1
|
Xia T, Han F, Wang Y, Xie X, Yuan C, Lu G, Xiao W, Tu B, Ren H, Gong W, Wang Y. Inhibition of CD53 Reduces the Formation of ROS-Induced Neutrophil Extracellular Traps and Protects Against Inflammatory Injury in Acute Pancreatitis. J Inflamm Res 2025; 18:3725-3739. [PMID: 40098997 PMCID: PMC11913036 DOI: 10.2147/jir.s507886] [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: 11/21/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background The tetraspanin CD53 transmembrane protein is vital in immune cells like B cells and T cells, playing a crucial role in various inflammatory conditions. However, its involvement in neutrophils regarding inflammation remains uncertain. This study aims to examine the impact of CD53 on neutrophil extracellular traps (NETs) formation. Methods Phorbol 12-myristate 13-acetate (PMA) was utilized to establish an in vitro classical NETs model to investigate the influence of CD53 on NETs formation and its regulatory mechanisms. Subsequently, the link between CD53 and acute pancreatitis (AP), a model of aseptic inflammatory responses connected to NETs, was verified. Peripheral blood neutrophils from clinical AP patients were collected to explore the role of CD53 in AP, while an AP mouse model induced by caerulein was employed to confirm the impact of CD53 inhibition on AP mice pancreatic tissue. Results Our study has shown that CD53 is significantly elevated in in vitro NETs models and neutrophils from AP patients. The expression of CD53 is closely related to the clinical prognosis of AP patients. At the same time, CD53 neutralizing antibody (Anti-CD53) can significantly inhibit the formation of NETs in vitro, inflammatory injury in AP mice and the formation of NETs in damaged tissues. Mechanistically, CD53 can modulate the PI3K/AKT pathway and promote the formation of NETs. Finally, targeted regulation of CD53 can effectively reduce inflammatory injury and NETs formation in damaged tissues of AP mice. Conclusion The results of this study mark the first confirmation that CD53 plays a crucial role in NETs formation. Targeting CD53 inhibition could potentially serve as a novel therapeutic approach for the treatment of AP.
Collapse
Affiliation(s)
- Tianqi Xia
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Fei Han
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Yaning Wang
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Xinyue Xie
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Chenchen Yuan
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Guotao Lu
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Weiming Xiao
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Bo Tu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hongbo Ren
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, People's Republic of China
| | - Weijuan Gong
- Pancreatic Center, Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
- Yangzhou Key Laboratory of Pancreatic Disease, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Yaodong Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou Key Laboratory of Integrated Traditional Chinese and Western Medicine of Digestive Diseases, Kunshan Affiliated Hospital of Yangzhou University, Kunshan, 215300, People's Republic of China
| |
Collapse
|
2
|
Fu T, Zhou J, Yang L, Wang J, Li H, Shan Y, Gao H, Xie C, Jiang D, Zhang L, Ma J, Pan Q, Xu M, Zhang M, Gu S. Neutrophil-induced pyroptosis promotes survival in patients with hepatoblastoma. Cancer Immunol Immunother 2025; 74:106. [PMID: 39932547 PMCID: PMC11813845 DOI: 10.1007/s00262-024-03922-z] [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: 07/02/2024] [Accepted: 12/11/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND Hepatoblastoma (HB) is the predominant hepatic malignancy among children. Despite therapeutic options for HB were gradually refined in recent years, patients with metastasis suffer from an unsatisfactory prognosis. Pyroptosis is a type of programmed and inflammatory necrosis. Neutrophils are crucial in innate immunity, which were shown to be associated with tumor progression. Our study strived to unravel the relationship between neutrophil-induced pyroptosis (NIP) and HB. METHODS The clinical and bulk RNA sequencing data of 38 patients with HB were obtained from Shanghai Children's Medical Center. We established NIP score based on the LASSO regression. The single-cell RNA sequencing data (GSE186975) were used for for key genes identification, cellular communication, and differentiation trajectories of neutrophils. KEGG, GO, GSVA, and ssGSEA enrichment were used to analyze biological functions, including neutrophil extracellular traps (NETs), NOD-like receptors pathway, neutrophil activation, neutrophil-mediated cytotoxicity, and others. RESULTS We constructed a NIP score based on the expression of three genes related to neutrophil and pyroptosis, namely ELANE, CASP1, and NOD2, which was positively correlated with a favorable prognosis of HB. Moreover, we clarified the function of ELANE in HB microenvironmwnt. Immunohistochemistry and transcriptome analysis unraveled a significant correlation between NETs and pyroptosis in HB, suggesting the key role of NETs-related neutrophils in inducing pyroptosis and prolonging survival. We also found upregulated tumor-promoting and immunosuppression-related pathways in the HB microenvironment. In addition, we clarified the growth trajectories and phenotypic changes of neutrophils in the immune microenvironment of HB, which can serve as potential targets for immunotherapy. CONCLUSIONS The novel NIP score for patients with HB shows high predictive value for survival. Moreover, we identified biological function, cellular communication, and growth trajectories of neutrophils in HB. Our findings broaden insights into the treatment of HB.
