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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (389)

Search Parameters:
Keywords = fibrotic lung disease

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5773 KB  
Article
Exploring the Cellular and Molecular Landscape of Idiopathic Pulmonary Fibrosis: Integrative Multi-Omics and Single-Cell Analysis
by Huanyu Jiang, Shujie Wang, Fanghui Zhong and Tao Shen
Biomedicines 2025, 13(9), 2135; https://doi.org/10.3390/biomedicines13092135 - 1 Sep 2025
Abstract
Background/Objectives: Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by lung scarring, impaired function, and high mortality. Effective therapies to reverse fibrosis are lacking. This study aims to uncover the molecular mechanisms of IPF, explore diagnostic biomarkers, and identify therapeutic targets. [...] Read more.
Background/Objectives: Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by lung scarring, impaired function, and high mortality. Effective therapies to reverse fibrosis are lacking. This study aims to uncover the molecular mechanisms of IPF, explore diagnostic biomarkers, and identify therapeutic targets. Methods: Multi-omics data were integrated to identify biomarkers with causal associations to IPF using Mendelian randomization and transcriptomic analysis. Machine learning was employed to construct a diagnostic model, and single-cell transcriptomic analysis determined gene expression patterns in fibrotic lung tissue. Results: Seven core genes (GREM1, UGT1A6, CDH2, TDO2, HS3ST1, ADGRF5, and MPO) were identified, showing strong diagnostic potential (AUC = 0.987, 95% CI: 0.972–0.987). These genes exhibited distinct distribution patterns in fibroblasts, endothelial cells, epithelial cells, macrophages, and dendritic cells. Conclusions: This study highlights key genes driving IPF, involved in pathways related to metabolism, immunity, and inflammation. However, their utility as fluid-based biomarkers remains unproven and requires protein-level validation in prospective cohorts. By integrating genomic, immunological, and cellular insights, it provides a framework for targeted therapies and advances mechanism-based precision medicine for IPF. Full article
(This article belongs to the Special Issue Advanced Research in Interstitial Lung Diseases)
Show Figures

Figure 1

34 pages, 2093 KB  
Review
Fibrotic Disease of the Skin and Lung: Shared Pathways, Environmental Drivers, and Therapeutic Opportunities in a Changing Climate
by Katerina Grafanaki, Alexandros Maniatis, Vasilina Sotiropoulou, Efstathia Pasmatzi and Argyris Tzouvelekis
Int. J. Mol. Sci. 2025, 26(17), 8394; https://doi.org/10.3390/ijms26178394 - 29 Aug 2025
Viewed by 107
Abstract
Fibrotic diseases of the skin and lung, such as systemic sclerosis, hypertrophic scars, keloids, and pulmonary fibrosis, share core molecular mechanisms despite their distinct anatomical settings. Central to their pathogenesis are persistent fibroblast activation, immune dysregulation, ECM remodeling, and failure of resolution pathways, [...] Read more.
Fibrotic diseases of the skin and lung, such as systemic sclerosis, hypertrophic scars, keloids, and pulmonary fibrosis, share core molecular mechanisms despite their distinct anatomical settings. Central to their pathogenesis are persistent fibroblast activation, immune dysregulation, ECM remodeling, and failure of resolution pathways, all modulated by an ever-changing environment and epigenetic regulation. Increasing evidence reveals that chronic injury from air pollution, ultraviolet radiation, climate stressors, and occupational hazards accelerates fibroinflammatory remodeling across these barrier organs. Moreover, shared signaling networks, including TGF-β, IL-4/IL-13, Wnt/β-catenin, and epigenetic regulators like miR-21 and miR-29, suggest convergent fibrotic programs may be subject to cross-organ therapeutic targeting. This review integrates recent insights into the exposome’s role in driving fibrosis, highlights novel RNA- and epigenetic-based interventions, and evaluates the repurposing of antifibrotic agents approved for pulmonary disease within dermatologic contexts. We emphasize the emerging concept of fibrosis-aware precision medicine and propose a unifying framework to guide integrated therapeutic strategies. In the face of global climate change and rising environmental insults, a cross-organ perspective on fibrosis offers a timely and translationally relevant approach to addressing this growing burden on human health. Full article
(This article belongs to the Special Issue Advanced Research of Skin Inflammation and Related Diseases)
Show Figures

