1
|
Fu H, Gao B, Zhou X, Hao Y, Liu C, Lan A, Tang J, Zhou F. DNA dioxygenase TET2 deficiency aggravates sepsis-induced acute lung injury by targeting ITGA10 via the PI3K/AKT signaling pathway. Cell Mol Biol Lett 2025; 30:60. [PMID: 40389853 PMCID: PMC12090539 DOI: 10.1186/s11658-025-00739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is a clinical condition with high morbidity and mortality, and impaired endothelial function is the main pathological characteristic. As a member of DNA demethylases, ten-eleven translocation protein 2 (TET2) is involved in a variety of biological processes. However, the role of TET2 in endothelial dysfunction of sepsis-induced ALI remains unclear. METHODS We used cecal ligation and puncture (CLP) to establish a sepsis-induced acute lung injury mouse model and screened out Tet2 from TET family proteins. The results suggested that Tet2 was obviously declined. We used lipopolysaccharide (LPS) to stimulate human pulmonary microvascular endothelial cells (HPMECs) as an in vitro model, and we found the expression of TET2 was also decreased. Then we used small interfering RNAs and adenovirus to knockdown or overexpress TET2 to investigate the effect of TET2 on the function of HPMECs. The changes in sepsis-induced ALI symptoms were also analyzed in Tet2-deficient mice generated by adeno-associated virus 6 (AAV6). Next, RNA sequencing and KEGG analysis were used to find the TET2-regulated downstream target genes and signaling pathways under LPS stimulation. Finally, the rescue experiments were performed to analyze the role of target genes and signaling pathways modulated by TET2 in LPS-treated HPMECs. RESULTS TET2 and 5-hmC levels were significantly decreased in both in vitro and in vivo models of sepsis-induced ALI. TET2 knockdown exacerbated the dysfunction and apoptosis of HPMECs induced by LPS. Conversely, TET2 overexpression significantly alleviated these dysfunctions and reduced apoptosis. Meanwhile, the lung injury of Tet2-deficient mice was aggravated by increased inflammation and apoptosis. RNA sequencing and subsequent experiments showed that TET2 overexpression could increase the expression of Integrin α10 (ITGA10) by reducing the methylation level of ITGA10 promoter. This, in turn, activated the PI3K-AKT signaling pathway. Knocking down ITGA10 weakened the beneficial effects of TET2 overexpression in LPS-stimulated endothelial cells. CONCLUSIONS In our study, we demonstrated that TET2 deficiency aggravates endothelial cell dysfunction and promotes acute lung injury by targeting ITGA10 via the PI3K-AKT pathway. These findings indicate that TET2 may be a promising therapeutic target for treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Hongxue Fu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bin Gao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yingting Hao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chang Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ailin Lan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingyi Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Fachun Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Cai Q, Jing C, Wang X, Xing X, Liu W. STEAP Proteins: Roles in disease biology and potential for therapeutic intervention. Int J Biol Macromol 2025; 309:142797. [PMID: 40185436 DOI: 10.1016/j.ijbiomac.2025.142797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Iron and copper are essential metal ions, and maintaining their metabolic balance is critical for organismal health. The Six-Transmembrane Epithelial Antigen of the Prostate (STEAP) protein family, comprising STEAP1, STEAP2, STEAP3, and STEAP4, plays a vital role in cellular metal homeostasis. These proteins are located on the cell membrane and are characterized by six transmembrane domains. With the exception of STEAP1, the STEAP proteins function as metal oxidoreductases due to their F420H2:NADP+ oxidoreductase (FNO)-like domain. However, STEAP1 contributes to metal metabolism through its heme group and interaction with other STEAP proteins. Beyond metal metabolism, STEAP proteins are involved in critical cellular processes, including the regulation of the cell cycle, proliferation, differentiation, and apoptosis. Notably, STEAP proteins are recognized as potential biomarkers and therapeutic targets in human cancers, particularly prostate cancer. This review outlines the structural features and functional roles of STEAP proteins in various diseases, including cancers, insulin resistance, non-alcoholic fatty liver disease (NAFLD), and benign prostatic hyperplasia, with a focus on their potential for therapeutic intervention.
