1
|
Zhou Y, Su W, Xu M, Zhang A, Li S, Guo H, Gong K, Lu K, Yu X, Zhu J, Zhu Q, Liu C. Maimendong decoction modulates the PINK1/Parkin signaling pathway alleviates type 2 alveolar epithelial cells senescence and enhances mitochondrial autophagy to offer potential therapeutic effects for idiopathic pulmonary fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119568. [PMID: 40037475 DOI: 10.1016/j.jep.2025.119568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/21/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Maimendong decoction (MMDD) originates from the ancient Chinese medical text Synopsis of the Golden Chamber and is a well-established remedy for treating lung diseases. It has demonstrated efficacy in the long-term clinical management of idiopathic pulmonary fibrosis (IPF); however, its underlying mechanisms remain unclear. AIM OF THE STUDY This study investigates whether MMDD alleviates IPF by reducing type 2 alveolar epithelial cell (AEC2) senescence and enhancing mitochondrial autophagy. It also explores whether these effects are mediated through the PTEN-induced putative kinase 1 (PINK1)/Parkinson juvenile disease protein 2 (Parkin) pathway. MATERIALS AND METHODS An IPF mouse model was established with bleomycin (BLM). Mice were administered MMDD, pirfenidone (PFD), or saline for 7 or 28 days. Body weight, lung coefficient, and lung appearance were monitored, and lung tissue pathology was assessed. The expression levels of p53, p21, p16, SA-β-gal activity, and senescence-associated secretory phenotype (SASP) markers were measured. Ultrastructural changes in AEC2 mitochondria were analyzed using transmission electron microscopy. Protein levels of autophagy markers sequestosome-1 and light chain 3 were assessed. The protein levels of PINK1, Parkin, and phosphorylated Parkin were further assessed using network pharmacology analysis and molecular docking technology. RESULTS MMDD alleviated BLM-induced IPF by improving body weight, lung appearance, and histopathological features. It reduced AEC2 senescence markers, including p53, p21, p16, SA-β-gal, and SASP, while enhancing mitochondrial autophagy and repairing mitochondrial damage. Network pharmacology and molecular docking identified PINK1 as a major target, and Western blot (WB) analysis confirmed that MMDD regulates the PINK1/Parkin signaling pathway in the treatment of IPF. CONCLUSIONS MMDD regulates the PINK1/Parkin signaling pathway, alleviates AEC2 senescence, and enhances mitochondrial autophagy, providing significant therapeutic potential for IPF treatment.
Collapse
Affiliation(s)
- Yuhe Zhou
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Wen Su
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Mengzhen Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Aijun Zhang
- Traditional Chinese Medicine Research Institute, Shandong Hongjitang Pharmaceutical Group Co, Ltd.Jinan, Jinan, 250100, China.
| | - Shaoli Li
- Jinan Lixia District People's Hospital, Jinan, 250014, China.
| | - Hong Guo
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Kai Gong
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Kaihui Lu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Xin Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Jiang Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Liu R, Wu T, Zhou W, Zhu A, Liao W, Ding K. A Novel Polysaccharide from the Flowers of Lilium lancifolium Alleviates Pulmonary Fibrosis In Vivo and In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7774-7787. [PMID: 40114341 DOI: 10.1021/acs.jafc.4c11703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Lily flowers are widely used in China for lung nourishment; however, their active ingredients remain unknown. To address this question, we isolated a novel polysaccharide (L005-B) from the flowers of Lilium lancifolium. Its backbone is comprised of Glcp, Galp, and 1,2-linked α-Rhap. The branch is composed of Xyl and T-α-Glcp residues substituted at the C-4 position of Rhap, along with portions of Glcp, Galp, Araf, and GlcpA residues substituted at the C-4 position of glucose or the C-3 position of galactose. Bioactivity study showed that L005-B alleviated fibrosis-associated protein (fibronectin, collagen, α-SMA) expression in TGF-β1-induced human fibroblast cells (MRC-5). Moreover, L005-B significantly inhibited the epithelial-mesenchymal transition of the human alveolar type II epithelial cell. More importantly, L005-B dramatically improved bleomycin-induced histopathological changes and attenuated the pulmonary index and hydroxyproline contents. Taken together, our findings revealed that L005-B may serve as a promising leading compound for the development of novel antipulmonary fibrosis therapeutics.
Collapse
Affiliation(s)
- Renjie Liu
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19Auquan Road, Beijing 100049, China
| | - Tong Wu
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19Auquan Road, Beijing 100049, China
| | - Wanqi Zhou
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Anming Zhu
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Wenfeng Liao
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19Auquan Road, Beijing 100049, China
| | - Kan Ding
- Carbohydrate Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19Auquan Road, Beijing 100049, China
- ZhongShan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan 528400, China
| |
Collapse
|
3
|
Allison MB, Catana C, Zhou IY, Caravan P, Montesi SB. Molecular Imaging of Pulmonary Fibrosis. J Nucl Med 2025; 66:502-505. [PMID: 40015916 PMCID: PMC11960603 DOI: 10.2967/jnumed.124.267852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Fibrosing lung diseases affect over 160,000 individuals in the United States alone and can carry a prognosis that is worse than many cancers. Antifibrotic treatments modify only the rate of fibrosis progression, and more effective therapies are urgently needed. Molecular imaging enables visualization of disease pathogenesis in progress. It provides a noninvasive means to monitor and quantify dysregulated molecular fibrotic pathways and shows great promise in aiding the diagnosis and disease activity monitoring of pulmonary fibrosis. Here, we review molecular imaging probes under development for use in pulmonary fibrosis. We provide our opinion on current challenges in translating preclinical molecular imaging probes into clinical successes, as well as future directions for expanding their use in drug development.
Collapse
Affiliation(s)
- Margaret B Allison
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ciprian Catana
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
- A.A. Martinos Center for Biomedical Imaging, Boston, Massachusetts; and
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Iris Y Zhou
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
- A.A. Martinos Center for Biomedical Imaging, Boston, Massachusetts; and
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter Caravan
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
- A.A. Martinos Center for Biomedical Imaging, Boston, Massachusetts; and
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts;
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
Liu Y, Zhang J, Hu Y, Liu Z, Yang Z, Jiao R, Liu X, Li X, Sang F. BI 1015550 Improves Silica-Induced Silicosis and LPS-Induced Acute Lung Injury in Mice. Molecules 2025; 30:1311. [PMID: 40142089 PMCID: PMC11946787 DOI: 10.3390/molecules30061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Silicosis is an interstitial lung disease (ILD) caused by prolonged inhalation of silica particles. Acute lung injury (ALI) is a critical clinical syndrome involving bilateral lung infiltration and acute hypoxic respiratory failure. However, there is currently no effective treatment for these two diseases. Previous research has established that cyclic adenosine monophosphate (cAMP) is pivotal in the pathogenesis of silicosis and acute lung injury. Phosphodiesterase 4 (PDE4) is a hydrolase enzyme of cAMP, and BI 1015550, as an inhibitor of PDE4B, is expected to be a candidate drug for treating both. BI 1015550 has shown certain anti-inflammatory and anti-fibrotic properties in systemic sclerosis-associated interstitial lung disease (SSc-ILD) and idiopathic pulmonary fibrosis (IPF), but there is a lack of research on silicosis and acute lung injury. In this research, we successfully synthesized BI 1015550 autonomously and demonstrated that it could significantly improve lung fibrosis and inflammation in a silica-induced silicosis mouse model. Furthermore, we found that BI 1015550 could also alleviate lung inflammation in a Lipopolysaccharide (LPS)-induced acute lung injury mouse model. The mechanism of action may involve the regulation of cAMP-related signaling pathways.
Collapse
Affiliation(s)
- Yuming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Jing Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China;
| | - Yayue Hu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Zhigang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Zhongyi Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Ran Jiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Xueze Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China;
| |
Collapse
|
5
|
Zhang W, Wang R, Guo R, Yi Z, Wang Y, Wang H, Li Y, Li X, Song J. The multiple biological activities of hyperoside: from molecular mechanisms to therapeutic perspectives in neoplastic and non-neoplastic diseases. Front Pharmacol 2025; 16:1538601. [PMID: 40098612 PMCID: PMC11911483 DOI: 10.3389/fphar.2025.1538601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
In recent years, hyperoside (quercetin 3-O-β-D-galactopyranoside) has garnered significant attention due to its diverse biological effects, which include vasoprotective, antioxidant, anti-inflammatory, and anti-tumor properties. Notably, hyperoside has shown remarkable potential in cancer therapy by targeting multiple mechanisms; it induces apoptosis, inhibits proliferation, blocks angiogenesis, and reduces the metastatic potential of cancer cells. Furthermore, hyperoside enhances the sensitivity of cancer cells to chemotherapy by modulating key signaling pathways. Beyond neoplastic diseases, hyperoside also presents promising therapeutic applications in managing non-cancerous conditions such as diabetes, Alzheimer's disease, and pulmonary fibrosis. This review comprehensively examines the molecular mechanisms underlying hyperoside's anti-cancer effects and highlights its role in the treatment of cancers, including lung and colorectal cancers. Additionally, it explores the latest research on hyperoside's potential in addressing non-neoplastic conditions, such as pulmonary fibrosis, diabetes, and Parkinson's disease. By summarizing current findings, this review underscores the unique therapeutic value of hyperoside and its potential as a multifunctional treatment in both neoplastic and non-neoplastic contexts.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Zhongquan Yi
- Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|
6
|
Naiel S, Dowdall N, Zhou Q, Ali P, Hayat A, Vierhout M, Wong EY, Couto R, Yépez B, Seifried B, Moquin P, Kolb MR, Ask K, Hoare T. Modulating pro-fibrotic macrophages using yeast beta-glucan microparticles prepared by Pressurized Gas eXpanded liquid (PGX) Technology®. Biomaterials 2025; 313:122816. [PMID: 39250864 DOI: 10.1016/j.biomaterials.2024.122816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Pro-fibrotic M2-like macrophages are widely implicated in the pathogenesis and progression of lung fibrosis due to their production of pro-fibrotic growth factors and cytokines. Yeast beta-glucan (YBG) microparticles have shown potential as immunomodulators that can convert macrophage polarization from a pro-fibrotic phenotype to an anti-fibrotic phenotype through the engagement of the Dectin-1 receptor. However, the processing conditions used to fabricate YBG microparticles can lead to unpredictable immunomodulatory effects. Herein, we report the use of Pressurized Gas eXpanded liquids (PGX) Technology® to fabricate YBG (PGX-YBG) microparticles with higher surface areas, lower densities, and smaller and more uniform size distributions compared to commercially available spray-dried YBGs. PGX-YBG is shown to activate Dectin-1 more efficiently in vitro while avoiding significant TLR 2/4 activation. Furthermore, PGX-YBG microparticles effectively modulate M2-like fibrosis-inducing murine and human macrophages into fibrosis-suppressing macrophages both in vitro as well as in ex vivo precision-cut murine lung slices, suggesting their potential utility as a therapeutic for addressing a broad spectrum of fibrotic end-point lung diseases.
Collapse
Affiliation(s)
- S Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - N Dowdall
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Q Zhou
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - P Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - A Hayat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - M Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - E Y Wong
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - R Couto
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - B Yépez
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - B Seifried
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - P Moquin
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - M R Kolb
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - K Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada.
| | - T Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
7
|
Liu Z, Zheng Q, Li Z, Huang M, Zhong C, Yu R, Jiang R, Dai H, Zhang J, Gu X, Xu Y, Li C, Shan S, Xu F, Hong Y, Ren T. Epithelial stem cells from human small bronchi offer a potential for therapy of idiopathic pulmonary fibrosis. EBioMedicine 2025; 112:105538. [PMID: 39753035 PMCID: PMC11754162 DOI: 10.1016/j.ebiom.2024.105538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a fibrosing interstitial pneumonia with restrictive ventilation. Recently, the structural and functional defects of small airways have received attention in the early pathogenesis of IPF. This study aimed to elucidate the characteristics of small airway epithelial dysfunction in patients with IPF and explore novel therapeutic interventions to impede IPF progression by targeting the dysfunctional small airways. METHODS Airway trees spanning the proximal-distal axis were harvested from control lungs and explanted lungs with end-stage IPF undergoing transplant. Qualified basal cells (BCs, p63/Krt5/ITGA6/NGFR) were expanded, and their cellular functions, feasibility, safety and efficacy for transplantation therapy in IPF were validated with experiments in vitro and mouse model. Single-cell RNA-sequencing was employed to elucidate the underlying mechanisms governing the BCs based therapy. Based upon these evidences, three patients with advanced IPF and small airway dysfunction received autologous-BCs transplantation. Post-transplantation assessments included lung function, exercise capacity and high resolution computed tomography (HRCT) scans were analyzed to quantify the clinical benefits conferred by the BCs transplantation. FINDINGS An overall landscape of senescent phenotype in airway epithelial cells and airway stem/progenitor cells along the proximal-distal axis of the airway tree in IPF were outlined. In contrast to the cells situated in distal airways, BCs located in small bronchi in IPF displayed a non-senescent phenotype, with comparable proliferative, differentiative capabilities, and similar transcriptomic profiles to normal controls. In a mouse model of pulmonary fibrosis, BCs exhibited promising protective efficacy and safety for transplantation therapy. Autologous BCs transplantation in three advanced IPF patients with small airway dysfunction yielded significant clinical improvements in pulmonary function, particularly evidence in lung volume and small airway function. INTERPRETATION Epithelia of small bronchi in IPF contain functional and expandable basal stem cells, which exert therapeutic benefits via bronchoscopic implantation. Our findings offer a potential for IPF treatment by targeting small airways. FUNDING National Natural Science Foundation of China (82430001, 81930001, and 81900059), Shanghai Shenkang Hospital Development Center (SHDC2020CR3063B), Department of Science and Technology of Shandong Province (2024HWYQ-058).
