1
|
Kikuchi K, Kazuma S, Yamakage M. Recombinant thrombomodulin and recombinant antithrombin attenuate pulmonary endothelial glycocalyx degradation and neutrophil extracellular trap formation in ventilator-induced lung injury in the context of endotoxemia. Respir Res 2024; 25:330. [PMID: 39227918 PMCID: PMC11373098 DOI: 10.1186/s12931-024-02958-0] [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: 06/06/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Vascular endothelial damage is involved in the development and exacerbation of ventilator-induced lung injury (VILI). Pulmonary endothelial glycocalyx and neutrophil extracellular traps (NETs) are endothelial protective and damaging factors, respectively; however, their dynamics in VILI and the effects of recombinant thrombomodulin and antithrombin on these dynamics remain unclear. We hypothesized that glycocalyx degradation and NETs are induced by VILI and suppressed by recombinant thrombomodulin, recombinant antithrombin, or their combination. METHODS VILI was induced in male C57BL/6J mice by intraperitoneal lipopolysaccharide injection (20 mg/kg) and high tidal volume ventilation (20 mL/kg). In the intervention groups, recombinant thrombomodulin, recombinant antithrombin, or their combination was administered at the start of mechanical ventilation. Glycocalyx degradation was quantified by measuring serum syndecan-1, fluorescence-labeled lectin intensity, and glycocalyx-occupied area in the pulmonary vascular lumen. Double-stranded DNA in the bronchoalveolar fluid and fluorescent areas of citrullinated histone H3 and myeloperoxidase were quantified as NET formation. RESULTS Serum syndecan-1 increased, and lectin fluorescence intensity decreased in VILI. Electron microscopy revealed decreases in glycocalyx-occupied areas within pulmonary microvessels in VILI. Double-stranded DNA levels in the bronchoalveolar lavage fluid and the fluorescent area of citrullinated histone H3 and myeloperoxidase in lung tissues increased in VILI. Recombinant thrombomodulin, recombinant antithrombin, and their combination reduced glycocalyx injury and NET marker levels. There was little difference in glycocalyx injury and NET makers between the intervention groups. CONCLUSION VILI induced glycocalyx degradation and NET formation. Recombinant thrombomodulin and recombinant antithrombin attenuated glycocalyx degradation and NETs in our VILI model. The effect of their combination did not differ from that of either drug alone. Recombinant thrombomodulin and antithrombin have the potential to be therapeutic agents for biotrauma in VILI.
Collapse
Affiliation(s)
- Kenichiro Kikuchi
- Department of Anesthesiology, School of Medicine, Sapporo Medical University, S-1, W-16 Chuo-ku, Sapporo, 060-8543, Hokkaido, Japan
| | - Satoshi Kazuma
- Department of Intensive Care Medicine, School of Medicine, Sapporo Medical University, S-1, W-16 Chuo-ku, Sapporo, 060-8543, Hokkaido, Japan.
| | - Michiaki Yamakage
- Department of Anesthesiology, School of Medicine, Sapporo Medical University, S-1, W-16 Chuo-ku, Sapporo, 060-8543, Hokkaido, Japan
| |
Collapse
|
2
|
Lou J, Zhang J, Deng Q, Chen X. Neutrophil extracellular traps mediate neuro-immunothrombosis. Neural Regen Res 2024; 19:1734-1740. [PMID: 38103239 PMCID: PMC10960287 DOI: 10.4103/1673-5374.389625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/14/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophil extracellular traps are primarily composed of DNA and histones and are released by neutrophils to promote inflammation and thrombosis when stimulated by various inflammatory reactions. Neutrophil extracellular trap formation occurs through lytic and non-lytic pathways that can be further classified by formation mechanisms. Histones, von Willebrand factor, fibrin, and many other factors participate in the interplay between inflammation and thrombosis. Neuro-immunothrombosis summarizes the intricate interplay between inflammation and thrombosis during neural development and the pathogenesis of neurological diseases, providing cutting-edge insights into post-neurotrauma thrombotic events. The blood-brain barrier defends the brain and spinal cord against external assaults, and neutrophil extracellular trap involvement in blood-brain barrier disruption and immunothrombosis contributes substantially to secondary injuries in neurological diseases. Further research is needed to understand how neutrophil extracellular traps promote blood-brain barrier disruption and immunothrombosis, but recent studies have demonstrated that neutrophil extracellular traps play a crucial role in immunothrombosis, and identified modulators of neuro-immunothrombosis. However, these neurological diseases occur in blood vessels, and the mechanisms are unclear by which neutrophil extracellular traps penetrate the blood-brain barrier to participate in immunothrombosis in traumatic brain injury. This review discusses the role of neutrophil extracellular traps in neuro-immunothrombosis and explores potential therapeutic interventions to modulate neutrophil extracellular traps that may reduce immunothrombosis and improve traumatic brain injury outcomes.