Collapse
Affiliation(s)
- Qiaomei Cai
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Chao Jing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Xiangling Xing
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China.
| | - Wancheng Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, PR China.
| |
Collapse
|
3
|
Gould S, Herron A, Davis J, Phillips M, Chakrabarti M, Evans CE. Control of inflammatory lung injury and repair by metabolic signaling in endothelial cells. Curr Opin Hematol 2025; 32:157-167. [PMID: 39450949 PMCID: PMC11949724 DOI: 10.1097/moh.0000000000000848] [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] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Sepsis-induced inflammatory lung injury includes acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). There are currently no effective treatments for ALI/ARDS, but clinical outcomes could be improved by inhibiting lung injury and/or promoting post-sepsis vascular repair. In this review, we describe studies of endothelial cell metabolic pathways in sepsis-induced ALI/ARDS and vascular repair and identify areas of research that deserve attention in future studies. We also describe studies of metabolic interventions that aim to inhibit ALI/ARDS and/or promote post-sepsis vascular repair, including those that target endothelial cell metabolites, endothelial cell metabolic signaling pathways, and endothelial cell metabolism. RECENT FINDINGS Endothelial cells are integral to both the injury and repair phases of ALI/ARDS. During the injury phase of ALI/ARDS, lung endothelial cell survival decreases, and lung endothelial cell-to-endothelial cell (EC-EC) junctions are weakened. During the repair phase after sepsis-induced lung injury, lung endothelial cell proliferation and lung EC-EC junction reannealing occur. These crucial aspects of ALI/ARDS and post-sepsis vascular repair, that is, endothelial cell viability, growth, and junction integrity, are controlled by a myriad of metabolites and metabolic signaling pathways in endothelial cells. SUMMARY Metabolic signaling pathways in endothelial cells represent a novel class of putative targets for the prevention and treatment of sepsis-induced inflammatory lung injury. Therapies that target metabolic signaling in endothelial cells are currently being explored as potential treatments for sepsis-induced inflammatory lung injury.
Collapse
Affiliation(s)
- Seth Gould
- Biomedical Engineering Program, University of South Carolina
| | - Ansley Herron
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina
| | - Jonathan Davis
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mollie Phillips
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mrinmay Chakrabarti
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Colin E. Evans
- Biomedical Engineering Program, University of South Carolina
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
- Institute on Cardiovascular Disease Research, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
4
|
Wang Y, Wang W, Zhang Y, Fleishman JS, Wang H. Targeting ferroptosis offers therapy choice in sepsis-associated acute lung injury. Eur J Med Chem 2025; 283:117152. [PMID: 39657462 DOI: 10.1016/j.ejmech.2024.117152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/06/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-associated acute lung injury (SALI) is a common complication of sepsis, consisting of a dysfunctional host response to infection-mediated heterogenous complexes. SALI is reported in up to 50 % of patients with sepsis and causes poor outcomes. Despite high incidence, there is a lack of understanding in its pathogenesis and optimal treatment. A better understanding of the molecular mechanisms underlying SALI may help produce better therapeutics. The effects of altered cell-death mechanisms, such as non-apoptotic regulated cell death (RCD) (i.e., ferroptosis), on the development of SALI are beginning to be discovered, while targeting ferroptosis as a meaningful target in SALI is increasingly being recognized. Here, we outline how a susceptible lung alveoli may develop SALI. Then we discuss the general mechanisms underlying ferroptosis, and how it contributes to SALI. We then outline the chemical structures of the emerging agents or compounds that can protect against SALI by inhibiting ferroptosis, summarizing their potential pharmacological effects. Finally, we highlight key limitations and possible strategies to overcome them. This review suggests that a detailed mechanistic and biological understanding of ferroptosis can foster the development of pharmacological antagonists in the treatment of SALI.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Yi Zhang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
5
|