Collapse
Affiliation(s)
- Zeyu Liu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qi Zheng
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhoubin Li
- Department of Lung Transplantation and Thoracic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Moli Huang
- Department of Bioinformatics, School of Biological and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Zhong
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ruize Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Rong Jiang
- Department of Bioinformatics, School of Biological and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Haotian Dai
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jingyuan Zhang
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaohua Gu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yongle Xu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chunwei Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shan Shan
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Feng Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
| | - Yue Hong
- School of Life and Health Sciences, Hainan University, Haikou, Hainan 570228, China; Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China.
| | - Tao Ren
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
8
|
Chang SY, Chang WH, Yang DC, Hong QS, Hsu SW, Wu R, Chen CH. Autologous precision-cut lung slice co-culture models for studying macrophage-driven fibrosis. Front Physiol 2025; 16:1526787. [PMID: 39958688 PMCID: PMC11825446 DOI: 10.3389/fphys.2025.1526787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
Precision-cut lung slices (PCLS) are commonly used as an ex vivo model to study lung fibrosis; however, traditional models lack immune cell infiltration, including the recruitment of monocytes and macrophages, which are critical for inflammation and fibrosis. To address this limitation, we developed novel autologous PCLS-immune co-culture models that better replicate the processes of inflammation, repair, and immune cell recruitment associated with fibrosis. Fibrotic responses to nicotine, cigarette smoke extract (CSE), and a fibrosis-inducing cocktail (FC) were first evaluated in PCLS containing only tissue-resident macrophages, with upregulation of α-SMA-expressing fibroblasts confirmed by immunofluorescence and Western blotting, and collagen deposition quantified using Sirius Red staining. To study macrophage recruitment, we employed an indirect co-culture model using transwells to approximate blood vessel function. Chemotactic studies revealed increased migration of autologous bone marrow-derived macrophages (BMDMs) toward and infiltration into CSE-injured PCLS. In a direct co-culture model simulating the repair phase of fibrosis, PCLS exposed to CSE and FC showed further increased collagen deposition in the presence of autologous BMDMs, but not heterologous ones. These findings suggest that our novel PCLS-immune co-culture models provide a platform for studying macrophage involvement in fibrosis and offer potential for developing macrophage-targeted therapeutic strategies in pulmonary fibrosis.
Collapse
Affiliation(s)
- So-Yi Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Wen-Hsin Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - David C. Yang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Qi-Sheng Hong
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Ssu-Wei Hsu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Reen Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Ching-Hsien Chen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| |
Collapse
|
9
|
Shadid A, Rich HE, DeVaughn H, Domozhirov A, Doursout MF, Weng-Mills T, Eckel-Mahan KL, Karmouty-Quintana H, Restrepo MI, Shivshankar P. Persistent microbial infections and idiopathic pulmonary fibrosis - an insight into non-typeable Haemophilus influenza pathogenesis. Front Cell Infect Microbiol 2024; 14:1479801. [PMID: 39760094 PMCID: PMC11695292 DOI: 10.3389/fcimb.2024.1479801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Interstitial lung disease (ILD) is characterized by chronic inflammation and scarring of the lungs, of which idiopathic pulmonary fibrosis (IPF) is the most devastating pathologic form. Idiopathic pulmonary fibrosis pathogenesis leads to loss of lung function and eventual death in 50% of patients, making it the leading cause of ILD-associated mortality worldwide. Persistent and subclinical microbial infections are implicated in the acute exacerbation of chronic lung diseases. However, while epidemiological studies have highlighted pollutants, gastric aspirate, and microbial infections as major causes for the progression and exacerbation of IPF, the role of persistent microbial infections in the pathogenesis of IPF remains unclear. In this review, we have focused on the role of persistent microbial infections, including viral, bacterial, and fungal infections, and their mechanisms of action in the pathogenesis of IPF. In particular, the mechanisms and pathogenesis of the Gram-negative bacteria Non-typeable Haemophilus influenzae (NTHi) in ILDs are discussed, along with growing evidence of its role in IPF, given its unique ability to establish persistent intracellular infections by leveraging its non-capsulated nature to evade host defenses. While antibiotic treatments are presumably beneficial to target the extracellular, interstitial, and systemic burden of pathogens, their effects are significantly reduced in combating pathogens that reside in the intracellular compartments. The review also includes recent clinical trials, which center on combinatorial treatments involving antimicrobials and immunosuppressants, along with antifibrotic drugs that help mitigate disease progression in IPF patients. Finally, future directions focus on mRNA-based therapeutics, given their demonstrated effectiveness across a wide range of clinical applications and feasibility in targeting intracellular pathogens.
Collapse
Affiliation(s)
- Anthony Shadid
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Haydn E. Rich
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Hunter DeVaughn
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Aleksey Domozhirov
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Marie- Françoise Doursout
- Department of Anesthesiology, Critical Care and Pain Medicine, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Tingting Weng-Mills
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Kristin L. Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, UTHealth-McGovern Medical School, Houston, TX, United States
| | - Marcos I. Restrepo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, South Texas Veterans Health Care System and the University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pooja Shivshankar
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for Prevention of Human Diseases, UTHealth-McGovern Medical School, Houston, TX, United States
| |
Collapse
|
10
|
Yang J, Lu D, Sun Y, Qiu M, Zhao T, Yan B, Wang S, Shao Z, Wang D, Li T, Xiao Q, Fu T. Cell Membrane Hybrid Liposome-Targeted Delivery of the Heat Shock Protein 90 C-Terminal Inhibitor for the Treatment of Idiopathic Pulmonary Fibrosis. ACS Pharmacol Transl Sci 2024; 7:4083-4095. [PMID: 39698274 PMCID: PMC11651165 DOI: 10.1021/acsptsci.4c00524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a grave challenge as it is characterized by high fatality rates and irreversible progression without effective clinical interventions available at present. Previous studies have demonstrated that inhibition of heat shock protein 90 (HSP90) by an N-terminal inhibitor disrupts its interaction with TGFβRII, leading to the instability of TGFβRII, thus blocking the role of transforming growth factor-β1 (TGF-β1), which could potentially ameliorate IPF symptoms. However, given that the broad spectrum of HSP90 N-terminal inhibitors may lead to unanticipated side effects, we hypothesize that C-terminal inhibitors of HSP90 can interfere with TGFβRII while minimizing adverse reactions. In this study, silybin, a C-terminal inhibitor of HSP90, was separated into monomers, and silybin A was screened for its superior efficacy against TGFβRII. To facilitate targeted therapy for treating IPF, a cell membrane hybrid liposome loaded with silybin A (Cm-A-Lip) was developed to deliver silybin A to lung fibroblasts through pulmonary drug delivery. A bleomycin-induced IPF mouse model was used to evaluate the efficacy of Cm-A-Lip. By examination of lung hydroxyproline content, wet weight, histology, and inflammatory factor expression, the results showed that pulmonary delivery of Cm-A-Lip could increase the drug retention time in lung tissue compared with intravenous injection. Furthermore, Cm-A-Lip exhibited superior antifibrotic activity relative to conventional liposmomes loaded with silybin A (A-Lip) while concurrently mitigating systemic inflammatory responses associated with silybin A administration, thus enhancing the overall safety profile.
Collapse
Affiliation(s)
| | | | - Yuping Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Qiu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianlong Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baofei Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Siting Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhitao Shao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Demei Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qingqing Xiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingming Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
11
|
Liu Y, Tang P, Peng S, Zhong J, Xu Z, Zhong J, Su J, Zhong Y, Hu K. [ 18F]AlF-CBP imaging of type I collagen for non-invasive monitoring of pulmonary fibrosis in preclinical models. Eur J Nucl Med Mol Imaging 2024; 52:22-35. [PMID: 39172179 DOI: 10.1007/s00259-024-06888-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE Pulmonary fibrosis is an irreversible scar-forming condition for which there is a lack of non-invasive and specific methods for monitoring its progression and therapy efficacy. However, the disease is known to be accompanied by collagen accumulation. Here, we developed a novel positron emission tomography (PET) probe targeting type I collagen to evaluate its utility for the non-invasive assessment of pulmonary fibrosis. METHODS We designed a 18F-labeled PET probe ([18F]AlF-CBP) to target type I collagen and evaluated its binding affinity, specificity and stability in vitro. PET with [18F]AlF-CBP, CT, histopathology, immunofluorescence, and biochemical indice were performed to assess and quantify type I collagen levels and pulmonary fibrosis progression and treatment in murine models. Dynamic PET/CT studies of [18F]AlF-CBP were conducted to assess lung fibrosis in non-human primate models. RESULTS [18F]AlF-CBP was successfully prepared, and in vitro and in vivo tests showed high stability (> 95%) and type I collagen specificity (IC50 = 0.36 µM). The lungs of the fibrotic murine model showed more elevated probe uptake and retention compared to the control group, and there was a positive correlation between the radioactivity uptake signals and the degree of fibrosis (CT: R2 = 0.89, P < 0.0001; hydroxyproline levels: R2 = 0.89, P < 0.0001). PET signals also correlated well with mean lung density in non-human primate models of pulmonary fibrosis (R2 = 0.84, P < 0.0001). CONCLUSION [18F]AlF-CBP PET imaging is a promising non-invasive method for specific monitoring of lung fibrosis progression and therapy efficacy.
Collapse
Affiliation(s)
- Yang Liu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Peipei Tang
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Simin Peng
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinmei Zhong
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zexin Xu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiawei Zhong
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Su
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Farooq H, Luehmann HP, Koenitzer JR, Heo GS, Sultan DH, Kulkarni DH, Gunsten SP, Sashti RM, Huang T, Keller AR, Lavine KJ, Atkinson JJ, Wingler LM, Liu Y, Brody SL. Molecular imaging in experimental pulmonary fibrosis reveals that nintedanib unexpectedly modulates CCR2 immune cell infiltration. EBioMedicine 2024; 110:105431. [PMID: 39515027 PMCID: PMC11582469 DOI: 10.1016/j.ebiom.2024.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis is a challenging clinical problem with lung pathology featuring immune cell infiltrates, fibroblast expansion, and matrix deposition. Molecular analysis of diseased lungs and preclinical models have uncovered C-C chemokine receptor type 2 (CCR2)+ monocyte egress from the bone marrow into the lung, where they acquire profibrotic activities. Current drug treatment is focused on fibroblast activity. Alternatively, therapeutic targeting and monitoring CCR2+ cells may be an effective patient management strategy. METHODS Inhibition of CCR2+ cells and, as a benchmark, the clinical antifibrotic agent, nintedanib, were used in mouse lung fibrosis models. Lungs were evaluated directly for CCR2+ cell infiltration and by non-invasive CCR2+ positron emission tomography imaging (CCR2-PET). FINDINGS Lung CCR2+ cells were significantly elevated in the bleomycin model as determined by tissue evaluation and CCR2-PET imaging. A protective treatment protocol with an oral CCR2 inhibitor was compared to oral nintedanib. While we expected disparate effects on CCR2+ cells, each drug similarly decreased lung CCR2+ cells and fibrosis. Chemotaxis assays showed nintedanib indirectly inhibited C-C motif chemokine 2 (CCL2)-mediated migration of CCR2+ cells. Even delayed therapeutic administration of nintedanib in bleomycin and the silicosis progressive fibrosis models decreased the accumulation of CCR2+ lung cells. In these treatments early CCR2-PET imaging predicted the later development of fibrosis. INTERPRETATION The inhibition of CCR2+ cell egress is likely a critical controller for stabilising lung fibrosis, as provided by nintedanib. Imaging with CCR2-PET may be useful to monitor nintedanib treatment responses, guide decision-making in the treatment of patients with progressive pulmonary fibrosis, and as a biomarker for drug development. FUNDING National Institutes of Health (NIH), R01HL131908 (SLB), R35HL145212 (YL), P41EB025815 (YL), K01DK133670 (DHK); Barnes Jewish Hospital Foundation (SLB).
Collapse
Affiliation(s)
- Hasan Farooq
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannah P Luehmann
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jeffrey R Koenitzer
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Deborah H Sultan
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Devesha H Kulkarni
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sean P Gunsten
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rekha M Sashti
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tao Huang
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Amanda R Keller
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Kory J Lavine
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jeffrey J Atkinson
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Laura M Wingler
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Steven L Brody
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
13
|
Pan C, Wei H, Chen B, Wu L, Song J, Zhang Q, Wu X, Liang G, Chen W, Wang Y, Xie Y. Inhalation of itraconazole mitigates bleomycin-induced lung fibrosis via regulating SPP1 and C3 signaling pathway pivotal in the interaction between phagocytic macrophages and diseased fibroblasts. J Transl Med 2024; 22:1058. [PMID: 39587675 PMCID: PMC11587652 DOI: 10.1186/s12967-024-05895-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) stands as a significant contributor to global mortality rates. Presently, there exists a dearth of effective anti-fibrotic treatments for this condition. While itraconazole (ITR) has exhibited potential in mitigating pulmonary fibrosis, its oral administration is hampered by unfavorable pharmacokinetics, which elevate the risk of adverse reactions, thus limiting its clinical utility. METHODS An inhalable formulation of ITR were engineered which aimed at enhancing its pulmonary dispersion. First, pharmacokinetics were conducted to investigate the blood concentration and tissue residue of ITR after inhalation administration. In addition, bleomycin induced mouse pulmonary fibrosis model was used to compare the therapeutic effects of ITR administered by inhalation and intragastric administration. Finally, single-cell RNA sequencing (scRNAseq) was used to explore the mechanism of ITR inhalation administration. RESULTS We found that a large amount of drugs accumulated in the lung tissue for a long time after inhalation administration, thus maximizing the therapeutic effect of drugs. Inhalation of ITR daily at for 21 days significantly attenuated bleomycin-induced lung fibrosis and inflammation in murine models. Additionally, our findings revealed that ITR inhalation diminished the proportion of diseased fibroblasts while promoting reparative fibroblast populations in the murine model. Furthermore, it effectively reversed the proportion of activated phagocytic macrophages. Mechanistically, ITR inhalation exerted its effects by regulating SPP1 and C3 signaling pathway pivotal in the interaction between phagocytic macrophages and diseased fibroblasts. CONCLUSIONS These insights into the molecular mechanisms underlying ITR's therapeutic effects on IPF underscore the favorable pharmacokinetic profile conferred by inhalation, thus presenting a promising formulation poised for clinical translation.
