cañadas, Amaya Blanco-Rivero, Barbara Olmeda, Jesús Pérez-Gil, Criselda Mendoza-Milla, José Luis Martinez-Vaquero, Yair Romero, David Atahualpa Contreras-Cruz, René de-la-Rosa Paredes, Sinuhé Ruiz-Salgado, Roberto Berra-Romani, Alonso Antonio Collantes-Gutiérrez, María Susana Pérez-Fernández, María Guadalupe Hernández-Linares and Gabriel Guerrero-Luna
Int. J. Mol. Sci. 2025, 26(15), 7132; https://doi.org/10.3390/ijms26157132 - 24 Jul 2025
Viewed by 275
Abstract
Pulmonary fibrosis (PF) is characterized by excessive collagen deposition and impaired lung function. Pulmonary surfactant may modulate fibroblast activity and offer therapeutic benefits. We developed a natural porcine pulmonary surfactant (NPPS) enriched with 1,2-dipalmitoyl-rac-glycero-3-phosphatidylethanolamine (PE) and evaluated its biophysical and biological properties. Biophysical [...] Read more.
Pulmonary fibrosis (PF) is characterized by excessive collagen deposition and impaired lung function. Pulmonary surfactant may modulate fibroblast activity and offer therapeutic benefits. We developed a natural porcine pulmonary surfactant (NPPS) enriched with 1,2-dipalmitoyl-rac-glycero-3-phosphatidylethanolamine (PE) and evaluated its biophysical and biological properties. Biophysical analysis showed that PE improved surfactant performance by increasing surface pressure and stability. In vitro, NPPS-PE reduced collagen expression and induced apoptosis in normal human lung fibroblasts; in addition, it decreased proliferation in fibroblasts stimulated with TGF-β. In vivo, NPPS-PE improved gas exchange and significantly reduced collagen deposition in bleomycin-treated mice. These findings suggest that NPPS-PE may be a promising therapeutic strategy for fibrosing lung diseases. Full article
(This article belongs to the Special Issue Molecular Pathways and Therapeutic Strategies for Fibrotic Conditions)
Show Figures

Figure 1

3 pages, 5950 KiB  
Correction
Correction: Mohammed et al. Alvespimycin Exhibits Potential Anti-TGF-β Signaling in the Setting of a Proteasome Activator in Rats with Bleomycin-Induced Pulmonary Fibrosis: A Promising Novel Approach. Pharmaceuticals 2023, 16, 1123
by Osama A. Mohammed, Mustafa Ahmed Abdel-Reheim, Lobna A. Saleh, Mohannad Mohammad S. Alamri, Jaber Alfaifi, Masoud I. E. Adam, Alshaimaa A. Farrag, AbdulElah Al Jarallah AlQahtani, Waad Fuad BinAfif, Abdullah A. Hashish, Sameh Abdel-Ghany, Elsayed A. Elmorsy, Hend S. El-wakeel, Ahmed S. Doghish, Rabab S. Hamad and Sameh Saber
Pharmaceuticals 2025, 18(7), 1011; https://doi.org/10.3390/ph18071011 - 7 Jul 2025
Viewed by 298
Abstract
In the original publication [...] Full article
Show Figures