Lai J, Yu S, Li X, Wei Q, Qin J. METTL14/IGF2BP2-MEDIATED M6A MODIFICATION OF STEAP1 AGGRAVATES ACUTE LUNG INJURY INDUCED BY SEPSIS. Shock 2025; 63:217-225. [PMID: 39193903 DOI: 10.1097/shk.0000000000002456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
ABSTRACT Background: Acute lung injury (ALI) is a severe complication of sepsis, characterized by inflammation, edema, and injury to alveolar cells, leading to high mortality rates. Septic ALI is a complex disease involving multiple factors and signaling pathways. STEAP family member 1 (STEAP1) has been reported to be upregulated in a sepsis-induced ALI model. However, the role of STEAP1 in the regulation of septic ALI is not yet fully understood. Methods: The study stimulated human pulmonary microvascular endothelial cells (HPMECs) using lipopolysaccharides (LPS) to establish an in vitro ALI model. The study used quantitative real-time polymerase chain reaction to measure mRNA expression, and western blotting assay or immunohistochemistry assay to analyze protein expression. Cell Counting Kit-8 assay was performed to assess cell viability. Flow cytometry was conducted to analyze cell apoptosis. Tube formation assay was used to analyze the tube formation rate of human umbilical vein endothelial cells. Enzyme-linked immunosorbent assays were used to measure the levels of interleukin 1beta and tumor necrosis factor alpha. The levels of Fe 2+ and reactive oxygen species were determined using colorimetric and fluorometric assays, respectively. The glutathione level was also determined using a colorimetric assay. m6A RNA immunoprecipitation assay, dual-luciferase reporter assay, and RNA immunoprecipitation assay were performed to identify the association of STEAP1 with methyltransferase 14, N6-adenosine-methyltransferase noncatalytic subunit (METTL14) and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2). The transcript half-life of STEAP1 was analyzed by actinomycin D assay. Finally, a rat model of polymicrobial sepsis was established to analyze the effects of STEAP1 knockdown on lung injury in vivo . Results: We found that the mRNA expression levels of STEAP1 and METTL14 were upregulated in the blood of ALI patients induced by sepsis compared to healthy volunteers. LPS treatment increased the protein levels of STEAP1 and METTL14 in HPMECs. STEAP1 depletion attenuated LPS-induced promoting effects on HPMECs' apoptosis, inflammatory response, and ferroptosis, as well as LPS-induced inhibitory effect on tube formation. We also found that METTL14 and IGF2BP2 stabilized STEAP1 mRNA expression through the m6A methylation modification process. Moreover, METTL14 silencing attenuated LPS-induced effects by decreasing STEAP1 expression in HPMECs, and STEAP1 silencing ameliorated cecal ligation and puncture-induced lung injury of mice. Conclusion: METTL14/IGF2BP2-mediated m6A modification of STEAP1 aggravated ALI induced by sepsis. These findings suggest potential therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Junhua Lai
- Intensive Care Unit, Liuzhou Worker's Hospital, Liuzhou City, Guangxi, China
| | | | | | | | | |
Collapse
|
6
|
Chen K, Lu S, Shi K, Ali MH, Liu J, Yin F, Yin W. Hyperoside attenuates sepsis-induced acute lung injury by Nrf2 activation and ferroptosis inhibition. Int Immunopharmacol 2025; 145:113734. [PMID: 39657533 DOI: 10.1016/j.intimp.2024.113734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening condition associated with high morbidity and mortality rates in intensive care units (ICUs). Emerging evidence from clinical studies suggests that compounds derived from traditional Chinese medicine (TCM) have shown promising therapeutic effects in treating sepsis-induced ALI. Hyperoside is a bioactive compound extracted from TCM. Prior studies reported that hyperoside exhibits potent anti-inflammatory, antioxidant, and organ-protective properties, however, the underlying mechanisms of its effects on ALI remain unclear. Hyperoside pretreatment significantly reduced inflammation, iron accumulation, and lipid peroxidation in the pulmonary tissues of ALI mice induced by CLP and in LPS-stimulated MLE-12 cells. In particular, hyperoside preferentially binds with Keap1 at Arg380 and Arg415, thereby inhibiting the ubiquitin-mediated degradation of nuclear Nrf2, promoting its translocation to the nucleus, and leading to upregulation of anti-ferroptosis gene expression. Moreover, the protective effects of hyperoside were significantly abrogated after Nrf2 expression was silenced or its activity was inhibited by chemical inhibitors, highlighting that Nrf2 is critically involved in the impact of hyperoside. This study confirms that hyperoside exhibits a therapeutically protective effect against sepsis-induced ALI by inhibiting ferroptosis through Nrf2-mediated signaling pathway. Hyperoside acts as an Nrf2 activator by preferentially binding to Arg380 and Arg415 of Keap1 and disrupting the Keap1/Nrf2 interaction.