Collapse
Affiliation(s)
- Caizhe Pan
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Hao Wei
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Bi Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Lei Wu
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Jiayao Song
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Qing Zhang
- School of of Computer Science and Engineering, Hubei Key Laboratory of Intelligent Robot, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Xinglong Wu
- School of of Computer Science and Engineering, Hubei Key Laboratory of Intelligent Robot, Wuhan Institute of Technology, Wuhan, 430205, China
| | | | - Wenhao Chen
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| | - Yingshuo Wang
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| | - Yicheng Xie
- Department of Pulmonology and Orthopedic Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| |
Collapse
|
14
|
Kadam AH, Schnitzer JE. Highly Calibrated Relationship Between Bleomycin Concentrations and Facets of the Active Phase Fibrosis in Classical Mouse Bleomycin Model. Int J Mol Sci 2024; 25:12300. [PMID: 39596365 PMCID: PMC11595013 DOI: 10.3390/ijms252212300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The mouse bleomycin model is useful in pre-clinical IPF research to understand pathophysiological mechanisms and pharmacological interventions. In the present study, we systematically investigated the effects of bleomycin at a 60-fold dose range on experimental features of lung fibrosis in the mouse bleomycin model. We analyzed the effect of intratracheal (i.t.) dosing of 0.05-3 U/kg bleomycin on disease phenotypes, including weight loss, morbidity and mortality, pulmonary inflammation, lung collagen content, various BALF biomarkers, and histology in a 14-day mouse model when the animals are in the active phase of fibrosis. In mice, challenge with 1-2 U/kg bleomycin doses induced significant and saturated responses on fibrotic endpoints, confirmed by collagen content, BALF biomarker levels, and marked weight loss compared to the normal control (NC). We observed 100% mortality in 3 U/kg of bleomycin-treated mice. In contrast, 0.05-0.5 U/kg bleomycin doses induced a dose-dependent fibrotic phenotype. The mice challenged with doses of 0.25-0.5 U/kg bleomycin showed optimum body weight loss, a significant increase in pulmonary inflammation, and the fibrotic phenotype compared to NC. Furthermore, we showed 0.25-0.5 U/kg bleomycin increases expression levels of (pro-) fibrotic cytokines, which are the mediators involved in the activation of myofibroblast during fibrogenesis (TGF-β1, IL-13, IL-6, WISP-1, VEGF), angiogenesis (VEGF), matrix remodeling (TIMP-1), and non-invasive lung function biomarker (CRP) compared to NC. A modified Ashcroft scale quantified that the fibrotic changes in the lungs were significantly higher in the lung of mice dosed at 0.25-0.5 U/kg > 0.1 U/kg bleomycin and non-significant in mice lung dosed at 0.05 U/kg bleomycin compared to NC. We demonstrated that the changes due to 0.25-0.5 U/kg i.t. bleomycin on protein biomarkers are enough to drive robust and detectable fibrotic pathology without mortality. The 0.1 U/kg has a moderate phenotype, and 0.05 U/kg had no detectable phenotype. The Goodness of Fit (r2) and Pearson correlation coefficient (r) analyses revealed a positive linear association between change evaluated in all experimental features of fibrosis and bleomycin concentrations (0.05-0.5 U/kg). Here, we provide an examination of a highly calibrated relationship between 60-fold bleomycin concentrations and a set of in vivo readouts that covers various facets of experimental fibrosis. Our study shows that there is a dose-dependent effect of bleomycin on the features of experimental fibrosis at <1 U/kg, whereas saturated responses are achieved at >1 U/kg. Our careful experimental observations, accuracy, and comprehensive data set provided meaningful insights into the effect of bleomycin dose(s) on the fibrotic phenotype, which is valuable in preclinical drug development and lung fibrosis research. In addition, we have presented a set of reproducible frameworks of endpoints that can be used for reliable assessment of the fibrotic phenotype, and in vivo therapeutic intervention(s) with improved accuracy.
Collapse
Affiliation(s)
| | - Jan E. Schnitzer
- Proteogenomics Research Institute for Systems Medicine (PRISM), 505 Coast Blvd. South, La Jolla, CA 92037, USA;
| |
Collapse
|
15
|
He Y, Yang F, Yang L, Yuan H, You Y, Chen Y, Wu X, Min H, Chen J, Li C. Mechanics-activated fibroblasts promote pulmonary group 2 innate lymphoid cell plasticity propelling silicosis progression. Nat Commun 2024; 15:9770. [PMID: 39532893 PMCID: PMC11557922 DOI: 10.1038/s41467-024-54174-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Crystalline silica (CS) particle exposure leads to silicosis which is characterized as progressive fibrosis. Fibroblasts are vital effector cells in fibrogenesis. Emerging studies have identified immune sentinel roles for fibroblasts in chronic disease, while their immune-modulatory roles in silicosis remain unclear. Herein, we show that group 2 innate lymphoid cell (ILC2) conversion to ILC1s is closely involved in silicosis progression, which is mediated by activated fibroblasts via interleukin (IL)-18. Mechanistically, Notch3 signaling in mechanics-activated fibroblasts modulates IL-18 production via caspase 1 activity. The mouse-specific Notch3 knockout in fibroblasts retards pulmonary fibrosis progression that is linked to attenuated ILC conversion. Our results indicate that activated fibroblasts in silicotic lungs are regulators of ILC2-ILC1 conversion, associated with silicosis progression via the Notch3-IL-18 signaling axis. This finding broadens our understanding of immune-modulatory mechanisms in silicosis, and indicates potential therapeutic targets for lung fibrotic diseases.
Collapse
Affiliation(s)
- Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Fan Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Lin Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Yinghui Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Xiulin Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
16
|
Araldi GL, Hwang YW, Raghu G. Development and Evaluation of ABI-171, a New Fluoro-Catechin Derivative, for the Treatment of Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2024; 25:11827. [PMID: 39519378 PMCID: PMC11546061 DOI: 10.3390/ijms252111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
The persistent challenge of idiopathic pulmonary fibrosis (IPF), characterized by disease progression and high mortality, underscores the urgent need for innovative therapeutic strategies. We have developed a novel small molecule-catechin derivative ABI-171-selectively targeting dual-specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) and proviral integration site for Moloney murine leukemia virus 1 (PIM1) kinases, crucial in the pathogenesis of fibrotic processes. We employed the Bleomycin-induced (intratracheal) mouse model of pulmonary fibrosis (PF) to evaluate the therapeutic efficacy of ABI-171. Mice with induced PF were treated QD with ABI-171, either prophylactically or therapeutically, using oral and intranasal routes. Pirfenidone (100 mg/kg, TID) and Epigallocatechin gallate (EGCG, 100 mg/kg, QD), a natural catechin currently in a Phase 1 clinical trial, were used as reference compounds. ABI-171, administered prophylactically, led to a significant reduction in hydroxyproline levels and fibrotic tissue formation compared to the control group. Treatment with ABI-171 improved body weight, indicating mitigation of disease-related weight loss. Additionally, ABI-171 demonstrated anti-inflammatory activity, reducing lymphocyte and neutrophil infiltration. In the therapeutic setting, ABI-171, administered 7 days post-induction, reduced mortality rates (p = 0.04) compared with the bleomycin and EGCG control groups. ABI-171 also ameliorated the severity of lung injuries assessed by improved Masson's trichrome scores when administered both orally and intranasally. ABI-171 significantly decreases bleomycin-induced PF and improves survival in mice, showcasing promising therapeutic potential beyond current medications like pirfenidone and EGCG for patients with IPF. Based on these results, further studies with ABI-171 are ongoing in preclinical studies.
Collapse
Affiliation(s)
- Gian Luca Araldi
- Avanti Biosciences, Inc., 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Yu-Wen Hwang
- Department of Molecular Biology, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
| | - Ganesh Raghu
- Center for Interstitial Lung Diseases, University of Washington Medical Center, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Song D, Li Z, Sun F, Wu K, Zhang K, Liu W, Liu K, An B, Wang Z, Zhao T, Chen H, Xiao L, Wang L, Xie L, Li W, Peng L, Hao J, Wu J, Dai H. Optimized administration of human embryonic stem cell-derived immunity-and-matrix regulatory cells for mouse lung injury and fibrosis. Stem Cell Res Ther 2024; 15:344. [PMID: 39380037 PMCID: PMC11462801 DOI: 10.1186/s13287-024-03945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Lung injury and pulmonary fibrosis (PF), frequently arising as sequelae of severe and acute lung disease, currently face a dearth of effective therapeutic potions. Mesenchymal stem cells (MSCs) with immunomodulatory and tissue repair functions have immense potential to treat lung injury and PF. However, the optimal route of administration, timing, and frequency of dosing remain elusive. Human embryonic stem cell-derived immunity-and-matrix-regulatory cells (IMRCs) have shown therapeutic potential for lung injury and PF. METHODS To ascertain the optimal therapeutic regimen for IMRCs in PF, we conducted an experimental study. Utilizing a mouse model of PF induced by bleomycin (BLM), IMRCs were administered via either a single or double intravenous (IV) or intratracheal (IT) injection on the first and seventh days post-BLM induction. RESULTS Our findings revealed that IV infusion of IMRCs surpassed IT infusion in enhancing survival rates, facilitating body weight recovery, and optimizing Ashcroft and Szapiel scores among the model mice. Notably, IV administration exhibited a more profound ability to mitigate lung inflammation and fibrosis. Moreover, earlier and more frequent administrations of IMRCs were found to be advantageous in enhancing their therapeutic effects. Specifically, early administration with two IV infusions significantly improved body weight, lung organ coefficient, pulmonary ventilation and diffusion functions, and PF. This was accompanied by an increase in alveolar type I and II epithelial cells and a suppression of macrophage infiltration via CD24. CONCLUSION Collectively, these results suggested that IMRCs infusion ameliorated lung injury by promoting lung regeneration and inhibiting macrophage infiltration in a route, time, and frequency-dependent manner.
Collapse
Affiliation(s)
- Dingyun Song
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
| | - Zhongwen Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Faguo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaiwei Wu
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Kan Zhang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
| | - Wenjing Liu
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Kaidi Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
| | - Bin An
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Tiemei Zhao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
| | - Huaiyong Chen
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China
| | - Li Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
- Respiratory Research Institute, Senior Department of Pulmonary & Critical Care Medicine, the 8th Medical Center of PLA General Hospital, Beijing, 100091, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese, PLA General Hospital, PLA General Hospital, Beijing, 100091, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liang Peng
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- National Stem Cell Resource Center, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huaping Dai
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
18
|
Petersen AG, Korntner SH, Bousamaki J, Oró D, Arraut AM, Pors SE, Salinas CG, Andersen MW, Madsen MR, Nie Y, Butts J, Roqueta‐Rivera M, Simonsen U, Hansen HH, Feigh M. Reproducible lung protective effects of a TGFβR1/ALK5 inhibitor in a bleomycin-induced and spirometry-confirmed model of IPF in male mice. Physiol Rep 2024; 12:e70077. [PMID: 39394052 PMCID: PMC11469938 DOI: 10.14814/phy2.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/30/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024] Open
Abstract
This study comprehensively validated the bleomycin (BLEO) induced mouse model of IPF for utility in preclinical drug discovery. To this end, the model was rigorously evaluated for reproducible phenotype and TGFβ-directed treatment outcomes. Lung disease was profiled longitudinally in male C57BL6/JRJ mice receiving a single intratracheal instillation of BLEO (n = 10-12 per group). A TGFβR1/ALK5 inhibitor (ALK5i) was profiled in six independent studies in BLEO-IPF mice, randomized/stratified to treatment according to baseline body weight and non-invasive whole-body plethysmography. ALK5i (60 mg/kg/day) or vehicle (n = 10-16 per study) was administered orally for 21 days, starting 7 days after intratracheal BLEO installation. BLEO-IPF mice recapitulated functional, histological and biochemical hallmarks of IPF, including declining expiratory/inspiratory capacity and inflammatory and fibrotic lung injury accompanied by markedly elevated TGFβ levels in bronchoalveolar lavage fluid and lung tissue. Pulmonary transcriptome signatures of inflammation and fibrosis in BLEO-IPF mice were comparable to reported data in IPF patients. ALK5i promoted reproducible and robust therapeutic outcomes on lung functional, biochemical and histological endpoints in BLEO-IPF mice. The robust lung fibrotic disease phenotype, along with the consistent and reproducible lung protective effects of ALK5i treatment, makes the spirometry-confirmed BLEO-IPF mouse model highly applicable for profiling novel drug candidates for IPF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaohui Nie
- Enanta PharmaceuticalsWatertownMassachusettsUSA
| | | | | | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of HealthAarhus UniversityAarhusDenmark
| | | | | |
Collapse
|
19
|
Bai Y, Gao L, Han T, Liang C, Zhou J, Liu Y, Guo J, Wu J, Hu D. 18β-glycyrrhetinic acid ameliorates bleomycin-induced idiopathic pulmonary fibrosis via inhibiting TGF-β1/JAK2/STAT3 signaling axis. J Steroid Biochem Mol Biol 2024; 243:106560. [PMID: 38917955 DOI: 10.1016/j.jsbmb.2024.106560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/25/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating and progressive lung disease with an unknown cause that has few treatment options. 18β-Glycyrrhetinic acid (18β-GA) is the main bioactive component in licorice, exhibiting anti-inflammatory and antioxidant effects, while also holding certain application value in the metabolism and regulation of steroids. In this study, we demonstrated that 18β-GA effectively alleviates bleomycin (BLM)-induced IPF by inhibiting the TGF-β1/JAK2/STAT3 signaling axis. In vivo experiments demonstrate that 18β-GA significantly attenuates pulmonary fibrosis progression by reducing lung inflammation, improving lung function, and decreasing collagen deposition. In vitro experiments reveal that 18β-GA inhibits the activation and migration of TGF-β1-induced fibroblasts. Furthermore, it regulates the expression of vimentin, N-cadherin and E-cadherin proteins, thereby inhibiting TGF-β1-induced epithelial-mesenchymal transition (EMT) in lung alveolar epithelial cells. Mechanistically, 18β-GA ameliorates pulmonary fibrosis by modulating the TGF-β1/JAK2/STAT3 signaling pathway in activated fibroblasts. Taken together, our findings demonstrate the potential and underlying mechanisms of 18β-GA in ameliorating IPF, emphasizing its potential as a novel therapeutic drug for the treatment of this devastating disease.