Figure 2

13 pages, 7485 KiB  
Article
Saroglitazar Ameliorates Pulmonary Fibrosis Progression in Mice by Suppressing NF-κB Activation and Attenuating Macrophage M1 Polarization
by Yawen Zhang, Jiaquan Lin, Xiaodong Han and Xiang Chen
Medicina 2025, 61(7), 1157; https://doi.org/10.3390/medicina61071157 - 26 Jun 2025
Viewed by 370
Abstract
Background and Objectives: Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal interstitial lung disease with limited therapeutic options. Current therapies (pirfenidone, nintedanib) exhibit modest efficacy and significant side effects, underscoring the need for novel strategies targeting early pathogenic drivers. Saroglitazar (SGZ), [...] Read more.
Background and Objectives: Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal interstitial lung disease with limited therapeutic options. Current therapies (pirfenidone, nintedanib) exhibit modest efficacy and significant side effects, underscoring the need for novel strategies targeting early pathogenic drivers. Saroglitazar (SGZ), a dual PPARα/γ agonist with anti-inflammatory properties approved for diabetic dyslipidemia, has not been explored for IPF. We aimed to investigate SGZ’s therapeutic potential in pulmonary fibrosis and elucidate its mechanisms of action. Materials and Methods: Using a bleomycin (BLM)-induced murine pulmonary fibrosis model, we administered SGZ therapeutically. A histopathological assessment (H&E, Masson’s trichrome, collagen I immunofluorescence), Western blotting, and qRT-PCR analyzed the fibrosis progression and inflammatory markers. Flow cytometry evaluated the macrophage polarization. In vitro studies used RAW264.7 macrophages stimulated with BLM/LPS and MRC-5 fibroblast co-cultures. The NF-κB/NLRP3 pathway activation was assessed through protein and gene expression. Results: SGZ significantly attenuated BLM-induced histopathological hallmarks, including alveolar wall thickening, collagen deposition, and inflammatory infiltration. Fibrotic markers (OPN, α-SMA) and pro-inflammatory cytokines (IL-1β, TNF-α, IL-6) were downregulated in the SGZ-treated mice. Mechanistically, SGZ suppressed the M1 macrophage polarization (reduced CD86+ populations) and inhibited the NF-κB/NLRP3 pathway activation in the alveolar macrophages. In the RAW264.7 cells, SGZ decreased the NLRP3 inflammasome components (ASC, cleaved IL-1β) and cytokine secretion. Co-cultures demonstrated that the SGZ-treated macrophage supernatants suppressed the fibroblast activation (α-SMA, collagen I) in MRC-5 cells. Conclusions: SGZ attenuates pulmonary fibrosis by suppressing macrophage-driven inflammation via NF-κB/NLRP3 inhibition and disrupting the macrophage–fibroblast crosstalk. These findings nominate SGZ as a promising candidate for preclinical optimization and future clinical evaluation in IPF. Full article
(This article belongs to the Special Issue Pulmonary Fibrosis: Current Understanding and Future Directions)
Show Figures

Figure 1

15 pages, 970 KiB  
Article
Potential Natural Blend Hydrosol TGLON Suppresses the Proliferation of Five Cancer Cell Lines and Also Ameliorates Idiopathic Pulmonary Fibrosis in a Mouse Model
by Wei-Hsiang Huang, Mei-Lin Chang, Ching-Che Lin, Chih-Peng Wang, Feng-Jie Tsai and Chih-Chien Lin
Pharmaceuticals 2025, 18(6), 872; https://doi.org/10.3390/ph18060872 - 11 Jun 2025
Viewed by 1499
Abstract
Background: Cancer and fibrotic diseases represent major global health challenges, underscoring the need for safe, multifunctional natural therapies. Although natural products possess notable anticancer properties, their clinical translation is often hindered by non-selective cytotoxicity toward normal cells. Moreover, their therapeutic potential against chronic [...] Read more.
Background: Cancer and fibrotic diseases represent major global health challenges, underscoring the need for safe, multifunctional natural therapies. Although natural products possess notable anticancer properties, their clinical translation is often hindered by non-selective cytotoxicity toward normal cells. Moreover, their therapeutic potential against chronic conditions such as idiopathic pulmonary fibrosis (IPF) remains insufficiently explored. This study aimed to evaluate the efficacy and safety of a natural hydrosol blend, The Greatest Love of Nature (TGLON), in inhibiting cancer cell proliferation and mitigating IPF. Methods: TGLON, composed of 12 steam-distilled plant hydrosols, was chemically characterized by gas chromatography–mass spectrometry (GC-MS). Its cytotoxicity was assessed using the MTT assay against five human cancer cell lines (A-549, HepG2, MCF-7, MKN-45, and MOLT-4) and normal human lung fibroblasts (MRC-5). In vivo safety and therapeutic efficacy were evaluated in Sprague Dawley rats and a bleomycin-induced IPF mouse model, following protocols approved by the Institutional Animal Care and Use Committee (IACUC). Results: TGLON maintained >90% viability in MRC-5 cells at an 80-fold dilution and significantly inhibited the proliferation of A-549 (41%), HepG2 (84%), MCF-7 (50%), MKN-45 (38%), and MOLT-4 (52%) cells. No signs of toxicity were observed in rats administered TGLON orally at 50% (v/v), 10 mL/kg. In mice, TGLON alleviated bleomycin-induced pulmonary inflammation and fibrosis. Conclusions: TGLON exhibited selective anticancer and anti-fibrotic activities under non-toxic conditions, supporting its potential as a bioactive agent for early-stage disease prevention and non-clinical health maintenance. Full article
(This article belongs to the Special Issue Advances in the Chemical-Biological Knowledge of Essential Oils)
Show Figures