Collapse
Affiliation(s)
- Kuida Chen
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Shipeng Lu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ke Shi
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China; Medical Research Center, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, China
| | - Mustafa Hussein Ali
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | - Wu Yin
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
7
|
Yuan Y, Wang Y, Yan Y, Kim E, Bai J, Zhao Y, Ma Q, Gu W, Song H. FBLN1 regulates ferroptosis in acute respiratory distress syndrome by reducing free ferrous iron by inhibiting the TGF-β/Smad pathway. PLoS One 2024; 19:e0314750. [PMID: 39671383 PMCID: PMC11643259 DOI: 10.1371/journal.pone.0314750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/17/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) / acute lung injury (ALI) is a serious medical disease characterized by pulmonary dysfunction and inflammation. This study aims to determine the main molecular modules linked to ARDS and investigate the role of Fibulin-1 (FBLN1) in regulating ferroptosis in ARDS. METHODS Weighted Gene Co-expression Network Analysis (WGCNA) was employed on the GSE263867 dataset to find key modules associated with ALI. Differentially expressed genes (DEGs) and protein-protein interaction (PPI) networks were analyzed. MLE-12 cells were treated with lipopolysaccharide (LPS) to induce ferroptosis. In vitro studies were conducted to investigate the effects of FBLN1 and Transforming Growth Factor Beta 1 (TGF-β) overexpression on cell viability, oxidative stress markers, and ferroptosis-related proteins. RESULTS WGCNA identified the turquoise module as significantly negatively correlated with ARDS. Five key overlapping genes (GRIA1, OGN, COL14A1, FBLN1, and COL6A3) were significantly downregulated in ARDS samples. LPS treatment induced ferroptosis in MLE-12 cells, indicated by increased malondialdehyde (MDA), lipid reactive oxygen species (ROS), and ferrous iron (Fe2⁺) levels, and decreased cell viability and glutathione (GSH) levels. FBLN1 overexpression partially reversed these effects. Additionally, FBLN1 inhibited the TGF-β/Smad signaling pathway, as shown by decreased TGF-β and p-Smad protein levels. TGF-β overexpression exacerbated LPS-induced oxidative stress and ferroptosis, reducing cell viability and GSH levels. FBLN1 overexpression counteracted this effect, suggesting antagonistic roles for FBLN1 and TGF-β in regulating ferroptosis. CONCLUSION This study highlights FBLN1 as a critical regulator of ferroptosis in ARDS. Targeting the TGF-β/Smad pathway to modulate FBLN1 expression offers a potential therapeutic strategy to alleviate oxidative stress and mitigate pulmonary injury in inflammatory lung diseases.
