1
|
Liu T, Jia H, Li X, Shi L, Wang J, Liu M, Liu H, Zhang T, Zhao Z, Zhao X, Zhao Z. CCTα and GVI iPLA2-induced aberrant phosphatidylcholine metabolism contributes to pulmonary inflammation and fibrosis. Int Immunopharmacol 2025; 156:114718. [PMID: 40286783 DOI: 10.1016/j.intimp.2025.114718] [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: 08/26/2024] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
To date, no comprehensive profiling of phosphatidylcholine (PC) and lysophosphatidylcholine (LPC) with pulmonary inflammation and fibrosis has been published. Our study aimed to analyze PC and LPC metabolism with the development and persistence of pulmonary inflammation and the progression to fibrosis; and their relationship. Mice and cell models exposed to bleomycin and/or transforming growth factor-β1 (TGF-β1) were developed; and porcine surrogates for pulmonary fibrosis were included. Histopathological, immunofluorescence and immunohistochemical staining, transmission electron microscopy, colorimetric, activity and immune complex (IC) assay, lipidomics analysis; and pharmacological intervention assay were used to analyze PC and LPC profile, pulmonary fibrosis and their relationship. Current evidence suggests that 16:0 20:5 PC is a conserved biomarker; and 16:0 18:1 PC, 16:0 18:2 PC; and 16:0 LPC are the potential targets for this disease. Specifically, 16:0 18:1 PC accumulation and exogenous treatment affected lung cell recruitment, migration, transformation, cross-talk, survival/death; and enhanced profibrotic factor release, IC and extracellular matrix (ECM) deposition, where CTP:phosphocholine cytidylyltransferase α (CCTα) and group VI Ca2+-independent phospholipase A2 (GVI iPLA2) play an important role, particularly in lung and spleen neutrophils, macrophages, and T lymphocytes. Overall, these results provide new insights into how the dysregulated PC metabolism, particularly for 16:0 18:1 PC, affects the development and persistence of lung inflammation and the progression to fibrosis, and thus may facilitate the discovery of biomarkers and targets for this disease.
Collapse
Affiliation(s)
- Tao Liu
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; College of Animal Husbandry and Veterinary Medicine, Xinyang Agriculture and Forestry University, Xinyang 464001, China
| | - Hong Jia
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xinsheng Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Lijun Shi
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jing Wang
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Meizhen Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Hailong Liu
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Tao Zhang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Zhenwen Zhao
- Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrometry Center, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xinghui Zhao
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhanzhong Zhao
- Laboratory of Pharmacobiology, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
2
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [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: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
3
|
Tong Z, Du X, Zhou Y, Jing F, Ma J, Feng Y, Lou S, Wang Q, Dong Z. Drp1-mediated mitochondrial fission promotes pulmonary fibrosis progression through the regulation of lipid metabolic reprogramming by ROS/HIF-1α. Cell Signal 2024; 117:111075. [PMID: 38311302 DOI: 10.1016/j.cellsig.2024.111075] [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: 10/11/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVE To confirm the mechanism of dynamic-related protein 1 (Drp1)-mediated mitochondrial fission through ROS/HIF-1α-mediated regulation of lipid metabolic reprogramming in the progression of pulmonary fibrosis (PF). METHODS A mouse model of PF was established by intratracheal instillation of bleomycin (BLM) (2.5 mg/kg). A PF cell model was constructed by stimulating MRC-5 cells with TGF-β (10 ng/mL). Pathological changes in the lung tissue and related protein levels were observed via tissue staining. The indicators related to lipid oxidation were detected by a kit, and lipid production was confirmed through oil red O staining. Inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). RT-qPCR, Western blotting and immunofluorescence staining were used to detect the expression of genes and proteins related to the disease. We used CCK-8 and EdU staining to confirm cell proliferation, flow cytometry was used to confirm apoptosis and ROS levels, α-SMA expression was detected by immunofluorescence staining, and mitochondria were observed by MitoTracker staining. RESULTS The BLM induced lung tissue structure and alveolar wall thickening in mice. Mitochondrial fission was observed in MRC-5 cells induced by TGF-β, which led to increased cell proliferation; decreased apoptosis; increased expression of collagen, α-SMA and Drp1; and increased lipid oxidation and inflammation. Treatment with the Drp1 inhibitor mdivi-1 or transfection with si-Drp1 attenuated the induction of BLM and TGF-β. For lipid metabolism, lipid droplets were formed in BLM-induced lung tissue and in TGF-β-induced cells, fatty acid oxidation genes and lipogenesis-related genes were upregulated, ROS levels in cells were increased, and the expression of HIF-1α was upregulated. Mdivi-1 treatment reversed TGF-β induction, while H2O2 treatment or OE-HIF-1α transfection reversed the effect of mdivi-1. CONCLUSION In PF, inhibition of Drp1 can prevent mitochondrial fission in fibroblasts and regulate lipid metabolism reprogramming through ROS/HIF-1α; thus, fibroblast activation was inhibited, alleviating the progression of PF.