Graphical abstract

20 pages, 3232 KB  
Review
Targeting Focal Adhesion Kinase in Lung Diseases: Current Progress and Future Directions
by Ziyu Wan, Zefeng Zhu, Pengbin Wang, Xuan Xu, Tianhao Ma, Huari Li, Lexing Li, Feng Qian and Wei Gu
Biomolecules 2025, 15(9), 1233; https://doi.org/10.3390/biom15091233 - 26 Aug 2025
Viewed by 430
Abstract
Focal adhesion kinase (FAK) is a crucial protein component of focal adhesions (FAs) and belongs to the cytoplasmic non-receptor protein tyrosine kinase family. FAK primarily regulates adhesion signaling and cell migration and is highly expressed in various tumors, including lung, liver, gastric, and [...] Read more.
Focal adhesion kinase (FAK) is a crucial protein component of focal adhesions (FAs) and belongs to the cytoplasmic non-receptor protein tyrosine kinase family. FAK primarily regulates adhesion signaling and cell migration and is highly expressed in various tumors, including lung, liver, gastric, and colorectal cancers, as well as in conditions such as acute lung injury (ALI) and pulmonary fibrosis (PF). Recent research on FAK and its small-molecule inhibitors has revealed that targeting FAK provides a novel approach for treating various lung diseases. FAK inhibitors can obstruct signaling pathways, demonstrating anti-tumor, anti-inflammatory, and anti-fibrotic effects. In lung cancer, FAK inhibitors suppress tumor growth and metastasis; in ALI, they exert protective effects by alleviating inflammatory responses and oxidative stress; and in pulmonary fibrosis, FAK inhibitors reduce fibroblast activation and inhibit collagen deposition. The findings demonstrate promising efficacy and an acceptable safety profile in preclinical models. However, these early-stage results require further validation through clinical studies. Additionally, the underlying mechanisms, as well as the toxic effects and side effects, necessitate further in-depth investigation. Some have progressed to clinical trials (Defactinib (Phase II), PF-562271 (Phase I), CEP-37440 (Phase I), PND-1186 (Phase I), GSK-2256098 (Phase II), BI-853520 (Phase I)), offering potential therapeutic targets for lung diseases. Collectively, these findings establish a foundational basis for the advancement of FAK inhibitor discovery. Emerging methodologies, such as PROTAC degraders and combination regimens, demonstrate significant potential for future research. Based on a comprehensive analysis of the relevant literature from 2015 to the present, this review briefly introduces the structure and function of FAK and discusses recent research advancements regarding FAK and its inhibitors in the context of pulmonary diseases. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

23 pages, 6706 KB  
Article
Oleuropein Ameliorates Bleomycin-Induced Pulmonary Fibrosis in Mice by Targeting TGF-β1 Signaling Pathway
by Liang Zhang, Zhigang Liu, Yayue Hu, Xueze Liu, Zhongyi Yang, Yuming Liu, Ran Jiao, Xiaoting Gu, Weidong Zhang, Xiaohe Li and Honggang Zhou
Biomolecules 2025, 15(9), 1211; https://doi.org/10.3390/biom15091211 - 22 Aug 2025
Viewed by 312
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by the accumulation of fibrotic tissue in the lungs, leading to impaired gas exchange and respiratory failure, with a poor prognosis and limited treatment options. Oleuropein, a compound extracted from olive [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by the accumulation of fibrotic tissue in the lungs, leading to impaired gas exchange and respiratory failure, with a poor prognosis and limited treatment options. Oleuropein, a compound extracted from olive leaves, demonstrates a range of pharmacological activities, including benefits for non-alcoholic fatty liver disease and cardiac fibrosis. This study investigates the therapeutic potential of oleuropein for IPF and its underlying mechanisms. We first established a bleomycin-induced mouse model of pulmonary fibrosis and evaluated the in vivo efficacy of oleuropein. Our findings demonstrated that oleuropein significantly alleviated lung fibrosis and improved pulmonary function. Through in vitro experiments, we found that oleuropein inhibited TGF-β1-induced fibroblast migration, activation, autophagy, and apoptotic resistance, and mechanistically, oleuropein could regulate the TGF-β1/Smad and TGF-β1/mTOR signaling pathways in fibroblasts. Additionally, molecular docking analysis indicated that FAP-α is a potential target of oleuropein, displaying strong binding affinity. The effects of oleuropein on fibroblasts were markedly disrupted in FAP-α knockout cells. In conclusion, oleuropein exerts its beneficial effects by targeting FAP-α and inhibiting TGF-β1-related signaling pathways, improving the pathological characteristics of pulmonary fibrosis in mouse models, and demonstrating promising application prospects for the treatment of IPF. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

