1
|
Liu A, Li S, Dong X, Qin X, Li Z. Farfarae Flos Mitigates Cigarette Smoking-Induced Lung Inflammation by Regulating the Lysophosphatidylcholine Biosynthesis and Tryptophan Metabolism. Biomed Chromatogr 2025; 39:e6072. [PMID: 39775926 DOI: 10.1002/bmc.6072] [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: 08/25/2024] [Revised: 12/01/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
An increased risk of developing respiratory diseases has been linked to exposure to cigarette smoking (CS). The flower buds of Tussilago farfara L., also known as Farfarae Flos (FF), can be used for the treatment of cough, bronchitis, and asthmatic disorders in China. In the present study, we used lung and fecal metabolomics, as well as the intestinal flora analysis, aimed to investigate the protective effect of FF against the CS exposure induced lung inflammation on mice. The results showed that FF administration could relieve the lung inflammation as demonstrated by lung index, interleukin-6 (IL-6), and interleukin-1β (IL-1β) levels, as well as the pulmonary pathological change. The lung metabolomics coupled with molecular docking showed that FF could alleviate lung inflammation by regulating lysophosphatidylcholine biosynthesis through the caffeoyl quinic acids distributed in the lung tissue. In addition, fecal metabolome coupled with 16S rRNA gene sequencing showed that FF could regulate the tryptophan metabolism by regulating the intestinal flora disorders. This study provided new insights of FF to relieve CS-induced pulmonary inflammation with the multimechanism.
Collapse
Affiliation(s)
- Aoqi Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Siyao Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Xianlong Dong
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Zhenyu Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| |
Collapse
|
2
|
Li J, Wu W, Kong X, Yang X, Li K, Jiang Z, Zhang C, Zou J, Liang Y. Roles of gut microbiome-associated metabolites in pulmonary fibrosis by integrated analysis. NPJ Biofilms Microbiomes 2024; 10:154. [PMID: 39702426 DOI: 10.1038/s41522-024-00631-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Lung diseases often coincide with imbalances in gut microbiota, but the role of gut microbiota in pulmonary fibrosis (PF) remains unclear. This study investigates the impact of gut microbiota and their metabolites on PF. Serum and lung tissues of normal, bleomycin (BLM)- and silica-induced mice showed significant differences in gut microbiota. L-Tryptophan was upregulated within pulmonary tissue and serum metabolites both in the BLM and Silica groups. The dominant gut microbiota associated with L-tryptophan metabolism included Lachnospiraceae_NK4A136_Group, Allobaculum, Alistipes, and Candidatus_Saccharimonas. L-Tryptophan promoted BLM- and silica-induced pathological damage in PF mice. L-Tryptophan promoted TGF-β1-induced EMT and fibroblast activation in vitro via activating the mTOR/S6 pathway. In conclusion, PF mice exhibited alterations in gut microbiota and serum and lung tissue metabolites. L-Tryptophan level was associated with changes in gut microbiota, and L-tryptophan promoted PF progression in both in vivo and in vitro models, potentially through activation of the mTOR/S6 pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Internal Medicine, Jiangxi Chest Hospital, The Third Affiliated Hospital of Nanchang Medical College, Key Laboratory of Health of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Wenqing Wu
- Medical Affairs, Johnson & Johnson Innovative Medicine, Beijing, 100025, China
| | - Xinyi Kong
- Department of Cardiovascular Intervention, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
| | - Xia Yang
- Department of Internal Medicine, Jiangxi Chest Hospital, The Third Affiliated Hospital of Nanchang Medical College, Key Laboratory of Health of Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Kui Li
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, 725000, Shaanxi Province, China
| | - Zicheng Jiang
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, 725000, Shaanxi Province, China
| | - Chunlan Zhang
- Department of Infectious Diseases, Wuming Hospital of Guangxi Medical University, Nanning, 530199, Guangxi, China
| | - Jun Zou
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, 530002, Guangxi, China.
| | - Ying Liang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, P.R. China.
| |
Collapse
|
3
|
Chen YZ, Zhao L, Wei W, Gu J, Liu ZH, Shan WY, Dong J, Li C, Qin LQ, Xu JY. The Effect of Metformin on Radiation-Induced Lung Fibrosis in Mice. Dose Response 2024; 22:15593258241308051. [PMID: 39664837 PMCID: PMC11632958 DOI: 10.1177/15593258241308051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction: Radiation-induced lung fibrosis (RILF) is a common complication of thoracic radiotherapy. Metformin has been suggested to have a radioprotective effect. Objective: This study explored the radioprotective effects of metformin on RILF and its mechanisms. Methods: C57BL/6J mice were randomly divided into control, ionizing radiation (IR), low-dose metformin (L-Met), and high-dose metformin (H-Met) groups. The IR, L-Met, and H-Met groups received 15 Gy chest irradiation. The L-Met and H-Met groups were administrated 100 or 200 mg/kg metformin from 3 days before irradiation and continued for 6 months. The mice were then sacrificed, and samples were collected for further analysis. Results: RILF was induced in the irradiated mice. Metformin improved lung pathology, inhibited collagen deposition, and reduced inflammatory factors such as high mobility group box 1 (HMGB1), interleukin-1 beta, interleukin-6, tumor necrosis factor alpha in lung tissue, lavage fluid, and serum. Western blot and quantitative real-time PCR analyses revealed that metformin downregulated HMGB1, toll-like receptor 4 (TLR4), and nuclear factor kappaB (NF-κB) expression. Additionally, metformin reversed the irradiation-induced reduction in the abundance of Lactobacillus and Lachnospiraceae at the genus level. Conclusion: Our findings indicated that metformin ameliorates RILF by downregulating the inflammatory-related HMGB1/TLR4/NF-κB pathway and improving intestinal flora disorder.