Collapse
Affiliation(s)
- Zhongkai Tong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Xuekui Du
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Fangxue Jing
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - JiangPo Ma
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325000, China
| | - Yingying Feng
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - Saiyun Lou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Second Clinical Medicine Faculty of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiong Wang
- Department of Respiratory Infection, Zhenhai Hospital of Traditional Chinese Medicine, Ningbo 315200, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China.
| |
Collapse
|
4
|
Fernández Requena B, Nadeem S, Reddy VP, Naidoo V, Glasgow JN, Steyn AJC, Barbas C, Gonzalez-Riano C. LiLA: lipid lung-based ATLAS built through a comprehensive workflow designed for an accurate lipid annotation. Commun Biol 2024; 7:45. [PMID: 38182666 PMCID: PMC10770321 DOI: 10.1038/s42003-023-05680-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024] Open
Abstract
Accurate lipid annotation is crucial for understanding the role of lipids in health and disease and identifying therapeutic targets. However, annotating the wide variety of lipid species in biological samples remains challenging in untargeted lipidomic studies. In this work, we present a lipid annotation workflow based on LC-MS and MS/MS strategies, the combination of four bioinformatic tools, and a decision tree to support the accurate annotation and semi-quantification of the lipid species present in lung tissue from control mice. The proposed workflow allowed us to generate a lipid lung-based ATLAS (LiLA), which was then employed to unveil the lipidomic signatures of the Mycobacterium tuberculosis infection at two different time points for a deeper understanding of the disease progression. This workflow, combined with manual inspection strategies of MS/MS data, can enhance the annotation process for lipidomic studies and guide the generation of sample-specific lipidome maps. LiLA serves as a freely available data resource that can be employed in future studies to address lipidomic alterations in mice lung tissue.
Collapse
Affiliation(s)
- Belén Fernández Requena
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, España
| | - Sajid Nadeem
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vineel P Reddy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adrie J C Steyn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Africa Health Research Institute, Durban, South Africa
- Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, España.
| | - Carolina Gonzalez-Riano
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, España.
| |
Collapse
|
5
|
O'Callaghan M, Duignan J, Tarling EJ, Waters DK, McStay M, O'Carroll O, Bridges JP, Redente EF, Franciosi AN, McGrath EE, Butler MW, Dodd JD, Fabre A, Murphy DJ, Keane MP, McCarthy C. Analysis of tissue lipidomics and computed tomography pulmonary fat attenuation volume (CT PFAV ) in idiopathic pulmonary fibrosis. Respirology 2023; 28:1043-1052. [PMID: 37642207 DOI: 10.1111/resp.14582] [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: 02/28/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND AND OBJECTIVE There is increasing interest in the role of lipids in processes that modulate lung fibrosis with evidence of lipid deposition in idiopathic pulmonary fibrosis (IPF) histological specimens. The aim of this study was to identify measurable markers of pulmonary lipid that may have utility as IPF biomarkers. STUDY DESIGN AND METHODS IPF and control lung biopsy specimens were analysed using a unbiased lipidomic approach. Pulmonary fat attenuation volume (PFAV) was assessed on chest CT images (CTPFAV ) with 3D semi-automated lung density software. Aerated lung was semi-automatically segmented and CTPFAV calculated using a Hounsfield-unit (-40 to -200HU) threshold range expressed as a percentage of total lung volume. CTPFAV was compared to pulmonary function, serum lipids and qualitative CT fibrosis scores. RESULTS There was a significant increase in total lipid content on histological analysis of IPF lung tissue (23.16 nmol/mg) compared to controls (18.66 mol/mg, p = 0.0317). The median CTPFAV in IPF was higher than controls (1.34% vs. 0.72%, p < 0.001) and CTPFAV correlated significantly with DLCO% predicted (R2 = 0.356, p < 0.0001) and FVC% predicted (R2 = 0.407, p < 0.0001) in patients with IPF. CTPFAV correlated with CT features of fibrosis; higher CTPFAV was associated with >10% reticulation (1.6% vs. 0.94%, p = 0.0017) and >10% honeycombing (1.87% vs. 1.12%, p = 0.0003). CTPFAV showed no correlation with serum lipids. CONCLUSION CTPFAV is an easily quantifiable non-invasive measure of pulmonary lipids. In this pilot study, CTPFAV correlates with pulmonary function and radiological features of IPF and could function as a potential biomarker for IPF disease severity assessment.