10 pages, 930 KB  
Case Report
Fibrotic Chronic Eosinophilic Pneumonia Treated with an Anti-IL-5 Monoclonal Antibody: A Case Report
by Umberto Semenzato, Daniele Previtero, Gioele Castelli, Eleonora Ruzzini, Elisabetta Cocconcelli, Mariaenrica Tinè, Roberto Padoan, Elisabetta Balestro, Simonetta Baraldo and Paolo Spagnolo
Biologics 2025, 5(3), 23; https://doi.org/10.3390/biologics5030023 - 14 Aug 2025
Viewed by 300
Abstract
Background: Chronic eosinophilic pneumonia (CEP) is a rare inflammatory lung disease typically responsive to glucocorticoids, but is prone to relapse and, in some cases, progressive deterioration. A subset of patients develops fibrosing CEP, a distinct phenotype characterized by irreversible parenchymal remodeling and declining [...] Read more.
Background: Chronic eosinophilic pneumonia (CEP) is a rare inflammatory lung disease typically responsive to glucocorticoids, but is prone to relapse and, in some cases, progressive deterioration. A subset of patients develops fibrosing CEP, a distinct phenotype characterized by irreversible parenchymal remodeling and declining lung function, for which no standard treatment exists. Although biologic therapies targeting interleukin-5 (IL-5) are effective in relapsing CEP, their role in fibrosing forms remains unclear. Case Presentation: We report the case of a 43-year-old man with idiopathic CEP initially treated with systemic glucocorticoids, which were discontinued due to severe adverse effects. Despite subsequent therapy with inhaled steroids and azathioprine, the disease relapsed and progressed to a fibrosing phenotype, as confirmed by radiologic and functional assessments. An off-label treatment with subcutaneous mepolizumab, 100 mg, every 4 weeks was started. After eight months of therapy, the patient achieved clinical stability, improved lung function, and the radiologic stabilization of fibrotic changes, without the need for any further treatment with a corticosteroid. Conclusions: This is, to the best of our knowledge, the first documented case of fibrosing CEP treated with an anti-IL-5 monoclonal antibody, highlighting its potential role as a steroid-sparing agent and immunomodulator even in the fibrotic phase of disease. Further research is warranted to define the place of biologics in the management of CEP with a fibrosing evolution and possible combinations with antifibrotic drugs. Full article
(This article belongs to the Section Monoclonal Antibodies)
Show Figures

Figure 1

29 pages, 2765 KB  
Review
Breathless Aftermath: Post-COVID-19 Pulmonary Fibrosis
by Dharanya Muthiah, Kishore Vaddadi and Lin Liu
Viruses 2025, 17(8), 1098; https://doi.org/10.3390/v17081098 - 9 Aug 2025
Viewed by 2251
Abstract
A significant number of individuals recovering from COVID-19 continue to experience persistent symptoms, collectively referred to as Post-Acute Sequelae of SARS-CoV-2 infection (PASC), or long COVID. Among these complications, post-COVID-19 pulmonary fibrosis (PC19-PF) is one of the most severe long-term outcomes, characterized by [...] Read more.
A significant number of individuals recovering from COVID-19 continue to experience persistent symptoms, collectively referred to as Post-Acute Sequelae of SARS-CoV-2 infection (PASC), or long COVID. Among these complications, post-COVID-19 pulmonary fibrosis (PC19-PF) is one of the most severe long-term outcomes, characterized by progressive lung scarring, chronic respiratory impairment, and reduced quality of life. Despite the increasing prevalence of PC19-PF, its underlying mechanisms remain poorly understood. In this review, we provide a comprehensive overview of PC19-PF, including its epidemiology, clinical manifestations, diagnostic strategies, and mechanistic insights. Additionally, we highlight the shared pathways between PC19-PF and other fibrotic lung diseases and discuss emerging therapeutic strategies. This review consolidates emerging insights from both clinical and experimental studies to advance our understanding of PC19-PF pathogenesis and guide the development of mechanism-based therapeutic approaches. Full article
(This article belongs to the Special Issue Lung Immunity to Viral Infections)
Show Figures

