1
|
Hu Y, Xie Y, Sun Y, Luo L, Wang H, Zhang R, Ge M. Anti-inflammatory effects of apigenin on LPS-induced mastitis in lactating SD rats through inhibiting TLR4/NF-κB signaling pathway. Cytokine 2025; 191:156944. [PMID: 40288318 DOI: 10.1016/j.cyto.2025.156944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/05/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Mastitis is an important disease of the mammary gland in all kinds of lactating mammals, endangering the development of animal husbandry and human health. Apigenin is one chemical constituent of Taraxacum and Philippine Violet Herb which are effective Chinese herbs for the treatment of mastitis. It is reported that apigenin possesses anti-inflammatory activity and other pharmacological effects. However, the attenuation of apigenin on mastitis has not yet been reported. The present study investigated the protection of apigenin against lipopolysaccharide (LPS)-induced mastitis in SD rats both in vivo and in vitro. The results suggested that apigenin relieved the lesions of mammary tissues induced by LPS, decreased mRNA and protein levels of pro-inflammatory cytokines:tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). Simultaneously, apigenin reduced the increasing content of myeloperoxidase (MPO) and Toll-like receptor 4 (TLR4), and phosphorylation of nuclear factor kappa B (NF-κB) induced by LPS. The results showed that apigenin was able to attenuate the LPS-induced mastitis in rats by inhibiting the TLR4/NF-κB signaling pathway in vivo and in vitro, which provides scientific references for further research.
Collapse
Affiliation(s)
- Yihan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Yingying Xie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China
| | - Yiming Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Linghuan Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China
| | - Haibin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Ruili Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China.
| | - Ming Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China.
| |
Collapse
|
2
|
Bhatt J, Ghigo A, Hirsch E. PI3K/Akt in IPF: untangling fibrosis and charting therapies. Front Immunol 2025; 16:1549277. [PMID: 40248697 PMCID: PMC12004373 DOI: 10.3389/fimmu.2025.1549277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic, progressive lung disease characterized by abnormal epithelial repair, persistent inflammation, and excessive extracellular matrix deposition, leading to irreversible scarring and respiratory failure. Central to its pathogenesis is the dysregulation of the PI3K/Akt signaling pathway, which drives fibroblast activation, epithelial-mesenchymal transition, apoptosis resistance, and cellular senescence. Senescent cells contribute to fibrosis through the secretion of pro-inflammatory and profibrotic factors in the senescence-associated secretory phenotype (SASP). Current antifibrotic therapies, Nintedanib and Pirfenidone, only slow disease progression and are limited by side effects, highlighting the need for novel treatments. This review focuses on the role of PI3K/Akt signaling in IPF pathogenesis, its intersection with inflammation and fibrosis, and emerging therapeutic approaches targeting molecules along this pathway.
Collapse
Affiliation(s)
- Janki Bhatt
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| |
Collapse
|
3
|
Zeng H, Zhou Y, Liu Z, Liu W. MiR-21-5p modulates LPS-induced acute injury in alveolar epithelial cells by targeting SLC16A10. Sci Rep 2024; 14:11160. [PMID: 38750066 PMCID: PMC11096310 DOI: 10.1038/s41598-024-61777-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome resulting from the invasion of the human body by bacteria and other pathogenic microorganisms. One of its most prevalent complications is acute lung injury, which places a significant medical burden on numerous countries and regions due to its high morbidity and mortality rates. MicroRNA (miRNA) plays a critical role in the body's inflammatory response and immune regulation. Recent studies have focused on miR-21-5p in the context of acute lung injury, but its role appears to vary in different models of this condition. In the LPS-induced acute injury model