Collapse
Affiliation(s)
- Marissa O'Callaghan
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - John Duignan
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Elizabeth J Tarling
- Division of Cardiology, University of California, Los Angeles, California, USA
| | - Darragh K Waters
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Megan McStay
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Orla O'Carroll
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - James P Bridges
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | | | - Alessandro N Franciosi
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Emmet E McGrath
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Marcus W Butler
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Jonathan D Dodd
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Aurelie Fabre
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland
| | - David J Murphy
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Michael P Keane
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Cormac McCarthy
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Fu Z, Yin H, Liu J, He Y, Song S, Peng X, Huang X, Lai Y, Li S, Luo Q, Su J, Yang P. Therapeutic effects of fatty acid binding protein 1 in mice with pulmonary fibrosis by regulating alveolar epithelial regeneration. BMJ Open Respir Res 2023; 10:e001568. [PMID: 37940355 PMCID: PMC10632910 DOI: 10.1136/bmjresp-2022-001568] [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: 12/03/2022] [Accepted: 10/20/2023] [Indexed: 11/10/2023] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis is a progressive fibrotic lung disease with limited therapeutic options and high lethality, related to alveolar type II epithelial (ATII) cell dysregulation, the abnormal repair of alveolar epithelial cells and activation of fibroblasts promote the development of pulmonary fibrosis. Fatty acid binding protein 1 (FABP1) was significantly downregulated in the fibrotic state by proteomics screening in our previous date, and the ATII cell dysregulation can be mediated by FABP1 via regulating fatty acid metabolism and intracellular transport. The aim of this study was to evaluate the role and potential mechanism of FABP1 in the development of pulmonary fibrosis. METHODS Proteomics screening was used to detect changes of the protein profiles in two different types (induced by bleomycin and silica, respectively) of pulmonary fibrosis models. The localisation of FABP1 in mouse lung was detected by Immunofluorescence and immunohistochemistry. Experimental methods such as lung pathology, micro-CT, western blotting, small animal imaging in vivo, EdU, etc were used to verify the role of FABP1 in pulmonary fibrosis. RESULTS The expression of FABP1 in the mouse lung was significantly reduced in the model of pulmonary fibrosis from our proteomic analysis and immunological methods, the double immunofluorescence staining showed that FABP1 was mainly localised in type II alveolar epithelial cells. Additionally, the expression of FABP1 was negatively correlated with the progression of pulmonary fibrosis. Further in vivo and in vitro experiments showed that overexpression of FABP1 alleviated pulmonary fibrosis by protecting alveolar epithelium from injury and promoting cell survival. CONCLUSION Our findings provide a proof-of-principle that FABP1 may represent an effective treatment for pulmonary fibrosis by regulating alveolar epithelial regeneration, which may be associated with the fatty acid metabolism in ATII cells.
Collapse
Affiliation(s)
- Zhenli Fu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hang Yin
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiani Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying He
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shengren Song
- Department of Respiratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaomin Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xihui Huang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunxin Lai
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qun Luo
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Su
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Penghui Yang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Huang P, Lee C, Lee L, Huang H, Huang Y, Lan J, Lee C. Surface-enhanced Raman scattering (SERS) by gold nanoparticle characterizes dermal thickening by collagen in bleomycin-treated skin ex vivo. Skin Res Technol 2023; 29:e13334. [PMID: 37231930 PMCID: PMC10316472 DOI: 10.1111/srt.13334] [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: 10/28/2022] [Accepted: 04/10/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE Current skin imaging modalities, including optical, electron, and confocal microscopy, mostly require tissue fixations that could damage proteins and biological molecules. Live tissue or cell imaging such as ultrasonography and optical coherent microscope may not adequately measure the dynamic spectroscopical changes. Raman spectroscopy has been adopted for skin imaging in vivo, mostly for skin cancer imaging. However, whether the epidermal and dermal thickening in skin could be measured and distinguished by conventional Ramen spectroscopy or the surface-enhanced Raman scattering (SERS), a rapid and label-free method for noninvasive measurement remains unknown. METHODS Human skin sections from patients of atopic dermatitis and keloid, which represent epidermal and dermal thickening, respectively, were measured by conventional Ramen spectroscopy. In mice, skin sections from imiquimod (IMQ)- and bleomycin (BLE)-treated mice, which reflect the epidermal and dermal thickening, respectively, were measured by SERS, that incorporates gold nanoparticles to generate surface plasma and enhance Raman signals. RESULTS Conventional Ramen spectroscopy failed to consistently show the Raman shift in human samples among the different groups. SERS successfully revealed a prominent peak around 1300 cm-1 in the IMQ-treated skin; and two significant peaks around 1100 and 1300 cm-1 in BLE-treated group. Further quantitative analysis showed 1100 cm-1 peak was significantly accentuated in the BLE-treated skin than that in control skin. SERS identified in vitro a similar 1100 cm-1 peak in solutions of collagen, the major dermal biological molecules. CONCLUSION SERS distinguishes the epidermal or dermal thickening in mouse skin with rapid and label-free measures. A prominent 1100 cm-1 SERS peak in the BLE-treated skin may result from collagen. SERS might help precision diagnosis in the future.
