1
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
2
|
Jain KG, Liu Y, Zhao R, Muire PJ, Zhang J, Zang QS, Ji HL. Humanized L184Q Mutated Surfactant Protein C Gene Alters Alveolar Type 2 Epithelial Cell Fate. Int J Mol Sci 2024; 25:8723. [PMID: 39201410 PMCID: PMC11354303 DOI: 10.3390/ijms25168723] [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: 06/13/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Alveolar type 2 epithelial (AT2) cells synthesize surfactant protein C (SPC) and repair an injured alveolar epithelium. A mutated surfactant protein C gene (SftpcL184Q, Gene ID: 6440) in newborns has been associated with respiratory distress syndrome and pulmonary fibrosis. However, the underlying mechanisms causing Sftpc gene mutations to regulate AT2 lineage remain unclear. We utilized three-dimensional (3D) feeder-free AT2 organoids in vitro to simulate the alveolar epithelium and compared AT2 lineage characteristics between WT (C57BL/6) and SftpcL184Q mutant mice using colony formation assays, immunofluorescence, flow cytometry, qRT-PCR, and Western blot assays. The AT2 numbers were reduced significantly in SftpcL184Q mice. Organoid numbers and colony-forming efficiency were significantly attenuated in the 3D cultures of primary SftpcL184Q AT2 cells compared to those of WT mice. Podoplanin (PDPN, Alveolar type 1 cell (AT1) marker) expression and transient cell count was significantly increased in SftpcL184Q organoids compared to in the WT mice. The expression levels of CD74, heat shock protein 90 (HSP90), and ribosomal protein S3A1 (RPS3A1) were not significantly different between WT and SftpcL184Q AT2 cells. This study demonstrated that humanized SftpcL184Q mutation regulates AT2 lineage intrinsically. This regulation is independent of CD74, HSP90, and RPS3A1 pathways.
Collapse
Affiliation(s)
- Krishan G. Jain
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Yang Liu
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Runzhen Zhao
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Preeti J. Muire
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Department of Orthopedics and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL 60153, USA;
- Departments of Pathology and Radiation Oncology, Loyola University Medical Center, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Hong-Long Ji
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
3
|
Tanese K, Ogata D. The role of macrophage migration inhibitory factor family and CD74 in the pathogenesis of melanoma. Exp Dermatol 2024; 33:e15122. [PMID: 38884501 DOI: 10.1111/exd.15122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Melanoma is an aggressive tumour with poor prognosis that arises from the malignant transformation of melanocytes. Over the past few decades, intense research into the pathogenesis of melanoma has led to the development of BRAF and immune checkpoint inhibitors, including antibodies against programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which have shown clinically significant efficacy. However, some tumours do not respond to these therapies initially or become treatment resistant. Most melanoma tissues appear to possess biological characteristics that allow them to evade these treatments, and identifying these characteristics is one of the major challenges facing cancer researchers. One such characteristic that has recently gained attention is the role of macrophage migration inhibitory factor (MIF) and its receptor CD74. This review outlines the cellular and molecular functions of CD74, MIF and their family of proteins. We then review their roles in tumours based on previous reports, highlight their pathological significance in melanoma and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keiji Tanese
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
4
|
Westmeier J, Brochtrup A, Paniskaki K, Karakoese Z, Werner T, Sutter K, Dolff S, Limmer A, Mittermüller D, Liu J, Zheng X, Koval T, Kaidashev I, Berger MM, Herbstreit F, Brenner T, Witzke O, Trilling M, Lu M, Yang D, Babel N, Westhoff T, Dittmer U, Zelinskyy G. Macrophage migration inhibitory factor receptor CD74 expression is associated with expansion and differentiation of effector T cells in COVID-19 patients. Front Immunol 2023; 14:1236374. [PMID: 37946732 PMCID: PMC10631787 DOI: 10.3389/fimmu.2023.1236374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused millions of COVID-19 cases and deaths worldwide. Severity of pulmonary pathologies and poor prognosis were reported to be associated with the activation non-virus-specific bystander T cells. In addition, high concentrations of the macrophage migration inhibitory factor (MIF) were found in serum of COVID-19 patients. We hypothesized that these two pathogenic factors might be related and analyzed the expression of receptors for MIF on T cells in COVID-19. T cells from PBMCs of hospitalized patients with mild and severe COVID-19 were characterized. A significantly higher proportion of CD4+ and CD8+ T cells from COVID-19 patients expressed CD74 on the cell surface compared to healthy controls. To induce intracellular signaling upon MIF binding, CD74 forms complexes with CD44, CXCR2, or CXCR4. The vast majority of CD74+ T cells expressed CD44, whereas expression of CXCR2 and CXCR4 was low in controls but increased upon SARS-CoV-2 infection. Hence, T cells in COVID-19 patients express receptors that render them responsive to MIF. A detailed analysis of CD74+ T cell populations revealed that most of them had a central memory phenotype early in infection, while cells with an effector and effector memory phenotype arose later during infection. Furthermore, CD74+ T cells produced more cytotoxic molecules and proliferation markers. Our data provide new insights into the MIF receptor and co-receptor repertoire of bystander T cells in COVID-19 and uncovers a novel and potentially druggable aspect of the immunological footprint of SARS-CoV-2.
Collapse
Affiliation(s)
- Jaana Westmeier
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annika Brochtrup
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Krystallenia Paniskaki
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Zehra Karakoese
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Werner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Limmer
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Pediatric Heart Surgery, Friedrich-Alexander- Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Mittermüller
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jia Liu
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xin Zheng
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Tetiana Koval
- Department of Infectious Diseases with Epidemiology, Poltava State Medical University, Poltava, Ukraine
| | - Igor Kaidashev
- Department of Internal Medicine №3 with Phthisiology, Poltava State Medical University, Poltava, Ukraine
| | - Marc Moritz Berger
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Frank Herbstreit
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Dongliang Yang
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Nina Babel
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Timm Westhoff
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University of Bochum, Herne, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| |
Collapse
|
5
|
Zhu BH, Lai HH, Wei CR, Shen Z, Sun Y, Zhu F, Wu GS. [Effects and mechanism of annexin A1-overexpressing human adipose-derived mesenchymal stem cells in the treatment of mice with acute respiratory distress syndrome]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2023; 39:456-464. [PMID: 37805755 DOI: 10.3760/cma.j.cn501225-20220408-00130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