Collapse
Affiliation(s)
- Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| |
Collapse
|
20
|
Zhao Y, Yang J, Zhang Q, Chen X, Liang W, Zheng Y, Huang J, Liao Y, Fu C, Huang T, Li X, Zheng Y, Bu J, Shen E. Fasting alleviates bleomycin-induced lung inflammation and fibrosis via decreased Tregs and monocytes. Adv Med Sci 2024; 69:303-311. [PMID: 38986767 DOI: 10.1016/j.advms.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/22/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF), a chronic and progressively worsening condition characterized by interstitial lung inflammation and fibrosis of unknown etiology, has a grim prognosis. The treatment options for IPF are limited and new therapeutic strategies are urgently needed. Dietary restriction can improve various inflammatory diseases, but its therapeutic effect on bleomycin (BLM)-induced pulmonary fibrosis mouse model remains unclear. This study aims to investigate whether intermittent fasting (IF) can alleviate BLM-induced pulmonary inflammation and fibrosis. METHODS Pulmonary fibrosis mouse models were induced by BLM. The IF group underwent 24-h fasting cycles for one week prior and three weeks following BLM administration. Meanwhile, the ad libitum feeding group had unrestricted access to food throughout the experiment. The evaluation focused on lung pathology via histological staining, qPCR analysis of collagen markers, and immune cell profiling through flow cytometry. RESULTS IF group significantly reduced inflammation and fibrosis in lung tissues of BLM-induced mice compared to ad libitum feeding group. qPCR results showed IF remarkably decreased the mRNA expression of Col 1a and Col 3a in the lungs of BLM-induced mouse models. IF also reduced the numbers of regulatory T cells (Tregs), T helper 17 (Th17) cells, monocytes, and monocyte-derived alveolar macrophages (MoAMs) in the lung tissues. CONCLUSIONS IF may improve BLM-induced pulmonary fibrosis by decreasing numbers of immune cells including Treg cells, Th17 cells, monocytes, and MoAMs in the lungs. This study offers experimental validation for dietary intervention as a viable treatment modality in IPF management.
Collapse
Affiliation(s)
- Yuyang Zhao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jingying Yang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Qi Zhang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China; The 903rd Hospital of the PLA, Hangzhou, Zhejiang, China
| | - Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Wenting Liang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yanling Zheng
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jijun Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yue Liao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Cheng Fu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Ting Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yu Zheng
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jin Bu
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Mohammed SM, Al-Saedi HFS, Mohammed AQ, Amir AA, Radi UK, Sattar R, Ahmad I, Ramadan MF, Alshahrani MY, Balasim HM, Alawadi A. Mechanisms of Bleomycin-induced Lung Fibrosis: A Review of Therapeutic Targets and Approaches. Cell Biochem Biophys 2024; 82:1845-1870. [PMID: 38955925 DOI: 10.1007/s12013-024-01384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Pulmonary toxicity is a serious side effect of some specific anticancer drugs. Bleomycin is a well-known anticancer drug that triggers severe reactions in the lungs. It is an approved drug that may be prescribed for the treatment of testicular cancers, Hodgkin's and non-Hodgkin's lymphomas, ovarian cancer, head and neck cancers, and cervical cancer. A large number of experimental studies and clinical findings show that bleomycin can concentrate in lung tissue, leading to massive oxidative stress, alveolar epithelial cell death, the proliferation of fibroblasts, and finally the infiltration of immune cells. Chronic release of pro-inflammatory and pro-fibrotic molecules by immune cells and fibroblasts leads to pneumonitis and fibrosis. Both fibrosis and pneumonitis are serious concerns for patients who receive bleomycin and may lead to death. Therefore, the management of lung toxicity following cancer therapy with bleomycin is a critical issue. This review explains the cellular and molecular mechanisms of pulmonary injury following treatment with bleomycin. Furthermore, we review therapeutic targets and possible promising strategies for ameliorating bleomycin-induced lung injury.
Collapse
Affiliation(s)
- Shaimaa M Mohammed
- Department of Pharmacy, Al- Mustaqbal University College, 51001, Hilla, Babylon, Iraq
| | | | | | - Ahmed Ali Amir
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ruaa Sattar
- Al-Hadi University College, Baghdad, 10011, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Halah Majeed Balasim
- Department of Medical Laboratory Technologies, Al Rafidain University College, Bagdad, Iraq
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq
- College of technical engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of technical engineering, the Islamic University of Babylon, Hilla, Iraq
| |
Collapse
|
22
|
Chakraborty A, Wang C, Hodgson-Garms M, Broughton BRS, Frith JE, Kelly K, Samuel CS. Induced pluripotent stem cell-derived mesenchymal stem cells reverse bleomycin-induced pulmonary fibrosis and related lung stiffness. Biomed Pharmacother 2024; 178:117259. [PMID: 39116786 DOI: 10.1016/j.biopha.2024.117259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterised by lung scarring and stiffening, for which there is no effective cure. Based on the immunomodulatory and anti-fibrotic effects of induced pluripotent stem cell (iPSC) and mesenchymoangioblast-derived mesenchymal stem cells (iPSCs-MSCs), this study evaluated the therapeutic effects of iPSCs-MSCs in a bleomycin (BLM)-induced model of pulmonary fibrosis. Adult male C57BL/6 mice received a double administration of BLM (0.15 mg/day) 7-days apart and were then maintained for a further 28-days (until day-35), whilst control mice were administered saline 7-days apart and maintained for the same time-period. Sub-groups of BLM-injured mice were intravenously-injected with 1×106 iPSC-MSCs on day-21 alone or on day-21 and day-28 and left until day-35 post-injury. Measures of lung inflammation, fibrosis and compliance were then evaluated. BLM-injured mice presented with lung inflammation characterised by increased immune cell infiltration and increased pro-inflammatory cytokine expression, epithelial damage, lung transforming growth factor (TGF)-β1 activity, myofibroblast differentiation, interstitial collagen fibre deposition and topology (fibrosis), in conjunction with reduced matrix metalloproteinase (MMP)-to-tissue inhibitor of metalloproteinase (TIMP) ratios and dynamic lung compliance. All these measures were ameliorated by a single or once-weekly intravenous-administration of iPSC-MSCs, with the latter reducing dendritic cell infiltration and lung epithelial damage, whilst promoting anti-inflammatory interleukin (IL)-10 levels to a greater extent. Proteomic profiling of the conditioned media of cultured iPSC-MSCs that were stimulated with TNF-α and IFN-γ, revealed that these stem cells secreted protein levels of immunosuppressive factors that contributed to the anti-fibrotic and therapeutic potential of iPSCs-MSCs as a novel treatment option for IPF.
Collapse
Affiliation(s)
- Amlan Chakraborty
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Division of Immunology, Immunity to Infection and Respiratory Medicine, The University of Manchester, Manchester, England, UK
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Margeaux Hodgson-Garms
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia
| | - Kilian Kelly
- Cynata Therapeutics Ltd, Cremorne, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
23
|
Gao L, Bai Y, Liang C, Han T, Liu Y, Zhou J, Guo J, Wu J, Hu D. Celastrol-Ligustrazine compound proven to be a novel drug candidate for idiopathic pulmonary fibrosis by intervening in the TGF-β1 mediated pathways-an experimental in vitro and vivo study. Mol Divers 2024:10.1007/s11030-024-10970-1. [PMID: 39207663 DOI: 10.1007/s11030-024-10970-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a disease characterized by pulmonary interstitial fibrosis and collagen proliferation, currently lacking effective therapeutic options. The combined use of Celastrol and Ligustrazine has been proved to synergistically improve the pathological processes of inflammation and fibrosis. In earlier studies, we designed and synthesized a Celastrol-Ligustrazine compound CL-001, though its role in IPF remains unclear. Here, the effects and mechanisms of CL-001 in bleomycin (BLM)-induced IPF were investigated. In vivo, CL-001 significantly improved lung function, reduced pulmonary inflammation, and decreased collagen deposition, thereby preventing the progression of IPF. In vitro, CL-001 concurrently inhibited both Smad-dependent and Smad-independent pathways, thereby suppressing TGF-β1-induced epithelial-mesenchymal transition (EMT) and epithelial cell migration. This inhibitory effect was superior to that of Celastrol or Ligustrazine administered alone. Additionally, CL-001 significantly increased the level of apoptosis and promoted the expression of apoptosis-related proteins (Caspase-8 and PARP), ultimately leading to widespread apoptosis in activated lung epithelial cells. In summary, CL-001 exhibits excellent anti-IPF effects both in vitro and in vivo, suggesting its potential as a novel candidate drug for IPF, warranting further development.
Collapse
Affiliation(s)
- Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China.
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
24
|
Mady B, Ibrahim HF, ElAziz Mm A, Basta M, Assem S, Ahmed Ali M, El Mottelib LMMAA. The potential ameliorating effect of vitamin E on bleomycin - induced lung fibrosis in adult albino rats. Int Immunopharmacol 2024; 136:112375. [PMID: 38823182 DOI: 10.1016/j.intimp.2024.112375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Lung fibrosis is a critical interstitial lung disease with poor prognosis. There is an urgent need to develop a proper and cost-effective therapeutic modality that can reverse and/or ameliorate lung fibrosis. Vitamin E is one of the widely investigated dietary antioxidants which has been linked to improvement of many health problems. The current study was conducted to evaluate the possible roles of vitamin E in prevention and treatment of bleomycin (BLM) induced lung fibrosis. Physiological, anatomical, histopathological and immunohistochemical studies were done to assess and compare between the structure and function of the lung tissue in lung fibrosis model, early and late treated groups with vitamin E. Furthermore, measurement of transforming growth factor-β(TGF-β), E-cadherin, Smad-3, BAX, BCL2, malondialdehyde (MDA), and superoxide dismutase (SOD) were done. The study revealed that administration of vitamin E helped to improve signs of lung fibrosis, as reflected by amelioration of structure and functions of lungs as well as the decrease in TGF-β levels and inhibition of α-SMA/collagen I profibrotic pathway. These findings highlight the importance of administration of vitamin E as a prophylactic agent prior to BLM therapy and as an adjuvant treatment in cases of lung fibrosis.
Collapse
Affiliation(s)
- Basma Mady
- Department of Human Anatomy and Embryology, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt.
| | - Heba F Ibrahim
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia; Department of Medical Histology and Cell Biology, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt
| | - Abd ElAziz Mm
- Department of Pathology, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt
| | - Marianne Basta
- Department of Physiology, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt
| | - Sara Assem
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt
| | - Manal Ahmed Ali
- Veterinarian, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt
| | - Lobna M M A Abd El Mottelib
- Department of Human Anatomy and Embryology, Faculty of Medicine, Alexandria University, Dr Fahmi Abdelmeguid St., Mowassah Campus, Alexandria 21561, Egypt; Department of Basic Medical Sciences, College of Dental Medicine, American University for Medical Sciences (AUMS), Eqaila, Block 6, Street 103, Building 2, Kuwait
| |
Collapse
|
25
|
Chen Z, Xie W, Tang S, Lin M, Ren L, Huang X, Deng L, Qian R, Wang Z, Xiong D, Xie P, Liu W. Taraxerone exerts antipulmonary fibrosis effect through Smad signaling pathway and antioxidant stress response in a Sirtuin1-dependent manner. Phytother Res 2024; 38:3720-3735. [PMID: 38776174 DOI: 10.1002/ptr.8221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 05/24/2024]
Abstract
Idiopathic pulmonary fibrosis treatments are limited, often with severe side effects, highlighting the need for novel options. Taraxerone has diverse biomedical properties, but its mechanism remains unclear. This study investigates taraxerone's impact and the mechanisms involved in bleomycin-induced pulmonary fibrosis in mice. After establishing a pulmonary fibrosis mouse model, taraxerone was intraperitoneally injected continuously for 14-28 days. The in vivo antifibrotic and antioxidative stress effects of taraxerone were assessed. In vitro, the influence of taraxerone on transforming growth factor-β1-induced myofibroblast transformation and oxidative stress was investigated. Subsequently, quantitative polymerase chain reaction screened the histone deacetylase and Sirtuin family, and taraxerone's effects on SIRT1 were assessed. After SIRT1 siRNA treatment, changes in myofibroblast transformation and antioxidant capacity in response to taraxerone were observed. Acetylation and phosphorylation levels of Smad3 were evaluated. We also examined the binding levels of SIRT1 with Pho-Smad3 and Smad3, as well as the nuclear localization of Smad2/3. EX527 confirmed SIRT1's in vivo action in response to taraxerone. In vitro experiments suggested that taraxerone inhibited myofibroblast differentiation by activating SIRT1 and reducing oxidative stress. We also observed a new interaction between SIRT1 and the Smad complex. Taraxerone activates SIRT1, enabling it to bind directly to Smad3. This leads to reduced Smad complex phosphorylation and limited nuclear translocation. As a result, the transcription of fibrotic factors is reduced. In vivo validation confirms taraxerone's SIRT1-mediated antifibrotic effectiveness. This suggests that targeting SIRT1-mediated inhibition of myofibroblast differentiation could be a key strategy in taraxerone-based therapy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Ziwei Chen
- Xiangya Nursing School, Central South University, Changsha, China
- Laboratory Medicine Department, Xiangya Hospital, Central South University, Changsha, China
| | - Weixi Xie
- Xiangya Nursing School, Central South University, Changsha, China
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Miao Lin
- Xiangya Nursing School, Central South University, Changsha, China
| | - Lu Ren
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Lang Deng
- Xiangya Nursing School, Central South University, Changsha, China
| | - Rui Qian
- Xiangya Nursing School, Central South University, Changsha, China
| | - Zun Wang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Dayang Xiong
- Xiangya Nursing School, Central South University, Changsha, China
| | - Pingli Xie
- National Experimental Teaching Demonstration Center for Medical Function, Central South University, Changsha, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, China
| |
Collapse
|
26
|
Peng J, Wang Q, Sun R, Zhang K, Chen Y, Gong Z. Phospholipids of inhaled liposomes determine the in vivo fate and therapeutic effects of salvianolic acid B on idiopathic pulmonary fibrosis. J Control Release 2024; 371:1-15. [PMID: 38761856 DOI: 10.1016/j.jconrel.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/18/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Since phospholipids have an important effect on the size, surface potential and hardness of liposomes that decide their in vivo fate after inhalation, this research has systematically evaluated the effect of phospholipids on pulmonary drug delivery by liposomes. In this study, liposomes composed of neutral saturated/unsaturated phospholipids, anionic and cationic phospholipids were constructed to investigate how surface potential and the degree of saturation of fatty acid chains determined their mucus and epithelium permeability both in vitro and in vivo. Our results clearly indicated that liposomes composed of saturated neutral and anionic phospholipids possessed high stability and permeability, compared to that of liposomes composed of unsaturated phospholipids and cationic phospholipids. Furthermore, both in vivo imaging of fluorescence-labeled liposomes and biodistribution of salvianolic acid B (SAB) that encapsulated in liposomes were performed to estimate the effect of phospholipids on the lung exposure and retention of inhaled liposomes. Finally, inhaled SAB-loaded liposomes exhibited enhanced therapeutic effects in a bleomycin-induced idiopathic pulmonary fibrosis mice model via inhibition of inflammation and regulation on coagulation-fibrinolytic system. Such findings will be beneficial to the development of inhalable lipid-based nanodrug delivery systems for the treatment of respiratory diseases where inhalation is the preferred route of administration.