Collapse
Affiliation(s)
- Po‐Jung Huang
- Institute of Environmental EngineeringNational Sun Yat‐sen UniversityKaohsiungTaiwan
- Department of Chemical and Materials EngineeringNational Central UniversityTaoyuanTaiwan
| | - Chao‐Kuei Lee
- Department of PhotonicsNational Sun Yat‐Sen UniversityKaohsiungTaiwan
| | - Ling‐Hau Lee
- Department of DermatologyKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
- Department of DermatologyChang Gung University College of MedicineTaoyuanTaiwan
| | - Hsiang‐Fu Huang
- Department of PhotonicsNational Sun Yat‐Sen UniversityKaohsiungTaiwan
| | - Yi‐Hsuan Huang
- Department of PhotonicsNational Sun Yat‐Sen UniversityKaohsiungTaiwan
| | - Jia‐Chi Lan
- Department of PhotonicsNational Sun Yat‐Sen UniversityKaohsiungTaiwan
| | - Chih‐Hung Lee
- Department of DermatologyKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
- Department of DermatologyChang Gung University College of MedicineTaoyuanTaiwan
| |
Collapse
|
8
|
El Husseini K, Poté N, Jaillet M, Mordant P, Mal H, Frija-Masson J, Borie R, Cazes A, Crestani B, Mailleux A. [Adipocytes, adipokines and metabolic alterations in pulmonary fibrosis]. Rev Mal Respir 2023; 40:225-229. [PMID: 36740493 DOI: 10.1016/j.rmr.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by severe remodeling of the lung parenchyma, with an accumulation of activated myofibroblasts and extracellular matrix, along with aberrant cellular differentiation. Within the subpleural fibrous zones, ectopic adipocyte deposits often appear. In addition, alterations in lipid homeostasis have been associated with IPF pathophysiology. In this mini-review, we will discuss the potential involvement of the adipocyte secretome and its paracrine or endocrine-based contribution to the pathophysiology of IPF, via protein or lipid mediators in particular.
Collapse
Affiliation(s)
- K El Husseini
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France.
| | - N Poté
- Service d'anatomopathologie, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - M Jaillet
- Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - P Mordant
- Service de chirurgie vasculaire et thoracique, Hôpital Bichat, AP-HP, Paris, France
| | - H Mal
- Service de pneumologie B, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - J Frija-Masson
- Service de physiologie-explorations fonctionnelles respiratoires, Hôpital Bichat, AP-HP, Paris, France
| | - R Borie
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France
| | - A Cazes
- Service d'anatomopathologie, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - B Crestani
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - A Mailleux
- Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| |
Collapse
|
9
|
Zhang Y, Li T, Pan M, Wang W, Huang W, Yuan Y, Xie Z, Chen Y, Peng J, Li X, Meng Y. SIRT1 prevents cigarette smoking-induced lung fibroblasts activation by regulating mitochondrial oxidative stress and lipid metabolism. J Transl Med 2022; 20:222. [PMID: 35568871 PMCID: PMC9107262 DOI: 10.1186/s12967-022-03408-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/24/2022] [Indexed: 12/06/2022] Open
Abstract
BACKGROUND Cigarette smoking (CS) is a strong risk factor for idiopathic pulmonary fibrosis (IPF). It can activate lung fibroblasts (LF) by inducing redox imbalance. We previously showed that clearing mitochondrial reactive oxygen species (mtROS) protects against CS-induced pulmonary fibrosis. However, the precise mechanisms of mtROS in LF need further investigation. Here we focused on mtROS to elucidate how it was regulated by CS in LF and how it contributed to LF activation. METHODS We treated cells with 1% cigarette smoking extract (CSE) and examined mtROS level by MitoSOX™ indicator. And the effect of CSE on expression of SIRT1, SOD2, mitochondrial NOX4 (mtNOX4), fatty acid oxidation (FAO)-related protein PPARα and CPT1a and LF activation marker Collagen I and α-SMA were detected. Nile Red staining was performed to show cellular lipid content. Then, lipid droplets, autophagosome and lysosome were marked by Bodipy 493/503, LC3 and LAMP1, respectively. And lipophagy was evaluated by the colocalization of lipid droplets with LC3 and LAMP1. The role of autophagy on lipid metabolism and LF activation were explored. Additionally, the effect of mitochondria-targeted ROS scavenger mitoquinone and SIRT1 activator SRT1720 on mitochondrial oxidative stress, autophagy flux, lipid metabolism and LF activation were investigated in vitro and in vivo. RESULTS We found that CS promoted mtROS production by increasing mtNOX4 and decreasing SOD2. Next, we proved mtROS inhibited the expression of PPARα and CPT1a. It also reduced lipophagy and upregulated cellular lipid content, suggesting lipid metabolism was disturbed by CS. In addition, we showed both insufficient FAO and lipophagy resulted from blocked autophagy flux caused by mtROS. Moreover, we uncovered decreased SIRT1 was responsible for mitochondrial redox imbalance. Furthermore, we proved that both SRT1720 and mitoquinone counteracted the effect of CS on NOX4, SOD2, PPARα and CPT1a in vivo. CONCLUSIONS We demonstrated that CS decreased SIRT1 to activate LF through dysregulating lipid metabolism, which was due to increased mtROS and impaired autophagy flux. These events may serve as therapeutic targets for IPF patients.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Li
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Miaoxia Pan
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhui Huang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yafei Yuan
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhanzhan Xie
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yixin Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Peng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou, China.