Figure 1

11 pages, 311 KB  
Article
Characteristics and Outcomes of Diffuse Interstitial Pneumonias Discovered in the ICU: A Retrospective Monocentric Study—The “IPIC” (Interstitial Pneumonia in Intensive Care) Study
by Damien Eckert, Julien Bermudez, Marc Leone, Mathieu Di Bisceglie and Florent Montini
Diagnostics 2025, 15(16), 1995; https://doi.org/10.3390/diagnostics15161995 - 9 Aug 2025
Viewed by 361
Abstract
Background/Objectives: Interstitial lung disease (ILD) is a heterogenous group of disorders characterised by an association of inflammatory and fibrotic abnormalities of the lung. Acute respiratory failure (ARF) may represent the initial picture of the disease. This study aims to highlight the diagnosis [...] Read more.
Background/Objectives: Interstitial lung disease (ILD) is a heterogenous group of disorders characterised by an association of inflammatory and fibrotic abnormalities of the lung. Acute respiratory failure (ARF) may represent the initial picture of the disease. This study aims to highlight the diagnosis of ILD in the intensive care unit (ICU) and to describe the epidemiological, prognostic, and imaging features of patients diagnosed for the first time with ILD in the ICU. Methods: We conducted a single-centre retrospective study. We screened all 2459 patients admitted to our ICU from October 2017 to February 2020. The inclusion criteria consisted of the ILD diagnosis criteria. For each patient, clinical data and lung computed tomography scan patterns were analysed. The selected cases were then reviewed by an expert team at the tertiary care teaching hospital of Marseille (Hôpital Nord, Marseille, France). Results: During the study period, 26 ICU patients were diagnosed with ILD and 20 cases were confirmed by the expert team. The most frequent diagnoses were idiopathic ILD (n = 7, 35%), auto-immune disease-related ILD (n = 7, 35%), exposure-related ILD (n = 3, 15%), and carcinomatous lymphangitis (n = 3, 15%). Fifteen patients were men (75%), with a mean age of 70 (62–72) years. The median SOFA score was 4 (3–7), and 16 (80%) patients received invasive mechanical ventilation. The mean ratio of the oxygen pressure to the fraction of inspired oxygen was 174 (148–198) mmHg. The ICU mortality rate of our cohort was significantly higher than the average ICU mortality (65% vs. 26%, p < 0.003). The mortality rate was lower among the subgroup of auto-immune disease-related ILD (57%). Conclusions: We conducted a single-centre cohort study of patients diagnosed with ILD in the ICU. This rare cause of ARF was associated with poor outcome in the ICU, but auto-immune disease-related ILD seemed to have a better prognosis. High-resolution lung CT and identification of lesion patterns are the cornerstones of the diagnosis. Improved knowledge of ILD and multidisciplinary discussion (MDD) involving radiologists, pneumologists, and intensivists may result in an earlier diagnosis and eventually improved treatments. Full article
Show Figures

Figure 1

22 pages, 9750 KB  
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
Viewed by 490
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