Collapse
Affiliation(s)
- Jianqing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Qin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Runbin Sun
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ke Zhang
- The Key and Characteristic Laboratory of Modern Pathogenicity Biology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
27
|
He B, Zeng Q, Tian Y, Luo Y, Liao M, Huang W, Wu B, Luo Z, Huang X, Liu W, Tang S. PGC1-Alpha/Sirt3 Signaling Pathway Mediates the Anti-Pulmonary Fibrosis Effect of Hirudin by Inhibiting Fibroblast Senescence. Biomedicines 2024; 12:1436. [PMID: 39062010 PMCID: PMC11274105 DOI: 10.3390/biomedicines12071436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrotic lung disease for which there is a lack of effective pharmacological treatments. Hirudin, a natural peptide extracted from leeches, has been used for broad pharmacological purposes. In this study, we investigated the therapeutic effects of hirudin on IPF and its related mechanism of action. By constructing a mouse model of pulmonary fibrosis and treating it with hirudin in vivo, we found that hirudin exerted anti-fibrotic, anti-oxidative, and anti-fibroblast senescence effects. Moreover, using an in vitro model of stress-induced premature senescence in primary mouse lung fibroblasts and treating with hirudin, we observed inhibition of fibroblast senescence and upregulation of PGC1-alpha and Sirt3 expression. However, specific silencing of PGC1-alpha or Sirt3 suppressed the anti-fibroblast senescence effect of hirudin. Thus, the PGC1-alpha/Sirt3 pathway mediates the anti-fibroblast senescence effect of hirudin, potentially serving as a molecular mechanism underlying its anti-fibrosis and anti-oxidative stress effects exerted on the lungs.
Collapse
Affiliation(s)
- Bin He
- School of Nursing, Hunan University of Medicine, Huaihua 418000, China; (B.H.); (Y.T.); (W.H.); (B.W.)
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Qian Zeng
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Yumei Tian
- School of Nursing, Hunan University of Medicine, Huaihua 418000, China; (B.H.); (Y.T.); (W.H.); (B.W.)
| | - Yuyang Luo
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Minlin Liao
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Wenjie Huang
- School of Nursing, Hunan University of Medicine, Huaihua 418000, China; (B.H.); (Y.T.); (W.H.); (B.W.)
| | - Bin Wu
- School of Nursing, Hunan University of Medicine, Huaihua 418000, China; (B.H.); (Y.T.); (W.H.); (B.W.)
| | - Ziqiang Luo
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (Q.Z.); (Y.L.); (M.L.); (X.H.)
| |
Collapse
|
28
|
Buccardi M, Grandi A, Ferrini E, Buseghin D, Villetti G, Civelli M, Sverzellati N, Aliverti A, Pennati F, Stellari FF. Micro-CT-assisted identification of the optimal time-window for antifibrotic treatment in a bleomycin mouse model of long-lasting pulmonary fibrosis. Sci Rep 2024; 14:14792. [PMID: 38926490 PMCID: PMC11208517 DOI: 10.1038/s41598-024-65030-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a debilitating and fatal lung disease characterized by the excessive formation of scar tissue and decline of lung function. Despite extensive research, only two FDA-approved drugs exist for IPF, with limited efficacy and relevant side effects. Thus, there is an urgent need for new effective therapies, whose discovery strongly relies on IPF animal models. Despite some limitations, the Bleomycin (BLM)-induced lung fibrosis mouse model is widely used for antifibrotic drug discovery and for investigating disease pathogenesis. The initial acute inflammation triggered by BLM instillation and the spontaneous fibrosis resolution that occurs after 3 weeks are the major drawbacks of this system. In the present study, we applied micro-CT technology to a longer-lasting, triple BLM administration fibrosis mouse model to define the best time-window for Nintedanib (NINT) treatment. Two different treatment regimens were examined, with a daily NINT administration from day 7 to 28 (NINT 7-28), and from day 14 to 28 (NINT 14-28). For the first time, we automatically derived both morphological and functional readouts from longitudinal micro-CT. NINT 14-28 showed significant effects on morphological parameters after just 1 week of treatment, while no modulations of these biomarkers were observed during the preceding 7-14-days period, likely due to persistent inflammation. Micro-CT morphological data evaluated on day 28 were confirmed by lung histology and bronchoalveolar lavage fluid (BALF) cells; Once again, the NINT 7-21 regimen did not provide substantial benefits over the NINT 14-28. Interestingly, both NINT treatments failed to improve micro-CT-derived functional parameters. Altogether, our findings support the need for optimized protocols in preclinical studies to expedite the drug discovery process for antifibrotic agents. This study represents a significant advancement in pulmonary fibrosis animal modeling and antifibrotic treatment understanding, with the potential for improved translatability through the concurrent structural-functional analysis offered by longitudinal micro-CT.
Collapse
Affiliation(s)
- Martina Buccardi
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parma, Italy
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy
| | - Andrea Grandi
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy
| | - Erica Ferrini
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Davide Buseghin
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
- ANTHEM (AdvaNced Technologies for Human-centrEd Medicine), Spoke 3, Milan, Italy
| | - Gino Villetti
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy
| | - Maurizio Civelli
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy
| | | | - Andrea Aliverti
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | - Francesca Pennati
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | - Franco Fabio Stellari
- Experimental Pharmacology and Translational Science Department, Chiesi Farmaceutici S.P.A, 43122, Parma, Italy.
| |
Collapse
|
29
|
Lin JH, Liu CC, Liu CY, Hsu TW, Yeh YC, How CK, Hsu HS, Hung SC. Selenite selectively kills lung fibroblasts to treat bleomycin-induced pulmonary fibrosis. Redox Biol 2024; 72:103148. [PMID: 38603946 PMCID: PMC11017345 DOI: 10.1016/j.redox.2024.103148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/01/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) treatment is a critical unmet need. Selenium is an essential trace element for human life and an antioxidant that activates glutathione, but the gap between its necessity and its toxicity is small and requires special attention. Whether selenium can be used in the treatment of ILD remains unclear. METHODS We investigated the prophylactic and therapeutic effects of selenite, a selenium derivative, in ILD using a murine model of bleomycin-induced idiopathic pulmonary fibrosis (IPF). We further elucidated the underlying mechanism using in vitro cell models and examined their relevance in human tissue specimens. The therapeutic effect of selenite in bleomycin-administered mice was assessed by respiratory function and histochemical changes. Selenite-induced apoptosis and reactive oxygen species (ROS) production in murine lung fibroblasts were measured. RESULTS Selenite, administered 1 day (inflammation phase) or 8 days (fibrotic phase) after bleomycin, prevented and treated deterioration of lung function and pulmonary fibrosis in mice. Mechanistically, selenite inhibited the proliferation and induced apoptosis of murine lung fibroblasts after bleomycin treatment both in vitro and in vivo. In addition, selenite upregulated glutathione reductase (GR) and thioredoxin reductase (TrxR) in murine lung fibroblasts, but not in lung epithelial cells, upon bleomycin treatment. GR and TrxR inhibition eliminates the therapeutic effects of selenite. Furthermore, we found that GR and TrxR were upregulated in the human lung fibroblasts of IPF patient samples. CONCLUSIONS Selenite induces ROS production and apoptosis in murine lung fibroblasts through GR and TrxR upregulation, thereby providing a therapeutic effect in bleomycin-induced IPF.
Collapse
Affiliation(s)
- Jiun-Han Lin
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chen-Chi Liu
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Traumatology, Emergency Department, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Chao-Yu Liu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Thoracic Surgery, Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Tien-Wei Hsu
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Chen Yeh
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chorng-Kuang How
- Division of Traumatology, Emergency Department, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Han-Shui Hsu
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Shih-Chieh Hung
- Drug Development Center, Institute of Translational Medicine and New Drug Development, School of Medicine, Taiwan; College of Life Sciences, China Medical University, Taichung, Taiwan; Integrative Stem Cell Center, Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
30
|
Henneke I, Pilz C, Wilhelm J, Alexopoulos I, Ezaddoustdar A, Mukhametshina R, Weissmann N, Ghofrani HA, Grimminger F, Seeger W, Schermuly RT, Wygrecka M, Kojonazarov B. Microscopic computed tomography with AI-CNN-powered image analysis: the path to phenotype bleomycin-induced lung injury. Am J Physiol Cell Physiol 2024; 326:C1637-C1647. [PMID: 38646782 DOI: 10.1152/ajpcell.00708.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Bleomycin (BLM)-induced lung injury in mice is a valuable model for investigating the molecular mechanisms that drive inflammation and fibrosis and for evaluating potential therapeutic approaches to treat the disease. Given high variability in the BLM model, it is critical to accurately phenotype the animals in the course of an experiment. In the present study, we aimed to demonstrate the utility of microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation for rapid phenotyping of BLM mice. µCT was performed in freely breathing C57BL/6J mice under isoflurane anesthesia on days 7 and 21 after BLM administration. Terminal invasive lung function measurement and histological assessment of the left lung collagen content were conducted as well. µCT image analysis demonstrated gradual and time-dependent development of lung injury as evident by alterations in the lung density, air-to-tissue volume ratio, and lung aeration in mice treated with BLM. The right and left lung were unequally affected. µCT-derived parameters such as lung density, air-to-tissue volume ratio, and nonaerated lung volume correlated well with the invasive lung function measurement and left lung collagen content. Our study demonstrates the utility of AI-CNN-powered µCT image analysis for rapid and accurate phenotyping of BLM mice in the course of disease development and progression.NEW & NOTEWORTHY Microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation is a rapid and powerful tool for noninvasive phenotyping of bleomycin mice over the course of the disease. This, in turn, allows earlier and more reliable identification of therapeutic effects of new drug candidates, ultimately leading to the reduction of unnecessary procedures in animals in pharmacological research.
Collapse
Affiliation(s)
- Ingrid Henneke
- Experimental Lung Disease Models Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Christina Pilz
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Jochen Wilhelm
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Genomics and Bioinformatics Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Multyscale Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Aysan Ezaddoustdar
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Center for Infection and Genomics of the Lung (CIGL), Faculty of Medicine, Justus Liebig University (JLU), Giessen, Germany
| | - Regina Mukhametshina
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Small Animal Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Norbert Weissmann
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph T Schermuly
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Malgorzata Wygrecka
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Center for Infection and Genomics of the Lung (CIGL), Faculty of Medicine, Justus Liebig University (JLU), Giessen, Germany
- CSL Behring Innovation GmbH, Marburg, Germany
| | - Baktybek Kojonazarov
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Small Animal Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| |
Collapse
|
31
|
Alhakamy NA, Alamoudi AJ, Asfour HZ, Ahmed OAA, Abdel-Naim AB, Aboubakr EM. L-arginine mitigates bleomycin-induced pulmonary fibrosis in rats through regulation of HO-1/PPAR-γ/β-catenin axis. Int Immunopharmacol 2024; 131:111834. [PMID: 38493696 DOI: 10.1016/j.intimp.2024.111834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Pulmonary fibrosis is a chronic and progressively deteriorating lung condition that can be replicated in laboratory animals by administering bleomycin, a chemotherapeutic antibiotic known for its lung fibrosis-inducing side effects. L-arginine, a semi-essential amino acid, is recognized for its diverse biological functions, including its potential to counteract fibrosis. This study aimed to evaluate the antifibrotic properties of L-arginine on bleomycin-induced pulmonary fibrosis in rats. The administration of a single intratracheal dose of bleomycin resulted in visible and microscopic damage to lung tissues, an uptick in oxidative stress markers, and an elevation in inflammatory, apoptotic, and fibrotic indicators. A seven-day treatment with L-arginine post-bleomycin exposure markedly improved the gross and histological architecture of the lungs, prevented the rise of malondialdehyde and carbonyl content, and enhanced total antioxidant capacity alongside the activities of antioxidant enzymes. Also, L-arginine attenuated the expression of the pro-fibrotic factors, transforming growth factor-β and lactate dehydrogenase in bronchoalveolar lavage fluid. In the lung tissue, L-arginine reduced collagen deposition, hydroxyproline concentration, and mucus production, along with decreasing expression of α-smooth muscle actin, tumor necrosis factor-α, caspase-3, matrix metalloproteinase-9, and β-catenin. Moreover, it boosted levels of nitric oxide and upregulated the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), heme oxygenase-1 (HO-1), and E-cadherin and downregulating the expression of β-catenin. These findings suggest that L-arginine has preventive activities against bleomycin-induced pulmonary fibrosis. This effect can be attributed to the increased production of nitric oxide, which modulates the HO-1/PPAR-γ/β-catenin axis.