Collapse
Affiliation(s)
- Tingyi Fu
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jiquan Zhou
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Liyuan Yang
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jing Wang
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center Affiliated to Shanghai, Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, People's Republic of China
| | - Yuhua Shan
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Hongxiang Gao
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Chenjie Xie
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Dapeng Jiang
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Lei Zhang
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Ji Ma
- Department of Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Qiuhui Pan
- Department of Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Min Xu
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center Affiliated to Shanghai, Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, People's Republic of China.
| | - Song Gu
- Department of General Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
3
|
Tang N, Li W, Shang H, Yang Z, Chen Z, Shi G. Irisin-mediated KEAP1 degradation alleviates oxidative stress and ameliorates pancreatitis. Immunol Res 2025; 73:37. [PMID: 39821708 DOI: 10.1007/s12026-024-09588-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Oxidative stress (OS) injury is pivotal in acute pancreatitis (AP) pathogenesis, contributing to inflammatory cascades. Irisin, a ubiquitous cytokine, exhibits antioxidant properties. However, the role of irisin in AP remains inconclusive. Our study aims to elucidate irisin expression in AP patients and investigate its mechanism of action to propose a novel treatment strategy for AP. Serum irisin levels in 65 AP patients were quantified using an enzyme-linked immunosorbent assay and correlated with disease severity scores. Core genes implicated in AP-related oxidative stress were identified and screened via bioinformatics analysis. The therapeutic efficacy of irisin in AP was confirmed using a murine cerulein-induced AP model. The intrinsic mechanism of irisin's antioxidative stress action was investigated and verified in pancreatic AR42J cells (Supplementary Fig. 1). Common targets shared by irisin and AP were further validated using a molecular docking model which was constructed for virtual docking analysis. This study investigated alterations in redox status in AP and found a significant reduction in serum irisin levels, correlating inversely with AP severity. In a murine AP model, we showed that irisin triggers an antioxidative stress program via the KEAP1 gene; this process helps reestablish redox balance by decreasing the buildup of reactive oxygen species (ROS) and suppressing the secretion of inflammatory mediators within pancreatic tissues Notably, increased KEAP1 expression counteracted the antioxidative effects of irisin. Our findings unveil a novel therapeutic mechanism for AP, wherein irisin inhibits KEAP1 to alleviate OS. Increasing irisin levels in vivo presents a promising strategy for AP treatment.
Collapse
Affiliation(s)
- Nan Tang
- Dalian Medical University, Dalian, Liaoning, China
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Wendi Li
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hezhen Shang
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zengyin Chen
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China.
