1
|
Jing R, Xie X, Liao X, He S, Mo J, Dai H, Hu Z, Pan L. Transforming growth factor-β1 is associated with inflammatory resolution via regulating macrophage polarization in lung injury model mice. Int Immunopharmacol 2024; 142:112997. [PMID: 39217883 DOI: 10.1016/j.intimp.2024.112997] [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: 01/07/2024] [Revised: 05/21/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Ventilation is the main respiratory support therapy for acute respiratory distress syndrome, which triggers acute lung injury (ALI). Macrophage polarization is vital for the resolution of inflammation and tissue injury. We hypothesized that transforming growth factor (TGF)-β1 may attenuate inflammation and ventilator-induced ALI by promoting M2 macrophage polarization. METHODS C57BL/6 mice received 4-hour ventilation and extubation to observe the resolution of lung injury and inflammation. Lung vascular permeability, inflammation, and histological changes in the lungs were evaluated by bronchoalveolar lavage analysis, enzyme linked immunosorbent assay, hematoxylin and eosin staining, as well as transmission electron microscope. TGF-β1 cellular production and macrophage subsets were analyzed by flow cytometry. The relative expressions of targeted proteins and genes were measured by immunofuorescence staining, Western blot, and quantitative polymerase chain reaction. RESULTS High tidal volume-induced injury and inflammation were resolved at 3 days of post-ventilation (PV3d) to PV10d, with increased elastic fibers, proteoglycans, and collagen content, as well as higher TGF-β1 levels. M1 macrophages were increased in the acute phase, whereas M2a macrophages began to increase from PV1d to PV3d, as well as increased M2c macrophages from PV3d to PV7d. A single dose of rTGF-β1 attenuated lung injury and inflammation at end of ventilation with polymorphonuclear leukocyte apoptosis, while nTAb pretreatment induced the abnormal elevation of TGF-β1 that aggravated lung injury and inflammation due to the significant inhibition of M1 macrophages polarized to M2a, M2b, and M2c macrophages. CONCLUSIONS Precise secretion of TGF-β1-mediated macrophage polarization plays a crucial role in the resolution of ventilator-induced inflammatory lung injury.
Collapse
Affiliation(s)
- Ren Jing
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Xianlong Xie
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China; Department of Critical Medicine, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Xiaoting Liao
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Sheng He
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Jianlan Mo
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Huijun Dai
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Zhaokun Hu
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China
| | - Linghui Pan
- Guangxi Clinical Research Center for Anesthesiology, Nanning 530021, PR China; Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning 530021, PR China; Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning 530021, PR China; Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning 530021, PR China.
| |
Collapse
|
2
|
Chauhan P, Bhardwaj N, Rajaura S, Chandra H, Singh A, Babu R, Gupta NJ. Benzo (A) pyrene exposure alters alveolar epithelial and macrophage cells diversity and induces antioxidant responses in lungs. Toxicol Rep 2024; 13:101777. [PMID: 39506978 PMCID: PMC11539143 DOI: 10.1016/j.toxrep.2024.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
This study was designed to investigate the toxic effects of benzo (a) pyrene (BaP) in the lungs. Mice were repeatedly treated orally with BaP (50 mg/kg body weight, twice a week for four weeks) to induce a tumour. After 4 months of BaP administration, tumours were visible beneath the skin. The histopathological section of the lungs shows congestion of pulmonary blood vessels, alveolar hyperplasia, and concurrent epithelial hyperplasia with infiltrates of inflammatory cells also seen. Thereafter, a single-cell suspension of lung tissues was stained with fluorescently conjugated antibodies for the demarcation of alveolar epithelial (anti-mouse CD74 and podoplanin) and macrophage (F4/80 and CD11b) cells and measured by flow cytometry. The expression of antioxidant genes was assessed by qRT-PCR. The number of alveolar epithelial cells 1 (AEC1) increased, but the number of alveolar epithelial cells 2 (AEC2) and transitional alveolar epithelial cells (TAEC) was significantly decreased in tumour-bearing mice. The proportion of CD11b+ alveolar macrophages (AM) and interstitial macrophages (IM) was increased, but the proportion of F4/80+ AM cells was reduced. The BaP administration significantly increased the ROS production in alveolar cells. The relative expression levels of antioxidant genes (SOD1, catalase, GPX1, and HIF-1α) were increased, but NRF2 expression was decreased in BaP-treated alveolar cells. The expression of anti-inflammatory (NF-κB) was also significantly increased. In conclusion, BaP exposure induced an inflammatory response, altered alveolar epithelial cell and macrophage diversity, and increased antioxidant responses in the lungs.