Collapse
Affiliation(s)
- Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulmohsin J Alamoudi
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Hani Z Asfour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
| | - Osama A A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam M Aboubakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
| |
Collapse
|
32
|
Chen L, Lin H, Qin L, Zhang G, Huang D, Chen P, Zhang X. Identification and validation of mutual hub genes in idiopathic pulmonary fibrosis and rheumatoid arthritis-associated usual interstitial pneumonia. Heliyon 2024; 10:e28088. [PMID: 38571583 PMCID: PMC10987927 DOI: 10.1016/j.heliyon.2024.e28088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Objectives The study aims at exploring common hub genes and pathways in idiopathic pulmonary fibrosis (IPF) and rheumatoid arthritis-associated usual interstitial pneumonia (RA-UIP) through integrated bioinformatics analyses. Methods The GSE199152 dataset containing lung tissue samples from IPF and RA-UIP patients was acquired from the Gene Expression Omnibus (GEO) database. The identification of overlapping differentially expressed genes (DEGs) in IPF and RA-UIP was carried out through R language. Protein-protein interaction (PPI) network analysis and module analysis were applied to filter mutual hub genes in the two diseases. Enrichment analyses were also conducted to analyze the possible biological functions and pathways of the overlapped DEGs and hub genes. The diagnostic value of key genes was assessed with R language, and the expressions of these genes in pulmonary cells of IPF and rheumatoid arthritis-associated interstitial lung disease (RA-ILD) patients were analyzed with single cell RNA-sequencing (scRNA-seq) datasets. The expression levels of hub genes were validated in blood samples from patients, specimens of human lung fibroblasts, lung tissue samples from mice, as well as external GEO datasets. Results Four common hub genes (THBS2, TIMP1, POSTN, and CD19) were screened. Enrichment analyses showed that the abnormal expressions of DEGs and hub genes may be connected with the onset of IPF and RA-UIP by regulating the progression of fibrosis. ScRNA-seq analyses illustrated that for both IPF and RA-ILD patients, THBS2, TIMP1, and POSTN were mainly expressed in lung fibroblasts, while CD19 was uniquely high-expressed in B cells. The qRT-PCR and immunohistochemistry (IHC) results verified that the expression levels of hub genes were mostly in accordance with the findings obtained from the bioinformatics analyses. Conclusion Though IPF and RA-UIP are distinct diseases, they may to some extent have mutual pathogenesis in the development of fibrosis. THBS2, TIMP1, POSTN, and CD19 may be the potential biomarkers of IPF and RA-UIP, and intervention on related pathways of these genes could offer new strategies for the precision treatment of IPF and RA-UIP.
Collapse
Affiliation(s)
- Liangyu Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
- Department of Respiratory and Critical Care Medicine, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Haobo Lin
- Department of Rheumatology, Guangdong Provincial People's Hospital, Guangzhou, China
- Guangdong Academy of Medical Sciences, Guangzhou, China
- Southern Medical University, Guangzhou, China
| | - Linmang Qin
- Department of Rheumatology, Guangdong Provincial People's Hospital, Guangzhou, China
- Guangdong Academy of Medical Sciences, Guangzhou, China
- Southern Medical University, Guangzhou, China
| | - Guangfeng Zhang
- Department of Rheumatology, Guangdong Provincial People's Hospital, Guangzhou, China
- Guangdong Academy of Medical Sciences, Guangzhou, China
- Southern Medical University, Guangzhou, China
| | - Donghui Huang
- Department of Respiratory and Critical Care Medicine, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Peisheng Chen
- Department of Respiratory and Critical Care Medicine, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Xiao Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
- Department of Rheumatology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
33
|
Mahmutovic Persson I, Fransén Petterson N, Liu J, in ‘t Zandt R, Carvalho C, Örbom A, Olsson LE, von Wachenfeldt K. In vivo MRI and PET imaging in a translational ILD mouse model expressing non-resolving fibrosis and bronchiectasis-like pathology after repeated systemic exposure to bleomycin. Front Med (Lausanne) 2024; 11:1276420. [PMID: 38654839 PMCID: PMC11035813 DOI: 10.3389/fmed.2024.1276420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 03/11/2024] [Indexed: 04/26/2024] Open
Abstract
Drug-induced interstitial lung disease (ILD) is crucial to detect early to achieve the best treatment outcome. Optimally, non-invasive imaging biomarkers can be used for early detection of disease progression and treatment follow-up. Therefore, reliable in vivo models are warranted in new imaging biomarker development to accelerate better-targeted treatment options. Single-dose bleomycin models have, for a long time, served as a reference model in fibrosis and lung injury research. Here, we aimed to use a clinically more relevant animal model by systemic exposure to bleomycin and assessing disease progression over time by combined magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging. Methods C57BL/6 mice received bleomycin (i.p. 35iU/kg) or saline as control twice per week for 4 weeks. Mice were monitored until 2 weeks after cessation of bleomycin administration (w4 + 1 and w4 + 2), referred to as the resting period. MRI scans were performed in weeks 3 and 4 and during the resting weeks. [18F]FDG-PET was performed at the last week of dosing (w4) and 2 weeks after the last dosing (w4 + 2). Lung tissue sections were stained with Masson's trichrome and evaluated by modified Ashcroft scoring. Lung volume and lesion volumes were assessed using MRI, as well as 3D mapping of the central airways. Results and discussion Bleomycin-challenged mice showed increased lung weights (p < 0.05), while total lung volume was unchanged (w4 and onward). Histology analysis demonstrated fibrotic lesions emanating from the distal parts of the lung. Fibrosis progression was visualized by MRI with significantly increased high signal in bleomycin-exposed lungs compared to controls (p < 0.05). In addition, a significant increase in central airway diameter (p < 0.01) was displayed in bleomycin-exposed animals compared to controls and further continued to dilate as the disease progressed, comparing the bleomycin groups over time (p < 0.05-0.001). Lung [18F]FDG uptake was significantly elevated in bleomycin-exposed mice compared to controls (p < 0.05). Conclusion Non-invasive imaging displayed progressing lesions in the lungs of bleomycin-exposed mice, using two distinct MRI sequences and [18F]FDG-PET. With observed fibrosis progression emanating from distal lung areas, dilation of the central airways was evident. Taken together, this chronic bleomycin-exposure model is translationally more relevant for studying lung injury in ILD and particularly in the context of DIILD.
Collapse
Affiliation(s)
- Irma Mahmutovic Persson
- Medical Radiation Physics, Institution of Translational Medicine, Lund University, Malmö, Sweden
- Lund University BioImaging Centre (LBIC), Medical Faculty, Lund University, Lund, Sweden
| | | | | | - René in ‘t Zandt
- Lund University BioImaging Centre (LBIC), Medical Faculty, Lund University, Lund, Sweden
| | | | - Anders Örbom
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lars E. Olsson
- Medical Radiation Physics, Institution of Translational Medicine, Lund University, Malmö, Sweden
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Malmö, Sweden
| | | |
Collapse
|
34
|
Hennion N, Chenivesse C, Humez S, Gottrand F, Desseyn JL, Gouyer V. [Idiopathic pulmonary fibrosis: Desperately seeking a model]. Rev Mal Respir 2024; 41:274-278. [PMID: 38480096 DOI: 10.1016/j.rmr.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 04/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and fatal lung disease of which the origin and development mechanisms remain unknown. The few available pharmacological treatments can only slow the progression of the disease. The development of curative treatments is hampered by the absence of experimental models that can mimic the specific pathophysiological mechanisms of IPF. The aim of this mini-review is to provide an overview of the most commonly used experimental animal models in the study of IPF and to underline the urgent need to seek out new, more satisfactory models.
Collapse
Affiliation(s)
- N Hennion
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| | - C Chenivesse
- Inserm, CNRS, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Centre de Référence Constitutif des Maladies Pulmonaires Rares, Université de Lille, CHU de Lille, Lille, France
| | - S Humez
- Department of Pathology, Université de Lille, CHU de Lille, Lille, France; Inserm, CNRS, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut Pasteur de Lille, Université de Lille, CHU de Lille, Lille, France
| | - F Gottrand
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| | - J-L Desseyn
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France.
| | - V Gouyer
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| |
Collapse
|
35
|
Zhang J, Zhang Y, Chen Q, Qi Y, Zhang X. The XPO1 inhibitor selinexor ameliorates bleomycin-induced pulmonary fibrosis in mice via GBP5/NLRP3 inflammasome signaling. Int Immunopharmacol 2024; 130:111734. [PMID: 38422768 DOI: 10.1016/j.intimp.2024.111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Pulmonary fibrosis is an irreversible and progressive lung disease with limited treatments available. Selinexor (Sel), an orally available, small-molecule, selective inhibitor of XPO1, exhibits notable antitumor, anti-inflammatory and antiviral activities. However, its potential role in treating pulmonary fibrosis is unknown. C57BL/6J mice were used to establish a pulmonary fibrosis model by intratracheal administration of bleomycin (BLM). Subsequently, Sel was administered intraperitoneally. Our data demonstrated that Sel administration ameliorated BLM-induced pulmonary fibrosis by increasing mouse body weights; reducing H&E staining, Masson staining scores, and shadows in mouse lung computed tomography (CT) images, decreasing the total cell and neutrophil counts in the lung and bronchoalveolar lavage fluid (BALF); and decreasing the levels of TGF-β1. We next confirmed that Sel reduced the deposition of extracellular matrix (ECM) components in the lungs of BLM-induced pulmonary fibrosis mice. We showed that collagen I, alpha-smooth muscle actin (α-SMA), and hydroxyproline levels and the mRNA levels of Col1a1, Eln, Fn1, Ctgf, and Fgf2 were reduced. Mechanistically, tandem mass tags (TMT)- based quantitative proteomics analysis revealed a significant increase in GBP5 in the lungs of BLM mice but a decrease in that of BLM + Sel mice; this phenomenon was confirmed by western blotting and RT-qPCR. NLRP3 inflammasome signaling was significantly enriched in both the BLM group and BLM + Sel group based on GO and KEGG analyses of differentially expressed proteins between the groups. Furthermore, Sel reduced the expression of NLRP3, cleaved caspase 1, and ASC in vivo and in vitro, and decreased the levels of IL-1β, IL-18, and IFN-r in lung tissue and BALF. SiRNA-GBP5 inhibited NLRP3 signaling in vitro, and overexpression of GBP5 inhibited the protective effect of Sel against BLM-induced cellular injury. Taken together, our findings indicate that Sel ameliorates BLM-induced pulmonary fibrosis by targeting GBP5 via NLRP3 inflammasome signaling. Thus, the XPO1 inhibitor - Sel might be a potential therapeutic agent for pulmonary fibrosis.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003 Zhengzhou, Henan, China
| | - Yihua Zhang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003 Zhengzhou, Henan, China; Xinxiang Medical University, 453003 Xinxiang, Henan, China
| | - Qi Chen
- Henan University People's Hospital, 450003 Zhengzhou, Henan, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003 Zhengzhou, Henan, China; Henan University People's Hospital, 450003 Zhengzhou, Henan, China.
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003 Zhengzhou, Henan, China; Xinxiang Medical University, 453003 Xinxiang, Henan, China.
| |
Collapse
|
36
|
Xie W, Deng L, Qian R, Huang X, Liu W, Tang S. Curculigoside Attenuates Endoplasmic Reticulum Stress-Induced Epithelial Cell and Fibroblast Senescence by Regulating the SIRT1-P300 Signaling Pathway. Antioxidants (Basel) 2024; 13:420. [PMID: 38671868 PMCID: PMC11047561 DOI: 10.3390/antiox13040420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
The senescence of alveolar epithelial cells (AECs) and fibroblasts plays a pivotal role in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a condition lacking specific therapeutic interventions. Curculigoside (CCG), a prominent bioactive constituent of Curculigo, exhibits anti-osteoporotic and antioxidant activities. Our investigation aimed to elucidate the anti-senescence and anti-fibrotic effects of CCG in experimental pulmonary fibrosis and delineate its underlying molecular mechanisms. Our findings demonstrate that CCG attenuates bleomycin-induced pulmonary fibrosis and lung senescence in murine models, concomitantly ameliorating lung function impairment. Immunofluorescence staining for senescence marker p21, alongside SPC or α-SMA, suggested that CCG's mitigation of lung senescence correlates closely with the deceleration of senescence in AECs and fibroblasts. In vitro, CCG mitigated H2O2-induced senescence in AECs and the natural senescence of primary mouse fibroblasts. Mechanistically, CCG can upregulate SIRT1 expression, downregulating P300 expression, enhancing Trim72 expression to facilitate P300 ubiquitination and degradation, reducing the acetylation levels of antioxidant enzymes, and upregulating their expression levels. These actions collectively inhibited endoplasmic reticulum stress (ERS) and alleviated senescence. Furthermore, the anti-senescence effects and mechanisms of CCG were validated in a D-galactose (D-gal)-induced progeroid model. This study provides novel insights into the mechanisms underlying the action of CCG in cellular senescence and chronic diseases, offering potential avenues for the development of innovative drugs or therapeutic strategies.