19 pages, 967 KB  
Review
Hematologic and Immunologic Overlap Between COVID-19 and Idiopathic Pulmonary Fibrosis
by Gabriela Mara, Gheorghe Nini, Stefan Marian Frenț and Coralia Cotoraci
J. Clin. Med. 2025, 14(15), 5229; https://doi.org/10.3390/jcm14155229 - 24 Jul 2025
Viewed by 591
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral infections and chronic lung disease, this review offers a focused analysis of the shared hematologic and immunologic mechanisms between COVID-19 and IPF. Our aim is to better understand how SARS-CoV-2 infection may worsen disease progression in IPF and identify converging pathophysiological pathways that may inform clinical management. We conducted a narrative synthesis of the peer-reviewed literature from PubMed, Scopus, and Web of Science, focusing on clinical, experimental, and pathological studies addressing immune and coagulation abnormalities in both COVID-19 and IPF. Both diseases exhibit significant overlap in inflammatory and fibrotic signaling, particularly via the TGF-β, IL-6, and TNF-α pathways. COVID-19 amplifies coagulation disturbances and endothelial dysfunction already present in IPF, promoting microvascular thrombosis and acute exacerbations. Myeloid cell overactivation, impaired lymphocyte responses, and fibroblast proliferation are central to this shared pathophysiology. These synergistic mechanisms may accelerate fibrosis and increase mortality risk in IPF patients infected with SARS-CoV-2. This review proposes an integrative framework for understanding the hematologic and immunologic convergence of COVID-19 and IPF. Such insights are essential for refining therapeutic targets, improving prognostic stratification, and guiding early interventions in this high-risk population. Full article
(This article belongs to the Special Issue Chronic Lung Conditions: Integrative Approaches to Long-Term Care)
Show Figures

Figure 1

9 pages, 418 KB  
Review
The Occult Cascade That Leads to CTEPH
by Charli Fox and Lavannya M. Pandit
BioChem 2025, 5(3), 22; https://doi.org/10.3390/biochem5030022 - 23 Jul 2025
Viewed by 276
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare, progressive form of pre-capillary pulmonary hypertension characterized by persistent, organized thromboemboli in the pulmonary vasculature, leading to vascular remodeling, elevated pulmonary artery pressures, right heart failure, and significant morbidity and mortality if untreated. Despite advances, [...] Read more.
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare, progressive form of pre-capillary pulmonary hypertension characterized by persistent, organized thromboemboli in the pulmonary vasculature, leading to vascular remodeling, elevated pulmonary artery pressures, right heart failure, and significant morbidity and mortality if untreated. Despite advances, CTEPH remains underdiagnosed due to nonspecific symptoms and overlapping features with other forms of pulmonary hypertension. Basic Methodology: This review synthesizes data from large international registries, epidemiologic studies, translational research, and multicenter clinical trials. Key methodologies include analysis of registry data to assess incidence and risk factors, histopathological examination of lung specimens, and molecular studies investigating endothelial dysfunction and inflammatory pathways. Diagnostic modalities and treatment outcomes are evaluated through observational studies and randomized controlled trials. Recent Advances and Affected Population: Research has elucidated that CTEPH arises from incomplete resolution of pulmonary emboli, with subsequent fibrotic transformation mediated by dysregulated TGF-β/TGFBI signaling, endothelial dysfunction, and chronic inflammation. Affected populations are typically older adults, often with prior venous thromboembolism, splenectomy, or prothrombotic conditions, though up to 25% have no history of acute PE. The disease burden is substantial, with delayed diagnosis contributing to worse outcomes and higher societal costs. Microvascular arteriopathy and PAH-like lesions in non-occluded vessels further complicate the clinical picture. Conclusions: CTEPH is now recognized as a treatable disease, with multimodal therapies—surgical endarterectomy, balloon pulmonary angioplasty, and targeted pharmacotherapy—significantly improving survival and quality of life. Ongoing research into molecular mechanisms and biomarker-driven diagnostics promises earlier identification and more personalized management. Multidisciplinary care and continued translational investigation are essential to further reduce mortality and optimize outcomes for this complex patient population. Full article
(This article belongs to the Special Issue Feature Papers in BioChem, 2nd Edition)
Show Figures