of A549 cells, there is differential expression, but the specific mechanism remains unclear. Therefore, our aim is to investigate the changes in the expression of miR-21-5p and SLC16A10 in a type II alveolar epithelial cell injury model induced by LPS and explore the therapeutic effects of their targeted regulation. A549 cells were directly stimulated with 10 µg/ml of LPS to construct a model of LPS-induced cell injury. Cells were collected at different time points and the expression of interleukin 1 beta (IL-1β), tumor necrosis factor-α (TNF-α) and miR-21-5p were measured by RT-qPCR and western blot. Then miR-21-5p mimic transfection was used to up-regulate the expression of miR-21-5p in A549 cells and the expression of IL-1β and TNF-α in each group of cells was measured by RT-qPCR and western blot. The miRDB, TargetScan, miRWalk, Starbase, Tarbase and miR Tarbase databases were used to predict the miR-21-5p target genes and simultaneously, the DisGeNet database was used to search the sepsis-related gene groups. The intersection of the two groups was taken as the core gene. Luciferase reporter assay further verified SLC16A10 as the core gene with miR-21-5p. The expression of miR-21-5p and SLC16A10 were regulated by transfection or inhibitors in A549 cells with or without LPS stimulation. And then the expression of IL-1β and TNF-α in A549 cells was tested by RT-qPCR and western blot in different groups, clarifying the role of miR-21-5p-SLC16A10 axis in LPS-induced inflammatory injury in A549 cells. (1) IL-1β and TNF-α mRNA and protein expression significantly increased at 6, 12, and 24 h after LPS stimulation as well as the miR-21-5p expression compared with the control group (P < 0.05). (2) After overexpression of miR-21-5p in A549 cells, the expression of IL-1β and TNF-α was significantly reduced after LPS stimulation, suggesting that miR-21-5p has a protection against LPS-induced injury. (3) The core gene set, comprising 51 target genes of miR-21-5p intersecting with the 1448 sepsis-related genes, was identified. This set includes SLC16A10, TNPO1, STAT3, PIK3R1, and FASLG. Following a literature review, SLC16A10 was selected as the ultimate target gene. Dual luciferase assay results confirmed that SLC16A10 is indeed a target gene of miR-21-5p. (4) Knocking down SLC16A10 expression by siRNA significantly reduced the expression of IL-1β and TNF-α in A549 cells after LPS treatment (P < 0.05). (5) miR-21-5p inhibitor increased the expression levels of IL-1β and TNF-α in A549 cells after LPS stimulation (P < 0.05). In comparison to cells solely transfected with miR-21-5p inhibitor, co-transfection of miR-21-5p inhibitor and si-SLC6A10 significantly reduced the expression of IL-1β and TNF-α (P < 0.05). MiR-21-5p plays a protective role in LPS-induced acute inflammatory injury of A549 cells. By targeting SLC16A10, it effectively mitigates the inflammatory response in A549 cells induced by LPS. Furthermore, SLC16A10 holds promise as a potential target for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Huanan Zeng
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang, 110001, Liaoning, China
| | - Yuqing Zhou
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang, 110001, Liaoning, China
| | - Zhi Liu
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang, 110001, Liaoning, China.
| | - Wei Liu
- Emergency Department, The First Hospital of China Medical University, No.155 of North Street Nanjing, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
4
|
Sachetto ATA, Mackman N. Commentary: Macrophage IL-1β-positive microvesicles exhibit thrombo-inflammatory properties and are detectable in patients with active juvenile idiopathic arthritis. Front Immunol 2024; 15:1397527. [PMID: 38655252 PMCID: PMC11035834 DOI: 10.3389/fimmu.2024.1397527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Affiliation(s)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
5
|