Collapse
Affiliation(s)
- Weixi Xie
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Lang Deng
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Rui Qian
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
- The School of Nursing, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
37
|
Li C, Feng X, Li S, He X, Luo Z, Cheng X, Yao J, Xiao J, Wang X, Wen D, Liu D, Li Y, Zhou H, Ma L, Lin T, Cai X, Lin Y, Guo L, Yang M. Tetrahedral DNA loaded siCCR2 restrains M1 macrophage polarization to ameliorate pulmonary fibrosis in chemoradiation-induced murine model. Mol Ther 2024; 32:766-782. [PMID: 38273656 PMCID: PMC10928155 DOI: 10.1016/j.ymthe.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lethal disease in the absence of demonstrated efficacy for preventing progression. Although macrophage-mediated alveolitis is determined to participate in myofibrotic transition during disease development, the paradigm of continuous macrophage polarization is still under-explored due to lack of proper animal models. Here, by integrating 2.5 U/kg intratracheal Bleomycin administration and 10 Gy thorax irradiation at day 7, we generated a murine model with continuous alveolitis-mediated fibrosis, which mimics most of the clinical features of our involved IPF patients. In combination with data from scRNA-seq of patients and a murine IPF model, a decisive role of CCL2/CCR2 axis in driving M1 macrophage polarization was revealed, and M1 macrophage was further confirmed to boost alveolitis in leading myofibroblast activation. Multiple sticky-end tetrahedral framework nucleic acids conjunct with quadruple ccr2-siRNA (FNA-siCCR2) was synthesized in targeting M1 macrophages. FNA-siCCR2 successfully blocked macrophage accumulation in pulmonary parenchyma of the IPF murine model, thus preventing myofibroblast activation and leading to the disease remitting. Overall, our studies lay the groundwork to develop a novel IPF murine model, reveal M1 macrophages as potential therapeutic targets, and establish new treatment strategy by using FNA-siCCR2, which are highly relevant to clinical scenarios and translational research in the field of IPF.
Collapse
Affiliation(s)
- Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaorong Feng
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Zeli Luo
- Department of Pulmonary and Critical Care Medicine, Wenjiang Hospital of Sichuan Provincial People's, Chengdu 611138, China
| | - Xia Cheng
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Jie Yao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaofei Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingke Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Duanya Liu
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Yanfei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tongyu Lin
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; College of Biomedical Engineering, Sichuan University, Chengdu 610041, China.
| | - Lu Guo
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Mu Yang
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China.
| |
Collapse
|
38
|
Montesi SB, Gomez CR, Beers M, Brown R, Chattopadhyay I, Flaherty KR, Garcia CK, Gomperts B, Hariri LP, Hogaboam CM, Jenkins RG, Kaminski N, Kim GHJ, Königshoff M, Kolb M, Kotton DN, Kropski JA, Lasky J, Magin CM, Maher TM, McCormick M, Moore BB, Nickerson-Nutter C, Oldham J, Podolanczuk AJ, Raghu G, Rosas I, Rowe SM, Schmidt WT, Schwartz D, Shore JE, Spino C, Craig JM, Martinez FJ. Pulmonary Fibrosis Stakeholder Summit: A Joint NHLBI, Three Lakes Foundation, and Pulmonary Fibrosis Foundation Workshop Report. Am J Respir Crit Care Med 2024; 209:362-373. [PMID: 38113442 PMCID: PMC10878386 DOI: 10.1164/rccm.202307-1154ws] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023] Open
Abstract
Despite progress in elucidation of disease mechanisms, identification of risk factors, biomarker discovery, and the approval of two medications to slow lung function decline in idiopathic pulmonary fibrosis and one medication to slow lung function decline in progressive pulmonary fibrosis, pulmonary fibrosis remains a disease with a high morbidity and mortality. In recognition of the need to catalyze ongoing advances and collaboration in the field of pulmonary fibrosis, the NHLBI, the Three Lakes Foundation, and the Pulmonary Fibrosis Foundation hosted the Pulmonary Fibrosis Stakeholder Summit on November 8-9, 2022. This workshop was held virtually and was organized into three topic areas: 1) novel models and research tools to better study pulmonary fibrosis and uncover new therapies, 2) early disease risk factors and methods to improve diagnosis, and 3) innovative approaches toward clinical trial design for pulmonary fibrosis. In this workshop report, we summarize the content of the presentations and discussions, enumerating research opportunities for advancing our understanding of the pathogenesis, treatment, and outcomes of pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Christian R. Gomez
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael Beers
- Pulmonary and Critical Care Division, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert Brown
- Program in Neurotherapeutics, University of Massachusetts Chan Medical School, Worchester, Massachusetts
| | | | | | - Christine Kim Garcia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine and
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Cory M. Hogaboam
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - R. Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Grace Hyun J. Kim
- Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, and
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Melanie Königshoff
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Martin Kolb
- Division of Respirology, McMaster University, Hamilton, Ontario, Canada
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph Lasky
- Pulmonary Fibrosis Foundation, Chicago, Illinois
- Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Chelsea M. Magin
- Department of Bioengineering
- Department of Pediatrics
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
| | - Toby M. Maher
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | | | | | | | - Anna J. Podolanczuk
- Division of Pulmonary and Critical Care, Weill Cornell Medical College, New York, New York
| | - Ganesh Raghu
- Division of Pulmonary, Sleep and Critical Care Medicine, University of Washington, Seattle, Washington
| | - Ivan Rosas
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas; and
| | - Steven M. Rowe
- Department of Medicine and
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - David Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Cathie Spino
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - J. Matthew Craig
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care, Weill Cornell Medical College, New York, New York
| |
Collapse
|
39
|
Han MM, Tang L, Huang B, Li XN, Fang YF, Qi L, Duan BW, Yao YT, He YJ, Xing L, Jiang HL. Inhaled nanoparticles for treating idiopathic pulmonary fibrosis by inhibiting honeycomb cyst and alveoli interstitium remodeling. J Control Release 2024; 366:732-745. [PMID: 38242209 DOI: 10.1016/j.jconrel.2024.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with high mortality. The Food and Drug Administration-approved drugs, nintedanib and pirfenidone, could delay progressive fibrosis by inhibiting the overactivation of fibroblast, however, there was no significant improvement in patient survival due to low levels of drug accumulation and remodeling of honeycomb cyst and interstitium surrounding the alveoli. Herein, we constructed a dual drug (verteporfin and pirfenidone)-loaded nanoparticle (Lip@VP) with the function of inhibiting airway epithelium fluidization and fibroblast overactivation to prevent honeycomb cyst and interstitium remodeling. Specifically, Lip@VP extensively accumulated in lung tissues via atomized inhalation. Released verteporfin inhibited the fluidization of airway epithelium and the formation of honeycomb cyst, and pirfenidone inhibited fibroblast overactivation and reduced cytokine secretion that promoted the fluidization of airway epithelium. Our results indicated that Lip@VP successfully rescued lung function through inhibiting honeycomb cyst and interstitium remodeling. This study provided a promising strategy to improve the therapeutic efficacy for IPF.
Collapse
Affiliation(s)
- Meng-Meng Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Bin Huang
- Department of Lung Transplantation, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xue-Na Li
- College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Yue-Fei Fang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Qi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bo-Wen Duan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ya-Ting Yao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Jing He
- School of Pharmaceutical Sciences & Institute of Materia Medica Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; College of Pharmacy, Yanbian University, Yanji 133002, China; Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
40
|
Chen Y, Li Z, Ji G, Wang S, Mo C, Ding B. Lung regeneration: diverse cell types and the therapeutic potential. MedComm (Beijing) 2024; 5:e494. [PMID: 38405059 PMCID: PMC10885188 DOI: 10.1002/mco2.494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Lung tissue has a certain regenerative ability and triggers repair procedures after injury. Under controllable conditions, lung tissue can restore normal structure and function. Disruptions in this process can lead to respiratory system failure and even death, causing substantial medical burden. The main types of respiratory diseases are chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and acute respiratory distress syndrome (ARDS). Multiple cells, such as lung epithelial cells, endothelial cells, fibroblasts, and immune cells, are involved in regulating the repair process after lung injury. Although the mechanism that regulates the process of lung repair has not been fully elucidated, clinical trials targeting different cells and signaling pathways have achieved some therapeutic effects in different respiratory diseases. In this review, we provide an overview of the cell type involved in the process of lung regeneration and repair, research models, and summarize molecular mechanisms involved in the regulation of lung regeneration and fibrosis. Moreover, we discuss the current clinical trials of stem cell therapy and pharmacological strategies for COPD, IPF, and ARDS treatment. This review provides a reference for further research on the molecular and cellular mechanisms of lung regeneration, drug development, and clinical trials.
Collapse
Affiliation(s)
- Yutian Chen
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Zhen Li
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gaili Ji
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shaochi Wang
- Department of Translational MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Bi‐Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
41
|
Chen H, Liu C, Zhan Y, Wang Y, Hu Q, Zeng Z. Alpinetin ameliorates bleomycin-induced pulmonary fibrosis by repressing fibroblast differentiation and proliferation. Biomed Pharmacother 2024; 171:116101. [PMID: 38228032 DOI: 10.1016/j.biopha.2023.116101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 01/18/2024] Open
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial lung disease with a poor prognosis. Alpinetin (ALP), derived from Alpinia katsumadai Hayata, has shown potential as a therapeutic measure of various diseases. However, the utilization of ALP in managing pulmonary fibrosis and its underlying mechanisms are still not fully understood. METHODS A well-established mouse model of pulmonary fibrosis induced by bleomycin (BLM) was used in this study. The antifibrotic effects of ALP on histopathologic manifestations and expression levels of fibrotic markers were examined. Subsequently, the impact of ALP on fibroblast differentiation, proliferation, apoptosis, and associated signaling pathways was investigated to elucidate the underlying mechanisms. RESULTS In the present study, we observed that ALP effectively mitigated BLM-induced pulmonary fibrosis in mice, as evidenced by histopathological manifestations and the expression levels of fibrotic markers. Furthermore, the in vitro experiments demonstrated that ALP treatment attenuated the ability of fibroblasts to differentiate into myofibroblasts. Mechanically, our findings provided evidence that ALP suppressed fibroblast-to-myofibroblast differentiation by repressing TGF-β/ALK5/Smad signaling pathway. ALP was found to possess the capability of inhibiting fibroblast proliferation and promoting apoptosis of fibroblasts induced by TGF-β. CONCLUSION In general, ALP may exert therapeutic effects on pulmonary fibrosis by modulating the differentiation, proliferation, and apoptosis of fibroblasts. Although its safety has been demonstrated in mice, further studies are required to investigate the efficacy of ALP in treatment of patients with IPF.
Collapse
Affiliation(s)
- Huilong Chen
- Department and Institute of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changyu Liu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Zhan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yi Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiongjie Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilin Zeng
- Department and Institute of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
42
|
Hua Q, Ren L. The SIRT1/Nrf2 signaling pathway mediates the anti-pulmonary fibrosis effect of liquiritigenin. Chin Med 2024; 19:12. [PMID: 38238857 PMCID: PMC10795230 DOI: 10.1186/s13020-024-00886-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/07/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND At present, the treatment options available for idiopathic pulmonary fibrosis are both limited and often come with severe side effects, emphasizing the pressing requirement for innovative therapeutic alternatives. Myofibroblasts, which hold a central role in pulmonary fibrosis, have a close association with the Smad signaling pathway induced by transforming growth factor-β1 (TGF-β1) and the transformation of myofibroblasts driven by oxidative stress. Liquiritigenin, an active compound extracted from the traditional Chinese herb licorice, boasts a wide array of biomedical properties, such as anti-fibrosis and anti-oxidation. The primary objective of this study was to examine the impact of liquiritigenin on bleomycin-induced pulmonary fibrosis in mice and the underlying mechanisms. METHODS The anti-pulmonary fibrosis and anti-oxidant effects of liquiritigenin in vivo were tested by HE staining, Masson staining, DHE staining and bio-chemical methods. In vitro, primary mouse lung fibroblasts were treated with TGF-β1 with or without liquiritigenin, the effects of liquiritigenin in inhibiting differentiation of myofibroblasts and facilitating the translocation of Nrf2 were valued using Quantitative real-time polymerase chain reaction (Q-PCR), western blotting and immunofluorescence. Nrf2 siRNA and SIRT1 siRNA were used to investigate the mechanism underlies liquiritigenin's effect in inhibiting myofibroblast differentiation. RESULTS Liquiritigenin displayed a dose-dependent reduction effect in bleomycin-induced fibrosis. In laboratory experiments, it was evident that liquiritigenin possessed the ability to enhance and activate sirtuin1 (SIRT1), thereby facilitating the nuclear translocation of Nrf2 and mitigating the oxidative stress-induced differentiation of primary mouse myofibroblasts. Moreover, our investigation unveiled that SIRT1 not only regulated myofibroblast differentiation via Nrf2-mediated antioxidant responses against oxidative stress but also revealed liquiritigenin's activation of SIRT1, enabling direct binding to Smad. This led to decreased phosphorylation of the Smad complex, constrained nuclear translocation, and suppressed acetylation of the Smad complex, ultimately curtailing the transcription of fibrotic factors. Validation in live subjects provided substantial evidence for the anti-fibrotic efficacy of liquiritigenin through the SIRT1/Nrf2 signaling pathway. CONCLUSIONS Our findings imply that targeting myofibroblast differentiation via the SIRT1/Nrf2 signaling pathway may constitute a pivotal strategy for liquiritigenin-based therapy against pulmonary fibrosis.