Figure 1

28 pages, 944 KB  
Review
Amphiregulin in Fibrotic Diseases and Cancer
by Tae Rim Kim, Beomseok Son, Chun Geun Lee and Han-Oh Park
Int. J. Mol. Sci. 2025, 26(14), 6945; https://doi.org/10.3390/ijms26146945 - 19 Jul 2025
Cited by 1 | Viewed by 913
Abstract
Fibrotic disorders pose a significant global health burden due to limited treatment options, creating an urgent need for novel therapeutic strategies. Amphiregulin (AREG), a low-affinity ligand for the epidermal growth factor receptor (EGFR), has emerged as a key mediator of fibrogenesis through dual [...] Read more.
Fibrotic disorders pose a significant global health burden due to limited treatment options, creating an urgent need for novel therapeutic strategies. Amphiregulin (AREG), a low-affinity ligand for the epidermal growth factor receptor (EGFR), has emerged as a key mediator of fibrogenesis through dual signaling pathways. Unlike high-affinity EGFR ligands, AREG induces sustained signaling that activates downstream effectors and promotes the integrin-mediated activation of transforming growth factor (TGF)-β. This enables both canonical and non-canonical EGFR signaling pathways that contribute to fibrosis. Elevated AREG expression correlates with disease severity across multiple organs, including the lungs, kidneys, liver, and heart. The therapeutic targeting of AREG has shown promising antifibrotic and anticancer effects, suggesting a dual-benefit strategy. The increasing recognition of the shared mechanisms between fibrosis and cancer further supports the development of unified treatment approaches. The inhibition of AREG has been shown to sensitize fibrotic tumor microenvironments to chemotherapy, enhancing combination therapy efficacy. Targeted therapies, such as Self-Assembled-Micelle inhibitory RNA (SAMiRNA)-AREG, have demonstrated enhanced specificity and favorable safety profiles in preclinical studies and early clinical trials. Personalized treatment based on AREG expression may improve clinical outcomes, establishing AREG as a promising precision medicine target for both fibrotic and malignant diseases. This review aims to provide a comprehensive understanding of AREG biology and evaluate its therapeutic potential in fibrosis and cancer. Full article
Show Figures

Figure 1

14 pages, 1329 KB  
Article
Cytokine Profiles of Bronchoalveolar Lavage in Patients with Interstitial Lung Diseases and Non-Allergic Asthma
by Dana Greif Lenarčič, Urska Bidovec Stojković, Pia Kristanc, Peter Kopač, Mateja Marc Malovrh, Izidor Kern, Katarina Osolnik and Peter Korošec
Int. J. Mol. Sci. 2025, 26(14), 6831; https://doi.org/10.3390/ijms26146831 - 16 Jul 2025
Viewed by 477
Abstract
Diagnosing and prognosing immune-mediated airway diseases, like hypersensitivity pneumonitis (HP) and sarcoidosis, is complicated due to their overlapping symptoms and the lack of definitive biomarkers. Hence, we wanted to compare bronchoalveolar lavage (BAL) cytokine and chemokine profiles from 92 patients with different immune-mediated [...] Read more.
Diagnosing and prognosing immune-mediated airway diseases, like hypersensitivity pneumonitis (HP) and sarcoidosis, is complicated due to their overlapping symptoms and the lack of definitive biomarkers. Hence, we wanted to compare bronchoalveolar lavage (BAL) cytokine and chemokine profiles from 92 patients with different immune-mediated and inflammatory airway diseases, namely, HP, sarcoidosis, non-allergic asthma, amiodarone lung, and EGPA. We also compared pulmonary function parameters, BAL’s cellularity, and lymphocyte immunophenotypes. We found significant differences across all measured lung functions (VC, VC%, FEV1, FEV1%, and Tiff%) and in the number of macrophages, lymphocytes, neutrophils, and eosinophils. Furthermore, we showed significant differences in CD4, CD8, and CD4/8 across all included ILDs and OLDs; however, no significant differences were found in CD3, CD19, NK, or NKT. We identified nine biomarkers (IL-1β, IL-6, IL-8, IL-13, VEGF, angiogenin, C4a, RANTES, and MCP-1) that significantly differ in the BAL of patients with HP and sarcoidosis and showed that RANTES and IL-6 are associated with fibrotic outcome. We have demonstrated that interstitial and obstructive lung diseases differ in cytokine and cellular lung imprint, which may, in the future, enable the determination of the disease subtype and thus the identification of targets for the treatment of individuals or subgroups within diseases. Full article
Show Figures