Collapse
Affiliation(s)
- Jianbo Lou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
3
|
Yamamoto H, Ishida Y, Zhang S, Osako M, Nosaka M, Kuninaka Y, Ishigami A, Iwahashi Y, Aragane M, Matsumoto L, Kimura A, Kondo T. Protective roles of thrombomodulin in cisplatin-induced nephrotoxicity through the inhibition of oxidative and endoplasmic reticulum stress. Sci Rep 2024; 14:14004. [PMID: 38890434 PMCID: PMC11189513 DOI: 10.1038/s41598-024-64619-y] [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: 08/30/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Cisplatin is an effective chemotherapeutic agent widely used for the treatment of various solid tumors. However, cisplatin has an important limitation in its use; currently, there is no method to ameliorate cisplatin-induced acute kidney injury (AKI). Thrombomodulin (TM) is well known not only for its role as a cofactor in the clinically important natural anticoagulation pathway but also for its anti-inflammatory properties. Here, we investigated the effects of TM in cisplatin-induced AKI. In mice intraperitoneally injected with 15 mg/kg cisplatin, TM (10 mg/kg) or PBS was administered intravenously at 24 h after cisplatin injection. TM significantly attenuated cisplatin-induced nephrotoxicity with the suppressed elevation of blood urea nitrogen and serum creatinine, and reduced histological damages. Actually, TM treatment significantly alleviated oxidative stress-induced apoptosis by reducing reactive oxygen species (ROS) levels in cisplatin-treated renal proximal tubular epithelial cells (RPTECs) in vitro. Furthermore, TM clarified cisplatin-induced apoptosis by reducing caspase-3 levels. In addition, TM attenuated the endoplasmic reticulum (ER) stress signaling pathway in both renal tissues and RPTECs to protect the kidneys from cisplatin-induced AKI. These findings suggest that TM is a potential protectant against cisplatin-induced nephrotoxicity through suppressing ROS generation and ER stress in response to cisplatin.
Collapse
Affiliation(s)
- Hiroki Yamamoto
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| | - Siying Zhang
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Miyu Osako
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Akiko Ishigami
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuya Iwahashi
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Miki Aragane
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Lennon Matsumoto
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| |
Collapse
|
4
|
Moriyama E, Nadatani Y, Higashimori A, Otani K, Ominami M, Fukunaga S, Hosomi S, Tanaka F, Taira K, Fujiwara Y, Watanabe T. Neutrophil extracellular trap formation and its implications in nonsteroidal anti-inflammatory drug-induced small intestinal injury. J Gastroenterol Hepatol 2024; 39:1123-1133. [PMID: 38576269 DOI: 10.1111/jgh.16543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/01/2024] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND AND AIM Nonsteroidal anti-inflammatory drugs (NSAIDs) damage the small intestine via neutrophil infiltration driven by the mucosal invasion of enterobacteria. The antimicrobial function of neutrophils is partially dependent on neutrophil extracellular traps (NETs). Excessive NET formation has been associated with several inflammatory diseases. Here, we aimed to investigate the role of NETs in NSAID-induced small intestinal damage using human samples and an experimental mouse model. METHODS Human small intestine specimens were obtained from NSAID users during double-balloon enteroscopy. Wild-type, protein arginine deiminase 4 (PAD4) knockout, and antibiotic-treated mice were administered indomethacin to induce small intestinal injury. The expression of NET-associated proteins, including PAD4, citrullinated histone H3 (CitH3), cell-free DNA, and myeloperoxidase (MPO), was evaluated. RESULTS The double-positive stained area with CitH3 and MPO, which is specific for neutrophil-derived extracellular traps, was significantly high in the injured small intestinal mucosa of NSAID users. In a mouse model, small intestinal damage developed at 6 h after indomethacin administration, accompanied by increased mRNA levels of interleukin-1β and keratinocyte chemoattractant and elevated NET-associated protein levels of PAD4, CitH3, and MPO in small intestine and serum levels of cell-free DNA. Both genetic deletion and pharmacological inhibition of PAD4 attenuated this damage by reducing the mRNA expression of inflammatory cytokines and NET-associated proteins. Furthermore, mice pretreated with antibiotics showed resistance to indomethacin-induced small intestinal damage, with less NET formation. CONCLUSION These results suggest that NETs aggravate NSAID-induced small intestinal injury. Therefore, NET inhibition could be a potential treatment for NSAID-induced small intestinal injury.