Collapse
Affiliation(s)
- Yu-Zhong Chen
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Yancheng Municipal Center for Disease Control and Prevention, Yancheng, China
| | - Lin Zhao
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Wei
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jia Gu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhen-Hua Liu
- Department of Radiotherapy, The Yancheng Clinical College of Xuzhou Medical University, The First people’s Hospital of Yancheng, Yancheng, China
| | - Wen-Yue Shan
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jie Dong
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Li
- Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Farh MEA, Kim HJ, Kim SY, Lee JH, Lee H, Cui R, Han S, Kim DW, Park S, Lee YJ, Lee YS, Sohn I, Cho J. Transcriptional Changes in Radiation-Induced Lung Injury: A Comparative Analysis of Two Radiation Doses for Preclinical Research. Int J Mol Sci 2024; 25:3766. [PMID: 38612576 PMCID: PMC11011446 DOI: 10.3390/ijms25073766] [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: 01/31/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
In a recent stereotactic body radiation therapy animal model, radiation pneumonitis and radiation pulmonary fibrosis were observed at around 2 and 6 weeks, respectively. However, the molecular signature of this model remains unclear. This study aimed to examine the molecular characteristics at these two stages using RNA-seq analysis. Transcriptomic profiling revealed distinct transcriptional patterns for each stage. Inflammatory response and immune cell activation were involved in both stages. Cell cycle processes and response to type II interferons were observed during the inflammation stage. Extracellular matrix organization and immunoglobulin production were noted during the fibrosis stage. To investigate the impact of a 10 Gy difference on fibrosis progression, doses of 45, 55, and 65 Gy were tested. A dose of 65 Gy was selected and compared with 75 Gy. The 65 Gy dose induced inflammation and fibrosis as well as the 75 Gy dose, but with reduced lung damage, fewer inflammatory cells, and decreased collagen deposition, particularly during the inflammation stage. Transcriptomic analysis revealed significant overlap, but differences were observed and clarified in Gene Ontology and KEGG pathway analysis, potentially influenced by changes in interferon-gamma-mediated lipid metabolism. This suggests the suitability of 65 Gy for future preclinical basic and pharmaceutical research connected with radiation-induced lung injury.
Collapse
Affiliation(s)
- Mohamed El-Agamy Farh
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
- Drug Development Team, ARONTIER, Co., Ltd., Seoul 06735, Republic of Korea;
| | - Hyun-Jin Kim
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Sang-Yeon Kim
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Jae-Hee Lee
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Hajeong Lee
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Ronglan Cui
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Soorim Han
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Dong Wook Kim
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Sunjoo Park
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| | - Yoon-Jin Lee
- Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea;
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Science, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Insuk Sohn
- Drug Development Team, ARONTIER, Co., Ltd., Seoul 06735, Republic of Korea;
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.E.-A.F.); (H.-J.K.); (S.-Y.K.); (J.-H.L.); (H.L.); (R.C.); (S.H.); (D.W.K.); (S.P.)
| |
Collapse
|
5
|
Bian B, Ge C, Wu F, Fan Y, Kong J, Li K, Bian H, Miao Q. Wogonin Attenuates Bleomycin-Induced Pulmonary Fibrosis and Oxidative Stress Injury via the MAPK Signaling Pathway. Biol Pharm Bull 2024; 47:2165-2172. [PMID: 39756931 DOI: 10.1248/bpb.b24-00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Idiopathic pulmonary fibrosis (PF) is an irreversible and chronic inflammatory condition with limited therapeutic options and a high mortality rate. We aimed to determine the possible role and mechanisms of wogonin (WGN) on PF. A rat model of PF was established with intratracheally administrated with bleomycin (BLM), followed by intravenously injecting with WGN and weekly body weight measurements for four weeks. Hematoxylin-eosin (H&E) and Masson's trichrome staining were implemented for histopathological analysis. In addition, the levels of fibrotic proteins and indicators of the mitogen-activated protein kinase (MAPK) pathway were assessed with Western blot. RT-quantitative (q)PCR experiment was conducted to investigate the fibrotic proteins' mRNA expression. Ultimately, the concentrations of glutathione peroxidase (GSH-PX), malonaldehyde (MDA), and superoxide dismutase (SOD) were ascertained with appropriate kits. The results showed that WGN administration significantly reversed BLM-induced body weight reduction, alleviated pathological fibrosis, and reduced the Ashcroft score and the lung wet-to-dry weight ratio. Additionally, WGN suppressed the rise of fibrotic protein levels in BLM-treated rat's lung tissues. Furthermore, WGN attenuated BLM-stimulated oxidative stress, as evidenced by the increased GSH-PX and SOD levels and decreased MDA levels in vivo. Finally, wogonin supplements significantly lowered the extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK phosphorylation levels in the BLM-treated rat's lung tissues. In conclusion, our study proved that PF induced by BLM administration can be mitigated by WGN treatment via suppressing the MAPK pathway, indicating that WGN is a candidate therapeutic agent for managing PF.
Collapse
Affiliation(s)
- Bo Bian
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Chang Ge
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Fanwu Wu
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Yiling Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| | | | - Kai Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Hua Bian
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Qing Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| |
Collapse
|
6
|
Han X, Wu W, Wang S. Krüppel-like factor 15 counteracts endoplasmic reticulum stress and suppresses lung fibroblast proliferation and extracellular matrix accumulation. Tissue Cell 2023; 84:102183. [PMID: 37531874 DOI: 10.1016/j.tice.2023.102183] [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: 01/10/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
The incidence of pulmonary fibrosis is on the rise, and existing treatments have limited efficacy in improving patient survival. The purpose of this study was to reveal the potential of Krüppel-like factor (KLF)15 activation in alleviating pulmonary fibrosis. Transforming growth factor beta (TGF-β) was utilized to induce lung fibroblasts to establish an in vitro model of pulmonary fibrosis. The impacts of TGF-β and KLF15 level on cell proliferation, migration, extracellular matrix (ECM) accumulation, and endoplasmic reticulum stress (ERS) were assessed. Additionally, tunicamycin, an ERS agonist, was used to investigate the role of ERS in KLF15 regulation. The results showed that KLF15 was dropped in response to TGF-β treatment. However, KLF15 overexpression reduced cell proliferation, migration, ECM accumulation, and ERS, alleviating the effects of TGF-β stimulation. Subsequent treatment with tunicamycin diminished the effects of KLF15 overexpression, demonstrating that ERS mediated the modulation of KLF15. KLF15 acts against ERS and suppresses excessive proliferation and ECM accumulation in lung fibroblast. These findings suggest that activating KLF15 is a promising strategy for alleviating pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiang Han
- Department of Emergency, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, China.