Li Q, Xiao C, Gu J, Chen X, Yuan J, Li S, Li W, Gao D, Li L, Liu Y, Shen F. 6-Gingerol ameliorates alveolar hypercoagulation and fibrinolytic inhibition in LPS-provoked ARDS via RUNX1/NF-κB signaling pathway. Int Immunopharmacol 2024; 128:111459. [PMID: 38181675 DOI: 10.1016/j.intimp.2023.111459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolytic inhibition play a central role in refractory hypoxemia in acute respiratory distress syndrome (ARDS), but it lacks effective drugs for prevention and treatment of this pathophysiology. Our previous experiment confirmed that RUNX1 promoted alveolar hypercoagulation and fibrinolytic inhibition through NF-κB pathway. Other studies demonstrated that 6-gingerol regulated inflammation and metabolism by inhibiting the NF-κB signaling pathway. We assume that 6-gingerol would ameliorate alveolar hypercoagulation and fibrinolytic inhibition via RUNX1/ NF-κB pathway in LPS-induced ARDS. METHODS Rat ARDS model was replicated through LPS inhalation. Before LPS inhalation, the rats were intraperitoneally treated with different doses of 6-gingerol or the same volume of normal saline (NS) for 12 h, and then intratracheal inhalation of LPS for 24 h. In cell experiment, alveolar epithelial cell type II (AECII) was treated with 6-gingerol for 6 h and then with LPS for another 24 h. RUNX1 gene was down-regulated both in pulmonary tissue and in cells. Tissue factor (TF), plasminogen Activator Inhibitor 1(PAI-1) and thrombin were determined by Wester-blot (WB), qPCR or by enzyme-linked immunosorbent (ELISA). Lung injury score, pulmonary edema and pulmonary collagen III in rat were assessed. NF-κB pathway were also observed in vivo and in vitro. The direct binding capability of 6-gingerol to RUNX1 was confirmed by using Drug Affinity Responsive Target Stability test (DARTS). RESULTS 6-gingerol dose-dependently attenuated LPS-induced lung injury and pulmonary edema. LPS administration caused excessive TF and PAI-1 expression both in pulmonary tissue and in AECII cell and a large amount of TF, PAI-1 and thrombin in bronchial alveolar lavage fluid (BALF), which all were effectively decreased by 6-gingerol treatment in a dose-dependent manner. The high collagen Ⅲ level in lung tissue provoked by LPS was significantly abated by 6-gingerol. 6-gingerol was seen to dramatically inhibit the LPS-stimulated activation of NF-κB pathway, indicated by decreases of p-p65/total p65, p-IKKβ/total IKKβ, and also to suppress the RUNX1 expression. RUNX1 gene knock down or RUNX1 inhibitor Ro5-3335 significantly enhanced the efficacies of 6-gingerol in vivo and in vitro, but RUNX1 over expression remarkably impaired the effects of 6-gingerol on TF, PAI-1 and on NF-κB pathway. DARTS result showed that 6-gingerol directly bond to RUNX1 molecules. CONCLUSIONS Our experimental data demonstrated that 6-gingerol ameliorates alveolar hypercoagulation and fibrinolytic inhibition via RUNX1/NF-κB pathway in LPS-induced ARDS. 6-gingerol is expected to be an effective drug in ARDS.
Collapse
Affiliation(s)
- Qing Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Chuan Xiao
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - JiaRun Gu
- Emergency department, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Xianjun Chen
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Jia Yuan
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Shuwen Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Wei Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Daixiu Gao
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Lu Li
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Ying Liu
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Feng Shen
- Department of Intensive Care Unit, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
6
|
Xiao C, Li Q, Xiao J, Chen X, Yuan J, Li S, Li W, Gao D, Li L, Liu Y, Shen F. miR-9 targeting RUNX1 improves LPS-induced alveolar hypercoagulation and fibrinolysis inhibition through NF-κB inactivation in ARDS. Int Immunopharmacol 2023; 120:110318. [PMID: 37201407 DOI: 10.1016/j.intimp.2023.110318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a clinical and pathophysiological complex syndrome with high mortality. Alveolar hypercoagulation and fibrinolytic inhibition constitute the core part of the pathophysiology of ARDS. miR-9 (microRNA-9a-5p) plays an important role in the pathogenesis of ARDS, but whether it regulates alveolar pro-coagulation and fibrinolysis inhibition in ARDS remains to be