Collapse
Affiliation(s)
- Eiji Moriyama
- Department of Gastroenterology, Graduate School of Medicine, Osaka city University, Osaka, Japan
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Higashimori
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Koji Otani
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Masaki Ominami
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shusei Fukunaga
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Koichi Taira
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Toshio Watanabe
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
5
|
Yin L, Bing Z, Zheng Y, Pan Y, Dong Y, Wang J, Luo R, Zhao Y, Dou H, Hou Y. Oroxylin A-induced Trained Immunity Promotes LC3-associated Phagocytosis in Macrophage in Protecting Mice Against Sepsis. Inflammation 2024:10.1007/s10753-024-02033-2. [PMID: 38739341 DOI: 10.1007/s10753-024-02033-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
Sepsis is defined as a dysregulated host response to infection that leads to multiorgan failure. Innate immune memory, i.e., "trained immunity", can result in stronger immune responses and provide protection against various infections. Many biological agents, including β-glucan, can induce trained immunity, but these stimuli may cause uncontrolled inflammation. Oroxylin A (OA) is an active flavonoid compound that is derived from Scutellaria baicalensis. OA is an agonist for inducing trained immunity in vivo and in vitro, and β-glucan was used as a positive control. The protective effects of OA-induced trained immunity were evaluated in mouse models that were established by either lipopolysaccharide (LPS) administration or caecal ligation and puncture (CLP). The expression of inflammatory factors and signaling pathway components involved in trained immunity was evaluated in vitro using qRT‒PCR, western blotting (WB) and enzyme-linked immunosorbent assay (ELISA). Flow cytometry and confocal microscopy were used to examine reactive oxygen species (ROS) levels and phagocytosis in trained macrophages. A PCR array was used to screen genes that were differentially expressed in trained macrophages. Here, we revealed that OA alleviated sepsis via trained immunity. OA-treated macrophages displayed increased glycolysis and mTOR phosphorylation, and mTOR inhibitors suppressed OA-induced trained immunity by effectively reprogramming macrophages. The PCR array revealed key genes in the mTOR signaling pathway in OA-treated macrophages. Furthermore, OA targeted the Dectin-1-syk axis to promote LC3-associated phagocytosis (LAP) by trained macrophages, thereby enhancing the ability of these macrophages to protect against infection. This ability could be transferred to a new host via the adoptive transfer of peritoneal macrophages. This study is the first to provide new insights into the potential of OA-induced trained immunity to be used as a strategy to protect mice against sepsis by promoting LAP by macrophages.
Collapse
Affiliation(s)
- Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Ziqian Bing
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yaojun Zheng
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuchen Pan
- Jiangsu International Laboratory of Immunity and Metabolism, The Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yue Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jiali Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Renjie Luo
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yue Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| |
Collapse
|
6
|
Chu C, Wang X, Chen F, Yang C, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Li J, Ding W. Neutrophil extracellular traps aggravate intestinal epithelial necroptosis in ischaemia-reperfusion by regulating TLR4/RIPK3/FUNDC1-required mitophagy. Cell Prolif 2024; 57:e13538. [PMID: 37691112 PMCID: PMC10771116 DOI: 10.1111/cpr.13538] [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: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Neutrophil extracellular trap (NET) has been confirmed to be related to gut barrier injury during intestinal ischaemia-reperfusion (II/R). However, the specific molecular regulatory mechanism of NETs in II/R-induced intestinal barrier damage has yet to be fully elucidated. Here, we reported increased NETs infiltration accompanied by elevated inflammatory cytokines, cellular necroptosis and tight junction disruption in the intestine of human II/R patients. Meanwhile, NETs aggravated Caco-2 intestinal epithelial cell necroptosis, impairing the monolayer barrier in vitro. Moreover, Pad4-deficient mice were used further to validate the role of NETs in II/R-induced intestinal injury. In contrast, NET inhibition via Pad4 deficiency alleviated intestinal inflammation, attenuated cellular necroptosis, improved intestinal permeability, and enhanced tight junction protein expression. Notably, NETs prevented FUN14 domain-containing 1 (FUNDC1)-required mitophagy activation in intestinal epithelial cells, and stimulating mitophagy attenuated NET-associated mitochondrial dysfunction, cellular necroptosis, and intestinal damage. Mechanistically, silencing Toll-like receptor 4 (TLR4) or receptor-interacting protein kinase 3 (RIPK3) via shRNA relieved mitophagy limitation, restored mitochondrial function and reduced NET-induced necroptosis in Caco-2 cells, whereas this protective effect was reversed by TLR4 or RIPK3 overexpression. The regulation of TLR4/RIPK3/FUNDC1-required mitophagy by NETs can potentially induce intestinal epithelium necroptosis.