| | - Weiqin Wu
- Department of Emergency, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Shuming Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, China.
| |
Collapse
|
7
|
The Role of Platelets in the Pathogenesis and Pathophysiology of Adenomyosis. J Clin Med 2023; 12:jcm12030842. [PMID: 36769489 PMCID: PMC9918158 DOI: 10.3390/jcm12030842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Widely viewed as an enigmatic disease, adenomyosis is a common gynecological disease with bewildering pathogenesis and pathophysiology. One defining hallmark of adenomyotic lesions is cyclic bleeding as in eutopic endometrium, yet bleeding is a quintessential trademark of tissue injury, which is invariably followed by tissue repair. Consequently, adenomyotic lesions resemble wounds. Following each bleeding episode, adenomyotic lesions undergo tissue repair, and, as such, platelets are the first responder that heralds the subsequent tissue repair. This repeated tissue injury and repair (ReTIAR) would elicit several key molecular events crucial for lesional progression, eventually leading to lesional fibrosis. Platelets interact with adenomyotic cells and actively participate in these events, promoting the lesional progression and fibrogenesis. Lesional fibrosis may also be propagated into their neighboring endometrial-myometrial interface and then to eutopic endometrium, impairing endometrial repair and causing heavy menstrual bleeding. Moreover, lesional progression may result in hyperinnervation and an enlarged uterus. In this review, the role of platelets in the pathogenesis, progression, and pathophysiology is reviewed, along with the therapeutic implication. In addition, I shall demonstrate how the notion of ReTIAR provides a much needed framework to tether to and piece together many seemingly unrelated findings and how it helps to make useful predictions.
Collapse
|
8
|
Immunophenotypic and molecular characterization of pancreatic neuroendocrine tumors producing serotonin. Mod Pathol 2022; 35:1713-1722. [PMID: 35739266 DOI: 10.1038/s41379-022-01110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/08/2022]
Abstract
Serotonin producing pancreatic neuroendocrine tumors (SP-PanNET) account for 0.58-1.4% of all pancreatic neuroendocrine tumors (PanNET). They may present with atypical symptoms, such as acute pancreatitis and are often radiologically characterized by main pancreatic duct dilatation. SP-PanNET are well differentiated neuroendocrine tumors (NET) distinct from classical PanNET by atypical serotonin secretion and abundant dense stroma deposition, like serotonin producing ileal NET leading in some cases to difficulties to reliably distinguish SP-PanNET from ileal NET metastases. The biology and molecular profile of SP-PanNET remain poorly characterized and the cell of origin within the pancreas is unclear. To address these questions, we analyzed a large cohort of SP-PanNET by immunohistochemistry (n = 29; ATRX, DAXX, MENIN, Islet1, PAX6, PDX1, ARX, CDX2), whole genome copy number array (Oncoscan™) and a large NGS panel (NovoPM™) (n = 10), FISH (n = 13) and RNA sequencing (n = 24) together with 21 ileal NET and 29 nonfunctioning PanNET (NF-PanNET). These analyses revealed a unique genomic profile with frequent isolated loss of chromosome 1 (14 cases-61%) and few pathogenic mutations (KMT2C in 2 cases, ARID1A in 1 case). Unsupervised RNAseq-based clustering showed that SP-PanNET were closer to NF-PanNET than ileal NET with an exclusive beta cell-like signature. SP-PanNET showed TGF-β pathway activation signatures associated with extracellular matrix remodeling and similar signature were reproduced in vitro when pancreatic stellate cells were exposed to serotonin. SP-PanNET immunohistochemical profile resemble that of ileal NET except for PDX1 and PAX6 expression to a lesser extend suggesting that these two markers may be useful to diagnose SP-PanNET. Taken together, this suggests that SP-PanNET are a very specific PanNET entity with a peculiar biology leading to the characteristic fibrotic aspect.
Collapse
|
9
|
Interdonato L, D’amico R, Cordaro M, Siracusa R, Fusco R, Peritore AF, Gugliandolo E, Crupi R, Coaccioli S, Genovese T, Impellizzeri D, Di Paola R, Cuzzocrea S. Aerosol-Administered Adelmidrol Attenuates Lung Inflammation in a Murine Model of Acute Lung Injury. Biomolecules 2022; 12:biom12091308. [PMID: 36139146 PMCID: PMC9496587 DOI: 10.3390/biom12091308] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Acute lung injury (ALI) is a common and devastating clinical disorder with a high mortality rate and no specific therapy. The pathophysiology of ALI is characterized by increased alveolar/capillary permeability, lung inflammation, oxidative stress and structural damage to lung tissues, which can progress to acute respiratory distress syndrome (ARDS). Adelmidrol (ADM), an analogue of palmitoylethanolamide (PEA), is known for its anti-inflammatory and antioxidant functions, which are mainly due to down-modulating mast cells (MCs) and promoting endogenous antioxidant defense. The aim of this study is to evaluate the protective effects of ADM in a mice model of ALI, induced by intratracheal administration of lipopolysaccharide (LPS) at the dose of 5 mg/kg. ADM 2% was administered by aerosol 1 and 6 h after LPS instillation. In this study, we clearly demonstrated that ADM reduced lung damage and airway infiltration induced by LPS instillation. At the same time, ADM counteracted the increase in MC number and the expression of specific markers of MC activation, i.e., chymase and tryptase. Moreover, ADM reduced oxidative stress by upregulating antioxidant enzymes as well as modulating the Nf-kB pathway and the resulting pro-inflammatory cytokine release. These results suggest that ADM could be a potential candidate in the management of ALI.