Figure 1

17 pages, 4283 KiB  
Article
SPHK1-S1p Signaling Drives Fibrocyte-Mediated Pulmonary Fibrosis: Mechanistic Insights and Therapeutic Potential
by Fei Lu, Gaoming Wang, Xiangzhe Yang, Jing Luo, Haitao Ma, Liangbin Pan, Yu Yao and Kai Xie
Pharmaceuticals 2025, 18(6), 859; https://doi.org/10.3390/ph18060859 - 9 Jun 2025
Viewed by 631
Abstract
Background: Pulmonary fibrosis (PF) is a progressive interstitial lung disease characterized by chronic inflammation and excessive extracellular matrix deposition, with fibrocytes playing a pivotal role in fibrotic remodeling. This study aimed to identify upstream molecular mechanisms regulating fibrocyte recruitment and activation, focusing on [...] Read more.
Background: Pulmonary fibrosis (PF) is a progressive interstitial lung disease characterized by chronic inflammation and excessive extracellular matrix deposition, with fibrocytes playing a pivotal role in fibrotic remodeling. This study aimed to identify upstream molecular mechanisms regulating fibrocyte recruitment and activation, focusing on the SPHK1 pathway as a potential therapeutic target. Methods: We utilized Mendelian Randomization and phenome-wide association analyses on genes involved in sphingolipid metabolism to identify potential regulators of idiopathic pulmonary fibrosis (IPF). A bleomycin-induced mouse model was employed to examine the role of the SPHK1-S1P axis in fibrocyte recruitment, using SKI-349 to target SPHK1 and FTY720 to antagonize S1PR1. Results: Our analyses revealed SPHK1 as a significant genetic driver of IPF. Targeting SPHK1 and S1PR1 led to a marked reduction in fibrocyte accumulation, collagen deposition, and histopathological fibrosis. Additionally, PAXX and RBKS were identified as downstream effectors of SPHK1. Our protein–protein interaction mapping indicated potential therapeutic synergies with existing anti-fibrotic drug targets. Conclusions: Our findings establish the SPHK1-S1P-S1PR1 axis as a key regulator of fibrocyte-mediated pulmonary fibrosis and support SPHK1 as a promising therapeutic target. Full article
Show Figures

Figure 1

15 pages, 6359 KiB  
Article
An Elastase Inhibitor ShSPI from Centipede Attenuates Bleomycin-Induced Pulmonary Fibrosis
by Xi Lian, Bin Liu, Dan Li, Xinyao Wang, Chengbo Long, Xing Feng, Qiong Liao and Mingqiang Rong
Toxins 2025, 17(5), 213; https://doi.org/10.3390/toxins17050213 - 24 Apr 2025
Viewed by 641
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by the fibrotic thickening of the alveolar walls, resulting in compromised gas exchange, restricted ventilation, and respiratory failure. It has been indicated that elastase inhibitors reduced the severity of IPF by neutralizing excessive [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by the fibrotic thickening of the alveolar walls, resulting in compromised gas exchange, restricted ventilation, and respiratory failure. It has been indicated that elastase inhibitors reduced the severity of IPF by neutralizing excessive elastase levels in the lungs. ShSPI is an elastase inhibitor derived from centipede toxin. The present study evaluates the therapeutic effects of ShSPI in a bleomycin-induced idiopathic pulmonary fibrosis model. According to the results, ShSPI markedly reduced the weight loss, showing the improvement of health status in bleomycin-induced mice. Its robust antifibrotic effects were evidenced by the mitigation of alveolar structural damage, reduction in inflammatory cell infiltration, inhibition of collagen deposition, and suppression of fibrotic nodule formation. ShSPI effectively attenuated inflammatory responses by downregulating pro-inflammatory factors (IL-6, IL-1β, and MCP-1) and upregulating the anti-inflammatory factor interleukin-10 (IL-10). After delivered via inhalation, ShSPI exhibited favorable pharmacokinetic properties. It could be detected at 8 h at doses of 1 mg/kg and achieved maximum plasma concentrations (Cmax) of 188.00 ± 64.40 ng/mL in vivo. At high doses (160 mg/kg), ShSPI maintained a strong safety profile, with no detectable toxicity observed. This feature shows the therapeutic potential of ShSPI in the treatment of idiopathic pulmonary fibrosis and provides valuable evidence for its development as a novel peptide-based therapy. Full article
(This article belongs to the Special Issue Animals Venom in Drug Discovery: A Valuable Therapeutic Tool)
Show Figures