Collapse
Affiliation(s)
- Chengnan Chu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Xinyu Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Fang Chen
- Division of Trauma and Acute Care Surgery, Jinling Hospital, School of MedicineSoutheast UniversityNanjingJiangsu ProvinceChina
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical EngineeringNanjing University of Science and TechnologyNanjingJiangsu ProvinceChina
| | - Weiqi Xu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Chenyang Wang
- Key Laboratory of Intestinal Injury, Research Institute of General Surgery, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingJiangsuChina
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical EngineeringNanjing University of Science and TechnologyNanjingJiangsu ProvinceChina
| | - Jieshou Li
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu ProvinceChina
- Division of Trauma and Acute Care Surgery, Jinling Hospital, School of MedicineSoutheast UniversityNanjingJiangsu ProvinceChina
| |
Collapse
|
7
|
Zhang W, Zhou B, Yang X, Zhao J, Hu J, Ding Y, Zhan S, Yang Y, Chen J, Zhang F, Zhao B, Deng F, Lin Z, Sun Q, Zhang F, Yao Z, Liu W, Li C, Liu KX. Exosomal circEZH2_005, an intestinal injury biomarker, alleviates intestinal ischemia/reperfusion injury by mediating Gprc5a signaling. Nat Commun 2023; 14:5437. [PMID: 37673874 PMCID: PMC10482849 DOI: 10.1038/s41467-023-41147-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a severe clinical condition without optimal diagnostic markers nor clear molecular etiological insights. Plasma exosomal circular RNAs (circRNAs) are valuable biomarkers and therapeutic targets for various diseases, but their role in intestinal I/R injury remains unknown. Here we screen the expression profile of circRNAs in intestinal tissue exosomes collected from intestinal I/R mice and identify circEZH2_005 as a significantly downregulated exosomal circRNA. In parallel, circEZH2_005 is also reduced in the plasma of clinical cardiac surgery patients who developed postoperative intestinal I/R injury. Exosomal circEZH2_005 displays a significant diagnostic value for intestinal injury induced by I/R. Mechanistically, circEZH2_005 is highly expressed in intestinal crypt cells. CircEZH2_005 upregulation promotes the proliferation of Lgr5+ stem cells by direct interaction with hnRNPA1, and enhanced Gprc5a stability, thereby alleviating I/R-induced intestinal mucosal damage. Hence, exosomal circEZH2_005 may serve as a biomarker for intestinal I/R injury and targeting the circEZH2_005/hnRNPA1/Gprc5a axis may be a potential therapeutic strategy for intestinal I/R injury.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Bowei Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jin Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jingjuan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yuqi Ding
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shuteng Zhan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yifeng Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jun Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Fu Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Bingcheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zebin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Qishun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Fangling Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zhiwen Yao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Weifeng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
8
|
Turan A, Zhang L, Tarique M, Ulker V, Arguc FN, Badal D, Yolcu ES, Shirwan H. Engineering pancreatic islets with a novel form of thrombomodulin protein to overcome early graft loss triggered by instant blood-mediated inflammatory reaction. Am J Transplant 2023; 23:619-628. [PMID: 36863480 PMCID: PMC10318623 DOI: 10.1016/j.ajt.2023.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
The instant blood-mediated inflammatory reaction (IBMIR) is initiated by innate immune responses that cause substantial islet loss after intraportal transplantation. Thrombomodulin (TM) is a multifaceted innate immune modulator. In this study, we report the generation of a chimeric form of thrombomodulin with streptavidin (SA-TM) for transient display on the surface of islets modified with biotin to mitigate IBMIR. SA-TM protein expressed in insect cells showed the expected structural and functional features. SA-TM converted protein C into activated protein C, blocked phagocytosis of xenogeneic cells by mouse macrophages and inhibited neutrophil activation. SA-TM was effectively displayed on the surface of biotinylated islets without a negative effect on their viability or function. Islets engineered with SA-TM showed improved engraftment and established euglycemia in 83% of diabetic recipients when compared with 29% of recipients transplanted with SA-engineered islets as control in a syngeneic minimal mass intraportal transplantation model. Enhanced engraftment and function of SA-TM-engineered islets were associated with the inhibition of intragraft proinflammatory innate cellular and soluble mediators of IBMIR, such as macrophages, neutrophils, high-mobility group box 1, tissue factor, macrophage chemoattractant protein-1, interleukin-1β, interleukin-6, tumor necrosis factor-α, interferon-γ. Transient display of SA-TM protein on the islet surface to modulate innate immune responses causing islet graft destruction has clinical potential for autologous and allogeneic islet transplantation.
Collapse
Affiliation(s)
- Ali Turan
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Lei Zhang
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Mohammad Tarique
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Vahap Ulker
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Feyza Nur Arguc
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Darshan Badal
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA
| | - Esma S Yolcu
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA.
| | - Haval Shirwan
- Department of Child Health,University of Missouri,Columbia,Missouri,USA; Department of Molecular Microbiology and Immunology,University of Missouri,Columbia,Missouri,USA; NextGen Precision Health,University of Missouri,Columbia,Missouri,USA.
| |
Collapse
|
9
|
Matsuura R, Doi K, Rabb H. Acute kidney injury and distant organ dysfunction-network system analysis. Kidney Int 2023; 103:1041-1055. [PMID: 37030663 DOI: 10.1016/j.kint.2023.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
Acute kidney injury (AKI) occurs in about half of critically ill patients and associates with high in-hospital mortality, increased long-term mortality post-discharge and subsequent progression to chronic kidney disease. Numerous clinical studies have shown that AKI is often complicated by dysfunction of distant organs, which is a cause of the high mortality associated with AKI. Experimental studies have elucidated many mechanisms of AKI-induced distant organ injury, which include inflammatory cytokines, oxidative stress and immune responses. This review will provide an update on evidence of organ crosstalk and potential therapeutics for AKI-induced organ injuries, and present the new concept of a systemic organ network to balance homeostasis and inflammation that goes beyond kidney-crosstalk with a single distant organ.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, the University of Tokyo Hospital
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, the University of Tokyo Hospital.