Collapse
Affiliation(s)
- Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Ramona D’amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy
| | - Stefano Coaccioli
- General Medical Clinic and Medical Therapy, Rheumatology and Medical Therapy of the Pain, University of Perugia, “Polo di Terni”, “AO Santa Maria” of Terni, 06129 Perugia, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
- Correspondence: ; Tel.: +39-090-676-5208
| | - Rosanna Di Paola
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| |
Collapse
|
10
|
Shi M, Cui H, Shi J, Mei Y. mmu-microRNA-92a-3p attenuates pulmonary fibrosis by modulating Cpeb4-mediated Smad2/3 signaling pathway. Funct Integr Genomics 2022; 22:1297-1306. [PMID: 35909199 DOI: 10.1007/s10142-022-00879-z] [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: 09/26/2021] [Revised: 04/02/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022]
Abstract
Pulmonary fibrosis (PF) is a chronic lung disorder, in which the mechanism of mmu-microRNA (miR)-92a-3p is not elucidated clearly. The present work was proposed to disclose mmu-miR-92a-3p-focused mechanism in PF with cytoplasmic polyadenylation element-binding protein 4 (Cpeb4)/Smad2/3 axis. PF was induced in mice by the intratracheal injection of bleomycin (BLM). Then, the BLM-treated mice were injected with mmu-miR-92a-3p- and/or Cpeb4-related adenovirus vectors. mmu-miR-92a-3p, Cpeb4, and Smad2/3 expression in lung tissues were examined. Alveolar cell apoptosis and collagen deposition in lung tissues and inflammatory factors in serum were observed. The interaction between mmu-miR-92a-3p and Cpeb4 was explored. Lowly expressed mmu-miR-92a-3p and highly expressed Cpeb4 and Smad2/3 were manifested in BLM-induced PF mice. BLM-induced PF mice exhibited enhanced inflammation, alveolar cell apoptosis, and collagen deposition, which would be attenuated by upregulating mmu-miR-92a-3p or downregulating Cpeb4. mmu-miR-92a-3p targeted Cpeb4. Upregulating mmu-miR-92a-3p or downregulating Cpeb4 inactivated the Smad2/3 signaling pathway in BLM-induced PF mice. It is elaborated that mmu-miR-92a-3p attenuates the process of PF by modulating Cpeb4-mediated Smad2/3 signaling pathway, renewing the molecular mechanism of PF.
Collapse
Affiliation(s)
- Mengkun Shi
- Department of Cardiothoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Putuo District, Shanghai, 200092, China
| | - Huixia Cui
- Department of Medical Institution Conducting Clinical Trials for Human Used Drug, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046099, Shanxi, China
| | - Jialun Shi
- Department of Cardiothoracic Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046099, Shanxi, China
| | - Yunqing Mei
- Department of Cardiothoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Putuo District, Shanghai, 200092, China.
| |
Collapse
|
11
|
Animal models of drug-induced pulmonary fibrosis: an overview of molecular mechanisms and characteristics. Cell Biol Toxicol 2021; 38:699-723. [PMID: 34741237 DOI: 10.1007/s10565-021-09676-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease characterized by progressive loss of pulmonary function. Drug-induced interstitial lung disease has been reported as a severe adverse effect of some drugs, such as bleomycin, amiodarone, and methotrexate. Based on good characteristics, drug-induced pulmonary fibrosis (PF) animal model has played a key role in our understanding of the molecular mechanisms of PF pathogenesis and recapitulates the specific pathology in patients and helps develop therapeutic strategies. Here, we summarize the mechanisms and characteristics of given fibrotic drug-induced animal models for PFs. Together with the key publications describing these models, this brief but detailed overview would be helpful for the pharmacological research with animal models of PFs. Potential mechanisms underlying drug induced lung toxicity.
Collapse
|
12
|
Xiong Y, Cui X, Zhou Y, Chai G, Jiang X, Ge G, Wang Y, Sun H, Che H, Nie Y, Zhao P. Dehydrocostus lactone inhibits BLM-induced pulmonary fibrosis and inflammation in mice via the JNK and p38 MAPK-mediated NF-κB signaling pathways. Int Immunopharmacol 2021; 98:107780. [PMID: 34118645 DOI: 10.1016/j.intimp.2021.107780] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and irreversible inflammatory disease with a high mortality rate and limited therapeutic options. This study explored the potential role and mechanisms of Dehydrocostus lactone (DHL) in the inflammatory and fibrotic responses in a bleomycin (BLM) induced model. Treatment with DHL significantly reduced pathological injury and fibrosis, the secretion of BLM-induced pro-fibrotic mediators TGF-β and α-SMA, and components of the extracellular matrix (fibronectin). Additionally, in the early stages of inflammation, DHL administration inhibited the infiltration of inflammatory cells and downregulated the expression of TGF-β, TNF-α, and IL-6, indicating that DHL treatment effectively alleviated BLM-induced pulmonary fibrosis and inflammation in a dose-dependent manner. Furthermore, BLM induced the production of IL-33 in vivo, which initiated and progressed pulmonary fibrosis by activating macrophages and enhancing the production of IL-13 and TGF-β. In contrast, a significant decrease in the expression of IL-33 after DHL treatment in vitro showed that DHL strongly reduced IL-13 and TGF-β. Regarding the mechanism, BLM-induced phosphorylation of JNK, p38 MAPK, and NF-κB were significantly reduced after DHL treatment, which further led to the down-regulation of IL-33 expression, thereby decreasing IL-13 and TGF-β. Collectively, our data suggested that DHL could exert its anti-fibrosis effect via inhibiting the early inflammatory response by downregulating the JNK/p38 MAPK-mediated NF-κB signaling pathway to suppress macrophage activation. Therefore, DHL has therapeutic potential for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yue Xiong
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xiaochuan Cui
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Yanjun Zhou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Gaoshang Chai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xiufeng Jiang
- Department of Respiratory and Critical Care Medicine, Wuxi Fifth People's Hospital, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Guizhi Ge
- The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Jiangsu, PR China
| | - Yue Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Hongxu Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Huilian Che
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
13
|
Masood A, Jacob M, Gu X, Abdel Jabar M, Benabdelkamel H, Nizami I, Li L, Dasouki M, Abdel Rahman AM. Distinctive metabolic profiles between Cystic Fibrosis mutational subclasses and lung function. Metabolomics 2021; 17:4. [PMID: 33394183 DOI: 10.1007/s11306-020-01760-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a lethal multisystemic disease of a monogenic origin with numerous mutations. Functional defects in the cystic fibrosis transmembrane conductance receptor (CFTR) protein based on these mutations are categorised into distinct classes having different clinical presentations and disease severity. OBJECTIVES The present study aimed to create a comprehensive metabolomic profile of altered metabolites in patients with CF, among different classes and in relation to lung function. METHODS A chemical isotope labeling liquid chromatography-mass spectrometry metabolomics was used to study the serum metabolic profiles of young and adult CF (n = 39) patients and healthy controls (n = 30). Comparisons were made at three levels, CF vs. controls, among mutational classes of CF, between CF class III and IV, and correlated the lung function findings. RESULTS A distinctive metabolic profile was observed in the three analyses. 78, 20, and 13 significantly differentially dysregulated metabolites were identified in the patients with CF, among the different classes and between class III and IV, respectively. The significantly identified metabolites included amino acids, di-, and tri-peptides, glutathione, glutamine, glutamate, and arginine metabolism. The top significant metabolites include 1-Aminopropan-2-ol, ophthalmate, serotonin, cystathionine, and gamma-glutamylglutamic acid. Lung function represented by an above-average FEV1% level was associated with decreased glutamic acid and increased guanosine levels. CONCLUSION Metabolomic profiling identified alterations in different amino acids and dipeptides, involved in regulating glutathione metabolism. Two metabolites, 3,4-dihydroxymandelate-3-O-sulfate and 5-Aminopentanoic acid, were identified in common between the three anlayses and may represent as highly sensitive biomarkers for CF.