Collapse
Affiliation(s)
- Pooja Chauhan
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Nitin Bhardwaj
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Sumit Rajaura
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Harish Chandra
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Ashutosh Singh
- Department of Biochemistry, Lucknow University, Lucknow, Uttar Pradesh, India
| | - Ram Babu
- Department of Botany, Kirori Mal College, New Delhi, India
| | - Neelu Jain Gupta
- Department of Zoology, CCS University Campus Meerut, Uttar Pradesh, India
| |
Collapse
|
3
|
Cui X, Fu J. Reinitiating lung development: a novel approach in the management of bronchopulmonary dysplasia. Respir Res 2024; 25:384. [PMID: 39449014 PMCID: PMC11515458 DOI: 10.1186/s12931-024-02996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the predominant chronic lung disease in preterm infants, linked with various adverse long-term outcomes. Multiple prenatal and postnatal risk factors can impede lung development, leading to BPD. Current management of BPD relies heavily on pharmacotherapies and alterations in ventilatory strategies. However, these interventions only mitigate BPD symptoms without addressing underlying alveolar, vascular, structural, and functional deficiencies. Given the retarded lung development in infants with BPD and the limitations of existing modalities, new therapeutic approaches are imperative. The induced differentiation of stem/progenitor cells and the spatiotemporal expression patterns of growth factors associated with lung developmental processes are critical for lung development reactivation in BPD, which focuses on stimulating pulmonary vasculogenesis and alveolarization. This review summarizes the process of lung development and offers a comprehensive overview of advancements in therapies designed to reinitiate lung development in BPD. Furthermore, we assessed the potential of these therapies for maintaining lung homeostasis and effectively restoring pulmonary structure and function through stem/progenitor cells and growth factors, which have been widely researched.
Collapse
Affiliation(s)
- Xuewei Cui
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
4
|
Wang X, Cao L, Liu S, Zhou Y, Zhou J, Zhao W, Gao S, Liu R, Shi Y, Shao C, Fang J. The critical roles of IGFs in immune modulation and inflammation. Cytokine 2024; 183:156750. [PMID: 39243567 DOI: 10.1016/j.cyto.2024.156750] [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: 06/03/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Insulin-like growth factors (IGFs) are crucial for embryonic and postnatal growth and development, influencing cell survival, metabolism, myogenesis, and cancer progression. Many studies have demonstrated that IGFs also play prominent roles in the modulation of both innate and adaptive immune systems during inflammation. Strikingly, IGFs dictate the phenotype and functional properties of macrophages and T cells. Furthermore, the interplay between IGFs and inflammatory cytokines may generate tissue-protective properties during inflammation. Herein, we review the recent advances on the dialogue between immune cells and IGFs, especially zooming in on the significance of immunomodulatory properties in inflammatory conditions, cancer and autoimmune diseases. The investigation of IGFs may have broad clinical implications.
Collapse
Affiliation(s)
- Xin Wang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lijuan Cao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yipeng Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiarui Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wenxuan Zhao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shengqi Gao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yufang Shi
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Changshun Shao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Jiankai Fang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Sha HX, Liu YB, Qiu YL, Zhong WJ, Yang NSY, Zhang CY, Duan JX, Xiong JB, Guan CX, Zhou Y. Neutrophil extracellular traps trigger alveolar epithelial cell necroptosis through the cGAS-STING pathway during acute lung injury in mice. Int J Biol Sci 2024; 20:4713-4730. [PMID: 39309425 PMCID: PMC11414388 DOI: 10.7150/ijbs.99456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/25/2024] [Indexed: 09/25/2024] Open
Abstract
Extensive loss of alveolar epithelial cells (AECs) undergoing necroptosis is a crucial mechanism of acute lung injury (ALI), but its triggering mechanism needs to be thoroughly investigated. Neutrophil extracellular traps (NETs) play a significant role in ALI. However, the effect of NETs on AECs' death has not been clarified. Our study found that intratracheal instillation of NETs disrupted lung tissue structure, suggesting that NETs could induce ALI in mice. Moreover, we observed that NETs could trigger necroptosis of AECs in vivo and in vitro. The phosphorylation levels of RIPK3 and MLKL were increased in MLE12 cells after NETs treatment (P < 0.05). Mechanistically, NETs taken up by AECs through endocytosis activated the cGAS-STING pathway and triggered AECs necroptosis. The expression of cGAS, STING, TBK1 and IRF3 were increased in MLE12 cells treated with NETs (P < 0.05). Furthermore, the cGAS inhibitor RU.521 inhibited NETs-triggered AECs necroptosis and alleviated the pulmonary damage induced by NETs in mice. In conclusion, our study demonstrates that NETs taken up by AECs via endocytosis can activate the cGAS-STING pathway and trigger AECs necroptosis to promote ALI in mice. Our findings indicate that targeting the NETs/cGAS-STING/necroptosis pathway in AECs is an effective strategy for treating ALI.