Collapse
Affiliation(s)
- Yaping Yuan
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Youbo Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yufeng Yan
- Department of Neurosurgery, Jinshan Hospital, Fudan University, Zhujing Town, Jinshan District, Shanghai, China
| | - Edward Kim
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
| | - Jin Bai
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- CRT Medical Union, Time International, Beijing, China
| | - Yang Zhao
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- CRT Medical Union, Time International, Beijing, China
| | - Qinyun Ma
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenchao Gu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Haihan Song
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| |
Collapse
|
8
|
Zhang W, Jiang L, Tong X, He H, Zheng Y, Xia Z. Sepsis-Induced Endothelial Dysfunction: Permeability and Regulated Cell Death. J Inflamm Res 2024; 17:9953-9973. [PMID: 39628705 PMCID: PMC11612565 DOI: 10.2147/jir.s479926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Endothelial cells (ECs) are an important cell type typically affected in sepsis, resulting in compromised barrier function and various forms of regulated cell death (RCD). However, the precise mechanisms underlying sepsis-induced EC damage remain unclear. This review summarizes the recent research progress on factors and mechanisms that may affect the permeability and RCD of ECs under septic conditions, including glycocalyx, damage-associated molecular patterns, and various forms of RCD in ECs, such as apoptosis, pyroptosis, ferroptosis, and autophagy. This review offers important insights into the underlying mechanisms of endothelial dysfunction in sepsis, aiming to contribute to developing small-molecule targeted clinical therapies.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Luofeng Jiang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Xirui Tong
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Heng He
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Zhaofan Xia
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
- Research Unit of Key Techniques for Treatment of burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
9
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
10
|
Xu Y, Qu X, Liang M, Huang D, Jin M, Sun L, Chen X, Liu F, Qiu Z. Focus on the role of calcium signaling in ferroptosis: a potential therapeutic strategy for sepsis-induced acute lung injury. Front Med (Lausanne) 2024; 11:1457882. [PMID: 39355841 PMCID: PMC11442327 DOI: 10.3389/fmed.2024.1457882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
By engaging in redox processes, ferroptosis plays a crucial role in sepsis-induced acute lung injury (ALI). Although iron stimulates calcium signaling through the stimulation of redox-sensitive calcium pathways, the function of calcium signals in the physiological process of ferroptosis in septic ALI remains unidentified. Iron homeostasis disequilibrium in ferroptosis is frequently accompanied by aberrant calcium signaling. Intracellular calcium overflow can be a symptom of dysregulation of the cellular redox state, which is characterized by iron overload during the early phase of ferroptosis. This can lead to disruptions in calcium homeostasis and calcium signaling. The mechanisms controlling iron homeostasis and ferroptosis are reviewed here, along with their significance in sepsis-induced acute lung injury, and the potential role of calcium signaling in these processes is clarified. We propose that the development of septic acute lung injury is a combined process involving the bidirectional interaction between iron homeostasis and calcium signaling. Our goal is to raise awareness about the pathophysiology of sepsis-induced acute lung injury and investigate the relationship between these mechanisms and ferroptosis. We also aimed to develop calcium-antagonistic therapies that target ferroptosis in septic ALI and improve the quality of survival for patients suffering from acute lung injury.
Collapse
Affiliation(s)
- Yifei Xu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintian Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghao Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minyan Jin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianhai Chen
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fen Liu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhanjun Qiu
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
Batista-Silva JP, Gomes D, Sousa SF, Sousa Â, Passarinha LA. Advances in structure-based drug design targeting membrane protein markers in prostate cancer. Drug Discov Today 2024; 29:104130. [PMID: 39103143 DOI: 10.1016/j.drudis.2024.104130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Prostate cancer (PCa) is one of the leading cancers in men and the lack of suitable biomarkers or their modulators results in poor prognosis. Membrane proteins (MPs) have a crucial role in the development and progression of PCa and can be attractive therapeutic targets. However, experimental limitations in targeting MPs hinder effective biomarker and inhibitor discovery. To overcome this barrier, computational methods can yield structural insights and screen large libraries of compounds, accelerating lead identification and optimization. In this review, we examine current breakthroughs in computer-aided drug design (CADD), with emphasis on structure-based approaches targeting the most relevant membrane-bound PCa biomarkers.