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
4
|
Wen H, Yano N, Zhao T, Wei L, Zhao TC. The protective effect of irisin against hemorrhagic injury is mediated by PI3K and p38 pathways in hemorrhage/resuscitation. J Pharmacol Exp Ther 2025; 392:100027. [PMID: 39892988 DOI: 10.1124/jpet.124.002238] [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: 03/28/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
The objective of this study was to investigate whether phosphoinositide 3-kinase (PI3K) and p38 mitogen-activated kinase contribute to the protection of irisin during hemorrhage/resuscitation. Experimental groups were divided based on the different treatments during resuscitation as follows: (1) hemorrhage: adult male CD-1 mice were subjected to hemorrhage at a mean arterial blood pressure of 35-45 mm Hg for 60 minutes, followed by resuscitation with shed blood and lactated Ringer's solution (n = 13); (2) hemorrhage + irisin: receiving irisin (5 μg/kg; n = 13); (3) hemorrhage + irisin + PI3K inhibitor: receiving both Ly294002 (1 mg/kg, i.v.) and irisin (n = 6); and (4) hemorrhage + irisin + p38 inhibitor: receiving SB202190 (1 mg/kg, i.v.) and irisin (n = 6). Compared with hemorrhage/resuscitation control, irisin improved cardiac function and the recovery of hemodynamics in association with the decreased systemic interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-α, which were completely abrogated by PI3K or p38 inhibitions. Furthermore, the inhibition of PI3K or p38 abolished irisin-induced reduction of the inflammatory cell infiltration and terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling-positive apoptosis in the cardiac and skeletal muscles. Irisin reduced TNF-α and IL-6 expression in cardiac and skeletal muscles, which was abrogated by the inhibition of PI3K or p38. Irisin-treated hemorrhage increases the phosphorylation of PI3K and p38 in both cardiac and skeletal muscles, which was mitigated by the inhibition of PI3K or p38. PI3K and p38 play an important role in modulating the protective effect of irisin during the hemorrhage/resuscitation. SIGNIFICANCE STATEMENT: This study has identified a critical pathway in the regulation of trauma/hemorrhage by using a preclinical trauma model, in which irisin, as a hormone factor, stimulates PI3K and p38 pathways to induce protection against traumatic conditions. The study holds promise for developing a new therapeutic strategy to target irisin and its pathways related to PI3K and p38 to treat trauma and its comorbidities to reduce mortality for clinical implications.
Collapse
Affiliation(s)
- Huai Wen
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Naohiro Yano
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Thomas Zhao
- Department of Biology, Boston University, Boston, Massachusetts
| | - Lei Wei
- Department of Orthopedics, Rhode Island Hospital, Brown University, Providence, Rhode Island
| | - Ting C Zhao
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island; Department of Plastic Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
5
|
Dong X, Luo W, Wang Y, Zhu Q, Yuan C, Xiao W, Gong W, Lu G, Shi X, Li J. Role and mechanism of myonectin in severe acute pancreatitis: a crosstalk between skeletal muscle and pancreas. Skelet Muscle 2024; 14:29. [PMID: 39627746 PMCID: PMC11613728 DOI: 10.1186/s13395-024-00363-1] [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: 07/22/2024] [Accepted: 11/24/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is characterized by high mortality rates and various complications, including skeletal muscle atrophy, which significantly exacerbates its outcomes. Despite its clinical relevance, the mechanistic understanding of the relationship between skeletal muscle and the pancreas in SAP remains limited. Our study aimed to elucidate this "organ crosstalk" and its potential implications. METHODS We established an SAP mouse model through pancreatic duct ligation (PDL) and evaluated pancreatic necrosis, skeletal muscle atrophy, and myonectin expression levels. Recombinant myonectin protein was administered in vivo and in vitro to assess its effects on acinar cell necrosis. Mechanistic insights were gained through RNA-seq data analysis and experimental validation. Serum samples from AP patients and healthy controls were collected to investigate the correlation between serum myonectin levels and disease severity. RESULTS The mouse model exhibited severe pancreatic necrosis, skeletal muscle atrophy, and elevated myonectin levels, with myonectin administration exacerbating model severity. We identified iron accumulation-induced ferroptosis as a key pathway contributing to myonectin-mediated acinar cell necrosis. A total of 22 healthy controls and 52 patients with varying degrees of AP were included in the serum samples and clinical data (36.5% females, age 49.79 ± 16.53). Analysis of serum samples revealed significantly higher myonectin levels in AP patients, correlating with disease severity (R = 0.28, P = 0.041). CONCLUSIONS Our findings underscore the significant role of myonectin in SAP progression and its potential as a prognostic marker for disease severity in AP patients. This study contributes to a deeper understanding of the pathophysiology of SAP and highlights potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Xiaowu Dong
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weiwei Luo
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yaodong Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, Jiangsu, China
| | - Qingtian Zhu
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chenchen Yuan
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weiming Xiao
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weijuan Gong
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guotao Lu
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
- Laboratory of Intensive Care of Yangzhou, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xiaolei Shi
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Jin Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Shanxi Medical University, 56 Xin Jian Road, Tai Yuan, Shan Xi, China.