Figure 1

18 pages, 8645 KB  
Article
CIC-Rearranged Sarcoma: A Clinical and Pathological Study of a Peculiar Entity
by Ward Maaita, Nabil Hasasna, Sameer Yaser, Yacob Saleh, Ramiz Abu-Hijlih, Wafa Asha, Hadeel Halalsheh, Samer Abdel Al, Maysa Al-Hussaini and Omar Jaber
Diagnostics 2025, 15(14), 1758; https://doi.org/10.3390/diagnostics15141758 - 11 Jul 2025
Viewed by 865
Abstract
Background: CIC-rearranged sarcoma is a rare and aggressive type of undifferentiated round cell tumor characterized by CIC gene fusion, most commonly CIC::DUX4. This study presents a series of eleven cases, highlighting their clinicopathological features. Methods: Pathology records (2019 to 2024) [...] Read more.
Background: CIC-rearranged sarcoma is a rare and aggressive type of undifferentiated round cell tumor characterized by CIC gene fusion, most commonly CIC::DUX4. This study presents a series of eleven cases, highlighting their clinicopathological features. Methods: Pathology records (2019 to 2024) were searched using “sarcoma with CIC”, identifying eleven cases, of which seven referred cases were initially misdiagnosed. Pathological and clinical analysis was conducted. Treatment was dictated upon multidisciplinary panel discussion based on tumor stage. Follow-up data (1–25 months) was available for all patients. Results: The cohort included six males and five females, with a median age of 43 years (range;14–53), with nine in soft tissue and two in bone. Tumor size ranged from 3.5 cm to 20.0 cm (mean: 9.8 cm). Most cases showed sheets of undifferentiated round- to oval-shaped cells. Two cases showed an Ewing-like pattern, and one case showed spindle cells in a fibrotic stroma transitioning to epithelioid cells. Necrosis was present in nine cases, and mitotic count ranged from 2 to 38/ 10HPFs (mean = 14.2). CD99 was positive in (10/11) cases and WT-1 in (6/9). NKX2.2, S100, and MDM2 were positive in rare cases. CIC::DUX4 fusion was detected in four cases. FISH for CIC gene rearrangement was positive in seven cases, two of them confirmed by methylation analysis. Metastasis at diagnosis was common (n = 8), primarily in the lungs, with later metastasis to the brain and bone. At time of final analysis, eight patients died within a median of 10 months (range: 1–19 months), while three were alive, two with stable disease (for a period of 6 and 25 months) and one with progression after 10 months. Significant correlation was seen between overall survival and the presence of metastasis at diagnosis (p value = 0.03). Conclusions: CIC-rearranged sarcomas are rare, high-grade tumors with predilection for soft tissue. Misdiagnosis is frequent, necessitating molecular confirmation. These tumors are treatment-resistant, often present with lung metastasis, and carry a poor prognosis, especially with initial metastasis. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

17 pages, 390 KB  
Article
The Role of Serum Prolidase Activity, MMP-1, MMP-7, and TGF-β Values in the Prediction of Early Fibrosis in Patients with Moderate to Severe COVID-19
by Didem Dogu Zengin, Dilek Ergun, Burcu Yormaz, Recai Ergun, Halil Guven, Muslu Kazim Korez, Halil Ozer, Ali Unlu, Baykal Tulek and Fikret Kanat
Viruses 2025, 17(7), 954; https://doi.org/10.3390/v17070954 - 6 Jul 2025
Viewed by 580
Abstract
Background: This study aims to identify predictive factors for pulmonary fibrosis development in COVID-19 patients by analyzing thorax CT (computed tomography) findings, serum prolidase activity, MMP-1, MMP-7, TGF-β values, laboratory findings, and demographic characteristics. Materials and methods: The investigation involved 68 patients, both [...] Read more.
Background: This study aims to identify predictive factors for pulmonary fibrosis development in COVID-19 patients by analyzing thorax CT (computed tomography) findings, serum prolidase activity, MMP-1, MMP-7, TGF-β values, laboratory findings, and demographic characteristics. Materials and methods: The investigation involved 68 patients, both male and female, aged 18 years and older, who were volunteers and had been diagnosed with confirmed COVID-19. The pulmonologist and the radiologist evaluated the thorax CT by consensus. Patients were evaluated in two categories, group 1 and group 2, based on the status of fibrotic changes, and 3-month fibrosis scores were calculated. Findings in both lungs were calculated and noted for the lobes, considering lobar spread. Correlations between quantitative parameters were assessed with Spearman’s rho correlation coefficient. Comparisons between independent samples were evaluated using either the independent sample t-test or the Mann–Whitney U test. We evaluated the relationship between categorical variables using the Pearson chi-square test and Fisher’s exact test. Results: Serum prolidase activity, MMP-1, MMP-7, and TGF-β biomarkers were not statistically significant among groups. LDH was found to be significantly high in the group with fibrotic changes. Additionally, the group with fibrotic changes also had higher levels of fibrinogen. The percentage of neutrophils, the severity of the disease, muscle–joint pain and fatigue symptoms, and the length of hospitalization stay were correlated with the total scores of fibrosis at the third month. In the group with fibrotic changes, the duration of muscle–joint pain and fatigue symptoms and the length of hospitalization were longer than in the other group. Conclusions: The group with fibrotic changes showed an increase in biomarkers. However, this increase did not reach a statistically significant level, suggesting that the third month may be an early period for these changes. The group with fibrotic changes showed high levels of LDH, one of the most important laboratory parameters of pulmonary fibrosis risk factors, along with fibrinogen, suggesting that these parameters are valuable in predicting pulmonary fibrosis. Patients with fibrotic changes can experience specific symptoms, commonly seen in COVID-19. Full article
(This article belongs to the Special Issue SARS-CoV-2, COVID-19 Pathologies, Long COVID, and Anti-COVID Vaccines)
Show Figures