Objective: To explore the effects and mechanism of annexin A1 (ANXA1)-overexpressing human adipose-derived mesenchymal stem cells (AMSCs) in the treatment of mice with acute respiratory distress syndrome (ARDS). Methods: The experimental study method was adopted. After the adult AMSCs were identified by flow cytometry, the 3rd passage cells were selected for the follow-up experiments. According to the random number table (the same grouping method below), the cells were divided into ANXA1-overexpressing group transfected with plasmid containing RNA sequences of ANXA1 gene and no-load control group transfected with the corresponding no-load plasmid. The other cells were divided into ANXA1-knockdown group transfected with plasmid containing small interfering RNA sequences of ANXA1 gene and no-load control group transfected with the corresponding no-load plasmid. At post transfection hour (PTH) 72, the fluorescence expression was observed under a fluorescence microscope imaging system, and the protein and mRNA expressions of ANXA1 were detected by Western blotting and real-time fluorescence quantitative reverse transcription polymerase chain reaction respectively (with the sample numbers being 3). Fifty male C57BL/6J mice aged 6-8 weeks were divided into sham injury group, ARDS alone group, normal cell group, ANXA1-overexpressing group, and ANXA1-knockdown group, with 10 mice in each group. Mice in the last 4 groups were treated with endotoxin/lipopolysaccharide to make ARDS lung injury model, and mice in sham injury group were simulated to cause false injury. Immediately after injury, mice in sham injury group and ARDS alone group were injected with normal saline through the tail vein, while mice in normal cell group, ANXA1-overexpressing group, and ANXA1-knockdown group were injected with normal AMSCs, ANXA1-overexpressing AMSCs, and ANXA1-knockdown AMSCs, correspondingly. At post injection hour (PIH) 24, 5 mice in each group were selected, the Evans blue staining was performed to observe the gross staining of the right lung tissue, and the absorbance value of bronchoalveolar lavage fluid (BALF) supernatant of left lung was detected by microplate reader to evaluate the pulmonary vascular permeability. Three days after injection, the remaining 5 mice in each group were taken, the right lung tissue was collected for hematoxylin-eosin staining to observe the pathological changes and immunohistochemical staining to observe the CD11b and F4/80 positive macrophages, and the levels of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), and IL-1β in BALF supernatant of left lung were determined by enzyme-linked immunosorbent assay. Data were statistically analyzed with paired sample t test, one-way analysis of variance, and least significant difference test. Results: At PTH 72, AMSCs in both ANXA1-overexpressing group and ANXA1-knockdown group expressed higher fluorescence intensity than AMSCs in corresponding no-load control group, respectively. At PTH 72, compared with those in corresponding no-load control group, the protein and mRNA expressions of ANXA1 in ANXA1-overexpressing group were significantly increased (wth t values of 249.80 and 6.56, respectively, P<0.05), while the protein and mRNA expressions of ANXA1 in ANXA1-knockdown group were significantly decreased (wth t values of 176.50 and 18.18, respectively, P<0.05). At PIH 24, compared with those in sham injury group (with the absorbance value of BALF supernatant being 0.041±0.009), the lung tissue of mice in ARDS alone group was obviously blue-stained and the absorbance value of BALF supernatant (0.126±0.022) was significantly increased (P<0.05). Compared with those in ARDS alone group, the degree of blue-staining in lung tissue of mice was significantly reduced in normal cell group or ANXA1-overexpressing group, and the absorbance values of BALF supernatant (0.095±0.020 and 0.069±0.015) were significantly decreased (P<0.05), but the degree of blue-staining in lung tissue and the absorbance value of BALF supernatant (0.109±0.016, P>0.05) of mice in ANXA1-knockdown group had no significant change. Compared with that in normal cell group, the absorbance value of BALF supernatant of mice in ANXA1-overexpressing group was significantly decreased (P<0.05). Three days after injection, the lung tissue structure of mice in ARDS alone group was significantly damaged compared with that in sham injury group. Compared with those in ARDS alone group, hemorrhage, infiltration of inflammatory cells, alveolar collapse, and interstitial widening in the lung tissue of mice were significantly alleviated in normal cell group and ANXA1-overexpressing group, while no significant improvement of above-mentioned lung tissue manifestation was observed in ANXA1-knockdown group. Three days after injection, the numbers of CD11b and F4/80 positive macrophages in the lung tissue of mice in ARDS alone group were significantly increased compared with those in sham injury group. Compared with those in ARDS alone group, the numbers of CD11b and F4/80 positive macrophages in lung tissue of mice in normal cell group, ANXA1-overexpressing group, and ANXA1-knockdown group reduced, with the most significant reduction in ANXA1-overexpressing group. Three days after injection, compared with those in sham injury group, the levels of TNF-α, IL-6, and IL-1β in BALF supernatant of mice in ARDS alone group were significantly increased (P<0.05). Compared with those in ARDS alone group, the levels of TNF-α, IL-6, and IL-1β in BALF supernatant of mice in normal cell group and ANXA1-overexpressing group, as well as the level of IL-1β in BALF supernatant of mice in ANXA1-knockdown group were significantly decreased (P<0.05). Compared with that in normal cell group, the level of TNF-α in BALF supernatant of mice was significantly decreased in ANXA1-overexpressing group (P<0.05) but significantly increased in ANXA1-knockdown group (P<0.05). Conclusions: Overexpression of ANXA1 can optimize the efficacy of AMSCs in treating ARDS and enhance the effects of these cells in inhibiting inflammatory response and improving pulmonary vascular permeability, thereby alleviating lung injury of mice with ARDS.
Collapse
Affiliation(s)
- B H Zhu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - H H Lai
- Department of Burn Injury, Zhujiang Hospital Affiliated to Southern Medical University, Guangzhou 510280, China
| | - C R Wei
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Z Shen
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Y Sun
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - F Zhu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - G S Wu
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| |
Collapse
|
6
|
Zhao CQ, Wang C, Liu MM, Cao M, Peng J, Kong DQ, Ren XT, Liu R, Hai CX, Zhang XD. Single-cell transcriptomes reveal heterogeneity of chlorine-induced mice acute lung injury and the inhibitory effect of pentoxifylline on ferroptosis. Sci Rep 2023; 13:6833. [PMID: 37100790 PMCID: PMC10131515 DOI: 10.1038/s41598-023-32093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
To investigate the effect of pentoxifylline (PTX) on Chlorine (Cl2)-induced acute lung injury (ALI) by single-cell RNA sequencing (scRNA-seq). Female BALB/c mice were exposed to Cl2 at 400 ppm for 15 min. H&E staining was used to observe the degree of lung injury. scRNA-seq was conducted to analysis of normal and Cl2-exposed mice lung tissues. Immunofluorescence was used to observe genes of interest. Thirty-two mice were randomly divided into four groups: Control, Cl2, Cl2+Fer-1, Cl2+PTX. TEM, WB and ELISA were used to detect ferroptosis-related indicators. The 5, 8, 10, 12, 16, 20 clusters were epithelial cells and 4, 15, 18, 19, 21 clusters were endothelial cells. Pseudo-time analysis revealed the differentiation trajectory of epithelial cells and key regulatory genes (Gclc, Bpifa1, Dnah5 and Dnah9) during the process of injury. Cell-cell communication analysis identified several important receptor-ligand complexes (Nrp1-Vegfa, Nrp2-Vegfa, Flt1-Vegfa and Flt4-Vegfa). Ferroptosis were found up-regulated in epithelial and endothelial cells by GSVA analysis. Highly expressed genes to which closely related ferroptosis were found by SCENIC analysis. PTX could significantly decrease the levels of MDA and abnormal high expression of solute carrier family 7 member 11 (SLC7A11, the key transporter of cystine) as well as increase the expression of GSH/GSSG and glutathione peroxidase 4 (GPX4) (p < 0.05). This study revealed novel molecular features of Cl2-induced ALI. PTX may be a potential specific drug by inhibiting the process of ferroptosis in epithelial and endothelial cells.
Collapse
Affiliation(s)
- Chen-Qian Zhao
- Department of Medical Experiment Center, Shaanxi University of Chinese Medicine, Xi'an, 712046, Xianyang, China
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Chong Wang
- Department of Medical Experiment Center, Shaanxi University of Chinese Medicine, Xi'an, 712046, Xianyang, China
| | - Meng-Meng Liu
- Department of Health Service, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, China
| | - Meng Cao
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - De-Qin Kong
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Xiao-Ting Ren
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Rui Liu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Chun-Xu Hai
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China.