| | - Ying Meng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Extracellular Lipids in the Lung and Their Role in Pulmonary Fibrosis. Cells 2022; 11:cells11071209. [PMID: 35406772 PMCID: PMC8997955 DOI: 10.3390/cells11071209] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Lipids are major actors and regulators of physiological processes within the lung. Initial research has described their critical role in tissue homeostasis and in orchestrating cellular communication to allow respiration. Over the past decades, a growing body of research has also emphasized how lipids and their metabolism may be altered, contributing to the development and progression of chronic lung diseases such as pulmonary fibrosis. In this review, we first describe the current working model of the mechanisms of lung fibrogenesis before introducing lipids and their cellular metabolism. We then summarize the evidence of altered lipid homeostasis during pulmonary fibrosis, focusing on their extracellular forms. Finally, we highlight how lipid targeting may open avenues to develop therapeutic options for patients with lung fibrosis.
Collapse
|
11
|
Efficacy and Safety of Dahuang Zhechong Pill in Silicosis: A Randomized Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4354054. [PMID: 34840587 PMCID: PMC8616670 DOI: 10.1155/2021/4354054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/19/2021] [Accepted: 10/23/2021] [Indexed: 12/26/2022]
Abstract
Background There is no effective therapy for silicosis, and Dahuang Zhechong pill (DHZCP), an ancient Chinese medicine prescription, may have a therapeutic effect on silicosis. This study aims to verify the efficacy and safety of DHZCP in silicosis. Methods This is a randomized controlled clinical trial done at Panzhihua Second People's Hospital (Panzhihua City, Sichuan Province, China). Participants diagnosed with silicosis were recruited and randomized to the conventional treatment group (CG) or DHZCP combined with the conventional treatment group (DG). Forced vital capacity % predicted (FVC%), diffusing capacity of the lung for carbon monoxide % predicted (DLCO%), six-minute walk distance (6MWD), peripheral oxygen (SpO2), King's Brief Interstitial Lung Disease Questionnaire (K-BILD), and safety outcomes were measured at baseline and 9 weeks. Results Fifty-six participants (28 in each group) completed the study, and 53 of them (26 in DG and 27 in CG) completed pulmonary function. At 9 weeks, compared with no DHZCP, DHZCP treatment was associated with significant improvements in FVC% (mean ± SD, 95%CI) (8.2 ± 3.9, 0.3 to 16.0), DLCO% (8.6 ± 3.5, 1.5 to 15.7), SpO2 (3.8 ± 0.7, 2.3 to 5.2), and K-BILD total score (6.0 ± 2.3, 1.4 to 10.7). And, there were no statistical differences of safety outcomes between the two groups. Eight patients accepting DHZCP developed mild diarrhea during the first week, which subsequently resolved on its own. Conclusion DHZCP could improve the pulmonary function, the quality of life, and the exercise capacity of silicosis patients.
Collapse
|
12
|
Nambiar S, Clynick B, How BS, King A, Walters EH, Goh NS, Corte TJ, Trengove R, Tan D, Moodley Y. There is detectable variation in the lipidomic profile between stable and progressive patients with idiopathic pulmonary fibrosis (IPF). Respir Res 2021; 22:105. [PMID: 33836757 PMCID: PMC8033725 DOI: 10.1186/s12931-021-01682-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease characterized by fibrosis and progressive loss of lung function. The pathophysiological pathways involved in IPF are not well understood. Abnormal lipid metabolism has been described in various other chronic lung diseases including asthma and chronic obstructive pulmonary disease (COPD). However, its potential role in IPF pathogenesis remains unclear. Methods In this study, we used ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) to characterize lipid changes in plasma derived from IPF patients with stable and progressive disease. We further applied a data-independent acquisition (DIA) technique called SONAR, to improve the specificity of lipid identification. Results Statistical modelling showed variable discrimination between the stable and progressive subjects, revealing differences in the detection of triglycerides (TG) and phosphatidylcholines (PC) between progressors and stable IPF groups, which was further confirmed by mass spectrometry imaging (MSI) in IPF tissue. Conclusion This is the first study to characterise lipid metabolism between stable and progressive IPF, with results suggesting disparities in the circulating lipidome with disease progression.