| |
Collapse
|
6
|
Wei X, Weng Z, Xu X, Yao J. Exploration of a miRNA-mRNA network shared between acute pancreatitis and Epstein-Barr virus infection by integrated bioinformatics analysis. PLoS One 2024; 19:e0311130. [PMID: 39546499 PMCID: PMC11567522 DOI: 10.1371/journal.pone.0311130] [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: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 11/17/2024] Open
Abstract
Acute pancreatitis (AP) stands out as a primary cause of hospitalization within gastrointestinal ailments, attributed to diverse factors, including Epstein-Barr virus (EBV) infection. Nevertheless, the common miRNAs and genes shared between AP and EBV infection remain unclear. In the present study, four datasets GSE194331, GSE42455, GSE45918 and GSE109220 were selected and downloaded from the Gene Expression Omnibus (GEO) database. Differential expression analysis was performed to screen for differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs). Target genes of overlapping DEMs were predicted, and intersections with overlapping DEGs were used to construct a miRNA-mRNA network. In addition, the enrichment analysis, drug prediction, diagnostic accuracy assessment, competitive endogenous RNA (ceRNA) network construction, transcription factor (TF)-miRNA-mRNA network construction, and immune cell infiltration analysis were also carried out. We found a total of 111 genes and 8 miRNAs shared between AP and EBV infection. A miRNA-mRNA network was constructed, which comprised 5 miRNAs and 10 genes exhibiting robust diagnostic performance. Histone deacetylase (HDAC) inhibitor was identified as a novel therapeutic intervention from drug prediction analysis. The results of immune cell infiltration analysis revealed that a consistent and significant difference could be found on activated B cell in AP and EBV-infected individuals in comparison to the controls. Taken together, our work, for the first time, revealed a miRNA-mRNA network shared between AP and EBV infection, thereby enriching a deeper comprehension of the intricate molecular mechanisms and potential therapeutic targets entwined in these two pathological conditions.
Collapse
Affiliation(s)
- Xing Wei
- Department of Infectious Disease, The Nantong First People’s Hospital and The Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Zhen Weng
- MOE Engineering Center of Hematological Disease, Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xia Xu
- Department of Gastroenterology, The Second People’s Hospital of Nantong and The Affiliated Rehabilitation Hospital of Nantong University, Nantong, China
| | - Jian Yao
- Department of Infectious Disease, The Nantong First People’s Hospital and The Affiliated Hospital 2 of Nantong University, Nantong, China
| |
Collapse
|
7
|
Zhang H, Wang Z, Li J, Jia Y, Li F. Timing, initiation and function: An in-depth exploration of the interaction network among neutrophil extracellular traps related genes in acute pancreatitis. Int Immunopharmacol 2024; 141:112923. [PMID: 39137629 DOI: 10.1016/j.intimp.2024.112923] [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: 06/23/2024] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Exogenous inhibition of neutrophil extracellular traps (NETs) was believed to alleviate acute pancreatitis (AP). This study aimed to comprehensively explore the key biological behavior of NETs including timing and pathogenesis in AP by integrating of single cell RNA sequencing(scRNA-seq) and bulk RNA-seq. METHODS Differentially expressed NETs-related genes and the hub genes of NETs were screened by bulk RNA-seq. ScRNA-seq was used to identify the cell types in pancreas of AP mice and to depict the transcriptomic maps in neutrophils. The mouse AP models were build to verify the timing of initiation of NETs and underlying pathogenesis of damage on pancreas acinar cells. RESULTS Tlr4 and Ccl3 were screened for hub genes by bulk RNA-seq. The trajectory analysis of neutrophils showed that high expression of Ccl3, Cybb and Padi4 can be observed in the middle stage during AP. Macrophages might be essential in the biological behavior of neutrophils and NETs. Through animal models, we presented that extensive NETs structures were formed at mid-stage of inflammation, accompanied by more serious pancreas and lung damage. NETs might promote necroptosis and macrophage infiltration in AP, and the damage on pancreatic injury could be regulated by Tlr4 pathway. Ccl3 was considered to recruit neutrophils and promote NETs formation. CONCLUSION The findings explored the underlying timing and pathogenesis of NETs in AP for the first time, which provided gene targets for further studies.