Collapse
Affiliation(s)
- Han-Xi Sha
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yan-Ling Qiu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jian-Bing Xiong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| |
Collapse
|
6
|
Mou K, Chan SMH, Vlahos R. Musculoskeletal crosstalk in chronic obstructive pulmonary disease and comorbidities: Emerging roles and therapeutic potentials. Pharmacol Ther 2024; 257:108635. [PMID: 38508342 DOI: 10.1016/j.pharmthera.2024.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a multifaceted respiratory disorder characterized by progressive airflow limitation and systemic implications. It has become increasingly apparent that COPD exerts its influence far beyond the respiratory system, extending its impact to various organ systems. Among these, the musculoskeletal system emerges as a central player in both the pathogenesis and management of COPD and its associated comorbidities. Muscle dysfunction and osteoporosis are prevalent musculoskeletal disorders in COPD patients, leading to a substantial decline in exercise capacity and overall health. These manifestations are influenced by systemic inflammation, oxidative stress, and hormonal imbalances, all hallmarks of COPD. Recent research has uncovered an intricate interplay between COPD and musculoskeletal comorbidities, suggesting that muscle and bone tissues may cross-communicate through the release of signalling molecules, known as "myokines" and "osteokines". We explored this dynamic relationship, with a particular focus on the role of the immune system in mediating the cross-communication between muscle and bone in COPD. Moreover, we delved into existing and emerging therapeutic strategies for managing musculoskeletal disorders in COPD. It underscores the development of personalized treatment approaches that target both the respiratory and musculoskeletal aspects of COPD, offering the promise of improved well-being and quality of life for individuals grappling with this complex condition. This comprehensive review underscores the significance of recognizing the profound impact of COPD on the musculoskeletal system and its comorbidities. By unravelling the intricate connections between these systems and exploring innovative treatment avenues, we can aspire to enhance the overall care and outcomes for COPD patients, ultimately offering hope for improved health and well-being.
Collapse
Affiliation(s)
- Kevin Mou
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Huang L, Chen W, Tan Z, Huang Y, Gu X, Liu L, Zhang H, Shi Y, Ding J, Zheng C, Guo Z, Yu B. Mrc1 + macrophage-derived IGF1 mitigates crystal nephropathy by promoting renal tubule cell proliferation via the AKT/Rb signaling pathway. Theranostics 2024; 14:1764-1780. [PMID: 38389846 PMCID: PMC10879870 DOI: 10.7150/thno.89174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/11/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: The present understanding of the cellular characteristics and communications in crystal nephropathy is limited. Here, molecular and cellular studies combined with single-cell RNA sequencing (scRNA-seq) were performed to investigate the changes in cell components and their interactions in glyoxylate-induced crystallized kidneys to provide promising treatments for crystal nephropathy. Methods: The transcriptomes of single cells from mouse kidneys treated with glyoxylate for 0, 1, 4, or 7 days were analyzed via 10× Genomics, and the single cells were clustered and characterized by the Seurat pipeline. The potential cellular interactions between specific cell types were explored by CellChat. Molecular and cellular findings related to macrophage-to-epithelium crosstalk were validated in sodium oxalate (NaOx)-induced renal tubular epithelial cell injury in vitro and in glyoxylate-induced crystal nephropathy in vivo. Results: Our established scRNA atlas of glyoxylate-induced crystalline nephropathy contained 15 cell populations with more than 40000 single cells, including relatively stable tubular cells of different segments, proliferating and injured proximal tubular cells, T cells, B cells, and myeloid and mesenchymal cells. In this study, we found that Mrc1+ macrophages, as a subtype of myeloid cells, increased in both the number and percentage within the myeloid population as crystal-induced injury progresses, and distinctly express IGF1, which induces the activation of a signal pathway to dominate a significant information flow towards injured and proliferating tubule cells. IGF1 promoted the repair of damaged tubular epithelial cells induced by NaOx in vitro, as well as the repair of damaged tubular epithelial cells and the recovery of disease outcomes in glyoxylate-induced nephrolithic mice in vivo. Conclusion: After constructing a cellular atlas of glyoxylate-induced crystal nephropathy, we found that IGF1 derived from Mrc1+ macrophages attenuated crystal nephropathy through promoting renal tubule cell proliferation via the AKT/Rb signaling pathway. These findings could lead to the identification of potential therapeutic targets for the treatment of crystal nephropathy.