Collapse
Affiliation(s)
- João P Batista-Silva
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Chemistry Department, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2819-516 Caparica, Portugal
| | - Diana Gomes
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Chemistry Department, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2819-516 Caparica, Portugal
| | - Sérgio F Sousa
- LAQV/REQUIMTE, BioSIM - Department of Medicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ângela Sousa
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - Luís A Passarinha
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal; UCIBIO-Applied Molecular Biosciences Unit, Chemistry Department, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2819-516 Caparica, Portugal; Laboratório de Fármaco-Toxicologia-UBIMedical, University of Beira Interior, 6200-284 Covilhã, Portugal.
| |
Collapse
|
12
|
Wu Z, Xi Q, Zhao Q, Zhu S. GDF11 OVEREXPRESSION ALLEVIATES SEPSIS-INDUCED LUNG MICROVASCULAR ENDOTHELIAL BARRIER DAMAGE BY ACTIVATING SIRT1/NOX4 SIGNALING TO INHIBIT FERROPTOSIS. Shock 2024; 62:245-254. [PMID: 38920138 DOI: 10.1097/shk.0000000000002391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
ABSTRACT Sepsis is a lethal clinical syndrome, and acute lung injury (ALI) is the earliest and most serious complication. We aimed to explore the role of growth differentiation factor 11 (GDF11) in sepsis-induced dysfunction of lung microvascular endothelial barrier in vivo and in vitro to elucidate its potential mechanism related to sirtuin 1 (SIRT1)/NADPH oxidase 4 (NOX4) signaling. Cecal ligation and puncture (CLP)-induced sepsis mice and lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cells (PMECs) were used in this study. Histopathological changes in lung tissues were tested by hematoxylin-eosin staining. Lung wet-to-dry weight ratio and inflammatory factors contents in bronchoalveolar lavage fluid were assessed. Evens blue index, trans-epithelial electrical resistance, and expression of zona occludens 1 (ZO-1), occludin-1, and claudin-1 were used to evaluate alveolar barrier integrity. Reactive oxygen species, lipid peroxidation, and ferroptosis markers were analyzed. Iron deposition in the lung tissues was assessed using Prussian blue staining. Intracellular Fe 2+ level was detected using FerroOrange staining. Additionally, expression of GDF11, SIRT1, and NOX4 was estimated with western blot. Then, EX527, a SIRT1 inhibitor, was employed to treat GDF11-overexpressed PMECs with LPS stimulation to clarify the regulatory mechanism. Results showed that GDF11 overexpression attenuated sepsis-induced pathological changes and inflammation and maintained alveolar barrier integrity. Moreover, GDF11 overexpression inhibited ferroptosis, upregulated SIRT1 expression and downregulated NOX4 expression. Additionally, EX527 treatment relieved the impacts of GDF11 overexpression on ferroptosis and destruction of integrity of human pulmonary microvascular endothelial cells exposed to LPS. Taken together, GDF11 overexpression could alleviate sepsis-induced lung microvascular endothelial barrier damage by activating SIRT1/NOX4 signaling to inhibit ferroptosis. Our findings potentially provide new molecular target for clinical therapy of ALI.