Figure 1

19 pages, 5826 KiB  
Article
Combination Treatment of Timosaponin BII and Pirfenidone Attenuated Pulmonary Fibrosis Through Anti-Inflammatory and Anti-Fibrotic Process in Rodent Pulmonary Fibrosis Model and Cellular Epithelial–Mesenchymal Transition Model
by Xuebin Shen, Yueyue Zheng, Hui Yang, Li Liu, Lizhen Yu, Yuanxiang Zhang, Xiaojun Song, Yuqing He, Runze Jin, Jianhao Jiao, Zhihui Gu, Kefeng Zhai, Sihui Nian and Limin Liu
Molecules 2025, 30(8), 1821; https://doi.org/10.3390/molecules30081821 - 18 Apr 2025
Viewed by 692
Abstract
Pulmonary fibrosis (PF) is a progressive lung disease with a poor prognosis. Pirfenidone (PFD) can slow down the decline of lung function, but defects in efficacy and accompanying side effects limit its application; hence, implementing methods including combination therapy might be a viable [...] Read more.
Pulmonary fibrosis (PF) is a progressive lung disease with a poor prognosis. Pirfenidone (PFD) can slow down the decline of lung function, but defects in efficacy and accompanying side effects limit its application; hence, implementing methods including combination therapy might be a viable option. Given this, we hypothesized that combining timosaponin BII (TS BII) with PFD might offer a more effective treatment approach. Bleomycin-induced rodent PF model and TGF-β1-induced cellular epithelial–mesenchymal transition (EMT) model were applied in the study. The results showed that the combination of TS BII and PFD was more effective in reducing the production of IL-1β, TNF-α, collagen fibers, hydroxyproline, and MDA. Moreover, the combination treatment could better restore levels SOD and GSH-Px. In addition, TS BII combined with PFD could downregulate the expression of NF-κB and the ratio of p-IκBα/IκBα, and modulate the aberrant expression of epithelial–mesenchymal transition markers. In addition, the combination treatment could regulate the intestinal flora of PF mice. It is worth noting that among the above results, there were significant differences (p < 0.05) between the combination group and either the TS BII or PFD monotherapy group. These findings indicate that the combination of TS BII and PFD has a synergistic effect in the treatment of PF and represents a promising treatment strategy. Full article
Show Figures