Figure 1

24 pages, 6515 KB  
Article
The Effects of Different Types of Exercise on Pulmonary Inflammation and Fibrosis in Mice with Type 2 Diabetes Mellitus
by Haoyang Gao, Xiaotong Ma, Ze Wang, Danlin Zhu, Yifan Guo, Linlin Zhao and Weihua Xiao
Cells 2025, 14(13), 1026; https://doi.org/10.3390/cells14131026 - 4 Jul 2025
Viewed by 886
Abstract
Background: Diabetic lung disease, characterized by inflammation and fibrosis, is an emerging chronic complication of type 2 diabetes mellitus (T2DM). However, systematic studies on the effects of exercise interventions remain limited. This study aimed to investigate the impact of different exercise types (swimming, [...] Read more.
Background: Diabetic lung disease, characterized by inflammation and fibrosis, is an emerging chronic complication of type 2 diabetes mellitus (T2DM). However, systematic studies on the effects of exercise interventions remain limited. This study aimed to investigate the impact of different exercise types (swimming, resistance training, and high-intensity interval training [HIIT]) on pulmonary inflammation and fibrosis in T2DM mice, and to explore underlying molecular mechanisms. Methods: A T2DM mouse model was established by a high-fat diet (HFD) combined with streptozotocin (STZ) induction. Mice were randomly divided into sedentary control, swimming, resistance training, and HIIT groups, and underwent 8 weeks of exercise intervention. After the intervention, body composition was assessed. Lung histopathological changes were evaluated by hematoxylin&eosin (HE) and Masson staining. Inflammatory cytokines, fibrosis markers, and the expression of the TGF-β1/Smad signaling pathway were detected. Macrophage infiltration and polarization were also analyzed. Results: Exercise intervention improved body composition and reduced oxidative stress in T2DM mice. All three exercise modalities downregulated inflammatory cytokine expression, inhibited macrophage activation and M1 polarization, and promoted M2 polarization. Additionally, exercise improved lung tissue structure, reduced collagen deposition, and decreased the expression of fibrosis-related markers. Furthermore, anti-fibrotic effects were mediated by suppression of the TGF-β1/Smad signaling pathway and inhibition of epithelial-mesenchymal transition (EMT). Among the interventions, HIIT demonstrated the strongest inhibitory effect on the TGF-β1/Smad pathway, while swimming showed the most significant anti-inflammatory benefits. Conclusions: Different types of exercise effectively alleviate pulmonary inflammation and fibrosis in T2DM mice. These effects are closely related to the inhibition of oxidative stress, regulation of macrophage polarization, and suppression of TGF-β1/Smad signaling activation, with swimming and HIIT demonstrating superior protective benefits. Full article
Show Figures

Figure 1

Back to TopTop