Collapse
Affiliation(s)
- Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, PR China; Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, PR China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, PR China; Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, PR China
| | - Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, PR China; Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, PR China
| | - Yuchen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, PR China; Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, PR China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, PR China; Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, PR China.
| |
Collapse
|
8
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024; 38:5067-5087. [PMID: 39105461 DOI: 10.1002/ptr.8311] [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: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Jin Y, Wang Y, Ma X, Li H, Zhang M. Identification of NET formation and the renoprotective effect of degraded NETs in lupus nephritis. Am J Physiol Renal Physiol 2024; 327:F637-F654. [PMID: 39205658 PMCID: PMC11483074 DOI: 10.1152/ajprenal.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
To explore molecular biomarkers associated with the pathophysiology and therapy of lupus nephritis (LN), we conducted a joint analysis of transcriptomic data from 40 peripheral blood mononuclear cells (PBMCs) (GSE81622) and 21 kidney samples (GSE112943) from the Gene Expression Omnibus database using bioinformatics. A total of 976 and 2,427 differentially expressed genes (DEGs) were identified in PBMCs and renal tissues. Seven and two functional modules closely related to LN were identified. Further enrichment analysis revealed that the neutrophil activation pathway was highly active in both PBMCs and the kidney. Subsequently, 16 core genes closely associated with LN were verified by protein-protein interaction screening and quantitative PCR. In vitro cell models and MRL/lpr mouse models confirmed that the abnormal expression of these core genes was closely linked to neutrophil extracellular traps (NETs) generated by neutrophil activation, while degradation of NETs led to downregulation of core gene expression, thereby improving pathological symptoms of LN. Therefore, identification of patients with systemic lupus erythematosus exhibiting abnormal expression patterns for these core genes may serve as a useful indicator for kidney involvement. In addition, targeting neutrophils to modulate their activation levels and inhibit aberrant expression of these genes represents a potential therapeutic strategy for treating LN. NEW & NOTEWORTHY The mechanisms by which immune cells cause kidney injury in lupus nephritis are poorly understood. We integrated and analyzed the transcriptomic features of PBMCs and renal tissues from the GEO database to identify key molecular markers associated with neutrophil activation. We confirmed that neutrophil extracellular traps (NETs) formed by neutrophil activation promoted the upregulation of key genes in cell and animal models. Targeted degradation of NETs significantly ameliorated kidney injury in MRL/lpr mice.