Collapse
Affiliation(s)
- Linxi Huang
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Nephrology, PLA Navy No.905 Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Chen
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhuojing Tan
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226006, Jiangsu, China
| | - Yunxiao Huang
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xinji Gu
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lantian Liu
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hongxia Zhang
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yihan Shi
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiarong Ding
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chengjian Zheng
- Faculty of Pharmacy, Naval Medical University, Shanghai, China
| | - Zhiyong Guo
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bing Yu
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
8
|
Wang Y, Wei H, Song Z, Jiang L, Zhang M, Lu X, Li W, Zhao Y, Wu L, Li S, Shen H, Shu Q, Xie Y. Inhalation of panaxadiol alleviates lung inflammation via inhibiting TNFA/TNFAR and IL7/IL7R signaling between macrophages and epithelial cells. J Ginseng Res 2024; 48:77-88. [PMID: 38223829 PMCID: PMC10785239 DOI: 10.1016/j.jgr.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 01/16/2024] Open
Abstract
Background Lung inflammation occurs in many lung diseases, but has limited effective therapeutics. Ginseng and its derivatives have anti-inflammatory effects, but their unstable physicochemical and metabolic properties hinder their application in the treatment. Panaxadiol (PD) is a stable saponin among ginsenosides. Inhalation administration may solve these issues, and the specific mechanism of action needs to be studied. Methods A mouse model of lung inflammation induced by lipopolysaccharide (LPS), an in vitro macrophage inflammation model, and a coculture model of epithelial cells and macrophages were used to study the effects and mechanisms of inhalation delivery of PD. Pathology and molecular assessments were used to evaluate efficacy. Transcriptome sequencing was used to screen the mechanism and target. Finally, the efficacy and mechanism were verified in a human BALF cell model. Results Inhaled PD reduced LPS-induced lung inflammation in mice in a dose-dependent manner, including inflammatory cell infiltration, lung tissue pathology, and inflammatory factor expression. Meanwhile, the dose of inhalation was much lower than that of intragastric administration under the same therapeutic effect, which may be related to its higher bioavailability and superior pharmacokinetic parameters. Using transcriptome analysis and verification by a coculture model of macrophage and epithelial cells, we found that PD may act by inhibiting TNFA/TNFAR and IL7/IL7R signaling to reduce macrophage inflammatory factor-induced epithelial apoptosis and promote proliferation. Conclusion PD inhalation alleviates lung inflammation and pathology by inhibiting TNFA/TNFAR and IL7/IL7R signaling between macrophages and epithelial cells. PD may be a novel drug for the clinical treatment of lung inflammation.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hao Wei
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Department of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhen Song
- Department of Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Liqun Jiang
- Department of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Mi Zhang
- Department of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Xiao Lu
- Shenyang Pharmaceutical University, Shenyang, China
| | - Wei Li
- Shenyang Pharmaceutical University, Shenyang, China
| | - Yuqing Zhao
- Shenyang Pharmaceutical University, Shenyang, China
| | - Lei Wu
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shuxian Li
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huijuan Shen
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Shu
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yicheng Xie
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
9
|
Chen L, Yu T, Zhai Y, Nie H, Li X, Ding Y. Luteolin Enhances Transepithelial Sodium Transport in the Lung Alveolar Model: Integrating Network Pharmacology and Mechanism Study. Int J Mol Sci 2023; 24:10122. [PMID: 37373270 DOI: 10.3390/ijms241210122] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Luteolin (Lut), a natural flavonoid compound existing in Perilla frutescens (L.) Britton, has been proven to play a protective role in the following biological aspects: inflammatory, viral, oxidant, and tumor-related. Lut can alleviate acute lung injury (ALI), manifested mainly by preventing the accumulation of inflammation-rich edematous fluid, while the protective actions of Lut on transepithelial ion transport in ALI were seldom researched. We found that Lut could improve the lung appearance/pathological structure in lipopolysaccharide (LPS)-induced mouse ALI models and reduce the wet/dry weight ratio, bronchoalveolar protein, and inflammatory cytokines. Meanwhile, Lut upregulated the expression level of the epithelial sodium channel (ENaC) in both the primary alveolar epithelial type 2 (AT2) cells and three-dimensional (3D) alveolar epithelial organoid model that recapitulated essential structural and functional aspects of the lung. Finally, by analyzing the 84 interaction genes between Lut and ALI/acute respiratory distress syndrome using GO and KEGG enrichment of network pharmacology, we found that the JAK/STAT signaling pathway might be involved in the network. Experimental data by knocking down STAT3 proved that Lut could reduce the phosphorylation of JAK/STAT and enhance the level of SOCS3, which abrogated the inhibition of ENaC expression induced by LPS accordingly. The evidence supported that Lut could attenuate inflammation-related ALI by enhancing transepithelial sodium transport, at least partially, via the JAK/STAT pathway, which may offer a promising therapeutic strategy for edematous lung diseases.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xin Li
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
10
|
Yue M, Liu Y, Zhang P, Li Z, Zhou Y. Integrative Analysis Reveals the Diverse Effects of 3D Stiffness upon Stem Cell Fate. Int J Mol Sci 2023; 24:9311. [PMID: 37298263 PMCID: PMC10253631 DOI: 10.3390/ijms24119311] [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: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The origin of life and native tissue development are dependent on the heterogeneity of pluripotent stem cells. Bone marrow mesenchymal stem cells (BMMSCs) are located in a complicated niche with variable matrix stiffnesses, resulting in divergent stem cell fates. However, how stiffness drives stem cell fate remains unknown. For this study, we performed whole-gene transcriptomics and precise untargeted metabolomics sequencing to elucidate the complex interaction network of stem cell transcriptional and metabolic signals in extracellular matrices (ECMs) with different stiffnesses, and we propose a potential mechanism involved in stem cell fate decision. In a stiff (39~45 kPa) ECM, biosynthesis of aminoacyl-tRNA was up-regulated, and increased osteogenesis was also observed. In a soft (7~10 kPa) ECM, biosynthesis of unsaturated fatty acids and deposition of glycosaminoglycans were increased, accompanied by enhanced adipogenic/chondrogenic differentiation of BMMSCs. In addition, a panel of genes responding to the stiffness of the ECM were validated in vitro, mapping out the key signaling network that regulates stem cells' fate decisions. This finding of "stiffness-dependent manipulation of stem cell fate" provides a novel molecular biological basis for development of potential therapeutic targets within tissue engineering, from both a cellular metabolic and a biomechanical perspective.