Collapse
Affiliation(s)
- Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, PO. Box 2925 (98), Riyadh, 11461, Saudi Arabia
| | - Minnie Jacob
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Xinyun Gu
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Mai Abdel Jabar
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, PO. Box 2925 (98), Riyadh, 11461, Saudi Arabia
| | - Imran Nizami
- Lung Transplant Section, Organ Transplant Center, King Faisal Specialist Hospital and Research Center, Zahrawi Street, Al Maather, Riyadh, 11211, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Majed Dasouki
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Anas M Abdel Rahman
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia.
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh, Saudi Arabia.
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X7, Canada.
| |
Collapse
|
14
|
Löfdahl A, Tornling G, Wigén J, Larsson-Callerfelt AK, Wenglén C, Westergren-Thorsson G. Pathological Insight into 5-HT 2B Receptor Activation in Fibrosing Interstitial Lung Diseases. Int J Mol Sci 2020; 22:ijms22010225. [PMID: 33379351 PMCID: PMC7796180 DOI: 10.3390/ijms22010225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 11/29/2022] Open
Abstract
Interstitial lung disease (ILD) encompasses a heterogeneous group of more than 200 conditions, of which primarily idiopathic pulmonary fibrosis (IPF), idiopathic nonspecific interstitial pneumonia, hypersensitivity pneumonitis, ILD associated with autoimmune diseases and sarcoidosis may present a progressive fibrosing (PF) phenotype. Despite different aetiology and histopathological patterns, the PF-ILDs have similarities regarding disease mechanisms with self-sustaining fibrosis, which suggests that the diseases may share common pathogenetic pathways. Previous studies show an enhanced activation of serotonergic signaling in pulmonary fibrosis, and the serotonin (5-HT)2 receptors have been implicated to have important roles in observed profibrotic actions. Our research findings in support by others, demonstrate antifibrotic effects with 5-HT2B receptor antagonists, alleviating several key events common for the fibrotic diseases such as myofibroblast differentiation and connective tissue deposition. In this review, we will address the potential role of 5-HT and in particular the 5-HT2B receptors in three PF-ILDs: ILD associated with systemic sclerosis (SSc-ILD), ILD associated with rheumatoid arthritis (RA-ILD) and IPF. Highlighting the converging pathways in these diseases discloses the 5-HT2B receptor as a potential disease target for PF-ILDs, which today have an urgent unmet need for therapeutic strategies.
Collapse
Affiliation(s)
- Anna Löfdahl
- Lung Biology, Department of Experimental Medical Science, Lund University, BMC C12, 22184 Lund, Sweden; (J.W.); (A.-K.L.-C.); (G.W.-T.)
- Correspondence:
| | - Göran Tornling
- AnaMar AB, Medicon Village, Scheeletorget 1, 22381 Lund, Sweden; (C.W.); (G.T.)
- Respiratory Medicine Division, Department of Medicine Solna, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jenny Wigén
- Lung Biology, Department of Experimental Medical Science, Lund University, BMC C12, 22184 Lund, Sweden; (J.W.); (A.-K.L.-C.); (G.W.-T.)
| | - Anna-Karin Larsson-Callerfelt
- Lung Biology, Department of Experimental Medical Science, Lund University, BMC C12, 22184 Lund, Sweden; (J.W.); (A.-K.L.-C.); (G.W.-T.)
| | - Christina Wenglén
- AnaMar AB, Medicon Village, Scheeletorget 1, 22381 Lund, Sweden; (C.W.); (G.T.)
| | - Gunilla Westergren-Thorsson
- Lung Biology, Department of Experimental Medical Science, Lund University, BMC C12, 22184 Lund, Sweden; (J.W.); (A.-K.L.-C.); (G.W.-T.)