elucidated. We aimed to determine the contributing role of miR-9 on alveolar hypercoagulation and fibrinolysis inhibition in ARDS. METHODS In the ARDS animal model, we first observed the miR-9 and runt-related transcription factor 1 (RUNX1) expression in lung tissue, the effects of miR-9 on alveolar hypercoagulation and fibrinolytic inhibition in ARDS rats, and the efficacy of miR-9 on acute lung injury. In the cell, alveolar epithelial cells type II (AECII) were treated with LPS, and the levels of miR-9 and RUNX1 were detected. Then we observed the effects of miR-9 on procoagulant and fibrinolysis inhibitor factors in cells. Finally, we explored whether the efficacies of miR-9 were associated with RUNX1; we also preliminarily examined the miR-9 and RUNX1 levels in plasma in patients with ARDS. RESULTS In ARDS rats, miR-9 expression decreased, but RUNX1 expression increased in the pulmonary tissue of ARDS rats. miR-9 displayed to attenuate lung injury and pulmonary wet/dry ratio. Study results in vivo demonstrated that miR-9 ameliorated alveolar hypercoagulation and fibrinolysis inhibition and attenuated the collagen III expressions in tissue. miR-9 also inhibited NF-κB signaling pathway activation in ARDS. In LPS-induced AECII, the expression changes of both miR-9 and RUNX1 were similar to those in pulmonary tissue in the animal ARDS model. miR-9 effectively inhabited tissue factor (TF), plasma activator inhibitor (PAI-1) expressions, and NF-κB activation in LPS-treated ACEII cells. Besides, miR-9 directly targeted RUNX1, inhibiting TF and PAI-1 expression and attenuating NF-κB activation in LPS-treated AECII cells. Clinically, we preliminarily found that the expression of miR-9 was significantly reduced in ARDS patients compared to non-ARDS patients. CONCLUSION Our experimental data indicate that by directly targeting RUNX1, miR-9 improves alveolar hypercoagulation and fibrinolysis inhibition via suppressing NF-κB pathway activation in LPS-induced rat ARDS, implying that miR-9/RUNX1 is expected to be a new therapeutic target for ARDS treatment.
Collapse
Affiliation(s)
- Chuan Xiao
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Qing Li
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Jingjing Xiao
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, China.
| | - Xianjun Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Jia Yuan
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Shuwen Li
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Wei Li
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Daixiu Gao
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Lu Li
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Ying Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Feng Shen
- Department of Critical Care Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
7
|
Liu L, Lei Y, Chen W, Zhou Q, Zheng Z, Zeng G, Liu W, Feng P, Zhang Z, Yu L, Chen L. In vivo genome-wide CRISPR screening identifies ZNF24 as a negative NF-κB modulator in lung cancer. Cell Biosci 2022; 12:193. [PMID: 36457047 PMCID: PMC9717477 DOI: 10.1186/s13578-022-00933-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Systemic identification of tumor suppressor genes (TSGs) and elucidation of their signaling provide a new angle for understanding of tumorigenesis, which is important for developing successful treatment for lung cancer patients. In our current work, we conducted an in vivo screen for lung cancer TSGs through CRISPR/Cas9 mediated knockout of genes at genome-wide scale. We found that ZNF24 was a potent and clinically relevant TSG of lung cancer. Ectopic expression of ZNF24 arrested lung cancer cells in S phase. Mechanistically, ZNF24 bound to promoter region of P65 to negatively regulate its transcription and thereby the signaling activity of NF-κB pathway. This signaling cascade is clinically relevant. Importantly, we found that combinational inhibition of KRAS, NF-κB, and PD-1 effectively shrank autochthonous KrasG12D/ZNF24-/- lung cancers in transgenic mouse model. Our current work thus revealed an important role played by loss of function of ZNF24 in lung tumorigenesis and shed new light in precision medicine for a portion of lung cancer patients.