Collapse
Affiliation(s)
- Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
11
|
He S, Tian R, Zhang X, Yao Q, Chen Q, Liu B, Liao L, Gong Y, Yang H, Wang D. PPARγ inhibits small airway remodeling through mediating the polarization homeostasis of alveolar macrophages in COPD. Clin Immunol 2023; 250:109293. [PMID: 36934848 DOI: 10.1016/j.clim.2023.109293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/04/2023] [Accepted: 03/09/2023] [Indexed: 03/19/2023]
Abstract
The role of Peroxisome Proliferator-Activated Receptor-γ (PPARγ) in alveolar macrophages(AMs) polarization homeostasis is closely associated with airway remodeling in COPD, but the definite mechanism remains unclear. In this study, elevated percentage of M1-type AMs and the expression of functionally cytokines were found in COPD patients and mice, which closely related to the disease severity. PPARγ was markedly up-regulated in M2-type AMs and down-regulated in M1-type AMs, and was associated with disease severity in COPD. Co-cultured with M1- or M2-type AMs promoted the epithelial-mesenchymal transition (EMT) of airway epithelial cells and the proliferation of airway smooth muscle cells. Moreover, airway remodeling and functional damage were observed in both IL4R-/- COPD mice with runaway M1-type AMs polarization and TLR4-/- COPD mice with runaway M2-type AMs polarization. Cigarette extract (CS) or lipopolysaccharide (LPS) stimulated PPARγ-/- AMs showed more serious polarization disorder towards M1, as well as CS induced PPARγ-/- COPD mice, which led to more severe airway inflammation, lung function damage, and airway remodeling. Treatment with PPARγ agonist significantly improved the polarization disorder and function activity in CS/LPS stimulated-AMs by inhibiting the JAK-STAT, MAPK and NF-κB pathways, and alleviated the airway inflammation, restored the lung function and suppressed airway remodeling in CS induced-COPD mice. Our research demonstrates that polarization homeostasis of AMs mediated by PPARγ has the protective effect in airway remodeling, and may be a novel therapeutic target for the intervention and treatment of airway remodeling in COPD.
Collapse
Affiliation(s)
- Sirong He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Ruoyuan Tian
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xinying Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Qingmei Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Quan Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Bicui Liu
- Department of Respiratory and Critical Care Medicine, The Bishan Hospital of Chongqing, Chongqing 404000, PR China
| | - Lele Liao
- Department of Respiratory Medicine, The Second Hospital of Jiulongpo District, Chongqing 400050, PR China
| | - Yuxuan Gong
- International medical college, Chongqing Medical University, Chongqing 401334, PR China
| | - Hua Yang
- Respiratory Department, Minda Hospital of Hubei Minzu University, Enshi 445000, PR China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
12
|
Xue Y, Wang M, Han H. Interaction between alveolar macrophages and epithelial cells during Mycoplasma pneumoniae infection. Front Cell Infect Microbiol 2023; 13:1052020. [PMID: 37113130 PMCID: PMC10126420 DOI: 10.3389/fcimb.2023.1052020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
Mycoplasma pneumoniae, as one of the most common pathogens, usually causes upper respiratory tract infections and pneumonia in humans and animals. It accounts for 10% to 40% of community-acquired pneumonia in children. The alveolar epithelial cells (AECs) are the first barrier against pathogen infections, triggering innate immune responses by recruiting and activating immune cells when pathogens invade into the lung. Alveolar macrophages (AMs) are the most plentiful innate immune cells in the lung, and are the first to initiate immune responses with pathogens invasion. The cross-talk between the alveolar epithelium and macrophages is necessary to maintain physiological homeostasis and to eradicate invaded pathogen by regulating immune responses during Mycoplasma pneumoniae infections. This review summarizes the communications between alveolar macrophages and epithelial cells during Mycoplasma pneumoniae infections, including cytokines-medicated communications, signal transduction by extracellular vesicles, surfactant associated proteins-medicated signal transmission and establishment of intercellular gap junction channels.
Collapse
Affiliation(s)
- Yazhi Xue
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mengyao Wang
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hongbing Han
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Mccauley KB, Kukreja K, Jaffe AB, Klein AM. A map of signaling responses in the human airway epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.21.521460. [PMID: 36597531 PMCID: PMC9810218 DOI: 10.1101/2022.12.21.521460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Receptor-mediated signaling plays a central role in tissue regeneration, and it is dysregulated in disease. Here, we build a signaling-response map for a model regenerative human tissue: the airway epithelium. We analyzed the effect of 17 receptor-mediated signaling pathways on organotypic cultures to determine changes in abundance and phenotype of all epithelial cell types. This map recapitulates the gamut of known airway epithelial signaling responses to these pathways. It defines convergent states induced by multiple ligands and diverse, ligand-specific responses in basal-cell and secretory-cell metaplasia. We show that loss of canonical differentiation induced by multiple pathways is associated with cell cycle arrest, but that arrest is not sufficient to block differentiation. Using the signaling-response map, we show that a TGFB1-mediated response underlies specific aberrant cells found in multiple lung diseases and identify interferon responses in COVID-19 patient samples. Thus, we offer a framework enabling systematic evaluation of tissue signaling responses.