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine
| |
Collapse
|
10
|
Pterostilbene Confers Protection against Diquat-Induced Intestinal Damage with Potential Regulation of Redox Status and Ferroptosis in Broiler Chickens. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8258354. [PMID: 36733420 PMCID: PMC9889155 DOI: 10.1155/2023/8258354] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/15/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023]
Abstract
Oxidative stress causes damage to macromolecules, including proteins, DNA, and lipid, and has been recognized as a crucial driver of the onset and progression of several intestinal disorders. Pterostilbene, one of the natural antioxidants, has attracted considerable attention owing to its multiple biological activities. In the present study, we established an oxidative stress model in broiler chickens via injection with diquat to investigate whether pterostilbene could attenuate diquat-induced intestinal damage and reveal the underlying mechanisms. We found that diquat-induced decreases in the activities of superoxide dismutase and glutathione peroxidase and the level of reduced glutathione and the increase in hydrogen peroxide content in plasma and jejunum were significantly alleviated by pterostilbene (P < 0.05). Pterostilbene supplementation also decreased intestinal permeability and jejunal apoptosis rate, improved jejunal villus height and the ratio of villus height to crypt depth, and promoted the transcription and translation of jejunal tight junction proteins occludin and zona occludens 1 in diquat-challenged broilers (P < 0.05). Furthermore, pterostilbene reversed diquat-induced mitochondrial injury in the jejunum, as indicated by the decreased reactive oxygen species level and elevated activities of superoxide dismutase 2 and mitochondrial respiratory complexes (P < 0.05). Importantly, administering pterostilbene maintained iron homeostasis, inhibited lipid peroxidation, and regulated the expression of the markers of ferroptosis in the jejunum of diquat-exposed broilers (P < 0.05). The nuclear factor erythroid 2-related factor 2 signaling pathway in the jejunum of diquat-exposed broilers was also activated by pterostilbene (P < 0.05). In conclusion, our study provides evidence that pterostilbene alleviates diquat-induced intestinal mucosa injury and barrier dysfunction by strengthening antioxidant capacity and regulating ferroptosis of broiler chickens.
Collapse
|
11
|
Ngo ATP, Gollomp K. Building a better
NET
: Neutrophil extracellular trap targeted therapeutics in the treatment of infectious and inflammatory disorders. Res Pract Thromb Haemost 2022. [DOI: 10.1002/rth2.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Anh T. P. Ngo
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Kandace Gollomp
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
- Department of Pediatrics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
12
|
Zhang F, Li Y, Wu J, Zhang J, Cao P, Sun Z, Wang W. The role of extracellular traps in ischemia reperfusion injury. Front Immunol 2022; 13:1022380. [PMID: 36211432 PMCID: PMC9533173 DOI: 10.3389/fimmu.2022.1022380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
In response to strong signals, several types of immune cells release extracellular traps (ETs), which are web-like structures consisting of DNA decorated with various protein substances. This process is most commonly observed in neutrophils. Over the past two decades, ET formation has been recognized as a unique mechanism of host defense and pathogen destruction. However, the role of ETs in sterile inflammation has only been studied extensively in recent years. Ischemia reperfusion injury (IRI) is a type of sterile inflammatory injury. Several studies have reported that ETs have an important role in IRI in various organs. In this review, we describe the release of ETs by various types of immune cells and focus on the mechanism underlying the formation of neutrophil ETs (NETs). In addition, we summarize the role of ETs in IRI in different organs and their effects on tumors. Finally, we discuss the value of ETs as a potential therapeutic target for organ IRI and present possible challenges in conducting studies on IRI-related ETs as well as future research directions and prospects.
Collapse
Affiliation(s)
- Feilong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Yuqing Li
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Peng Cao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
- *Correspondence: Wei Wang,
| |
Collapse
|
13
|
Wan Y, Dong P, Zhu X, Lei Y, Shen J, Liu W, Liu K, Zhang X. Bibliometric and visual analysis of intestinal ischemia reperfusion from 2004 to 2022. Front Med (Lausanne) 2022; 9:963104. [PMID: 36052333 PMCID: PMC9426633 DOI: 10.3389/fmed.2022.963104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a common tissue-organ damage occurring in surgical practice. This study aims to comprehensively review the collaboration and impact of countries, institutions, authors, subject areas, journals, keywords, and critical literature on intestinal I/R injury from a bibliometric perspective, and to assess the evolution of clustering of knowledge structures and identify hot trends and emerging topics. Methods Articles and reviews related to intestinal I/R were retrieved through subject search from Web of Science Core Collection. Bibliometric analyses were conducted on Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio). Results A total of 1069 articles and reviews were included from 2004 to 2022. The number of articles on intestinal I/R injury gradually plateaued, but the number of citations increased. These publications were mainly from 985 institutions in 46 countries, led by China and the United States. Liu Kx published the most papers, while Chiu Cj had the largest number of co-citations. Analysis of the journals with the most outputs showed that most journals focused on surgical sciences, cell biology, and immunology. Macroscopic sketch and microscopic characterization of the entire knowledge domain were achieved through co-citation analysis. The roles of cell death, exosomes, intestinal flora, and anesthetics in intestinal I/R injury are the current and developing research focuses. The keywords "dexmedetomidine", "proliferation", and "ferroptosis" may also become new trends and focus of future research. Conclusion This study comprehensively reviews the research on intestinal I/R injury using bibliometric and visualization methods, and will help scholars better understand the dynamic evolution of intestinal I/R injury and provide directions for future research.