| | - Xiao-di Zhang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
7
|
Shin JJ, Fan W, Par-Young J, Piecychna M, Leng L, Israni-Winger K, Qing H, Gu J, Zhao H, Schulz WL, Unlu S, Kuster J, Young G, Liu J, Ko AI, Baeza Garcia A, Sauler M, Wisnewski AV, Young L, Orduña A, Wang A, Klementina O, Garcia AB, Hegyi P, Armstrong ME, Mitchell P, Ordiz DB, Garami A, Kang I, Bucala R. MIF is a common genetic determinant of COVID-19 symptomatic infection and severity. QJM 2023; 116:205-212. [PMID: 36222594 PMCID: PMC9620729 DOI: 10.1093/qjmed/hcac234] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Genetic predisposition to coronavirus disease 2019 (COVID-19) may contribute to its morbidity and mortality. Because cytokines play an important role in multiple phases of infection, we examined whether commonly occurring, functional polymorphisms in macrophage migration inhibitory factor (MIF) are associated with COVID-19 infection or disease severity. AIM To determine associations of common functional polymorphisms in MIF with symptomatic COVID-19 or its severity. METHODS This retrospective case-control study utilized 1171 patients with COVID-19 from three tertiary medical centers in the USA, Hungary and Spain, together with a group of 637 pre-pandemic, healthy control subjects. Functional MIF promoter alleles (-794 CATT5-8,rs5844572), serum MIF and soluble MIF receptor levels, and available clinical characteristics were measured and correlated with COVID-19 diagnosis and hospitalization. Experimental mice genetically engineered to express human high- or low-expression MIF alleles were studied for response to coronavirus infection. RESULTS In patients with COVID-19, there was a lower frequency of the high-expression MIF CATT7 allele when compared to healthy controls [11% vs. 19%, odds ratio (OR) 0.54 [0.41-0.72], P < 0.0001]. Among inpatients with COVID-19 (n = 805), there was a higher frequency of the MIF CATT7 allele compared to outpatients (n = 187) (12% vs. 5%, OR 2.87 [1.42-5.78], P = 0.002). Inpatients presented with higher serum MIF levels when compared to outpatients or uninfected healthy controls (87 ng/ml vs. 35 ng/ml vs. 29 ng/ml, P < 0.001, respectively). Among inpatients, circulating MIF concentrations correlated with admission ferritin (r = 0.19, P = 0.01) and maximum CRP (r = 0.16, P = 0.03) levels. Mice with a human high-expression MIF allele showed more severe disease than those with a low-expression MIF allele. CONCLUSIONS In this multinational retrospective study of 1171 subjects with COVID-19, the commonly occurring -794 CATT7MIF allele is associated with reduced susceptibility to symptomatic SARS-CoV-2 infection but increased disease progression as assessed by hospitalization. These findings affirm the importance of the high-expression CATT7MIF allele, which occurs in 19% of the population, in different stages of COVID-19 infection.
Collapse
Affiliation(s)
- Junghee J Shin
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | - Wei Fan
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | | | - Marta Piecychna
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | - Lin Leng
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | | | - Hua Qing
- Department of Medicine, Department of Immunobiology, New Haven, CT, USA
| | - Jianlei Gu
- Department of Pathology, New Haven, CT, USA
| | | | - Wade L Schulz
- Department of Medicine, Department of Immunobiology, New Haven, CT, USA
| | - Serhan Unlu
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | - John Kuster
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | | | - Jian Liu
- Department of Pathology, New Haven, CT, USA
| | | | | | - Maor Sauler
- Pulmonary, Critical Care, and Sleep Medicine, New Haven, CT, USA
| | | | | | - Antonio Orduña
- Microbiology Service. Hospital Clínico Universtario. Valladolid. Spain
| | - Andrew Wang
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
- Department of Medicine, Department of Immunobiology, New Haven, CT, USA
| | - Ocskay Klementina
- Universidad de Valladolid, Valladolid, Spain; University of Pécs, Pécs, Hungary. Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest Hungary
| | - Antonio Blesa Garcia
- Mucosal Immunology Lab. Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid-CSIC. Valladolid. Spain
| | - Peter Hegyi
- Universidad de Valladolid, Valladolid, Spain; University of Pécs, Pécs, Hungary. Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest Hungary
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | | | - David Bernardo Ordiz
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Centro de Investigaciones Biomédicas en Red de Enfermedades infecciosas (CIBERinfec). Madrid. Spain
| | - András Garami
- Universidad de Valladolid, Valladolid, Spain; University of Pécs, Pécs, Hungary. Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Insoo Kang
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
| | - Richard Bucala
- Sections of Rheumatology, Allergy and Immunology, New Haven, CT, USA
- Department of Pathology, New Haven, CT, USA
- Yale Schools of Medicine and Public Health, New Haven, CT, USA
| |
Collapse
|
8
|
Li QL, Tang J, Zhao L, Ruze A, Shan XF, Gao XM. The role of CD74 in cardiovascular disease. Front Cardiovasc Med 2023; 9:1049143. [PMID: 36712241 PMCID: PMC9877307 DOI: 10.3389/fcvm.2022.1049143] [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: 09/20/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Leukocyte differentiation antigen 74 (CD74), also known as invariant chain, is a molecular chaperone of major histocompatibility complex class II (MHC II) molecules involved in antigen presentation. CD74 has recently been shown to be a receptor for the macrophage migration inhibitory factor family proteins (MIF/MIF2). Many studies have revealed that CD74 plays an important role in cardiovascular disease. In this review, we summarize the structure and main functions of CD74 and then focus on the recent research progress on the role of CD74 in cardiovascular diseases. In addition, we also discuss potential treatment strategies that target CD74. Our systematic review of the role of CD74 in cardiovascular disease will fill some knowledge gaps in the field.
Collapse
Affiliation(s)
- Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Clinical Medical Research Institute of Xinjiang Medical University, Ürümqi, China,*Correspondence: Xiao-Ming Gao,
| |
Collapse
|
9
|
Hallmarks of Cancer Affected by the MIF Cytokine Family. Cancers (Basel) 2023; 15:cancers15020395. [PMID: 36672343 PMCID: PMC9856758 DOI: 10.3390/cancers15020395] [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: 11/25/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
New diagnostic methods and treatments have significantly decreased the mortality rates of cancer patients, but further improvements are warranted based on the identification of novel tumor-promoting molecules that can serve as therapeutic targets. The macrophage migration inhibitory factor (MIF) family of cytokines, comprising MIF and DDT (also known as MIF2), are overexpressed in almost all cancer types, and their high expressions are related to a worse prognosis for the patients. MIF is involved in 9 of the 10 hallmarks of cancer, and its inhibition by antibodies, nanobodies, or small synthetic molecules has shown promising results. Even though DDT is also proposed to be involved in several of the hallmarks of cancer, the available information about its pro-tumoral role and mechanism of action is more limited. Here, we provide an overview of the involvement of both MIF and DDT in cancer, and we propose that blocking both cytokines is needed to obtain the maximum anti-tumor response.
Collapse
|
10
|
Zan C, Yang B, Brandhofer M, El Bounkari O, Bernhagen J. D-dopachrome tautomerase in cardiovascular and inflammatory diseases-A new kid on the block or just another MIF? FASEB J 2022; 36:e22601. [PMID: 36269019 DOI: 10.1096/fj.202201213r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) as well as its more recently described structural homolog D-dopachrome tautomerase (D-DT), now also termed MIF-2, are atypical cytokines and chemokines with key roles in host immunity. They also have an important pathogenic role in acute and chronic inflammatory conditions, cardiovascular diseases, lung diseases, adipose tissue inflammation, and cancer. Although our mechanistic understanding of MIF-2 is relatively limited compared to the extensive body of evidence available for MIF, emerging data suggests that MIF-2 is not only a functional phenocopy of MIF, but may have differential or even oppositional activities, depending on the disease and context. In this review, we summarize and discuss the similarities and differences between MIF and MIF-2, with a focus on their structures, receptors, signaling pathways, and their roles in diseases. While mainly covering the roles of the MIF homologs in cardiovascular, inflammatory, autoimmune, and metabolic diseases, we also discuss their involvement in cancer, sepsis, and chronic obstructive lung disease (COPD). A particular emphasis is laid upon potential mechanistic explanations for synergistic or cooperative activities of the MIF homologs in cancer, myocardial diseases, and COPD as opposed to emerging disparate or antagonistic activities in adipose tissue inflammation, metabolic diseases, and atherosclerosis. Lastly, we discuss potential future opportunities of jointly targeting MIF and MIF-2 in certain diseases, whereas precision targeting of only one homolog might be preferable in other conditions. Together, this article provides an update of the mechanisms and future therapeutic avenues of human MIF proteins with a focus on their emerging, surprisingly disparate activities, suggesting that MIF-2 displays a variety of activities that are distinct from those of MIF.