Figure 1

17 pages, 7507 KiB  
Article
Inhibition of Transglutaminase 2 by a Selective Small Molecule Inhibitor Reduces Fibrosis and Improves Pulmonary Function in a Bleomycin Mouse Model
by Zhuo Wang, Sriniwas Sriram, Cynthia Ugwoke, Zoe Gale, Maral Tabrizi and Martin Griffin
Cells 2025, 14(7), 497; https://doi.org/10.3390/cells14070497 - 26 Mar 2025
Viewed by 883
Abstract
This paper investigates the ability of our selective small molecule TG2 inhibitor 1-155 in reducing fibrosis in a bleomycin-induced pulmonary fibrosis mouse model. Formulated as a fine stable suspension, 1-155 was delivered intranasally (IN) at 3 mg/kg via IN delivery once daily. It [...] Read more.
This paper investigates the ability of our selective small molecule TG2 inhibitor 1-155 in reducing fibrosis in a bleomycin-induced pulmonary fibrosis mouse model. Formulated as a fine stable suspension, 1-155 was delivered intranasally (IN) at 3 mg/kg via IN delivery once daily. It significantly inhibited collagen deposition in the lungs in the bleomycin-challenged mice. Compared to its vehicle control treatment, a significant reduction in a key myofibroblast marker α smooth muscle actin and TG2 was also detected in the 1-155-treated animals. Most importantly, 1-155 treatment significantly improved several key lung function parameters, such as cord compliance, vital capacity, and dynamic compliance, which are comparable to that found for the positive control nintedanib at a much higher dosage of 60 mg/kg twice daily via oral delivery. The 1-155-treated mice showed a trend in improvement of average body weight. For the first time, our study demonstrates the effectiveness of a selective small molecule TG2 inhibitor in reducing pulmonary fibrosis in a pre-clinical model. Importantly, we were able to correlate this effect of 1-155 with the improvement of animal lung function showing the potential of the use of TG2 inhibitors as a therapeutic treatment for fibrotic lung conditions like IPF. Full article
(This article belongs to the Special Issue Organ and Tissue Fibrosis: Molecular Signals and Cellular Mechanisms)
Show Figures

Figure 1

12 pages, 2735 KiB  
Article
PET Imaging of CD206 Macrophages in Bleomycin-Induced Lung Injury Mouse Model
by Volkan Tekin, Yujun Zhang, Clayton Yates, Jesse Jaynes, Henry Lopez, Charles Garvin, Benjamin M. Larimer and Suzanne E. Lapi
Pharmaceutics 2025, 17(2), 253; https://doi.org/10.3390/pharmaceutics17020253 - 14 Feb 2025
Viewed by 1132
Abstract
Background/Objectives: The identification of inflammatory mediators and the involvement of CD206 macrophages in anti-inflammatory responses, along with the synthesis of fibrotic mediators, are crucial for the diagnosis and treatment of Idiopathic Pulmonary Fibrosis (IPF). Methods: In this study, the assessment of [...] Read more.
Background/Objectives: The identification of inflammatory mediators and the involvement of CD206 macrophages in anti-inflammatory responses, along with the synthesis of fibrotic mediators, are crucial for the diagnosis and treatment of Idiopathic Pulmonary Fibrosis (IPF). Methods: In this study, the assessment of 68Ga-labeled linear and cyclic forms of the RP832c peptide, which demonstrate a specific affinity for CD206 macrophages, was performed to evaluate efficacy for CD206 imaging through PET/CT, biodistribution studies, and CD206 staining in a bleomycin-induced lung injury mouse model (BLM). This model serves as a representative framework for inflammation and fibrosis. Results: The findings reveal significant peak PET/CT signals (SUV means), ID/gram values, and CD206 staining scores in lung tissues at one week post bleomycin instillation, likely due to the heightened expression of CD206 in the bleomycin-induced lung injury model. In contrast, the healthy mice exhibited no detectable CD206 staining, lower PET signals, and reduced radiopharmaceutical accumulation in lung tissues at the same timepoint. Conclusions: These findings suggest that both linear and cyclic [68Ga]Ga-RP832c may function as promising PET imaging agents for CD206 macrophages, and thereby a strategy to non-invasively explore the role of macrophages during fibrogenesis. Full article
(This article belongs to the Special Issue Advances in Radiopharmaceuticals for Disease Diagnoses and Therapy)
Show Figures