Collapse
Affiliation(s)
- Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Xiaobing Zhu
- Department of Anesthesiology, Hospital of Traditional Chinese Medicine of Zhongshan City, Zhongshan, China
| | - Yuqiong Lei
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Shen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Weifeng Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Kexuan Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Xiyang Zhang
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Chen F, Liu Y, Shi Y, Zhang J, Liu X, Liu Z, Lv J, Leng Y. The emerging role of neutrophilic extracellular traps in intestinal disease. Gut Pathog 2022; 14:27. [PMID: 35733158 PMCID: PMC9214684 DOI: 10.1186/s13099-022-00497-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular reticular fibrillar structures composed of DNA, histones, granulins and cytoplasmic proteins that are delivered externally by neutrophils in response to stimulation with various types of microorganisms, cytokines and host molecules, etc. NET formation has been extensively demonstrated to trap, immobilize, inactivate and kill invading microorganisms and acts as a form of innate response against pathogenic invasion. However, NETs are a double-edged sword. In the event of imbalance between NET formation and clearance, excessive NETs not only directly inflict tissue lesions, but also recruit pro-inflammatory cells or proteins that promote the release of inflammatory factors and magnify the inflammatory response further, driving the progression of many human diseases. The deleterious effects of excessive release of NETs on gut diseases are particularly crucial as NETs are more likely to be disrupted by neutrophils infiltrating the intestinal epithelium during intestinal disorders, leading to intestinal injury, and in addition, NETs and their relevant molecules are capable of directly triggering the death of intestinal epithelial cells. Within this context, a large number of NETs have been reported in several intestinal diseases, including intestinal infections, inflammatory bowel disease, intestinal ischemia–reperfusion injury, sepsis, necrotizing enterocolitis, and colorectal cancer. Therefore, the formation of NET would have to be strictly monitored to prevent their mediated tissue damage. In this review, we summarize the latest knowledge on the formation mechanisms of NETs and their pathophysiological roles in a variety of intestinal diseases, with the aim of providing an essential directional guidance and theoretical basis for clinical interventions in the exploration of mechanisms underlying NETs and targeted therapies.
Collapse
Affiliation(s)
- Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yongqiang Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yajing Shi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jianmin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Xin Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zhenzhen Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jipeng Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yufang Leng
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China. .,Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
15
|
Jin B, Li G, Zhou L, Fan Z. Mechanism Involved in Acute Liver Injury Induced by Intestinal Ischemia-Reperfusion. Front Pharmacol 2022; 13:924695. [PMID: 35694264 PMCID: PMC9185410 DOI: 10.3389/fphar.2022.924695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Intestinal ischemia-reperfusion (I/R) is a common pathophysiological process, which can occur in many conditions such as acute enteric ischemia, severe burns, small intestinal transplantation, etc,. Ischemia-reperfusion of the intestine is often accompanied by distal organ injury, especially liver injury. This paper outlined the signal pathways and cytokines involved in acute liver injury induced by intestinal I/R: the NF-κB Signaling Pathway, the P66shc Signaling Pathway, the HMGB1 Signaling Pathway, the Nrf2-ARE Signaling Pathway, the AMPK-SIRT-1 Signaling Pathway and other cytokines, providing new ideas for the prevention and treatment of liver injury caused by reperfusion after intestinal I/R.
Collapse
Affiliation(s)
- Binghui Jin
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Guangyao Li
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Lin Zhou
- Department of Outpatient, the NO. 967 Hospital of PLA Joint Logistics Support Force, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Zhao J, Chen XD, Yan ZZ, Huang WF, Liu KX, Li C. Gut-Derived Exosomes Induce Liver Injury After Intestinal Ischemia/Reperfusion by Promoting Hepatic Macrophage Polarization. Inflammation 2022; 45:2325-2338. [PMID: 35701685 DOI: 10.1007/s10753-022-01695-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 12/17/2022]
Abstract
Liver injury induced by intestinal ischemia/reperfusion (I/R) is accompanied by the polarization of Kupffer cells, which are specialized macrophages located in the liver. However, the causes of hepatic macrophage polarization after intestinal I/R remain unknown. This study investigated whether gut-derived exosomes contribute to the pathogenesis of liver injury triggered by intestinal I/R in a murine model and explored the underlying mechanisms. Intestinal I/R models were established by temporally clamping the superior mesenteric arteries of mice. Exosomes were isolated from the intestinal tissue of mice that underwent intestinal I/R or sham surgery according to a centrifugation-based protocol. Exosomes were co-cultured with RAW 264.7 macrophages or injected intravenously in mice. Liposomal clodronate was administered intraperitoneally to deplete the macrophages. Macrophage polarization was determined by flow cytometry, immunohistochemistry, and quantitative polymerase chain reaction. Liver injury was assessed by histological morphology and increased serum aspartate aminotransferase and alanine aminotransferase levels. Exosomes from mice intestines subjected to I/R (IR-Exo) promoted macrophage activation in vitro. Intravenous injection of IR-Exo caused hepatic M1 macrophage polarization and led to liver injury in mice. Depleting macrophages ameliorated liver injury caused by intestinal I/R or the injection of IR-Exo. Furthermore, inhibiting exosome release improved intestinal injury, liver function, and survival rates of mice subjected to intestinal I/R. Our study provides evidence that gut-derived exosomes induce liver injury after intestinal I/R by promoting hepatic M1 macrophage polarization. Inhibition of exosome secretion could be a therapeutic target for preventing hepatic impairment after intestinal I/R.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Xiao-Dong Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Zheng-Zheng Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Wen-Fang Huang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China.