Collapse
Affiliation(s)
- Chunfang Zan
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Bishan Yang
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Markus Brandhofer
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Omar El Bounkari
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Jürgen Bernhagen
- Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany.,Deutsches Zentrum für Herz-Kreislauferkrankungen (DZHK), Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
11
|
Cui YR, Qu F, Zhong WJ, Yang HH, Zeng J, Huang JH, Liu J, Zhang MY, Zhou Y, Guan CX. Beneficial effects of aloperine on inflammation and oxidative stress by suppressing necroptosis in lipopolysaccharide-induced acute lung injury mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154074. [PMID: 35397283 DOI: 10.1016/j.phymed.2022.154074] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 02/22/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
RATIONALE Alveolar epithelial cell death, inflammation, and oxidative stress are typical features of acute lung injury (ALI). Aloperine (Alo), an alkaloid isolated from Sophora alopecuroides, has been reported to display various biological effects, such as anti-inflammatory, immunoregulatory, and anti-oxidant properties. In this study, we investigated the effects and mechanisms of Alo in treating a lipopolysaccharide (LPS)-induced ALI in a murine model. METHODS The effects of Alo in LPS-induced ALI were investigated in C57BL/6 mice. The RIPK1 inhibitor (Nec-1) and the RIPK3 inhibitor (GSK'872) were used to evaluate the relationship of necroptosis, NF-κB activation, and PDC subunits in LPS-treated mouse alveolar epithelial cells (MLE-12). Then the effects of Alo on necroptosis, inflammation, and oxidative stress of LPS-stimulated MLE-12 cells were evaluated. RESULTS Alo significantly attenuated histopathological lung injuries and reduced lung wet/dry ratio in LPS-induced ALI mice. Alo also remarkedly reduced total protein and neutrophils recruitment in bronchoalveolar lavage fluid of ALI mice. Meanwhile, Alo ameliorated the LPS-induced necroptosis in the lungs of ALI mice. The RIPK3 inhibitor GSK'872, but not the RIPK1 inhibitor Nec-1, reversed LPS-induced p65 phosphorylation and translocation to the nucleus in MLE-12 cells. GSK'872 also reversed the LPS-induced increase in ROS and binding of RIPK3 and PDC subunits in MLE-12 cells. Moreover, Alo down-regulated the levels of p-RIPK1, p-RIPK3, p-MLKL, p-p65, the translocation of p65 to the nucleus, and reduced the expression of IL-6 and IL-8 in LPS-stimulated MLE-12 cells. Alo also inhibited the binding of RIPK3 and PDC-E1α, PDC-E1β, PDC-E2, and PDC-E3 and the ROS production in LPS-treated MLE-12 cells. CONCLUSION The present study validated the beneficial effects of Alo on LPS-induced ALI , suggesting Alo may be a new drug candidate against ALI.
Collapse
Affiliation(s)
- Yan-Ru Cui
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China; Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Fei Qu
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Jie Zeng
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jun-Hao Huang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jie Liu
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Ming-Yue Zhang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
12
|
Fukuda Y, Bustos MA, Cho SN, Roszik J, Ryu S, Lopez VM, Burks JK, Lee JE, Grimm EA, Hoon DSB, Ekmekcioglu S. Interplay between soluble CD74 and macrophage-migration inhibitory factor drives tumor growth and influences patient survival in melanoma. Cell Death Dis 2022; 13:117. [PMID: 35121729 PMCID: PMC8816905 DOI: 10.1038/s41419-022-04552-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/12/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Soluble forms of receptors play distinctive roles in modulating signal-transduction pathways. Soluble CD74 (sCD74) has been identified in sera of inflammatory diseases and implicated in their pathophysiology; however, few relevant data are available in the context of cancer. Here we assessed the composition and production mechanisms, as well as the clinical significance and biological properties, of sCD74 in melanoma. Serum sCD74 levels were significantly elevated in advanced melanoma patients compared with normal healthy donors, and the high ratio of sCD74 to macrophage-migration inhibitory factor (MIF) conferred significant predictive value for prolonged survival in these patients (p = 0.0035). Secretion of sCD74 was observed primarily in melanoma cell lines as well as a THP-1 line of macrophages from monocytes and primary macrophages, especially in response to interferon-γ (IFN-γ). A predominant form that showed clinical relevance was the 25-KDa sCD74, which originated from the 33-KDa isoform of CD74. The release of this sCD74 was regulated by either a disintegrin and metalloproteinase-mediated cell-surface cleavage or cysteine-protease-mediated lysosomal cleavage, depending on cell types. Both recombinant and THP-1 macrophage-released endogenous sCD74 suppressed melanoma cell growth and induced apoptosis under IFN-γ stimulatory conditions via inhibiting the MIF/CD74/AKT-survival pathway. Our findings demonstrate that the interplay between sCD74 and MIF regulates tumor progression and determines patient outcomes in advanced melanoma.
Collapse
Affiliation(s)
- Yasunari Fukuda
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matias A Bustos
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Sung-Nam Cho
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jason Roszik
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Suyeon Ryu
- Department of Genome Sequencing, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Victor M Lopez
- Department of Genome Sequencing, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dave S B Hoon
- Department of Genome Sequencing, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
CD74 in Apoptotic Macrophages Is Associated with Inflammation, Plaque Progression and Clinical Manifestations in Human Atherosclerotic Lesions. Metabolites 2022; 12:metabo12010054. [PMID: 35050177 PMCID: PMC8781814 DOI: 10.3390/metabo12010054] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The aim of this study was to investigate whether CD74 levels in atherosclerotic lesions are associated with inflammation, apoptosis, plaque severity, and clinical symptoms among patients with carotid atherosclerosis. We further studied whether CD74 expression is associated with apoptosis in macrophages induced by 7ketocholesterol (7keto). Sixty-one carotid samples (39 males and 22 females) were immunostained with macrophages, smooth muscle cells, CD74, ferritin, TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling), and thrombin receptors. Double immunocytochemistry of CD74 and caspase 3 or CD74 and Annexin V was performed on THP-1 macrophages exposed to 7keto. In human carotid plaques, CD74 expression is lesion-dependently increased and is associated with necrotic core formation and plaque rupture, clinical symptoms, macrophage apoptosis, ferritin, and thrombin receptors. CD74 levels were inversely correlated to high-density lipoproteins and statin treatment, and positively correlated to triglycerides. In THP-1 macrophages, 7keto induced a significant increase in levels of CD74, ferritin, and apoptotic cell death. This study suggests that CD74 in apoptotic macrophages is linked to inflammation and thrombosis in progression of human atherosclerotic plaques, lipid metabolism, and clinical manifestation in atherosclerosis. Surface CD74 in apoptotic macrophages and ferritin production induced by oxidized lipids may contribute to inflammation and plaque vulnerability in atherosclerosis.