Collapse
Affiliation(s)
- Katherine B Mccauley
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Disease Area X, Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Aron B Jaffe
- Disease Area X, Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
- Current address: Chroma Medicine, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Chen R, Cao C, Liu H, Jiang W, Pan R, He H, Ding K, Meng Q. Macrophage Sprouty4 deficiency diminishes sepsis-induced acute lung injury in mice. Redox Biol 2022; 58:102513. [PMID: 36334381 PMCID: PMC9637958 DOI: 10.1016/j.redox.2022.102513] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Inflammation and oxidative stress play critical roles in sepsis-induced acute lung injury (ALI). Sprout4 (Spry4) is involved in regulating inflammation and tissue injury; however, its role and mechanism in sepsis-induced ALI remain elusive. METHODS Macrophage-specific Spry4 knockout (Spry4MKO), transgenic (Spry4MTG) mice and matched control littermates were generated and exposed to cecum ligation and puncture (CLP) surgery to establish bacterial sepsis-induced ALI. Bone marrow-derived macrophages (BMDMs) from Spry4MKO or Spry4MTG mice were isolated and subjected to lipopolysaccharide (LPS) stimulation to further validate the role of Spry4 in vitro. To verify the necessity of AMP-activated protein kinase (AMPK), Spry4 and AMPK double knockout mice and compound C were used in vivo and in vitro. BMDMs were treated with STO-609 to inhibit calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2). RESULTS We found that macrophage Spry4 was increased in CLP mice and positively correlated with sepsis-induced ALI. Macrophage Spry4 deficiency prevented, while macrophage Spry4 overexpression exacerbated sepsis-induced inflammation, oxidative stress and ALI in mice and BMDMs. Mechanistic studies revealed that macrophage Spry4 deficiency alleviated sepsis-induced ALI through activating CaMKK2/AMPK pathway. CONCLUSION Our study identify macrophage Spry4 as a promising predictive and therapeutic target of sepsis-induced ALI.
Collapse
Affiliation(s)
- Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Cao
- Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huimin Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Pan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - He He
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
15
|
Li X, Zhou Y, Yuan S, Zhou X, Wang L, Sun J, Yu L, Zhu J, Zhang H, Yang N, Dai S, Song P, Larsson SC, Theodoratou E, Zhu Y, Li X. Genetically predicted high IGF-1 levels showed protective effects on COVID-19 susceptibility and hospitalization: a Mendelian randomisation study with data from 60 studies across 25 countries. eLife 2022; 11:e79720. [PMID: 36250974 PMCID: PMC9576268 DOI: 10.7554/elife.79720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022] Open
Abstract
Background Epidemiological studies observed gender differences in COVID-19 outcomes, however, whether sex hormone plays a causal in COVID-19 risk remains unclear. This study aimed to examine associations of sex hormone, sex hormones-binding globulin (SHBG), insulin-like growth factor-1 (IGF-1), and COVID-19 risk. Methods Two-sample Mendelian randomization (TSMR) study was performed to explore the causal associations between testosterone, estrogen, SHBG, IGF-1, and the risk of COVID-19 (susceptibility, hospitalization, and severity) using genome-wide association study (GWAS) summary level data from the COVID-19 Host Genetics Initiative (N=1,348,701). Random-effects inverse variance weighted (IVW) MR approach was used as the primary MR method and the weighted median, MR-Egger, and MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) test were conducted as sensitivity analyses. Results Higher genetically predicted IGF-1 levels have nominally significant association with reduced risk of COVID-19 susceptibility and hospitalization. For one standard deviation increase in genetically predicted IGF-1 levels, the odds ratio was 0.77 (95% confidence interval [CI], 0.61-0.97, p=0.027) for COVID-19 susceptibility, 0.62 (95% CI: 0.25-0.51, p=0.018) for COVID-19 hospitalization, and 0.85 (95% CI: 0.52-1.38, p=0.513) for COVID-19 severity. There was no evidence that testosterone, estrogen, and SHBG are associated with the risk of COVID-19 susceptibility, hospitalization, and severity in either overall or sex-stratified TSMR analysis. Conclusions Our study indicated that genetically predicted high IGF-1 levels were associated with decrease the risk of COVID-19 susceptibility and hospitalization, but these associations did not survive the Bonferroni correction of multiple testing. Further studies are needed to validate the findings and explore whether IGF-1 could be a potential intervention target to reduce COVID-19 risk. Funding We acknowledge support from NSFC (LR22H260001), CRUK (C31250/A22804), SHLF (Hjärt-Lungfonden, 20210351), VR (Vetenskapsrådet, 2019-00977), and SCI (Cancerfonden).