| |
Collapse
|
15
|
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Inhibitor as a Novel Therapeutic Tool for Lung Injury. Int J Mol Sci 2020; 21:ijms21207761. [PMID: 33092214 PMCID: PMC7589767 DOI: 10.3390/ijms21207761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary fibrosis is a progressive disease characterized by lung remodeling due to excessive deposition of extracellular matrix. In this study, the bleomycin experimental model of pulmonary fibrosis was employed to investigate the anti-fibrotic and immunomodulatory activity of the inhibition of MALT1 protease activity. Mice received a single intra-tracheal administration of bleomycin (1 mg/kg) in the presence or absence of MI-2, a selective MALT1 inhibitor, (a dose of 30 mg/kg administered intra-peritoneally 1 h after bleomycin and daily until the end of the experiment). Seven days after bleomycin instillation mice were sacrificed and bronchoalveolar lavage fluid analysis, measurement of collagen content in the lung, histology, molecular analysis and immunohistochemistry were performed. To evaluate mortality and body weight gain a subset of mice was administered daily with MI-2 for 21 days. Mice that received MI-2 showed decreased weight loss and mortality, inflammatory cells infiltration, cytokines overexpression and tissue injury. Moreover, biochemical and immunohistochemical analysis displayed that MI-2 was able to modulate the excessive production of reactive oxygen species and the inflammatory mediator upregulation induced by bleomycin instillation. Additionally, MI-2 demonstrated anti-fibrotic activity by reducing transforming growth factor-β (TGF-β), α-smooth muscle actin (α-SMA) and receptor associated factor 6 (TRAF6) expression. The underlying mechanisms for the protective effect of MI-2 bleomycin induced pulmonary fibrosis may be attributed to its inhibition on NF-κB pathway. This is the first report showing the therapeutic role of MALT1 inhibition in a bleomycin model of pulmonary fibrosis, thus supporting further preclinical and clinical studies.
Collapse
|
16
|
Sunnaghatta Nagaraja S, Raviraj R, Selvakumar I, Dharmalingam D, Ramadas N, Chellappan DR, Ponnachipudhur Chinnaswamy P, Nagarajan D. Radiation-induced H3K9 tri-methylation in E-cadherin promoter during lung EMT: in vitro and in vivo approaches using vanillin. Free Radic Res 2020; 54:540-555. [PMID: 32842802 DOI: 10.1080/10715762.2020.1814274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Radiotherapy is an important treatment regime for lung cancer, worldwide. However, radiation-induced pneumonitis and fibrosis are the treatment-limiting toxicities among patients who have undergone radiotherapy. The epithelial cells via epithelial to mesenchymal transition [EMT] acquires mesenchymal phenotype, which ultimately leads to fibrosis. Many investigations are focussed on understanding the signalling pathways mediating in EMT, however, the role of histone methylation is less understood in radiation-induced lung EMT. In the present study, we analysed the effect of vanillin, an antioxidant, on histone methylation during radiation-induced EMT. The thoracic region of Wistar rats was irradiated with a fractionated dose of X-ray (3 Gy/day) for two weeks (total of 30 Gy). The irradiated animals were sacrificed at the 8th and 16th weeks and tissues were used for analyses. Our data showed that radiation decreased the level of antioxidant enzymes such as SOD, catalase and reduced glutathione that would ultimately enhance oxidative stress in the tissues. Histopathological analysis revealed that radiation increased the infiltration of inflammatory cells to the tissue injury site. Total global histone methylation was increased upon irradiation, which was effectively prevented by vanillin administration. Vanillin enhanced E-cadherin expression and decreased the mesenchymal markers N-cadherin and vimentin in the irradiated lung tissue. The ChIP-qPCR analysis suggested that snail expression in the nucleus might involve in the enrichment of suppressive marker H3K9me3 on the E-cadherin promoter. Finally, we suggested that vanillin administration decreased radiation-induced oxidative stress and EMT expression. Additionally, irradiation increased the H3K9 methylation status with nuclear translocation of snail during lung EMT.
Collapse
Affiliation(s)
| | - Raghavi Raviraj
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Ilakya Selvakumar
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | | | - Nirupama Ramadas
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | | | | | - Devipriya Nagarajan
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
17
|
Pan L, Lu Y, Li Z, Tan Y, Yang H, Ruan P, Li R. Ginkgo biloba Extract EGb761 Attenuates Bleomycin-Induced Experimental Pulmonary Fibrosis in Mice by Regulating the Balance of M1/M2 Macrophages and Nuclear Factor Kappa B (NF-κB)-Mediated Cellular Apoptosis. Med Sci Monit 2020; 26:e922634. [PMID: 32799214 PMCID: PMC7448693 DOI: 10.12659/msm.922634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background The aim of this study was to show whether the standardized Ginkgo biloba extract EGb761, a traditional Chinese medicine, has a therapeutic effect on pulmonary fibrosis (PF). Material/Methods Bleomycin (BLM) was used for establishing the PF mouse model. The mice were treated with a gradient of EGb761 for 28 days to determine an appropriate drug dose. On day 28, the effect of EGb761 on lung injury and inflammation was confirmed by hematoxylin and eosin and Masson staining and evaluated by pulmonary alveolitis and Ashcroft score. The balance of M1/M2 macrophages was evaluated with the respective markers inducible nitric oxide synthase and and interleukin-10 by real-time polymerase chain reaction. Furthermore, the expressions of fibrosis-associated protein α-smooth muscle actin (SMA), related inflammatory protein transforming growth factor (TGF)-β1, the apoptosis-related proteins B-cell lymphoma-associated X protein (Bax), B-cell lymphoma (Bcl)-2, caspase-3, caspase-9, and phosphorylated nuclear factor (NF)-κB (p65) were assessed by western blot. Results On day 28, PF was induced by treating with BLM, whereas EGb761 suppressed the PF of lung tissue. The BLM-induced imbalance of M1/M2 macrophages was reduced by EGb761. Furthermore, the increasing amounts of α-SMA and TGF-β1 induced by BLM were suppressed by EGb761. In addition, the protein or messenger ribonucleic acid expression levels of phosphorylated NF-κB (p65), caspase-3, and caspase-9 were upregulated, whereas Bax and Bcl-2 were downregulated. Treatment with EGb761 restored the levels of these proteins except for caspase-9. Conclusions This study illustrated the protective effect of EGb761 on BLM-induced PF by regulating the balance of M1/M2 macrophages and NF-κB (p65)-mediated apoptosis. The results demonstrated the potential clinical therapeutic effect of EGb761, providing a novel possibility for curing PF.