Collapse
Affiliation(s)
- Zhixiang Wu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | |
Collapse
|
13
|
Wen Z, Fan J, Zhan F, Li X, Li B, Lu P, Yao X, Shen Z, Liu Z, Wang C, Li X, Jin W, Zhang X, Qi Y, Wang X, Song M. The role of FPR2-mediated ferroptosis in formyl peptide-induced acute lung injury against endothelial barrier damage and protective effect of the mitochondria-derived peptide MOTS-c. Int Immunopharmacol 2024; 131:111911. [PMID: 38527401 DOI: 10.1016/j.intimp.2024.111911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Acute lung injury (ALI) has garnered significant attention in the field of respiratory and critical care due to its high mortality and morbidity, and limited treatment options. The role of the endothelial barrier in the development of ALI is crucial. Several bacterial pathogenic factors, including the bacteria-derived formyl peptide (fMLP), have been implicated in damaging the endothelial barrier and initiating ALI. However, the mechanism by which fMLP causes ALI remains unclear. In this study, we aim to explore the mechanisms of ALI caused by fMLP and evaluate the protective effects of MOTS-c, a mitochondrial-derived peptide. METHODS We established a rat model of ALI and a human pulmonary microvascular endothelial cell (HPMVEC) model of ALI by treatment with fMLP. In vivo experiments involved lung histopathology assays, assessments of inflammatory and oxidative stress factors, and measurements of ferroptosis-related proteins and barrier proteins to evaluate the severity of fMLP-induced ALI and the type of tissue damage in rats. In vitro experiments included evaluations of fMLP-induced damage on HPMVEC using cell activity assays, assessments of inflammatory and oxidative stress factors, measurements of ferroptosis-related proteins, endothelial barrier function assays, and examination of the key role of FPR2 in fMLP-induced ALI. We also assessed the protective effect of MOTS-c and investigated its mechanism on the fMLP-induced ALI in vivo and in vitro. RESULTS Results from both in vitro and in vivo experiments demonstrate that fMLP promotes the expression of inflammatory and oxidative stress factors, activates ferroptosis and disrupts the vascular endothelial barrier, ultimately contributing to the development and progression of ALI. Mechanistically, ferroptosis mediated by FPR2 plays a key role in fMLP-induced injury, and the Nrf2 and MAPK pathways are involved in this process. Knockdown of FPR2 and inhibition of ferroptosis can attenuate ALI induced by fMLP. Moreover, MOTS-c could protect the vascular endothelial barrier function by inhibiting ferroptosis and suppressing the expression of inflammatory and oxidative stress factors through Nrf2 and MAPK pathways, thereby alleviating fMLP-induced ALI. CONCLUSION Overall, fMLP disrupts the vascular endothelial barrier through FPR2-mediated ferroptosis, leading to the development and progression of ALI. MOTS-c demonstrates potential as a protective treatment against ALI by alleviating the damage induced by fMLP.
Collapse
Affiliation(s)
- Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jidan Fan
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Faliang Zhan
- Department of Cardiothoracic Surgery, Yili Friendship Hospital, Yining, Xinjiang Uyghur Autonomous Region 839300, China
| | - Xiaopei Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xin Yao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zhaoyang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Chufan Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Wanjun Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiao Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China; Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
14
|
Xu H, Sheng S, Luo W, Xu X, Zhang Z. Acute respiratory distress syndrome heterogeneity and the septic ARDS subgroup. Front Immunol 2023; 14:1277161. [PMID: 38035100 PMCID: PMC10682474 DOI: 10.3389/fimmu.2023.1277161] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute diffuse inflammatory lung injury characterized by the damage of alveolar epithelial cells and pulmonary capillary endothelial cells. It is mainly manifested by non-cardiogenic pulmonary edema, resulting from intrapulmonary and extrapulmonary risk factors. ARDS is often accompanied by immune system disturbance, both locally in the lungs and systemically. As a common heterogeneous disease in critical care medicine, researchers are often faced with the failure of clinical trials. Latent class analysis had been used to compensate for poor outcomes and found that targeted treatment after subgrouping contribute to ARDS therapy. The subphenotype of ARDS caused by sepsis has garnered attention due to its refractory nature and detrimental consequences. Sepsis stands as the most predominant extrapulmonary cause of ARDS, accounting for approximately 32% of ARDS cases. Studies indicate that sepsis-induced ARDS tends to be more severe than ARDS caused by other factors, leading to poorer prognosis and higher mortality rate. This comprehensive review delves into the immunological mechanisms of sepsis-ARDS, the heterogeneity of ARDS and existing research on targeted treatments, aiming to providing mechanism understanding and exploring ideas for accurate treatment of ARDS or sepsis-ARDS.
Collapse
Affiliation(s)
- Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiying Sheng
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weiwei Luo
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| |
Collapse
|