Collapse
Affiliation(s)
- Xinxuan Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yajing Zhou
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Shuai Yuan
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska InstitutetStockholmSweden
| | - Xuan Zhou
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Lijuan Wang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Jing Sun
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Lili Yu
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Jinghan Zhu
- The Second School of Clinical Medicine, Southern Medical UniversityGuangzhouChina
| | - Han Zhang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Nan Yang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Shuhui Dai
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Peige Song
- School of Public Health and Women's Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska InstitutetStockholmSweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala UniversityUppsalaSweden
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Yimin Zhu
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
16
|
Yan T, Li X, Nian T, Zhang X, He B, Bi K, Wang Z. Salidroside Inhibits Ischemia/Reperfusion-Induced Myocardial Apoptosis by Targeting Mir-378a-3p Via the Igf1r/Pi3k/Akt Signaling Pathway. Transplant Proc 2022; 54:1970-1983. [PMID: 35927086 DOI: 10.1016/j.transproceed.2022.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/03/2022] [Accepted: 05/22/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The present study aimed to investigate the protective effects and mechanism of salidroside (SAL) on hypoxia/reoxygenation (H/R)-induced cardiomyocyte apoptosis and myocardial ischemia/reperfusion (I/R) injury. METHODS We set up an H/R H9c2 cell model in vitro and an I/R rat model in vivo. Cell viability, apoptosis and histopathologic evaluation were conducted. RESULTS The cell viability of H/R-induced cardiomyocytes was increased by pretreatment of SAL, whereas the release of lactate dehydrogenase, reactive oxygen species production, and apoptosis were decreased accompanied with reduced Cleaved-caspase-3 and Bax, and increased Bcl-2 expressions. The SAL restored mitochondrial membrane potential both in vitro and in vivo, and improved electrocardiographic abnormality, and attenuated myocardial apoptosis and injury in I/R-induced rats. The transfection of miR-378a-3p inhibitor counteracted the effects of SAL-induced increase of cell viability and decrease of cell apoptosis and mitochondrial membrane potential. SAL reduced the expression of insulin-like growth factor 1 receptor (IGF1R), and increased the expressions of PI3K and Akt, however, these alterations were blocked by miR-378a-3p inhibitor. CONCLUSIONS miR-378a-3p might participate in the protective effect of SAL in I/R-induced myocardial apoptosis via the IGF1R/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Tingxu Yan
- Jiangsu Kangyuan Pharmaceutical Co, Ltd, Lianyungang, China; School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Xu Li
- Jiangsu Kangyuan Pharmaceutical Co, Ltd, Lianyungang, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Lianyungang, China
| | - Tingting Nian
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaozhuo Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bosai He
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| | - Zhenzhong Wang
- Jiangsu Kangyuan Pharmaceutical Co, Ltd, Lianyungang, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Lianyungang, China.
| |
Collapse
|
17
|
Feizollahi P, Matin S, Roghani SA, Mostafaei S, Safarzadeh E, Taghadosi M. Evaluation serum levels of Insulin Growth Factor-1 (IGF-1) and its association with clinical parameters in severe COVID-19. Inflammopharmacology 2022; 30:199-205. [PMID: 35098386 PMCID: PMC8801278 DOI: 10.1007/s10787-021-00908-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022]
Abstract
Background Severe coronavirus disease-2019 (COVID-19) is associated with dysregulated immune response and extreme inflammatory injury. Considering the role of insulin growth factor-1 (IGF-1) in immune-mediated and inflammatory reactions, this study was conducted to investigate the IGF-1 contribution to the pathogenesis of severe form of COVID-19. Material and methods Sixty-two patients with severe COVID-19 and 52 healthy subjects were enrolled in this study. The serum levels of IGF-1 were measured using a solid-phase enzyme-linked chemiluminescent immunoassay on an Immulite 2000 system (Siemens Healthcare Diagnostics. Result The serum levels of IGF-1 had no significant difference in COVID-19 patients compared to the healthy subjects (p = 0.359). There was a positive correlation between IGF-1 and age in the severe COVID-19 patients, while a negative correlation was observed for the serum levels of IGF-1 and age in the control group (r = 0.364, p = 0.036, r = − 0.536, p = 0.001, respectively). Moreover, IGF-1 was remarkably associated with hypertension, neurogenic disease, shock, and nausea in patients with the severe form of COVID-19 (p = 0.031, p = 0.044, p = 0.01, p = 0.03, respectively). Conclusion Our results pointed to the complex role of IGF-1 in the severe form of COVID-19, and its association with clinical parameters, and some risk factors in the severe form of COVID-19.
Collapse
Affiliation(s)
- Parisa Feizollahi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Somaieh Matin
- Department of Internal Medicine, Emam Khomeini Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
- Gastrointestinal and Liver Disease Research Center, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Askar Roghani
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shayan Mostafaei
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mahdi Taghadosi
- Department of Immunology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
18
|
Yang Y, Wang Y. Autocrine, Paracrine, and Endocrine Signals That Can Alter Alveolar Macrophages Function. Rev Physiol Biochem Pharmacol 2022; 186:177-198. [PMID: 36472676 DOI: 10.1007/112_2022_76] [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] [Indexed: 12/12/2022]
Abstract
Alveolar macrophages (AMs) are extremely versatile cells with complex functions involved in health or diseases such as pneumonia, asthma, and pulmonary alveolar proteinosis. In recent years, it has been widely identified that the different functions and states of macrophages are the results from the complex interplay between microenvironmental signals and macrophage lineage. Diverse and complicated signals to which AMs respond are mentioned when they are described individually or in a particular state of AMs. In this review, the microenvironmental signals are divided into autocrine, paracrine, and endocrine signals based on their secreting characteristics. This new perspective on classification provides a more comprehensive and systematic introduction to the complex signals around AMs and is helpful for understanding the roles of AMs affected by physiological environment. The existing possible treatments of AMs are also mentioned in it. The thorough understanding of AMs signals modulation may be contributed to the development of more effective therapies for AMs-related lung diseases.