Figure 1

21 pages, 6345 KiB  
Article
Integrating Metabolomics and Network Analyses to Explore Mechanisms of Geum japonicum var. chinense Against Pulmonary Fibrosis: Involvement of Arachidonic Acid Metabolic Pathway
by Junyan Ran, Qian Wang, Tao Lu, Xiuqing Pang, Shanggao Liao and Xun He
Int. J. Mol. Sci. 2025, 26(4), 1462; https://doi.org/10.3390/ijms26041462 - 10 Feb 2025
Cited by 1 | Viewed by 1211
Abstract
Pulmonary fibrosis (PF) emerges as a significant pulmonary sequelae in the convalescent phase of coronavirus disease 2019 (COVID-19), with current strategies neither specifically preventive nor therapeutic. Geum japonicum var. chinense (GJC) is used as a traditional Chinese medicine to effectively treat various respiratory [...] Read more.
Pulmonary fibrosis (PF) emerges as a significant pulmonary sequelae in the convalescent phase of coronavirus disease 2019 (COVID-19), with current strategies neither specifically preventive nor therapeutic. Geum japonicum var. chinense (GJC) is used as a traditional Chinese medicine to effectively treat various respiratory conditions. However, the protective effects of GJC against PF remains unclear. In the present study, the anti-PF effect of GJC aqueous extract was studied using a PF mouse model induced by bleomycin (BLM). To characterize the metabolite changes related to PF and reveal therapeutic targets for GJC aqueous extract, we performed metabolomic and network analysis on mice lungs. Finally, key targets were then validated by Western blotting. GJC aqueous extract effectively alleviated the onset and progression of lung fibrosis in PF mice by inhibiting inflammatory responses and regulating oxidative stress levels. Integrating serum metabolomics and network analyses showed the arachidonic acid (AA) pathway to be the most important metabolic pathway of GJC aqueous extract against PF. Further validation of AA pathway protein levels showed a significant rise in the levels of ALOX5, PTGS2, CYP2C9, and PLA2G2A in PF lungs. GJC aqueous extract treatment regulated the above changes in metabolic programming. In conclusion, GJC is a promising botanical drug to delay the onset and progression of PF mice. The primary mechanism of action is associated with the comprehensive regulation of metabolites and protein expression related to the AA metabolic pathway. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

18 pages, 6757 KiB  
Article
An FGF2-Derived Short Peptide Attenuates Bleomycin-Induced Pulmonary Fibrosis by Inhibiting Collagen Deposition and Epithelial–Mesenchymal Transition via the FGFR/MAPK Signaling Pathway
by Mengwei Wang, Yuanmeng Sun, Yanzhi Zhao, Xinyi Jiang, Teng Wang, Junye Xie, Xiuling Yu, Shujun Guo, Yibo Zhang, Xiaojia Chen and An Hong
Int. J. Mol. Sci. 2025, 26(2), 517; https://doi.org/10.3390/ijms26020517 - 9 Jan 2025
Viewed by 3716
Abstract
Following the COVID-19 pandemic, the prevalence of pulmonary fibrosis has increased significantly, placing patients at higher risk and presenting new therapeutic challenges. Current anti-fibrotic drugs, such as Nintedanib, can slow the decline in lung function, but their severe side effects highlight the urgent [...] Read more.
Following the COVID-19 pandemic, the prevalence of pulmonary fibrosis has increased significantly, placing patients at higher risk and presenting new therapeutic challenges. Current anti-fibrotic drugs, such as Nintedanib, can slow the decline in lung function, but their severe side effects highlight the urgent need for safer and more targeted alternatives. This study explores the anti-fibrotic potential and underlying mechanisms of an endogenous peptide (P5) derived from fibroblast growth factor 2 (FGF2), developed by our research team. Using a bleomycin-induced pulmonary fibrosis mouse model, we observed that P5 alleviated fibrosis by inhibiting collagen deposition, as confirmed by CT scans and histological staining. In TGF-β-induced cell models, P5 effectively suppressed collagen deposition and epithelial–mesenchymal transition (EMT). Transcriptome analysis highlighted pathways related to receptor binding, extracellular matrix organization, and cell adhesion, with KEGG analysis confirming FGFR/MAPK signaling inhibition as the primary mechanism underlying its anti-fibrotic effects. In summary, our study demonstrates that P5 significantly attenuates pulmonary fibrosis through the inhibition of EMT, collagen deposition, and FGFR/MAPK signaling, providing a promising therapeutic approach for fibrosis. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

49 pages, 2915 KiB  
Review
The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice
by Remo Castro Russo and Bernhard Ryffel
Cells 2024, 13(24), 2058; https://doi.org/10.3390/cells13242058 - 12 Dec 2024
Cited by 3 | Viewed by 4449
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system. Full article
Show Figures

Figure 1

Back to TopTop