Collapse
Affiliation(s)
- Yong Jin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yutong Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xu Ma
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hongbin Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Manling Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
10
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
11
|
Zhang Q, Xiang S, Chen X, Rong Y, Huang L, Chen Z, Yao K, Chen W, Deng C, Wang J. Irisin attenuates acute glaucoma-induced neuroinflammation by activating microglia-integrin αVβ5/AMPK and promoting autophagy. Int Immunopharmacol 2024; 138:112545. [PMID: 38955026 DOI: 10.1016/j.intimp.2024.112545] [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: 12/05/2023] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Neuroinflammation, characterized by microglial activation and the release of multiple inflammatory mediators, is a key factor in acute glaucomatous injury leading to retinal ganglion cell (RGC) death and ultimately irreversible vision loss. Irisin, a novel exercise-induced myokine, has demonstrated anti-inflammatory activity in ischemia/reperfusion injuries across multiple organs and has displayed a significant neuroprotective role in experimental stroke disease models. This study examined the protective impact of irisin and investigated its potential mechanism involved in this process utilizing an acute ocular hypertension (AOH)-induced retinal injury model in mice and a microglia inflammation model induced by lipopolysaccharide (LPS). There was a transient downregulation of irisin in the retina after AOH injury, with parallel emergence of retinal neuroinflammation and RGC death. Irisin attenuated retinal and optic nerve damage and promotes the phenotypic conversion of microglia from M1 to M2. Mechanistically, irisin significantly upregulated the expression of integrin αVβ5, p-AMPK, and autophagy-related markers. Integrin αVβ5 was highly expressed on microglia but hardly expressed on RGC. The integrin αVβ5 inhibitor cilengitide, the AMPK inhibitor dorsomorphin, and the autophagy inhibitor 3-Methyladenine (3-MA) blocked the neuroprotective effects of irisin. Our results suggest irisin attenuates acute glaucoma-induced neuroinflammation and RGC death by activating integrin αVβ5/AMPK in microglia and promoting autophagy. It should be considered a potential neuroprotective therapy for acute glaucoma.
Collapse
Affiliation(s)
- Qiuxiang Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Sifei Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lan Huang
- Department of Ophthalmology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443000, China
| | - Zhiqi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ke Yao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chaohua Deng
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Junming Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
12
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
13
|
Wang Z, Xu J, Mo L, Zhan R, Zhang J, Liu L, Jiang J, Zhang Y, Bai Y. The Application Potential of the Regulation of Tregs Function by Irisin in the Prevention and Treatment of Immune-Related Diseases. Drug Des Devel Ther 2024; 18:3005-3023. [PMID: 39050796 PMCID: PMC11268596 DOI: 10.2147/dddt.s465713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Irisin is a muscle factor induced by exercise, generated through the proteolytic cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC-5). Numerous studies have shown that irisin plays a significant role in regulating glucose and lipid metabolism, inhibiting oxidative stress, reducing systemic inflammatory responses, and providing neuroprotection. Additionally, irisin can exert immunomodulatory functions by regulating regulatory T cells (Tregs). Tregs are a highly differentiated subset of mature T cells that play a key role in maintaining self-immune homeostasis and are closely related to infections, inflammation, immune-related diseases, and tumors. Irisin exerts persistent positive effects on Treg cell functions through various mechanisms, including regulating Treg cell differentiation and proliferation, improving their function, modulating the balance of immune cells, increasing the production of anti-inflammatory cytokines, and enhancing metabolic functions, thereby helping to maintain immune homeostasis and prevent immune-related diseases. As an important myokine, irisin interacts with receptors on the cell membrane, activating multiple intracellular signaling pathways to regulate cell metabolism, proliferation, and function. Although the specific receptor for irisin has not been fully identified, integrins are considered potential receptors. Irisin activates various signaling pathways, including AMPK, MAPK, and PI3K/Akt, through integrin receptors, thereby exerting multiple biological effects. These research findings provide important clues for understanding the mechanisms of irisin's action and theoretical basis for its potential applications in metabolic diseases and immunomodulation. This article reviews the relationship between irisin and Tregs, as well as the research progress of irisin in immune-related diseases such as multiple sclerosis, myasthenia gravis, acquired immune deficiency syndrome, type 1 diabetes, sepsis, and rheumatoid arthritis. Studies have revealed that irisin plays an important role in immune regulation by improving the function of Tregs, suggesting its potential application value in the treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zhengjiang Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jiaqi Xu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Liqun Mo