Collapse
Affiliation(s)
- Lu Liu
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Yuxi Lei
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Wensheng Chen
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Qian Zhou
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Zongyao Zheng
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Guandi Zeng
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Wanting Liu
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Pengju Feng
- grid.258164.c0000 0004 1790 3548Department of Chemistry, Jinan University, Guangzhou, 510632 China
| | - Zhiyi Zhang
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| | - Lei Yu
- grid.24696.3f0000 0004 0369 153XBeijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Liang Chen
- grid.258164.c0000 0004 1790 3548MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
8
|
Yang W, Peng T, Shi C, Cui F, Chen M, Zhang T. The Mechanism of Delayed Ischemic Preconditioning in Alleviating Acute Ischemia/Reperfusion Renal Injury through Treg Mediated by Immature CD11c + Dendritic Cells. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:487-499. [PMID: 36590681 PMCID: PMC9798836 DOI: 10.1159/000527172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022]
Abstract
Introduction Renal ischemia-reperfusion injury (IRI) is one of the major causes of acute kidney injury, and its mechanism is complex involving multiple factors, while delayed ischemic preconditioning (DIPC) has a protective effect on the above process. In our previous study, we found that DIPC can exert its protection on renal IRI by inhibiting the maturation of dendritic cells (DCs), but the mechanism has not been clarified. This study aimed to investigate the protective mechanism of DIPC on renal IRI in mice through Treg mediated by immature DCs (imDCs). Methods The IRI mice model, DIPC treatment, and conditional CD11c+ DCs (CD11c-DTR) knockout mice were used to perform our study. The maturation and differentiation of DCs and Treg cells in the kidney and spleen were analyzed by flow cytometry. HE staining was used to evaluate the pathology of the kidney tissue. The level of creatinine (Cr), oxidative stress factors (SOD, MDA), and inflammatory factors (TNF-α, IL-10, IL-4) were also measured. Then, imDCs were co-cultured with HK-2 cells, and apoptosis was analyzed with flow cytometry and PI-Hoechst 33,342 fluorescence staining to assess the apoptosis rate of HK-2 cells under hypoxic-reoxygenated (H/R) conditions. Results DIPC could decrease renal Cr levels, alleviate pathological renal damage, and reduce oxidative stress and inflammation caused by IRI. Moreover, DIPC could decrease the number of mature DCs (mDCs) and increase Treg lymphocyte infiltration in the kidney tissue, while the reduction of DCs reversed this process. In addition, our in vitro experiment found that in the H/R model, the apoptosis of HK-2 cells decreased which were co-cultured with imDCs. Conclusion DIPC can regulate the differentiation of DCs into imDCs, thus affecting the differentiation level and distribution of Treg cells to exert its protective effect on renal IRI.
Collapse
Affiliation(s)
- Wenjuan Yang
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China,Wuxi People's Hospital, Wuxi, China,Ningxia Medical University, Yinchuan, China
| | - Tao Peng
- Ningxia Medical University, Yinchuan, China
| | - Chunli Shi
- Ningxia Medical University, Yinchuan, China,Department of Molecular Biology, Shanghai Centre for Clinical Laboratory, Shanghai, China
| | - Fang Cui
- Ningxia Medical University, Yinchuan, China
| | - Menghua Chen
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China,Ningxia Medical University, Yinchuan, China,*Menghua Chen,
| | - Ting Zhang
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China,Ningxia Medical University, Yinchuan, China,*Menghua Chen,
| |
Collapse
|
9
|
Al-Lamki RS, Wang J, Pober JS, Bradley JR. Co-Expression and Functional Interactions of Death Receptor 3 and E-Selectin in Clear Cell Renal Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:722-736. [PMID: 35063404 DOI: 10.1016/j.ajpath.2021.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Similar to the behavior of inflamed tubular epithelial cells, clear cell renal cell carcinoma (ccRCC) cells express death receptor 3 (DR3 or TNFSFR25) in situ, and expression increases with tumor grade. Surprisingly, E-selectin, which can be induced in endothelial cells by DR3 signaling, is also expressed by ccRCC cells and increases with tumor grade. In ccRCC organ cultures, addition of tumor necrosis factor-like 1A (TL1A or TNFSF15), the ligand for DR3, activates NF-κB and mitogen-activated protein kinases, induces both DR3 and E-selectin expression in an NF-κB-dependent manner, and promotes cell cycle entry. DR3 immunoprecipitated from ccRCC tissue contains sialyl Lewis X moieties (the ligand recognized by E-selectin), proximity ligation assays reveal DR3, and E-selectin interacts on ccRCC cells. Similar to that with the addition of TL1A, the addition of soluble E-selectin to ccRCC organ cultures activates NF-κB and mitogen-activated protein kinases in ccRCC cells and increases both DR3 and E-selectin expression and cell-cycle entry. In contrast, normal renal tubular epithelium, which poorly expresses DR3, is minimally responsive to either of these ligands. These data suggest a functional role for autocrine/paracrine DR3/E-selectin interactions in ccRCC and its progression, revealing a potential new target for therapeutic intervention.