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Deng F, Zhao BC, Yang X, Lin ZB, Sun QS, Wang YF, Yan ZZ, Liu WF, Li C, Hu JJ, Liu KX. The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microbes 2022; 13:1-21. [PMID: 33779497 PMCID: PMC8009132 DOI: 10.1080/19490976.2021.1902719] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Ferroptosis, a new type of cell death has been found to aggravate intestinal ischemia/reperfusion (I/R) injury. However, little is known about the changes of gut microbiota and metabolites in intestinal I/R and the role of gut microbiota metabolites on ferroptosis-induced intestinal I/R injury. This study aimed to establish a mouse intestinal I/R model and ileum organoid hypoxia/reoxygenation (H/R) model to explore the changes of the gut microbiota and metabolites during intestinal I/R and protective ability of capsiate (CAT) against ferroptosis-dependent intestinal I/R injury. Intestinal I/R induced disturbance of gut microbiota and significant changes in metabolites. We found that CAT is a metabolite of the gut microbiota and that CAT levels in the preoperative stool of patients undergoing cardiopulmonary bypass were negatively correlated with intestinal I/R injury. Furthermore, CAT reduced ferroptosis-dependent intestinal I/R injury in vivo and in vitro. However, the protective effects of CAT against ferroptosis-dependent intestinal I/R injury were abolished by RSL3, an inhibitor of glutathione peroxidase 4 (Gpx4), which is a negative regulator of ferroptosis. We also found that the ability of CAT to promote Gpx4 expression and inhibit ferroptosis-dependent intestinal I/R injury was abrogated by JNJ-17203212, an antagonist of transient receptor potential cation channel subfamily V member 1 (TRPV1). This study suggests that the gut microbiota metabolite CAT enhances Gpx4 expression and inhibits ferroptosis by activating TRPV1 in intestinal I/R injury, providing a potential avenue for the management of intestinal I/R injury.
Collapse
Affiliation(s)
- Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bing-Cheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ze-Bin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qi-Shun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Fan Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zheng-Zheng Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei-Feng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China,Jing-Juan Hu Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China,CONTACT Ke-Xuan Liu
| |
Collapse
|
18
|
Pan Y, Li J, Xia X, Wang J, Jiang Q, Yang J, Dou H, Liang H, Li K, Hou Y. β-glucan-coupled superparamagnetic iron oxide nanoparticles induce trained immunity to protect mice against sepsis. Am J Cancer Res 2022; 12:675-688. [PMID: 34976207 PMCID: PMC8692910 DOI: 10.7150/thno.64874] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Innate immune memory, also termed “trained immunity”, is thought to protect against experimental models of infection, including sepsis. Trained immunity via reprogramming monocytes/macrophages has been reported to result in enhanced inflammatory status and antimicrobial activity against infection in sepsis. However, a safe and efficient way to induce trained immunity remains unclear. Methods: β-glucan is a prototypical agonist for inducing trained immunity. Ferumoxytol, superparamagnetic iron oxide (SPIO) with low cytotoxicity, has been approved by FDA for clinical use. We synthesized novel nanoparticles BSNPs by coupling β-glucan with SPIO. BSNPs were further conjugated with fluorescein for quantitative analysis and trace detection of β-glucan on BSNPs. Inflammatory cytokine levels were measured by ELISA and qRT-PCR, and the phagocytosis of macrophages was detected by flow cytometry and confocal microscopy. The therapeutic effect of BSNPs was evaluated on the well-established sepsis mouse model induced by both clinical Escherichia coli (E. coli) and cecal ligation and puncture (CLP). Results: BSNPs were synthesized successfully with a 3:20 mass ratio of β-glucan and SPIO on BSNPs, which were mainly internalized by macrophages and accumulated in the lungs and livers of mice. BSNPs effectively reprogrammed macrophages to enhance the production of trained immunity markers and phagocytosis toward bacteria. BSNP-induced trained immunity protected mice against sepsis caused by E. coli and CLP and also against secondary infection. We found that BSNP treatment elevated Akt, S6, and 4EBP phosphorylation, while mTOR inhibitors decreased the trained immunity markers and phagocytosis enhanced by BSNPs. Furthermore, the PCR Array analysis revealed Igf1, Sesn1, Vegfa, and Rps6ka5 as possible key regulators of mTOR signaling during trained immunity. BSNP-induced trained immunity mainly regulated cellular signal transduction, protein modification, and cell cycle by modulating ATP binding and the kinase activity. Our results indicated that BSNPs induced trained immunity in an mTOR-dependent manner. Conclusion: Our data highlight that the trained immunity of macrophages is an effective strategy against sepsis and suggest that BSNPs are a powerful tool for inducing trained immunity to prevent and treat sepsis and secondary infections.