Collapse
|
14
|
Li L, Qu M, Yang L, Liu J, Wang Q, Zhong P, Zeng Y, Wang T, Xiao H, Liu D, Huang X, Wang J, Zhou J. Effects of Ultrashort Wave Therapy on Inflammation and Macrophage Polarization after Acute Lung Injury in Rats. Bioelectromagnetics 2021; 42:464-472. [PMID: 34130351 DOI: 10.1002/bem.22353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 03/25/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) features dysregulated pulmonary inflammation. Ultrashort waves (USWs) exert anti-inflammatory effects but no studies have evaluated their activity in ALI. Herein, we used an in vivo lipopolysaccharide (LPS)-induced ALI model to investigate whether the anti-inflammatory activity of USWs is mediated by altering the polarization of M1 to M2 macrophages. Twenty-four male Sprague-Dawley rats were randomly divided into control, untreated ALI, and ALI treated with USW groups (n = 8 in each group). ALI was induced by intratracheal LPS instillation. Rats in the USW group were treated for 15 min at 0, 4, and 8 h after a single LPS intratracheal instillation. Histopathologic examination, wet/dry lung weight ratio, enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction, and western blot analyses were performed to evaluate the degree of lung injury and to determine macrophage phenotypes. Histopathologic examination disclosed attenuation of ALI, with reduced alveolar hemorrhage and neutrophilic infiltration in the USW group. Serum levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were significantly decreased after USW therapy. Moreover, the messenger RNA (mRNA) expressions of TNF-α and IL-1β were significantly decreased in the USW group, whereas the mRNA expression of Arginase 1 (Arg1) and the protein expression of mannose receptor significantly increased in comparison with the untreated ALI group. We conclude that USW therapy may attenuate inflammation in LPS-induced ALI through the modulation of macrophage polarization. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Lan Li
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Mengjian Qu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Lu Yang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jing Liu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Qian Wang
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Peirui Zhong
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Yahua Zeng
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Ting Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Hao Xiao
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Danni Liu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Xiarong Huang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jinling Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jun Zhou
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| |
Collapse
|
15
|
Wirtz TH, Reuken PA, Jansen C, Fischer P, Bergmann I, Backhaus C, Emontzpohl C, Reißing J, Brandt EF, Koenen MT, Schneider KM, Schierwagen R, Brol MJ, Chang J, Zimmermann HW, Köse-Vogel N, Eggermann T, Kurth I, Stoppe C, Bucala R, Bernhagen J, Praktiknjo M, Stallmach A, Trautwein C, Trebicka J, Bruns T, Berres ML. Balance between macrophage migration inhibitory factor and sCD74 predicts outcome in patients with acute decompensation of cirrhosis. JHEP Rep 2020; 3:100221. [PMID: 33659891 PMCID: PMC7890204 DOI: 10.1016/j.jhepr.2020.100221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background & Aims Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine and an important regulator of innate immune responses. We hypothesised that serum concentrations of MIF are associated with disease severity and outcome in patients with decompensated cirrhosis and acute-on-chronic liver failure (ACLF). Methods Circulating concentrations of MIF and its soluble receptor CD74 (sCD74) were determined in sera from 292 patients with acute decompensation of cirrhosis defined as new onset or worsening of ascites requiring hospitalisation. Of those, 78 (27%) had ACLF. Short-term mortality was assessed 90 days after inclusion. Results Although serum concentrations of MIF and sCD74 did not correlate with liver function parameters or ACLF, higher MIF (optimum cut-off >2.3 ng/ml) and lower concentrations of sCD74 (optimum cut-off <66.5 ng/ml) both indicated poorer 90-day transplant-free survival in univariate analyses (unadjusted hazard ratio [HR] 2.01 [1.26-3.22]; p = 0.004 for MIF; HR 0.59 [0.38-0.92]; p = 0.02 for sCD74) and after adjustment in multivariable models. Higher MIF concentrations correlated with surrogates of systemic inflammation (white blood cells, p = 0.005; C-reactive protein, p = 0.05) and were independent of genetic MIF promoter polymorphisms. Assessment of MIF plasma concentrations in portal venous blood and matched blood samples from the right atrium in a second cohort of patients undergoing transjugular intrahepatic portosystemic shunt insertion revealed a transhepatic MIF gradient with higher concentrations in the right atrial blood. Conclusions Serum concentrations of MIF and its soluble receptor CD74 predict 90-day transplant-free survival in patients with acute decompensation of cirrhosis. This effect was independent of liver function and genetic predispositions, but rather reflected systemic inflammation. Therefore, MIF and sCD74 represent promising prognostic markers beyond classical scoring systems in patients at risk of ACLF. Lay summary Inflammatory processes contribute to the increased risk of death in patients with cirrhosis and ascites. We show that patients with high serum levels of the inflammatory cytokine macrophage migration inhibitory factor (MIF) alongside low levels of its binding receptor sCD74 in blood indicate an increased mortality risk in patients with ascites. The cirrhotic liver is a relevant source of elevated circulating MIF levels.
Collapse
Key Words
- ACLF, acute-on-chronic liver failure
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Acute-on-chronic liver failure
- Biomarker
- CRP, C-reactive protein
- CXCL10, C-X-C motif chemokine
- HCC, hepatocellular carcinoma
- Inflammation
- Liver cirrhosis
- MELD, model for end-stage liver disease
- MIF, macrophage migration inhibitory factor
- SBP, spontaneous bacterial peritonitis
- SDC, stable decompensated cirrhosis
- SHR, subdistribution hazard ratio
- SNP, single nucleotide polymorphism
- Survival
- TIPS, transjugular intrahepatic portosystemic shunt
- UDC, unstable decompensated cirrhosis
- WBC, white blood cell count
- sCD74, soluble receptor CD74
Collapse
Affiliation(s)
- Theresa H Wirtz
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Philipp A Reuken
- Department of Internal Medicine IV, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Christian Jansen
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Petra Fischer
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Irina Bergmann
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Christina Backhaus
- Institute of Human Genetics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Christoph Emontzpohl
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, Mc Govern Medical School, Houston, TX, USA
| | - Johanna Reißing
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Elisa F Brandt
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - M Teresa Koenen
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Kai M Schneider
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Robert Schierwagen
- Department of Internal Medicine 1, University Hospital, Goethe University, Frankfurt, Germany
| | - Maximilian J Brol
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Johannes Chang
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Henning W Zimmermann
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Nilay Köse-Vogel
- Department of Internal Medicine IV, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Thomas Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Christian Stoppe
- Department of Intensive Care Medicine, University Hospital Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig Maximilians-University (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (EXC 2145 SyNergy), Munich, Germany
| | | | - Andreas Stallmach
- Department of Internal Medicine IV, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Jonel Trebicka
- Department of Internal Medicine 1, University Hospital, Goethe University, Frankfurt, Germany
| | - Tony Bruns
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
16
|
Zhu B, Wu G, Wang C, Xiao Y, Jin J, Wang K, Jiang Y, Sun Y, Ben D, Xia Z. Soluble cluster of differentiation 74 regulates lung inflammation through the nuclear factor-κB signaling pathway. Immunobiology 2020; 225:152007. [PMID: 32962825 DOI: 10.1016/j.imbio.2020.152007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/31/2020] [Accepted: 08/11/2020] [Indexed: 11/30/2022]
Abstract
The soluble form of the migration inhibitory factor receptor cluster of differentiation 74 (sCD74) has previously been shown to be elevated during the development of acute lung injury (ALI) in mice. However, the biological role of increased sCD74 in ALI remans poorly understood. Synthesized recombinant sCD74 protein was administered to mice intratracheally. Pro-inflammatory genes in lung tissue and the inflammatory factors in bronchoalveolar lavage fluid (BALF) were analyzed using RT-PCR and ELISA, respectively. Additionally, RAW264.7, A549, and human umbilical vein endothelial cells (HUVEC) were treated with sCD74, and the resulting pro-inflammatory factor protein and gene expression levels were analyzed in the supernatants and cell lysates. Meanwhile, the level of nuclear factor (NF)-κB components in cell lysates after stimulating macrophages with sCD74 was also assessed. After administration of 0.5 mg/kg body weight sCD74 to mice, the expression of Tnfa, Mip2, and Il6 increased in lung tissues after 2 h by 2.1-, 3.4-, and 2.8-fold, respectively. Moreover, the BALF concentrations of TNF-α and MIP-2 reached maximal levels of 560 and 107 pg/mL at 8 h compared to those in the saline group, respectively. Similarly, TNFA, MIP2, and IL6 expression increased by 4.0-, 11.8-, and 2.6-fold, respectively, 2 h after stimulating macrophages with 1000 ng/mL sCD74. The levels of phospho-IκB and phospho-p65 were also significantly increased in the cytoplasm and nucleus of macrophages following sCD74 stimulation. Taken together, these results suggest that sCD74 is a critical cellular factor involved in the lung acute inflammatory response through nuclear factor-κB signaling.