Collapse
Affiliation(s)
- Yue Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
19
|
Peng J, Fan B, Bao C, Jing C. JMJD3 deficiency alleviates lipopolysaccharide‑induced acute lung injury by inhibiting alveolar epithelial ferroptosis in a Nrf2‑dependent manner. Mol Med Rep 2021; 24:807. [PMID: 34542160 DOI: 10.3892/mmr.2021.12447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/23/2021] [Indexed: 11/06/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a deadly illness which presents with severe hypoxemia as well as diffuse alveolar damage. Jumonji domain‑containing 3 (JMJD3), which belongs to the UTX/UTY JmjC‑domain protein subfamily, is involved in infection, development, aging and immune disorders. However, the role of JMJD3 in acute lung injury (ALI) is still unclear. The present study explored the roles and potential mechanisms of JMJD3 in ALI. Alveolar epithelial cell‑specific knockout of JMJD3 mice and A549 alveolar epithelial cells were used to investigate the function of JMJD3 in ALI. Lipopolysaccharide (LPS) was used to establish an in vivo and in vitro ALI model. The expression of JMJD3 in murine lung tissue and alveolar epithelial cells was detected. Pathological injury of lung tissue and alveolar epithelial cells was also investigated following inhibition of JMJD3. The results showed that JMJD3 expression was significantly increased in murine lung tissues and in A549 cells following LPS stimulation. JMJD3‑deficient mice in alveolar epithelial cells exhibited alleviated lung pathological injury and ferroptosis following h stimulation. Mechanistically, it was found that JMJD3 knockout could increase the expression of nuclear factor erythroid‑2‑related factor‑2 (Nrf2) in lung tissues challenged with h. However, Nrf2 overexpression by adenovirus could further enhance the anti‑ferroptotic effect from JMJD3 silence in h‑treated A549 cells. Taken together, the present study revealed that JMJD3 deficiency may relieve LPS‑induced ALI by blocking alveolar epithelial ferroptosis in a Nrf2‑dependent manner, which may serve as a novel therapeutic target against ALI.
Collapse
Affiliation(s)
- Junwei Peng
- Department of Cardiothoracic Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Bin Fan
- Department of Cardiothoracic Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Chuanming Bao
- Department of Cardiothoracic Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Chen Jing
- Department of Cardiothoracic Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| |
Collapse
|
20
|
Croasdell Lucchini A, Gachanja NN, Rossi AG, Dorward DA, Lucas CD. Epithelial Cells and Inflammation in Pulmonary Wound Repair. Cells 2021; 10:339. [PMID: 33562816 PMCID: PMC7914803 DOI: 10.3390/cells10020339] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/15/2021] [Accepted: 01/30/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory diseases are frequently characterised by epithelial injury, airway inflammation, defective tissue repair, and airway remodelling. This may occur in a subacute or chronic context, such as asthma and chronic obstructive pulmonary disease, or occur acutely as in pathogen challenge and acute respiratory distress syndrome (ARDS). Despite the frequent challenge of lung homeostasis, not all pulmonary insults lead to disease. Traditionally thought of as a quiescent organ, emerging evidence highlights that the lung has significant capacity to respond to injury by repairing and replacing damaged cells. This occurs with the appropriate and timely resolution of inflammation and concurrent initiation of tissue repair programmes. Airway epithelial cells are key effectors in lung homeostasis and host defence; continual exposure to pathogens, toxins, and particulate matter challenge homeostasis, requiring robust defence and repair mechanisms. As such, the epithelium is critically involved in the return to homeostasis, orchestrating the resolution of inflammation and initiating tissue repair. This review examines the pivotal role of pulmonary airway epithelial cells in initiating and moderating tissue repair and restitution. We discuss emerging evidence of the interactions between airway epithelial cells and candidate stem or progenitor cells to initiate tissue repair as well as with cells of the innate and adaptive immune systems in driving successful tissue regeneration. Understanding the mechanisms of intercellular communication is rapidly increasing, and a major focus of this review includes the various mediators involved, including growth factors, extracellular vesicles, soluble lipid mediators, cytokines, and chemokines. Understanding these areas will ultimately identify potential cells, mediators, and interactions for therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | | | - Christopher D. Lucas
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh Bioquarter, Edinburgh EH16 4TJ, UK; (A.C.L.); (N.N.G.); (A.G.R.); (D.A.D.)
| |
Collapse
|