Collapse
Affiliation(s)
- Shabarinath Nambiar
- Separation Science and Metabolomics Laboratory, Murdoch University, Murdoch, WA, Australia
| | - Britt Clynick
- School of Biomedical Science, University of Western Australia, Crawley, WA, Australia. .,Institute for Respiratory Health, Nedlands, WA, Australia.
| | - Bong S How
- Separation Science and Metabolomics Laboratory, Murdoch University, Murdoch, WA, Australia.,Metabolomics Australia, Murdoch University, Murdoch, WA, Australia
| | - Adam King
- Scientific Operations, Waters Corporation, Stamford Avenue, Wilmslow, SK9 4AX, UK
| | - E Haydn Walters
- Alfred Hospital, Melbourne, VIC, Australia.,University of Tasmania, Hobart, TAS, Australia.,University of Melbourne, Parkville, VIC, Australia.,Royal Hobart Hospital, Hobart, TAS, Australia
| | - Nicole S Goh
- Austin Hospital, Heidelberg, VIC, Australia.,Institute of Breathing and Sleep, Heidelberg, VIC, Australia
| | - Tamera J Corte
- University of Sydney, Camperdown, NSW, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Robert Trengove
- Separation Science and Metabolomics Laboratory, Murdoch University, Murdoch, WA, Australia.,Metabolomics Australia, Murdoch University, Murdoch, WA, Australia
| | - Dino Tan
- School of Biomedical Science, University of Western Australia, Crawley, WA, Australia.,Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yuben Moodley
- School of Biomedical Science, University of Western Australia, Crawley, WA, Australia.,Institute for Respiratory Health, Nedlands, WA, Australia.,Fiona Stanley Hospital, Murdoch, WA, Australia
| |
Collapse
|
13
|
Tejada MÁ, Santos-Llamas AI, Fernández-Ramírez MJ, Tarín JJ, Cano A, Gómez R. A Reassessment of the Therapeutic Potential of a Dopamine Receptor 2 Agonist (D2-AG) in Endometriosis by Comparison against a Standardized Antiangiogenic Treatment. Biomedicines 2021; 9:biomedicines9030269. [PMID: 33800198 PMCID: PMC8001569 DOI: 10.3390/biomedicines9030269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/26/2022] Open
Abstract
Dopamine receptor 2 agonists (D2-ags) have been shown to reduce the size of tumors by targeting aberrant angiogenesis in pathological tissue. Because of this, the use of a D2-ag was inferred for endometriosis treatment. When assayed in mouse models however, D2-ags have been shown to cause a shift of the immature vessels towards a more mature phenotype but not a significant reduction in the amount of vascularization and size of lesions. These has raised concerns on whether the antiangiogenic effects of these compounds confer a therapeutic value for endometriosis. In the belief that antiangiogenic effects of D2-ags in endometriosis were masked due to non-optimal timing of pharmacological interventions, herein we aimed to reassess the antiangiogenic therapeutic potential of D2-ags in vivo by administering compounds at a timeframe in which vessels in the lesions are expected to be more sensitive to antiangiogenic stimuli. To prove our point, immunodeficient (NU/NU) mice were given a D2-ag (cabergoline), anti-VEGF (CBO-P11) or vehicle (saline) compounds (n = 8 per group) starting 5 days after implantation of a fluorescently labeled human lesion. The effects on the size of the implants was estimated by monitoring the extent of fluorescence emitted by the lesion during the three-week treatment period. Subsequently mice were sacrificed and lesions excised and fixed for quantitative immunohistochemical/immunofluorescent analysis of angiogenic parameters. Lesion size, vascular density and innervation were comparable in D2-ag and anti-VEGF groups and significantly decreased when compared to control. These data suggest that D2-ags are as powerful as standard antiangiogenic compounds in interfering with angiogenesis and lesion size. Our preliminary study opens the way to further exploration of the mechanisms beneath the antiangiogenic effects of D2-ags for endometriosis treatment in humans.
Collapse
Affiliation(s)
- Miguel Á. Tejada
- Research Unit on Women’s Health-Institute of Health Research, INCLIVA, 46010 Valencia, Spain; (A.I.S.-L.); (J.J.T.)