Collapse
Affiliation(s)
- Banghui Zhu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Guosheng Wu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Chen Wang
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Yongqiang Xiao
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Jian Jin
- Department of Burn and Plastic Surgery, 903 Hospital of PLA, Hangzhou, 313000, Zhengjiang Province, PR China.
| | - Kangan Wang
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Yong Jiang
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Yu Sun
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Daofeng Ben
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| | - Zhaofan Xia
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, PR China.
| |
Collapse
|
17
|
Prevention of Oxygen-Induced Inflammatory Lung Injury by Caffeine in Neonatal Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3840124. [PMID: 32831996 PMCID: PMC7429812 DOI: 10.1155/2020/3840124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/26/2022]
Abstract
Background Preterm birth implies an array of respiratory diseases including apnea of prematurity and bronchopulmonary dysplasia (BPD). Caffeine has been introduced to treat apneas but also appears to reduce rates of BPD. Oxygen is essential when treating preterm infants with respiratory problems but high oxygen exposure aggravates BPD. This experimental study is aimed at investigating the action of caffeine on inflammatory response and cell death in pulmonary tissue in a hyperoxia-based model of BPD in the newborn rat. Material/Methods. Lung injury was induced by hyperoxic exposure with 80% oxygen for three (P3) or five (P5) postnatal days with or without recovery in ambient air until postnatal day 15 (P15). Newborn Wistar rats were treated with PBS or caffeine (10 mg/kg) every two days beginning at the day of birth. The effects of caffeine on hyperoxic-induced pulmonary inflammatory response were examined at P3 and P5 immediately after oxygen exposure or after recovery in ambient air (P15) by immunohistological staining and analysis of lung homogenates by ELISA and qPCR. Results Treatment with caffeine significantly attenuated changes in hyperoxia-induced cell death and apoptosis-associated factors. There was a significant decrease in proinflammatory mediators and redox-sensitive transcription factor NFκB in the hyperoxia-exposed lung tissue of the caffeine-treated group compared to the nontreated group. Moreover, treatment with caffeine under hyperoxia modulated the transcription of the adenosine receptor (Adora)1. Caffeine induced pulmonary chemokine and cytokine transcription followed by immune cell infiltration of alveolar macrophages as well as increased adenosine receptor (Adora1, 2a, and 2b) expression. Conclusions The present study investigating the impact of caffeine on the inflammatory response, pulmonary cell degeneration and modulation of adenosine receptor expression, provides further evidence that caffeine acts as an antioxidative and anti-inflammatory drug for experimental oxygen-mediated lung injury. Experimental studies may broaden the understanding of therapeutic use of caffeine in modulating detrimental mechanisms involved in BPD development.
Collapse
|
18
|
Associations of Serological Biomarkers of sICAM-1, IL-1 β, MIF, and su-PAR with 3-Month Mortality in Acute Exacerbation of Idiopathic Pulmonary Fibrosis. Mediators Inflamm 2020; 2020:4534272. [PMID: 32694925 PMCID: PMC7362287 DOI: 10.1155/2020/4534272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/30/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Objective To investigate prognostic values of serum biomarkers of soluble intercellular adhesion molecule 1 (sICAM-1), macrophage migration inhibitor factor (MIF), interleukin 1β (IL-1β), and soluble urokinase plasminogen activator receptor (su-PAR) in patients with acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF). Methods From August 2017 to November 2019, 122 consecutive IPF patients treated in our center were classified as stable IPF and AE-IPF based on the newly published international guidelines. Serum levels of four biomarkers at admission were measured by the enzyme-linked immunosorbent assay (ELISA). The primary endpoint was 3-month mortality. The log-rank test and logistic regression analysis were used to evaluate the effects of these biomarkers for survival in patients with AE-IPF. Cox proportional hazards analysis was performed to evaluate the prognostic values of serological biomarkers and clinical data. Results Eighty-one patients were diagnosed with stable IPF, and 41 AE-IPF patients were enrolled in the study. Serum levels of sICAM-1 (p < 0.001), IL-1β (p < 0.001), MIF (p < 0.001), and su-PAR (p < 0.001) in patients with IPF were significantly increased compared to those in healthy controls. All the four biomarkers were elevated in patients with AE-IPF compared to those with stable IPF. The 3-month mortality in AE-IPF was 56.1% (23/41). Increased levels of MIF (p = 0.01) and IL-1β (>5 pg/mL, p = 0.033) were independent risk factors for 3-month mortality in patients with AE-IPF. Conclusions We showed the higher serum levels of IL-1β, and MIF had prognostic values for 3-month mortality in AE-IPF. This study provided a clue to promote our understanding in the pathogenesis of the disease.
Collapse
|
19
|
Cheng WL, Kao YH, Chen YC, Lin YK, Chen SA, Chen YJ. Macrophage migration inhibitory factor increases atrial arrhythmogenesis through CD74 signaling. Transl Res 2020; 216:43-56. [PMID: 31669150 DOI: 10.1016/j.trsl.2019.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 09/04/2019] [Accepted: 10/02/2019] [Indexed: 01/23/2023]
Abstract
Macrophage migration inhibitory factor (MIF), a pleiotropic inflammatory cytokine, is highly expressed in patients with atrial fibrillation (AF). CD74 (major histocompatibility complex, class II invariant chain) is the main receptor for MIF. However, the role of the MIF/CD74 axis in atrial arrhythmogenesis is unclear. In this study, we investigated the effects of MIF/CD74 signaling on atrial electrophysiological characteristics and determined its underlying mechanisms. Confocal fluorescence microscopy, patch clamp, and western blot analysis were used to study calcium homeostasis, ionic currents, and calcium-related signaling in MIF-treated HL-1 atrial cardiomyocytes with or without anti-CD74 neutralized antibodies treatment. Furthermore, electrocardiographic telemetry recording and echocardiography were obtained from mice treated with MIF. Compared with controls, MIF-treated HL-1 myocytes had increased calcium transients, sarcoplasmic reticulum (SR) calcium content, Na+/Ca2+ exchanger (NCX) efflux rate, calcium leak, transient outward potassium current, and ultra-rapid delayed rectifier potassium current. Furthermore, MIF could induce expression of SR Ca2+ATPase, NCX, phosphorylation of ryanodine receptor 2 (RyR2), and activation of calcium/calmodulin kinase II (CaMKII) when compared with control cells. MIF-mediated electrical dysregulation and CaMKII-RyR2 signaling activation were attenuated through blocking of CD74. Moreover, MIF-injected mice had lesser left atrium fractional shortening, greater atrial fibrosis, and atrial ectopic beats than control (nonspecific immunoglobulin treated) or MIF combined with anti-CD74 neutralized antibody-treated mice. Consequently, our study on MIF/CD74 signaling has pointed out a new potential therapeutic intervention of AF patients with MIF elevation.