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Renshu Zhan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Yingying Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| | - Yiping Bai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
| |
Collapse
|
14
|
Liu Q, Zhu X, Guo S. From pancreas to lungs: The role of immune cells in severe acute pancreatitis and acute lung injury. Immun Inflamm Dis 2024; 12:e1351. [PMID: 39023414 PMCID: PMC11256889 DOI: 10.1002/iid3.1351] [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: 03/28/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is a potentially lethal inflammatory pancreatitis condition that is usually linked to multiple organ failure. When it comes to SAP, the lung is the main organ that is frequently involved. Many SAP patients experience respiratory failure following an acute lung injury (ALI). Clinicians provide insufficient care for compounded ALI since the underlying pathophysiology is unknown. The mortality rate of SAP patients is severely impacted by it. OBJECTIVE The study aims to provide insight into immune cells, specifically their roles and modifications during SAP and ALI, through a comprehensive literature review. The emphasis is on immune cells as a therapeutic approach for treating SAP and ALI. FINDINGS Immune cells play an important role in the complicated pathophysiology ofSAP and ALI by maintaining the right balance of pro- and anti-inflammatory responses. Immunomodulatory drugs now in the market have low thepeutic efficacy because they selectively target one immune cell while ignoring immune cell interactions. Accurate management of dysregulated immune responses is necessary. A critical initial step is precisely characterizing the activity of the immune cells during SAP and ALI. CONCLUSION Given the increasing incidence of SAP, immunotherapy is emerging as a potential treatment option for these patients. Interactions among immune cells improve our understanding of the intricacy of concurrent ALI in SAP patients. Acquiring expertise in these domains will stimulate the development of innovative immunomodulation therapies that will improve the outlook for patients with SAP and ALI.
Collapse
Affiliation(s)
- Qi Liu
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| | - Xiaomei Zhu
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| |
Collapse
|
15
|
Yin N, Xu B, Huang Z, Fu Y, Huang H, Fan J, Huang C, Mei Q, Zeng Y. Inhibition of Pck1 in intestinal epithelial cells alleviates acute pancreatitis via modulating intestinal homeostasis. FASEB J 2024; 38:e23618. [PMID: 38651689 DOI: 10.1096/fj.202400039r] [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: 01/06/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Intestinal barrier dysfunction usually occurred in acute pancreatitis (AP) but the mechanism remains unclear. In this study, RNA sequencing of ileum in L-arginine-induced AP mice demonstrated that phosphoenolpyruvate kinase 1 (Pck1) was significantly up-regulated. Increased Pck1 expression in intestinal epithelial cells (IECs) was further validated in ileum of AP mice and duodenum of AP patients. In AP mice, level of Pck1 was positively correlated with pancreatic and ileal histopathological scores, serum amylase activity, and intestinal permeability (serum diamine oxidase (DAO), D-lactate, and endotoxin). In AP patients, level of Pck1 had a positive correlation with Ranson scores, white blood cell count and C-reactive protein. Inhibition of Pck1 by 3-Mercaptopicolinic acid hydrochloride (3-MPA) alleviated pancreatic and ileal injuries in AP mice. AP + 3-MPA mice showed improved intestinal permeability, including less epithelial apoptosis, increased tight junction proteins (TJPs) expression, decreased serum DAO, D-lactate, endotoxin, and FITC-Dextran levels, and reduced bacteria translocation. Lysozyme secreted by Paneth cells and mucin2 (MUC2) secretion in goblet cells were also partly restored in AP + 3-MPA mice. Meanwhile, inhibition of Pck1 improved intestinal immune response during AP, including elevation of M2/M1 macrophages ratio and secretory immunoglobulin A (sIgA) and reduction in neutrophils infiltration. In vitro, administration of 3-MPA dramatically ameliorated inflammation and injuries of epithelial cells in enteroids treated by LPS. In conclusion, inhibition of Pck1 in IECs might alleviate AP via modulating intestinal homeostasis.