Collapse
Affiliation(s)
- Ling Pan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Yuehong Lu
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Zhanhua Li
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Yuping Tan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Hongmei Yang
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Ping Ruan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Ruixiang Li
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
18
|
Adelmidrol: A New Promising Antioxidant and Anti-Inflammatory Therapeutic Tool in Pulmonary Fibrosis. Antioxidants (Basel) 2020; 9:antiox9070601. [PMID: 32660140 PMCID: PMC7402091 DOI: 10.3390/antiox9070601] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Chronic pulmonary diseases are characterized by airway remodeling due to complex multicellular responses and the production of free oxygen radicals. They lead to a progressive decline of pulmonary functions. Adelmidrol is an analogue of palmitoylethanolamide (PEA), which is a well-known anti-inflammatory and anti-oxidant compound. In this study, we investigated the efficacy of adelmidrol (10 mg/Kg) for bleomycin-induced pulmonary fibrosis in mice. METHODS Bleomycin intratracheal administration was performed on the first day and for the following twenty-one days, mice were treated with adelmidrol (10 mg/Kg). RESULTS The survival rate and body weight gain were recorded daily. At the end of the experiment, adelmidrol-administered animals showed reduced airway infiltration by inflammatory cells, Myeloperoxidase (MPO) activity, and pro-inflammatory cytokine overexpression (IL,6 IL-1β, TNF-α, and TGF-1β). Moreover, adelmidrol treatment was able to manage the significant incapacity of antioxidants and elevation of the oxidant burden, as shown by the MDA, SOD, and GSH levels and decreased nitric oxide production. It was also able to significantly modulate the JAK2/STAT3 and IκBα/NF-kB pathway. Histologic examination of the lung tissues showed reduced sample injury, mast cell degranulation, chymase activity, and collagen deposition. CONCLUSIONS In sum, our results propose adelmidrol as a therapeutic approach in the treatment of pulmonary fibrosis.
Collapse
|
19
|
Tzouvelekis A, Karampitsakos T, Kourtidou S, Bouros E, Tzilas V, Katsaras M, Antonou C, Dassiou M, Bouros D. Impact of Depression on Patients With Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2020; 7:29. [PMID: 32118014 PMCID: PMC7020231 DOI: 10.3389/fmed.2020.00029] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/20/2020] [Indexed: 01/12/2023] Open
Abstract
Introduction: Depression is prevalent in patients with Idiopathic Pulmonary Fibrosis (IPF). The impact of depression on quality of life and its correlation with disease severity in patients with IPF has not been thoroughly evaluated on prospective studies. Patients and Methods: Between 2016 and 2017, we prospectively enrolled 101 patients (80 male, mean age (years) ± SD: 70.8 ± 8.1) with IPF (mean GAP score ± SD: 4.7 ± 1.8) without previous diagnosis of depression. Depressive symptoms were evaluated with Beck's depression inventory-II (BDI-II). Disease severity was evaluated with pulmonary function (FVC, DLCO) and exercise capacity measures. Symptom burden was assessed by cough and dyspnea scales. Health Related Quality of Life (HRQL) was assessed with two questionnaires. Results: Data for analysis was available from 98 patients (97%). Forty two patients (42.9%) presented with depressive symptoms scoring≥14. A significant association between depressive symptoms and measures of: 1) disease severity: a) GAP score: r = 0.32, p = 0.007, b) DLCO: r = -0.28, p = 0.007, c) 6MWD: r = -0.39, p = 0.017, 2) symptom burden: a) cough: r = -0.57, p < 0.001, b) dyspnea (Borg: r = 0.54, p < 0.001, mMRC: r = 0.55, p < 0.001, SOBQ: r = 0.57, p < 0.001 and 3) HRQL: a) SGRQ: (Total score: r = 0.68, p < 0.001, Activity Score: r = 0.60, p < 0.001, Impact score: r = 0.68, p < 0.001, Symptoms score: r = 0.60, p < 0.001, b) K-BILD: r = -0.66, p < 0.001), was identified. There was no statistically significant difference in BDI-II (p = 0.62) and SGRQ (p = 0.64) 1 year after treatment with antifibrotics. Conclusions: Patients with IPF and severe functional impairment tend to have increased risk for depression development and poor quality of life. Further prospective studies should investigate the role of antidepressant drug therapy in patients with IPF and comorbid depression.
Collapse
Affiliation(s)
- Argyris Tzouvelekis
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Karampitsakos
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Kourtidou
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Bouros
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilios Tzilas
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Matthaios Katsaras
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysoula Antonou
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Dassiou
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| | - Demosthenes Bouros
- First Academic Department of Pneumonology, Medical School, Hospital for Diseases of the Chest Sotiria, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Qiu M, An M, Bian M, Yu S, Liu C, Liu Q. Terrestrosin D from Tribulus terrestris attenuates bleomycin-induced inflammation and suppresses fibrotic changes in the lungs of mice. PHARMACEUTICAL BIOLOGY 2019; 57:694-700. [PMID: 31608748 PMCID: PMC6807862 DOI: 10.1080/13880209.2019.1672754] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/15/2019] [Accepted: 09/20/2019] [Indexed: 05/04/2023]
Abstract
Context: Terrestrosin D (TED), from Tribulus terrestris L. (Zygophyllaceae), exhibits anti-tumour and anti-inflammatory activities. However, its effects on bleomycin (BLM)-induced pulmonary inflammation and the subsequent fibrotic changes remain unclear. Objective: To examine the anti-inflammatory and anti-fibrotic effects of TED against BLM in murine pulmonary tissues. Materials and methods: Male SPF mice received saline (control), TED (10 mg/kg), BLM (2.5 mg/kg), or BLM (2.5 mg/kg) + TED (10 mg/kg) group. BLM was administered as a single intranasal inoculation, and TED was intraperitoneally administered once daily. After 2 and 6 weeks of treatment, cell number and differentiation (Giemsa staining) and TNF-α, IL-6, IL-8, TGF-β1, and PDGF-AB levels (ELISA) were determined in the bronchoalveolar lavage fluid (BALF). Hydroxyproline (Hyp) content in the left pulmonary tissue was also determined (ELISA). The right pulmonary tissue was H&E-stained and assessed for the severity of pulmonary fibrosis using the Ashcroft scoring method. Compared with the BLM group, TED decreased inflammatory cell infiltration; number of macrophages (p < 0.05), neutrophils (p < 0.05), lymphocytes (p < 0.05); percentage of macrophages in the monocyte-macrophage system (p < 0.05), and levels of TNF-α (p < 0.01), IL-6 (p < 0.01), IL-8 (p < 0.05), TGF-β1 (p < 0.05), and PDGF-AB (p < 0.05) in the BALF. TED also reduced Hyp content (p < 0.05) in the pulmonary tissue and attenuated the BLM-induced deterioration in lung histopathology. Discussion and conclusions: TED can inhibit BLM-induced inflammation and fibrosis in the lungs of mice, which may be related to reduced inflammatory and fibrotic markers. These results could be further tested in humans through clinical studies.