Collapse
Affiliation(s)
- Wan-Li Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ann Chen
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
20
|
Abu El-Asrar AM, Ahmad A, Siddiquei MM, De Zutter A, Allegaert E, Gikandi PW, De Hertogh G, Van Damme J, Opdenakker G, Struyf S. The Proinflammatory and Proangiogenic Macrophage Migration Inhibitory Factor Is a Potential Regulator in Proliferative Diabetic Retinopathy. Front Immunol 2019; 10:2752. [PMID: 31866994 PMCID: PMC6904364 DOI: 10.3389/fimmu.2019.02752] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
The macrophage migration inhibitory factor (MIF)/CD74 signaling pathway is strongly implicated in inflammation and angiogenesis. We investigated the expression of MIF and its receptor CD74 in proliferative diabetic retinopathy (PDR) to reveal a possible role of this pathway in the pathogenesis of PDR. Levels of MIF, soluble (s)CD74, soluble intercellular adhesion molecule-1 (sICAM-1) and vascular endothelial growth factor (VEGF) were significantly increased in the vitreous from patients with PDR compared to nondiabetic control samples. We detected significant positive correlations between the levels of MIF and the levels of sICAM-1 (r = 0.43; p = 0.001) and VEGF (r = 0.7; p < 0.001). Through immunohistochemical analysis of PDR epiretinal membranes, significant positive correlations were also found between microvessel density (CD31 expression) and the numbers of blood vessels expressing MIF (r = 0.56; p = 0.045) and stromal cells expressing MIF (r = 0.79; p = 0.001) and CD74 (r = 0.59; p = 0.045). Similar to VEGF, MIF was induced in Müller cells cultured under hypoxic conditions and MIF induced phosphorylation of ERK1/2 and VEGF production in Müller cells. Intravitreal administration of MIF in normal rats induced increased retinal vascular permeability and significant upregulation of phospho-ERK1/2, NF-κB, ICAM-1 and vascular cell adhesion molecule-1 expression in the retina. MIF induced migration and proliferation of human retinal microvascular endothelial cells. These results suggest that MIF/CD74 signaling is involved in PDR angiogenesis.
Collapse
Affiliation(s)
- Ahmed M Abu El-Asrar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ajmal Ahmad
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Alexandra De Zutter
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Eef Allegaert
- Laboratory of Histochemistry and Cytochemistry, KU Leuven, Leuven, Belgium
| | - Priscilla W Gikandi
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Gert De Hertogh
- Laboratory of Histochemistry and Cytochemistry, KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Autoimmune antibodies correlate with immune checkpoint therapy-induced toxicities. Proc Natl Acad Sci U S A 2019; 116:22246-22251. [PMID: 31611368 DOI: 10.1073/pnas.1908079116] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint (IC) therapy provides substantial benefits to cancer patients but can also cause distinctive toxicities termed immune-related adverse events (irAEs). Biomarkers to predict toxicities will be necessary to improve management of patients receiving IC therapy. We relied on serological analysis of recombinant cDNA expression libraries to evaluate plasma samples from patients treated with IC therapy and identified autoantibodies, both in pretreatment and on-treatment samples prior to the development of irAEs, which correlate with the development of immune-related hypophysitis (anti-GNAL and anti-ITM2B autoantibodies) and pneumonitis (anti-CD74 autoantibody). We developed an enzyme-linked immunosorbent assay and tested additional patient samples to confirm our initial findings. Collectively, our data suggest that autoantibodies may correlate with irAEs related to IC therapy, and specific autoantibodies may be detected early for the management of irAEs.
Collapse
|
22
|
Stoppe C, Averdunk L, Goetzenich A, Soppert J, Marlier A, Kraemer S, Vieten J, Coburn M, Kowark A, Kim BS, Marx G, Rex S, Ochi A, Leng L, Moeckel G, Linkermann A, El Bounkari O, Zarbock A, Bernhagen J, Djudjaj S, Bucala R, Boor P. The protective role of macrophage migration inhibitory factor in acute kidney injury after cardiac surgery. Sci Transl Med 2019; 10:10/441/eaan4886. [PMID: 29769287 DOI: 10.1126/scitranslmed.aan4886] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/22/2017] [Accepted: 04/13/2018] [Indexed: 12/20/2022]
Abstract
Acute kidney injury (AKI) represents the most frequent complication after cardiac surgery. Macrophage migration inhibitory factor (MIF) is a stress-regulating cytokine that was shown to protect the heart from myocardial ischemia-reperfusion injury, but its role in the pathogenesis of AKI remains unknown. In an observational study, serum and urinary MIF was quantified in 60 patients scheduled for elective conventional cardiac surgery with the use of cardiopulmonary bypass. Cardiac surgery triggered an increase in MIF serum concentrations, and patients with high circulating MIF (>median) 12 hours after surgery had a significantly reduced risk of developing AKI (relative risk reduction, 72.7%; 95% confidence interval, 12 to 91.5%; P = 0.03). Experimental AKI was induced in wild-type and Mif-/- mice by 30 min of ischemia followed by 6 or 24 hours of reperfusion, or by rhabdomyolysis. Mif-deficient mice exhibited increased tubular cell injury, increased regulated cell death (necroptosis and ferroptosis), and enhanced oxidative stress. Therapeutic administration of recombinant MIF after ischemia-reperfusion in mice ameliorated AKI. In vitro treatment of tubular epithelial cells with recombinant MIF reduced cell death and oxidative stress as measured by glutathione and thiobarbituric acid reactive substances in the setting of hypoxia. Our data provide evidence of a renoprotective role of MIF in experimental ischemia-reperfusion injury by protecting renal tubular epithelial cells, consistent with our observation that high MIF in cardiac surgery patients is associated with a reduced incidence of AKI.
Collapse
Affiliation(s)
- Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany.
| | - Luisa Averdunk
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany.,Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Arnaud Marlier
- Department of Nephrology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jil Vieten
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Ana Kowark
- Department of Anesthesiology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Bong-Song Kim
- Department of Plastic and Reconstructive Surgery, Hand Surgery, Burn Center, RWTH Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Akinobu Ochi
- Department of Nephropathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gilbert Moeckel
- Department of Nephropathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Critical Care Medicine and Pain Therapy, University Hospital Münster, Münster, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (EXC 1010 SyNergy), Munich, Germany
| | - Sonja Djudjaj
- Institute of Pathology and Department of Nephrology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital, RWTH Aachen, Aachen, Germany. .,Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
23
|
Soppert J, Kraemer S, Beckers C, Averdunk L, Möllmann J, Denecke B, Goetzenich A, Marx G, Bernhagen J, Stoppe C. Soluble CD74 Reroutes MIF/CXCR4/AKT-Mediated Survival of Cardiac Myofibroblasts to Necroptosis. J Am Heart Assoc 2018; 7:e009384. [PMID: 30371153 PMCID: PMC6201423 DOI: 10.1161/jaha.118.009384] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/09/2018] [Indexed: 01/03/2023]
Abstract
Background Although macrophage migration inhibitory factor ( MIF ) has been demonstrated to mediate cardioprotection in ischemia/reperfusion injury and antagonize fibrotic effects through its receptor, CD 74, the function of the soluble CD 74 receptor ectodomain ( sCD 74) and its interaction with circulating MIF have not been explored in cardiac disease. Methods and Results Cardiac fibroblasts were isolated from hearts of neonatal mice and differentiated into myofibroblasts. Co-treatment with recombinant MIF and sCD 74 induced cell death ( P<0.001), which was mediated by receptor-interacting serine/threonine-protein kinase ( RIP) 1/ RIP 3-dependent necroptosis ( P=0.0376). This effect was specific for cardiac fibroblasts and did not affect cardiomyocytes. Gene expression analyses using microarray and RT - qPCR technology revealed a 4-fold upregulation of several interferon-induced genes upon co-treatment of myofibroblasts with sCD 74 and MIF (Ifi44: P=0.011; Irg1: P=0.022; Clec4e: P=0.011). Furthermore, Western blot analysis confirmed the role of sCD 74 as a modulator of MIF signaling by diminishing MIF -mediated protein kinase B ( AKT) activation ( P=0.0197) and triggering p38 activation ( P=0.0641). We obtained evidence that sCD 74 inhibits MIF -mediated survival pathway through the C-X-C chemokine receptor 4/ AKT axis, enabling the induction of CD 74-dependent necroptotic processes in cardiac myofibroblasts. Preliminary clinical data revealed a lowered sCD 74/ MIF ratio in heart failure patients (17.47±10.09 versus 1.413±0.6244). Conclusions These findings suggest that treatment of cardiac myofibroblasts with sCD 74 and MIF induces necroptosis, offering new insights into the mechanism of myofibroblast depletion during scar maturation. Preliminary clinical data provided first evidence about a clinical relevance of the sCD 74/ MIF axis in heart failure, suggesting that these proteins may be a promising target to modulate cardiac remodeling and disease progression in heart failure.