Collapse
Affiliation(s)
- Rafia S Al-Lamki
- Department of Medicine, National Institute of Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom.
| | - Jun Wang
- Department of Medicine, National Institute of Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Jordan S Pober
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | - John R Bradley
- Department of Medicine, National Institute of Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
10
|
Mi YH, Xu MY. Trauma-induced pulmonary thromboembolism: What's update? Chin J Traumatol 2022; 25:67-76. [PMID: 34404569 PMCID: PMC9039469 DOI: 10.1016/j.cjtee.2021.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 02/04/2023] Open
Abstract
Trauma-induced pulmonary thromboembolism is the second leading cause of death in severe trauma patients. Primary fibrinolytic hyperactivity combined with hemorrhage and consequential hypercoagulability in severe trauma patients create a huge challenge for clinicians. It is crucial to ensure a safe anticoagulant therapy for trauma patients, but a series of clinical issues need to be answered first, for example, what are the risk factors for traumatic venous thromboembolism? How to assess and determine the status of coagulation dysfunction of patients? When is the optimal timing to initiate pharmacologic prophylaxis for venous thromboembolism? What types of prophylactic agents should be used? How to manage the anticoagulation-related hemorrhage and to determine the optimal timing of restarting chemoprophylaxis? The present review attempts to answer the above questions.
Collapse
|
11
|
Qian H, Yang H, Li X, Yang G, Zheng X, He T, Li S, Liu B, Wu Y, Cheng Y, Shen F. Andrographolide sulfonate attenuates alveolar hypercoagulation and fibrinolytic inhibition partly via NF-κB pathway in LPS-induced acute respiratory distress syndrome in mice. Biomed Pharmacother 2021; 143:112209. [PMID: 34649343 DOI: 10.1016/j.biopha.2021.112209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Alveolar hypercoagulation and fibrinolytic inhibition are important characteristics during acute respiratory distress syndrome (ARDS), and NF-κB p65 signaling pathway is involved to regulate these pathophysiologies. We hypothesize that targeting NF-κB signal pathway could ameliorate alveolar hypercoagulation and fibrinolyitc inhibition, thus attenuating lung injury in ARDS. PURPOSE We explore the efficacy and the potential mechanism of andrographolide sulfonate (Andro-S) on alveolar hypercoagulation and fibrinolytic inhibition in LPS-induced ARDS in mice. METHODS ARDS was made by lipopolysaccharide (LPS) inhalation in C57BLmice. Andrographolide sulfonate (2.5, 5 and 10 mg/kg) was intraperitoneally given to the mice (once a day for three consecutive days) before LPS administration. NEMO binding domain peptide (NBD), an inhibitor of NF-κB, was used as the positive control and it replaced Andro-S in mice of NBD group. Mice in normal control received saline instead of LPS. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected for analysis of alveolar coagulation, fibrinolytic inhibition as well as of pulmonary inflammatory response after 8 h of LPS inhalation. NF-κB signal pathway in lung tissue was simultaneously determined. RESULTS Andro-S dose-dependently inhibited tissue factor (TF) and plasminogen activator inhibitor (PAI)-1 expressions either in mRNA or in protein in lung tissue of ARDS mice, and it also decreased the concentrations of TF, PAI-1, thrombin-antithrombin complex (TAT), procollagen peptide type Ⅲ (PⅢP) while promoting the production of activated protein C (APC) in BALF. Meanwhile, Andro-S effectively inhibited inflammatory response (interleukin 1β and myeloperoxidase) induced by LPS. LPS stimulation dramatically activated NF-κB signal pathway, indicated by increased expressions of phosphorylation of p65 (p-p65), p-IKKα/β and p-IκBα and the higher p65-DNA binding activity, which were all dose-dependently reversed by Andro-S. Andro-S and NBD presented similar efficacies. CONCLUSIONS Andro-S treatment improves alveolar hypercoagulation and fibrinolytic inhibition and attenuates pulmonary inflammation in LPS-induced ARDS in mice partly through NF-κB pathway inactivation. The drug is expected to be an effective choice for ARDS.