- Correspondence: (M.Á.T.); (A.C.); (R.G.)
| | - Ana I. Santos-Llamas
- Research Unit on Women’s Health-Institute of Health Research, INCLIVA, 46010 Valencia, Spain; (A.I.S.-L.); (J.J.T.)
| | - María José Fernández-Ramírez
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario, 46010 Valencia, Spain;
- Department of Pediatrics and Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain
| | - Juan J. Tarín
- Research Unit on Women’s Health-Institute of Health Research, INCLIVA, 46010 Valencia, Spain; (A.I.S.-L.); (J.J.T.)
- Department of Cellular Biology, Functional Biology, and Physical Anthropology, University of Valencia, 46100 Burjassot, Spain
| | - Antonio Cano
- Research Unit on Women’s Health-Institute of Health Research, INCLIVA, 46010 Valencia, Spain; (A.I.S.-L.); (J.J.T.)
- Department of Pediatrics and Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.Á.T.); (A.C.); (R.G.)
| | - Raúl Gómez
- Research Unit on Women’s Health-Institute of Health Research, INCLIVA, 46010 Valencia, Spain; (A.I.S.-L.); (J.J.T.)
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.Á.T.); (A.C.); (R.G.)
| |
Collapse
|
14
|
Nojima Y, Takeda Y, Maeda Y, Bamba T, Fukusaki E, Itoh MN, Mizuguchi K, Kumanogoh A. Metabolomic analysis of fibrotic mice combined with public RNA-Seq human lung data reveal potential diagnostic biomarker candidates for lung fibrosis. FEBS Open Bio 2020; 10:2427-2436. [PMID: 32961634 PMCID: PMC7609803 DOI: 10.1002/2211-5463.12982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/06/2020] [Accepted: 09/15/2020] [Indexed: 01/22/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe lung disease with poor survival that warrants early and precise diagnosis for timely therapeutic intervention. Despite accumulating genomic, transcriptomic, proteomic, and lipidomic data on IPF, evidence from water‐soluble metabolomics is limited. To identify biomarkers for IPF from water‐soluble metabolomic data, we measured the levels of various metabolites in bronchoalveolar lavage fluid (BALF) and serum samples from a bleomycin‐induced murine pulmonary fibrotic model using gas chromatography/mass spectrometry. Thirty‐two of 73 BALF metabolites and 29 of 74 serum metabolites were annotated. We observed that the levels of proline and methionine were higher in BALF but lower in serum than those in the control. Furthermore, analysis of public RNA‐Seq data from the lungs of patients with IPF revealed that proline‐ and methionine‐related genes were significantly upregulated compared to those in the lungs of healthy controls. These results suggest that proline and methionine may be potential biomarkers for IPF and may help to deepen our understanding of the pathophysiology of the disease. Based on our results, we propose a model capable of recapitulating the proline and methionine metabolism of fibrotic lungs, thereby providing better means for studying the disease and developing novel therapeutic strategies for IPF.
Collapse
Affiliation(s)
- Yosui Nojima
- Laboratory of BioinformaticsArtificial Intelligence Center for Health and Biomedical Research (ArCHER)National Institutes of Biomedical Innovation, Health and NutritionOsakaJapan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical ImmunologyOsaka University Graduate School of MedicineJapan
| | - Yohei Maeda
- Department of Respiratory Medicine and Clinical ImmunologyOsaka University Graduate School of MedicineJapan
| | - Takeshi Bamba
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversityJapan
- Division of MetabolomicsMedical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Eiichiro Fukusaki
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversityJapan
| | - Mari N. Itoh
- Laboratory of BioinformaticsArtificial Intelligence Center for Health and Biomedical Research (ArCHER)National Institutes of Biomedical Innovation, Health and NutritionOsakaJapan
| | - Kenji Mizuguchi
- Laboratory of BioinformaticsArtificial Intelligence Center for Health and Biomedical Research (ArCHER)National Institutes of Biomedical Innovation, Health and NutritionOsakaJapan
- Institute for Protein ResearchOsaka UniversityJapan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical ImmunologyOsaka University Graduate School of MedicineJapan
| |
Collapse
|
15
|
Shimizu Y, Nakamura Y, Horibata Y, Fujimaki M, Hayashi K, Uchida N, Morita H, Arai R, Chibana K, Takemasa A, Sugimoto H. Imaging of lysophosphatidylcholine in an induced pluripotent stem cell-derived endothelial cell network. Regen Ther 2020; 14:299-305. [PMID: 32462058 PMCID: PMC7240204 DOI: 10.1016/j.reth.2020.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/01/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022] Open
Abstract