Collapse
Affiliation(s)
- Josefin Soppert
- Department of Intensive Care MedicineUniversity HospitalRWTH AachenAachenGermany
- Department of Thoracic, Cardiac and Vascular SurgeryUniversity HospitalRWTH AachenAachenGermany
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular SurgeryUniversity HospitalRWTH AachenAachenGermany
| | - Christian Beckers
- Department of Thoracic, Cardiac and Vascular SurgeryUniversity HospitalRWTH AachenAachenGermany
| | - Luisa Averdunk
- Department of Intensive Care MedicineUniversity HospitalRWTH AachenAachenGermany
| | - Julia Möllmann
- Department of Cardiology, Pneumology, Angiology and Internal Intensive CareUniversity HospitalRWTH AachenAachenGermany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research (IZKF)University HospitalRWTH AachenAachenGermany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular SurgeryUniversity HospitalRWTH AachenAachenGermany
| | - Gernot Marx
- Department of Intensive Care MedicineUniversity HospitalRWTH AachenAachenGermany
| | - Jürgen Bernhagen
- Department of Vascular BiologyInstitute for Stroke and Dementia Research (ISD)Ludwig‐Maximilians‐University (LMU) MunichMunichGermany
- German Center for Cardiovascular Research (DZHK)partner site Munich Heart AllianceMunichGermany
- Munich Cluster for Systems Neurology (EXC 1010 SyNergy)MunichGermany
| | - Christian Stoppe
- Department of Intensive Care MedicineUniversity HospitalRWTH AachenAachenGermany
| |
Collapse
|
24
|
Tang X, Sun L, Wang G, Chen B, Luo F. RUNX1: A Regulator of NF-kB Signaling in Pulmonary Diseases. Curr Protein Pept Sci 2018; 19:172-178. [PMID: 28990531 PMCID: PMC5876917 DOI: 10.2174/1389203718666171009111835] [Citation(s) in RCA: 28] [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: 06/30/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 02/05/2023]
Abstract
Runt-related transcription factor 1 (RUNX1), a member of the RUNX family, is one of the key regulatory proteins in vertebrates. RUNX1 is involved in embryonic development, hematopoiesis, angiogenesis, tumorigenesis and immune response. In the past few decades, studies mainly focused on the effect of RUNX1 on acute leukemia and cancer. Only few studies about the function of RUNX1 in the pathological process of pulmonary diseases have been reported. Recent studies have demonstrated that RUNX1 is highly expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung and that it plays a critical role in the lipopolysaccharide induced lung inflammation by regulating the NF-kB pathway. RUNX1 participates in the regulation of the NF-kB signaling pathway through interaction with IkB kinase complex in the cytoplasm or interaction with the NF-kB subunit P50. NF-kB is well-known signaling pathway necessary for inflammatory response in the lung. This review is to highlight the RUNX1 structure, isoforms and to present the mechanism that RUNX1 regulates NF-kB. This will illustrate the great potential role of RUNX1 in the inflammation signaling pathway in pulmonary diseases.
Collapse
Affiliation(s)
- Xiaoju Tang
- Department of Pulmonary and Critical Care Medicine
| | - Ling Sun
- Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- Department of Pulmonary and Critical Care Medicine
| | - Bojiang Chen
- Department of Pulmonary and Critical Care Medicine
| | - Fengming Luo
- Department of Pulmonary and Critical Care Medicine
| |
Collapse
|
25
|
Tang X, Sun L, Jin X, Chen Y, Zhu H, Liang Y, Wu Q, Han X, Liang J, Liu X, Liang Z, Wang G, Luo F. Runt-Related Transcription Factor 1 Regulates LPS-Induced Acute Lung Injury via NF-κB Signaling. Am J Respir Cell Mol Biol 2017; 57:174-183. [PMID: 28314106 DOI: 10.1165/rcmb.2016-0319oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Runt-related transcription factor 1 (RUNX1), a transcription factor expressed in multiple organs, plays important roles in embryonic development and hematopoiesis. Although RUNX1 is highly expressed in pulmonary tissues, its roles in lung function and homeostasis are unknown. We sought to assess the role of RUNX1 in lung development and inflammation after LPS challenge. Expression of RUNX1 was assessed in the developing and postnatal lung. RUNX1 was conditionally deleted in pulmonary epithelial cells. Pulmonary maturation was evaluated in the developing and postnatal lung, and lung inflammation was investigated in adult mice after LPS challenge. Interactions between RUNX1 and inflammatory signaling via NF-κB-IkB kinase β were assessed in vitro. RUNX1 was expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung. The RUNX1 gene was efficiently deleted from respiratory epithelial cells producing Runx1∆/∆ mice. Although lung maturation was delayed, Runx1∆/∆ mice survived postnatally and subsequent growth and maturation of the lung proceeded normally. Increased respiratory distress, inflammation, and proinflammatory cytokines were observed in the Runx1-deleted mice after pulmonary LPS exposure. RUNX1 deletion was associated with the activation of NF-κB in respiratory epithelial cells. RUNX1 was required for the suppression of NF-κB signaling pathway via inhibition of IkB kinase β in in vitro studies. RUNX1 plays a critical role in the lung inflammation after LPS-induced injury.
Collapse
Affiliation(s)
- Xiaoju Tang
- 1 Department of Respiratory Medicine.,2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Ling Sun
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Xiaodong Jin
- 3 Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hui Zhu
- 1 Department of Respiratory Medicine
| | - Yasha Liang
- 4 First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingbo Wu
- 1 Department of Respiratory Medicine
| | - Xing Han
- 5 Fourth People's Hospital of Sichuan Province, Chengdu, China
| | - Jianing Liang
- 6 Department of Respiratory Medicine, Fourth Military Medical University, Xian, China
| | - Xiaojing Liu
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | | | - Gang Wang
- 1 Department of Respiratory Medicine
| | | |
Collapse
|
26
|
Tilstam PV, Qi D, Leng L, Young L, Bucala R. MIF family cytokines in cardiovascular diseases and prospects for precision-based therapeutics. Expert Opin Ther Targets 2017; 21:671-683. [PMID: 28562118 DOI: 10.1080/14728222.2017.1336227] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with chemokine-like functions that increasingly is being studied in different aspects of cardiovascular disease. MIF was first identified as a proinflammatory and pro-survival mediator within the immune system, and a second structurally related MIF family member, D-dopachrome tautomerase (a.k.a. MIF-2), was reported recently. Both MIF family members are released by myocardium and modulate the manifestations of cardiovascular disease, specifically in myocardial ischemia. Areas covered: A scientific overview is provided for the involvement of MIF family cytokines in the inflammatory pathogenesis of atherosclerosis, myocardial infarction, and ischemia-reperfusion injury. We summarize findings of experimental, human genetic and clinical studies, and suggest therapeutic opportunities for modulating the activity of MIF family proteins that potentially may be applied in a MIF allele specific manner. Expert opinion: Knowledge of MIF, MIF-2 and their receptor pathways are under active investigation in different types of cardiovascular diseases, and novel therapeutic opportunities are being identified. Clinical translation may be accelerated by accruing experience with MIF-directed therapies currently in human testing in cancer and autoimmunity.
Collapse
Affiliation(s)
- Pathricia V Tilstam
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Dake Qi
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA.,b Department of Biomedical Sciences , Memorial University of Newfoundland , St. John's , Canada
| | - Lin Leng
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Lawrence Young
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Richard Bucala
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|