Collapse
Affiliation(s)
- Nuoming Yin
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Binqiang Xu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zehua Huang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yang Fu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Huizheng Huang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Junjie Fan
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chunlan Huang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qixiang Mei
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yue Zeng
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Wang S, Hu S, Pan Y. The emerging roles of irisin in vascular calcification. Front Endocrinol (Lausanne) 2024; 15:1337995. [PMID: 38405155 PMCID: PMC10884194 DOI: 10.3389/fendo.2024.1337995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Vascular calcification is a common accompanying pathological change in many chronic diseases, which is caused by calcium deposition in the blood vessel wall and leads to abnormal blood vessel function. With the progress of medical technology, the diagnosis rate of vascular calcification has explosively increased. However, due to its mechanism's complexity, no effective drug can relieve or even reverse vascular calcification. Irisin is a myogenic cytokine regulating adipose tissue browning, energy metabolism, glucose metabolism, and other physiological processes. Previous studies have shown that irisin could serve as a predictor for vascular calcification, and protect against hypertension, diabetes, chronic kidney disease, and other risk factors for vascular calcification. In terms of mechanism, it improves vascular endothelial dysfunction and phenotypic transformation of vascular smooth muscle cells. All the above evidence suggests that irisin plays a predictive and protective role in vascular calcification. In this review, we summarize the association of irisin to the related risk factors for vascular calcification and mainly explore the role of irisin in vascular calcification.
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Cardiology, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Siwang Hu
- The Orthopaedic Center, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Yuping Pan
- Department of Internal Medicine, Yuhuan Second People’s Hospital, Yuhuan, Zhejiang, China
| |
Collapse
|
17
|
Dong H, Lv X, Gao P, Hao Y. Potential role of irisin in lung diseases and advances in research. Front Pharmacol 2023; 14:1307651. [PMID: 38143500 PMCID: PMC10746167 DOI: 10.3389/fphar.2023.1307651] [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: 10/05/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Irisin, a myokine, is secreted by the movement of skeletal muscles. It plays an important role in metabolic homeostasis, insulin resistance, anti-inflammation, oxidative stress, and bone metabolism. Several studies have reported that irisin-related signaling pathways play a critical role in the treatment of various diseases, including obesity, cardiovascular disease, diabetes, and neurodegenerative disorders. Recently, the potential role of irisin in lung diseases, including chronic obstructive pulmonary disease, acute lung injury, lung cancer, and their associated complications, has received increasing attention. This article aims to explore the role of irisin in lung diseases, primarily focusing on the underlying molecular mechanisms, which may serve as a marker for the diagnosis as well as a potential target for the treatment of lung diseases, thus providing new strategies for their treatment.
Collapse
Affiliation(s)
| | | | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yuqiu Hao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
18
|
Zhang Y, Zhao L, Gao H, Zhai J, Song Y. Potential role of irisin in digestive system diseases. Biomed Pharmacother 2023; 166:115347. [PMID: 37625325 DOI: 10.1016/j.biopha.2023.115347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Digestive system diseases (DSD) are very complex conditions that severely threaten human health. Therefore, there is an urgent need to develop new pharmacological treatment strategies. Irisin, a myokine discovered in 2012, is produced by fibronectin type III domain-containing protein 5 (FNDC5), which is a transmembrane protein. Irisin is involved in promoting the browning of white adipose tissue, the regulation of energy metabolism, and the improvement of insulin resistance. Irisin is also an essential mediator of the inflammatory response, oxidative stress, and cell apoptosis. Recent studies have proved that irisin concentration is altered in DSD and exerts pivotal effects on the initiation, progression, and prognosis of these diseases through various mechanisms. Therefore, studying the expression and function of irisin may have great significance for the diagnosis and treatment of DSD. Here, we focus on irisin and explore the multiple molecular pathways targeted by irisin therapy. This review indicates that irisin can serve as a diagnostic marker or potential therapeutic agent for DSD. DATA AVAILABILITY: Not applicable.
Collapse
Affiliation(s)
- Yueming Zhang
- Department of Clinical Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Linxian Zhao
- Department of General Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Huan Gao
- Department of Clinical Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Jinghui Zhai
- Department of Clinical Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Yanqing Song
- Department of Pharmacy, Lequn Branch, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|