Introduction Vascular endothelial cell disorders are closely related to cardiovascular disease (CVD) and pulmonary diseases. Abnormal lipid metabolism in the endothelium leads to changes in cell signalling, and the expression of genes related to immunity and inflammation. It is therefore important to investigate the pathophysiology of vascular endothelial disorders in terms of lipid metabolism, using a disease model of endothelium. Methods Human induced pluripotent stem cell-derived endothelial cells (iECs) were cultured on a matrigel to form an iEC network. Lipids in the iEC network were investigated by matrix-assisted laser desorption/ionization (MALDI) time-of-flight (TOF) imaging mass spectrometry (IMS) analysis. Ion fragments obtained by mass spectrometry were analysed using an infusion method, involving precursor ion scanning with fragment ion. Results The MALDI TOF IMS analysis revealed co-localized intensity of peaks at m/z 592.1 and 593.1 in the iEC network. Tandem mass spectrometry (MS/MS) analysis by MALDI-imaging, in conjunction with precursor ion scanning using an infusion method with lipid extracts, identified that these precursor ions were lysophosphatidylcholine (LPC) (22:5) and its isotype. Conclusion The MALDI-imaging analysis showed that LPC (22:5) was abundant in an iEC network. As an in vitro test model for disease and potential therapy, present analysis methods using MALDI-imaging combined with, for example, mesenchymal stem cells (MSC) to a disease derived iEC network may be useful in revealing the changes in the amount and distribution of lipids under various stimuli.
Collapse
Affiliation(s)
- Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yusuke Nakamura
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yasuhiro Horibata
- Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Mio Fujimaki
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Keitaro Hayashi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Nobuhiko Uchida
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroko Morita
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Ryo Arai
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kazuyuki Chibana
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Akihiro Takemasa
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroyuki Sugimoto
- Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
16
|
Ricard-Blum S, Miele AE. Omic approaches to decipher the molecular mechanisms of fibrosis, and design new anti-fibrotic strategies. Semin Cell Dev Biol 2020; 101:161-169. [DOI: 10.1016/j.semcdb.2019.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
|
17
|
Kithur Mohamed S, Asif M, Nazari MV, Baharetha HM, Mahmood S, Yatim ARM, Abdul Majid AS, Abdul Majid AMS. Antiangiogenic activity of sophorolipids extracted from refined bleached deodorized palm olein. Indian J Pharmacol 2019; 51:45-54. [PMID: 31031467 PMCID: PMC6444841 DOI: 10.4103/ijp.ijp_312_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES: Sophorolipids (SLs) are a group of surface-active glycolipids produced by a type of nonpathogenic yeast Candida bombicola in the presence of vegetable oil through fermentation technology. SLs have shown antitumor activity; however, the mechanism of action underlying the anticancer activity of SLs is poorly understood. This work evaluated the anticancer activity of SLs fermented from palm oil by exploring its antiangiogenic activity. MATERIALS AND METHODS: The SLs that were fermented and further characterized for their biochemical activities. Cytotoxicity study was performed to assess cytostatic properties. A series of in vitro and ex vivo angiogenesis assay was also carried out. The relative fold change in the expression of p53 mRNA by SLs was also studied. RESULTS: Altogether, the data show that SLs derived from palm oil fermentation process inhibited neovascularization in the ex vivo tissue segments and also the endothelial cell proliferation between 50% and 65% inhibition as a whole. The palm oil derived SLs also caused downregulation of the suppression level of vascular endothelial growth factor and also upregulate the p53 mRNA level. The analytical studies revealed the presence of high amount of phenolic compounds but with relatively weak antioxidant activity. The gas chromatography-mass spectrometry studies revealed abundant amount of palmitic and oleic acid, the latter an established antiangiogenic agent, and the former being proangiogenic. CONCLUSION: Therefore, it can be concluded from this study that SLs derived from fermented palm oil have potent antiangiogenic activity which may be attributed by its oleic acid component.
Collapse
Affiliation(s)
- Shazmin Kithur Mohamed
- Department of Pharmacology, EMAN Testing and Research Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Muhammad Asif
- Department of Pharmacology, EMAN Testing and Research Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Mansoureh Vishkaei Nazari
- Department of Pharmacology, EMAN Testing and Research Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Hussein M Baharetha
- Department of Pharmacology, EMAN Testing and Research Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Syed Mahmood
- Department of Pharmaceutical Engineering, Faculty of Engineering Technology, University Malaysia Pahang, Gambang, Malaysia
| | - Abdul Rashid M Yatim
- Advanced Oleochemicals Technology Research Division, Malaysian Palm Oil Board, Kajang, Selangor, Malaysia
| | | | - Amin Malik Shah Abdul Majid
- Department of Pharmacology, EMAN Testing and Research Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia.,ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|