Collapse
Affiliation(s)
- Hong Qian
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China; Department of Intensive Care Unit, The Second People's Hospital of Guiyang, 550001, China.
| | - Huilin Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Xiang Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Guixia Yang
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Xinghao Zheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Tianhui He
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Shuwen Li
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Bo Liu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Yanqi Wu
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Yumei Cheng
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| | - Feng Shen
- Department of Intensive Care Unit, Guizhou Medical University Affiliated Hospital, Guiyang 550001, China.
| |
Collapse
|
12
|
PER2: a potential molecular marker for hematological malignancies. Mol Biol Rep 2021; 48:7587-7595. [PMID: 34642831 DOI: 10.1007/s11033-021-06751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/16/2021] [Indexed: 11/27/2022]
Abstract
Circadian rhythm is a periodic change of organism according to the law of external environment, which is manifested in metabolism, cell proliferation, physiology and behavior. In recent years, the role of circadian genes in the occurrence and progression of hematological malignancies have been continuously demonstrated. PER2 is the core component of the circadian rhythm playing an important role in regulating the circadian rhythm of the biological clock. This review summarizes the research progress of PER2 in hematological malignancies, especially leukemia, in order to better understand its role in hematological malignancies, and provide new ideas for clinical diagnosis and treatment.
Collapse
|
13
|
de Abreu JSS, Fernandes J. The contrast agent 2,3,5-triiodobenzoic acid (TIBA) induces cell death in tumor cells through the generation of reactive oxygen species. Mol Biol Rep 2021; 48:5199-5207. [PMID: 34196897 DOI: 10.1007/s11033-021-06524-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/25/2021] [Indexed: 11/28/2022]
Abstract
The 2,3,5-triiodobenzoic acid (TIBA) is an iodine contrast agent used for visualization of tissue in X-ray techniques. However, TIBA induces physiological complications like increase in oxygen reactive species (ROS), and consequently, contrast-induced nephropathies. TIBA's antitumor activity was demonstrated in lung cancer, but the subcellular mechanisms involving its activity in tumor cells are still unknown. Thus, the objective of this work was evaluate whether the anti-tumor activity of TIBA involves ROS increase, in tumor lines of non-small cell lung cancer (H460), chronic myeloid leukemia (K562), and its cytotoxicity in normal renal epithelial (VERO). The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide) assay was used for evaluation of cell viability, the H2DCFDA (cell-permeant 2',7'-dichlorodihydrofluorescein diacetate) fluorescent probe to evaluate ROS induction, cell cycle analysis was performed using flow cytometry to measure cell death, and immunofluorescence with annexin/7-AAD (7-amino-actinomycin D), to assess the association of cell death with the ROS generation. TIBA decreases cell viability in a dose-dependent manner for the H460 and K562. However, VERO cells showed less response to the drug, with 70% viable cells after 72 h of treatment in the highest concentration of the drug. While the tumor cells with only 20% viable cells. Besides, tumor cells exhibited higher DNA fragmentation, compared to the renal line (VERO with 5% of fragmented DNA, H460 with 26%, and 56% in K562). Finally, TIBA-induced ROS increase and apoptosis in all lines, which is significantly decreased after treatment with the antioxidant N-acetyl-cysteine (NAC). These data demonstrate the relationship between the increased cellular oxidative stress and the anti-tumor action of the TIBA.
Collapse
Affiliation(s)
- Jéssica Sodré Silva de Abreu
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Universidade Federal Do Rio de Janeiro (UFRJ), Rio de Janeiro, Duque de Caxias, Brazil
| | - Janaína Fernandes
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Universidade Federal Do Rio de Janeiro (UFRJ), Rio de Janeiro, Duque de Caxias, Brazil.
| |
Collapse
|