Collapse
Affiliation(s)
- Qingzhong Hua
- The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518101, Guangdong, China
| | - Lu Ren
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
43
|
Kaur R, Shaikh TB, Priya Sripadi H, Kuncha M, Vijaya Sarathi UVR, Kulhari H, Balaji Andugulapati S, Sistla R. Nintedanib solid lipid nanoparticles improve oral bioavailability and ameliorate pulmonary fibrosis in vitro and in vivo models. Int J Pharm 2024; 649:123644. [PMID: 38040396 DOI: 10.1016/j.ijpharm.2023.123644] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Nintedanib (NIN) and pirfenidone are the only approved drugs for the treatment of Idiopathic Pulmonary Fibrosis (IPF). However, NIN and pirfenidone have low oral bioavailability and limited therapeutic potential, requiring higher dosages to increase their efficacy, which causes significant liver and gastrointestinal toxicities. In this study, we aimed to develop nintedanib-loaded solid lipid nanoparticles (NIN-SLN) to improve the oral bioavailability and therapeutic potential against TGF-β-induced differentiation in IPF fibroblasts and bleomycin (BLM)-induced lung fibrosis in rat models. NIN-SLN was prepared using a double-emulsification method and characterization studies (Particle size, zeta potential, entrapment efficiency and other parameters) were performed using various techniques. NIN-SLN treatment significantly (p < 0.001) downregulated α-SMA and COL3A1 expression in TGF-β stimulated DHLF and LL29 cells. NIN-SLN showed a 2.87-fold increase in the bioavailability of NIN and also improved the NIN levels in lung tissues compared to NIN alone. Pharmacodynamic investigation revealed that NIN-SLN (50 mg/Kg) treatment significantly attenuated BLM-induced lung fibrosis by inhibiting epithelial-to-mesenchymal-transition (EMT), extracellular matrix remodelling, and collagen deposition compared to free NIN. Additionally, in the BLM model of fibrosis, NIN-SLN greatly improved the BLM-caused pathological changes, attenuated the NIN-induced gastrointestinal abnormalities, and significantly improved the lung functional indices compared to free NIN. Collectively, NIN-SLN could be a promising nanoformulation for the management of pulmonary fibrosis.
Collapse
Affiliation(s)
- Rajwinder Kaur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Taslim B Shaikh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Hari Priya Sripadi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Madhusudana Kuncha
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India
| | - U V R Vijaya Sarathi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India; Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India
| | - Hitesh Kulhari
- School of Nano Sciences, Central University of Gujarat, Gandhinagar 382 030, Gujarat, India.
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| | - Ramakrishna Sistla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
44
|
Zheng M, Liu K, Li L, Feng C, Wu G. Traditional Chinese medicine inspired dual-drugs loaded inhalable nano-therapeutics alleviated idiopathic pulmonary fibrosis by targeting early inflammation and late fibrosis. J Nanobiotechnology 2024; 22:14. [PMID: 38166847 PMCID: PMC10763202 DOI: 10.1186/s12951-023-02251-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a highly debilitating and fatal chronic lung disease that is difficult to cure clinically. IPF is characterized by a gradual decline in lung function, which leads to respiratory failure and severely affects patient quality of life and survival. Oxidative stress and chronic inflammation are believed to be important pathological mechanisms underlying the onset and progression of IPF, and the vicious cycle of NOX4-derived ROS, NLRP3 inflammasome activation, and p38 MAPK in pulmonary fibrogenesis explains the ineffectiveness of single-target or single-drug interventions. In this study, we combined astragaloside IV (AS-IV) and ligustrazine (LIG) based on the fundamental theory of traditional Chinese medicine (TCM) of "tonifying qi and activating blood" and loaded these drugs onto nanoparticles (AS_LIG@PPGC NPs) that were inhalable and could penetrate the mucosal barrier. Our results suggested that inhalation of AS_LIG@PPGC NPs significantly improved bleomycin-induced lung injury and fibrosis by regulating the NOX4-ROS-p38 MAPK and NOX4-NLRP3 pathways to treat and prevent IPF. This study not only demonstrated the superiority, feasibility, and safety of inhalation therapy for IPF intervention but also confirmed that breaking the vicious cycle of ROS and the NLRP3 inflammasome is a promising strategy for the successful treatment of IPF. Moreover, this successful nanoplatform is a good example of the integration of TCM and modern medicine.
Collapse
Affiliation(s)
- Meiling Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China
- Peking University People's Hospital, Beijing, 100032, China
| | - Kai Liu
- Division of Pulmonary and Critical Care Medicine, Kunming Children's Hospital, Kunming, 650000, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, National Clinical Research Center for Obstetric & Gynecologic Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100032, China.
| | - Cuiling Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China.
- Peking University People's Hospital, Beijing, 100032, China.
| | - Guanghao Wu
- School of Materials Science & Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
45
|
Wang Z, Liu M, Ai Y, Zheng S, Chen Y, Du H, Yuan S, Guo X, Yuan Y, Li G, Song J, Deng C. The compound artemisinin-hydroxychloroquine ameliorates bleomycin-induced pulmonary fibrosis in rats by inhibiting TGF-β1/Smad2/3 signaling pathway. Pulm Pharmacol Ther 2023; 83:102268. [PMID: 37967761 DOI: 10.1016/j.pupt.2023.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/27/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Pulmonary fibrosis (PF) is a lethal disease characterized by a progressive decline in lung function. Currently, lung transplantation remains the only available treatment for PF. However, both artemisinin (ART) and hydroxychloroquine (HCQ) possess potential antifibrotic properties. This study aimed to investigate the effects and mechanisms of a compound known as Artemisinin-Hydroxychloroquine (AH) in treating PF, specifically by targeting the TGF-β1/Smad2/3 pathway. To do this, we utilized an animal model of PF induced by a single tracheal drip of bleomycin (BLM) in Sprague-Dawley (SD) rats. The PF animal models were administered various doses of AH, and the efficacy and safety of AH were evaluated through pulmonary function testing, blood routine tests, serum biochemistry tests, organ index measurements, and pathological examinations. Additionally, Elisa, western blotting, and qPCR techniques were employed to explore the potential molecular mechanisms of AH in treating PF. Our findings reveal that AH effectively and safely alleviate PF by inhibiting BLM-induced specific inflammation, reducing extracellular matrix (ECM) deposition, and interfering with the TGF-β1/Smad2/3 signaling pathway. Notably, the windfall for this study is that the inhibition of ECM may initiate self-healing in the BLM-induced PF animal model. In conclusion, AH shows promise as a potential therapeutic drug for PF, as it inhibits disease progression through the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Zhaojia Wang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Min Liu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Ying Ai
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Shaoqin Zheng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China; Institute of Science and Technology, Guangzhou University of Chinese Medicine, 26 Chentai Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Yingyi Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Hujun Du
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Shijia Yuan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Xueying Guo
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Yueming Yuan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China; Institute of Science and Technology, Guangzhou University of Chinese Medicine, 26 Chentai Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Guoming Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
46
|
Cui Y, Yang Z, Lv Z, Lei J. Disruption of extracellular redox balance drives persistent lung fibrosis and impairs fibrosis resolution. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166842. [PMID: 37558008 DOI: 10.1016/j.bbadis.2023.166842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/13/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023]
Abstract
Lung fibrosis is a devastating outcome of various diffuse parenchymal lung diseases. Despite rigorous research efforts, the mechanisms that propagate its progressive and nonresolving nature remain enigmatic. Oxidative stress has been implicated in the pathogenesis of lung fibrosis. However, the role of extracellular redox state in disease progression and resolution remains largely unexplored. Here, we show that compartmentalized control over extracellular reactive oxygen species (ROS) by aerosolized delivery of recombinant extracellular superoxide dismutase (ECSOD) suppresses an established bleomycin-induced fibrotic process in mice. Further analysis of publicly available microarray, RNA-seq and single-cell RNAseq datasets reveals a significant decrease in ECSOD expression in fibrotic lung tissues that can be spontaneously restored during fibrosis resolution. Therefore, we investigate the effect of siRNA-mediated ECSOD depletion during the established fibrotic phase on the self-limiting nature of the bleomycin mouse model. Our results demonstrate that in vivo knockdown of ECSOD in mouse fibrotic lungs impairs fibrosis resolution. Mechanistically, we demonstrate that transforming growth factor (TGF)-β1 downregulates endogenous ECSOD expression, leading to the accumulation of extracellular superoxide via Smad-mediated signaling and the activation of additional stores of latent TGF-β1. In addition, depletion of endogenous ECSOD during the fibrotic phase in the bleomycin model induces an apoptosis-resistant phenotype in lung fibroblasts through unrestricted Akt signaling. Taken together, our data strongly support the critical role of extracellular redox state in fibrosis persistence and resolution. Based on these findings, we propose that compartment-specific control over extracellular ROS may be a potential therapeutic strategy for managing fibrotic lung disorders.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China.
| | - Zeran Yang
- Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China
| | - Jianfeng Lei
- Medical Imaging Laboratory, Research Core Facilities, Capital Medical University, Beijing 100069, People's Republic of China
| |
Collapse
|
47
|
Ye X, Zhang M, Gu H, Liu M, Zhao Y, Shi Y, Wu S, Jiang C, Ye X, Zhu H, Li Q, Huang X, Cao M. Animal models of acute exacerbation of pulmonary fibrosis. Respir Res 2023; 24:296. [PMID: 38007420 PMCID: PMC10675932 DOI: 10.1186/s12931-023-02595-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive scarring interstitial lung disease with an unknown cause. Some patients may experience acute exacerbations (AE), which result in severe lung damage visible on imaging or through examination of tissue samples, often leading to high mortality rates. However, the etiology and pathogenesis of AE-IPF remain unclear. AE-IPF patients exhibit diffuse lung damage, apoptosis of type II alveolar epithelial cells, and an excessive inflammatory response. Establishing a reliable animal model of AE is critical for investigating the pathogenesis. Recent studies have reported a variety of animal models for AE-IPF, each with its own advantages and disadvantages. These models are usually established in mice with bleomycin-induced pulmonary fibrosis, using viruses, bacteria, small peptides, or specific drugs. In this review, we present an overview of different AE models, hoping to provide a useful resource for exploring the mechanisms and targeted therapies for AE-IPF.
Collapse
Affiliation(s)
- Xu Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Mingrui Zhang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Gu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Mengying Liu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yichao Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanchen Shi
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shufei Wu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Jiang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huihui Zhu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Li
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinmei Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| | - Mengshu Cao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| |
Collapse
|
48
|
Chang A, Van Ry PM, Raghu G. Idiopathic pulmonary fibrosis: aligning murine models to clinical trials in humans. THE LANCET. RESPIRATORY MEDICINE 2023; 11:953-955. [PMID: 37914467 DOI: 10.1016/s2213-2600(23)00325-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Pam M Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Ganesh Raghu
- University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
49
|
Young ON, Bourke JE, Widdop RE. Catch your breath: The protective role of the angiotensin AT 2 receptor for the treatment of idiopathic pulmonary fibrosis. Biochem Pharmacol 2023; 217:115839. [PMID: 37778444 DOI: 10.1016/j.bcp.2023.115839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease whereby excessive deposition of extracellular matrix proteins (ECM) ultimately leads to respiratory failure. While there have been advances in pharmacotherapies for pulmonary fibrosis, IPF remains an incurable and irreversible disease. There remains an unmet clinical need for treatments that reverse fibrosis, or at the very least have a more tolerable side effect profile than currently available treatments. Transforming growth factor β1(TGFβ1) is considered the main driver of fibrosis in IPF. However, as our understanding of the role of the pulmonary renin-angiotensin system (PRAS) in the pathogenesis of IPF increases, it is becoming clear that targeting angiotensin receptors represents a potential novel treatment strategy for IPF - in particular, via activation of the anti-fibrotic angiotensin type 2 receptor (AT2R). This review describes the current understanding of the pathophysiology of IPF and the mediators implicated in its pathogenesis; focusing on TGFβ1, angiotensin II and related peptides in the PRAS and their contribution to fibrotic processes in the lung. Preclinical and clinical assessment of currently available AT2R agonists and the development of novel, highly selective ligands for this receptor will also be described, with a focus on compound 21, currently in clinical trials for IPF. Collectively, this review provides evidence of the potential of AT2R as a novel therapeutic target for IPF.
Collapse
Affiliation(s)
- Olivia N Young
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jane E Bourke
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
50
|
Wang X, Li S, Liu J, Sun W, Zhao H, Han Q, Liu Y, Cao X, Li Q, Jin Y, Guo X, Ren G. Evaluation of prevention and treatment effects of fibroblast growth factor-21 in BLM-induced pulmonary fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3299-3313. [PMID: 37256335 PMCID: PMC10230495 DOI: 10.1007/s00210-023-02540-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/17/2023] [Indexed: 06/01/2023]
Abstract
Pulmonary fibrosis is a progressive and fatal fibrotic lung disease and associated with a high mortality rate. In the study, the prevention and treatment effects of fibroblast growth factor-21 (FGF-21) in bleomycin (BLM)-induced pulmonary fibrosis were investigated in vivo and vitro. In the prevention of pulmonary fibrosis studies, the results showed that interdict of FGF-21 could reduce the related gene and protein expression levels of pulmonary fibrosis. In addition, FGF-21 significantly reduced both the aggregation of inflammatory cells and deposition of collagen in the lung by histopathology. In therapy of pulmonary fibrosis studies, the results indicated that treatment with FGF-21 resulted in an amelioration of the pulmonary fibrosis in mice with reductions of the pathological score, collagen deposition and transforming growth factor (TGF)-β and α-smooth muscle actin (α-SMA) expressions in the lung tissues at fibrotic stage, and late administration was also able to reduce the degree of pulmonary fibrosis and even better than these in the prevention group. Furthermore, BLM-induced THP-1 macrophage model was verified using FGF-21; the result showed that FGF-21 decreased the related gene expression level of pulmonary fibrosis. FGF-21 may have preventive and therapeutic effects on BLM-induced pulmonary fibrosis via inhibiting myofibroblast differentiation and inflammatory. Thus, FGF-21 represents a potential drug for the prevention and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiangxiang Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, 150010, China
| | - Shuang Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Jinmiao Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Wenying Sun
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Han Zhao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Qing Han
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yijia Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaolin Cao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Qianhui Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yuhan Jin
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.
- Research Center of Genetic Engineering of Pharmaceuticals of Heilongjiang Province, Northeast Agricultural University, Harbin, 150030, China.
- Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|