Collapse
Affiliation(s)
- Min Qiu
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Ming An
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Mengni Bian
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Shunbang Yu
- School of Health Sciences, University of Newcastle, Newcastle, Australia
| | - Changxiao Liu
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Quanli Liu
- Department of Pharmacy, Baotou Medical College, Baotou, China
| |
Collapse
|
21
|
Abstract
The rate-limiting enzyme in serotonin synthesis is tryptophan hydroxylase (TPH). There are two independent serotonin systems in the body characterized by two isoforms of TPH, TPH1 and TPH2. While TPH2 synthesizes serotonin in the brain, TPH1 is expressed in the gut and in other peripheral tissues and supplies platelets in the circulation with serotonin. This duality of the serotonin system is enforced by the blood-brain barrier which is impermeable for serotonin. In the brain serotonin acts as neurotransmitter and is a main target for the treatment of psychiatric disorders. In the periphery it is released by platelets at the site of activation and elicits numerous physiological effects. TPH1 deficient mice were shown to be protected from diverse diseases including hemostatic, inflammatory, fibrotic, gastrointestinal, and metabolic disorders and therefore serotonin synthesis inhibition emerged as a reasonable therapeutic paradigm. Recently the first TPH inhibitor, telotristat ethyl, came on the market for the treatment of carcinoid syndrome. This review summarizes the state of development and the therapeutic opportunities of such compounds.
Collapse
Affiliation(s)
- Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany; University of Lübeck, Institute for Biology, Ratzeburger Allee 160, 23562 Lübeck, Germany; Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Berlin, Germany.
| |
Collapse
|
22
|
Nie Y, Zhang D, Qian F, Wu Y. Baccatin III ameliorates bleomycin-induced pulmonary fibrosis via suppression of TGF-β1 production and TGF-β1-induced fibroblast differentiation. Int Immunopharmacol 2019; 74:105696. [PMID: 31229901 DOI: 10.1016/j.intimp.2019.105696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and generally lethal lung disease with a high mortality rate. Current therapeutic drugs exhibit limited efficacy but severe adverse effects. Paclitaxel has been identified to exert both anti-inflammatory and anti-fibrosis activity. Baccatin III (BAC), an important precursor of paclitaxel, has been identified as exhibiting immunomodulatory activity with decisively lower toxicity. However, its effects on pulmonary fibrosis remain unknown. In this study, the role of BAC in bleomycin (BLM)-induced pulmonary fibrosis and inflammation in mice was investigated in addition to elucidation of its mechanism of action. Our results demonstrated that administration of BAC in a dose-dependent manner reduced inflammatory infiltration, secretion of the pro-fibrotic mediator TGF-β1 and deposition of collagen and other components of the extracellular matrix (ECM), including alpha smooth muscle actin (α-SMA) and fibronectin. Administration of BAC to treat isolated macrophages stimulated with IL-13, known to activate macrophages, the principal source of TGF-β1, resulted in markedly reduced TGF-β1 expression from macrophages. The AKT/STAT6 signaling pathway was shown to be involved in this process. In addition, we have provided in vitro evidence that BAC inhibits TGF-β1-induced fibroblast differentiation via the Smad2/3 signaling pathway. Furthermore, intratracheal injection of rTGF-β1 significantly exacerbated the degree of fibrosis which was down-regulated by treatment with BAC. Taken together, our data suggest that BAC exerts a protective effect against lung fibrosis and may serve as a potential therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China
| | - Dan Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yaxian Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China.
| |
Collapse
|
23
|
Yang L, Chen PP, Luo M, Shi WL, Hou DS, Gao Y, Xu SF, Deng J. Inhibitory effects of total ginsenoside on bleomycin-induced pulmonary fibrosis in mice. Biomed Pharmacother 2019; 114:108851. [DOI: 10.1016/j.biopha.2019.108851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 01/18/2023] Open
|
24
|
Platelets in Systemic Sclerosis: the Missing Link Connecting Vasculopathy, Autoimmunity, and Fibrosis? Curr Rheumatol Rep 2019; 21:15. [PMID: 30830444 DOI: 10.1007/s11926-019-0815-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Platelets are no longer recognized solely as cell fragments regulating hemostasis. They have pleiotropic functions and they are linked directly or indirectly with the three cornerstones of systemic sclerosis (SSc): vasculopathy, autoimmunity, and fibrosis. In this review, we summarize the current knowledge on the potential role of platelets in the pathogenesis of SSc. RECENT FINDINGS Experimental evidence suggests that vasculopathy, a universal and early finding in SSc, may activate platelets which subsequently release several profibrotic mediators such as serotonin and transforming growth factor β (TGFβ). Platelets may also cross-react with the endothelium leading to the release of molecules, such as thymic stromal lymphopoietin (TSLP), that may trigger fibrosis or sustain vascular damage. Finally, activated platelets express CD40L and provide costimulatory help to B cells, something that may facilitate the breach in immune tolerance. Preclinical studies point to the direction that platelets are actively involved in SSc pathogenesis. Targeting platelets may be an attractive therapeutic approach in SSc.
Collapse
|