Collapse
|
19
|
NETosis in ischemic/reperfusion injuries: An organ-based review. Life Sci 2021; 290:120158. [PMID: 34822798 DOI: 10.1016/j.lfs.2021.120158] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 10/19/2022]
Abstract
Neutrophil extracellular trap (NETosis), the web-like structures induced by neutrophil death, is an important inflammatory mechanism of the immune system leading to reactive oxygen species production/coagulopathy, endothelial dysfunction, atherosclerosis, and ischemia. NETosis exerts its role through different mechanisms such as triggering Toll-like receptors, inflammatory cytokines, platelet aggregation, neutrophil activation/infiltration, and vascular impairment. NETosis plays a key role in the prognosis of coronary artery disease, ischemic injury of kidney, lung, gastrointestinal tract and skeletal muscles. In this review, we explored the molecular mechanisms involved in NETosis, and ischemic/reperfusion injuries in body organs.
Collapse
|
20
|
Zhenzhen L, Wenting L, Jianmin Z, Guangru Z, Disheng L, Zhiyu Z, Feng C, Yajing S, Yingxiang H, Jipeng L, Zhanhai W, Yan Z, Xin L, Yongqiang L, Yufang L. miR-146a-5p/TXNIP axis attenuates intestinal ischemia-reperfusion injury by inhibiting autophagy via the PRKAA/mTOR signaling pathway. Biochem Pharmacol 2021; 197:114839. [PMID: 34774846 DOI: 10.1016/j.bcp.2021.114839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 11/02/2022]
Abstract
Autophagy is being increasingly recognized as an important regulator of intestinal ischemia-reperfusion(I/R)injury, but its exact role is still debated. Emerging evidence suggests that miR-146a-5p is involved in the initiation and development of I/R injury, but its role in intestinal I/R injury remains unclear. The present study generated an intestinal I/R mouse model and an oxygen glucose deprivation/reoxygenation (OGD/R) Caco-2 cell model and found that autophagy was increased and contributed to the intestinal injury and cell death induced by I/R and OGD/R. In addition, in both I/R and OGD/R models, the miR-146a-5p expression level was decreased and accompanied by an increase in TXNIP expression. By transfecting cells with an miR-146a-5p inhibitor or mimic, we observed that miR-146a-5p inhibits autophagy during OGD/R by targeting TXNIP; this was confirmed by the dual luciferase reporter gene assay. Additionally, through overexpression and knockdown cell lines, we established that TXNIP regulates autophagy during intestinal I/R via the PRKAA/mTOR pathway. The interaction between TXNIP and p-PRKAA was verified by immunofluorescence co-localization and immunoprecipitation assays. Moreover, we confirmed that TXNIP is indispensable for miR-146a-5p-mediated cell protection. Finally, we observed that miR-146a-5p overexpression inhibits autophagy and attenuates intestinal I/R injury via the PRKAA/mTOR pathway by targeting TXNIP in vivo. In conclusion, this study highlights the role of miR-146a-5p in regulating autophagy by targeting TXNIP, suggesting that miR-146a-5p may be a novel drug target for intestinal I/R therapy.
Collapse
Affiliation(s)
- Liu Zhenzhen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Leng Wenting
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Jianmin
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Guangru
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Disheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhao Zhiyu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Chen Feng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shi Yajing
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hao Yingxiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Lv Jipeng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wan Zhanhai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Xin
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Yongqiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Leng Yufang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China; The Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
21
|
Netting Gut Disease: Neutrophil Extracellular Trap in Intestinal Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5541222. [PMID: 34712384 PMCID: PMC8548149 DOI: 10.1155/2021/5541222] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/04/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022]
Abstract
Many gut disease etiologies are attributed to the presence of robust inflammatory cell recruitment. The recruitment of neutrophils plays a vital role in inflammatory infiltration. Neutrophils have various antimicrobial effector mechanisms, including phagocytosis, oxidative burst, and degranulation. It is suggested that neutrophils could release neutrophil extracellular traps (NETs) to kill pathogens. However, recent evidence indicates that neutrophil infiltration within the gut is associated with disrupted local immunological microenvironment and impaired epithelial barrier. Growing evidence implies that NETs are involved in the progression of many diseases, including cancer, diabetes, thrombosis, and autoimmune disease. Increased NET formation was found in acute or chronic conditions, including infection, sterile inflammation, cancer, and ischemia/reperfusion injury (IRI). Here, we present a comprehensive review of recent advances in the understanding of NETs, focusing on their effects in gut disease. We also discuss NETs as a potential therapeutic target in gut disease.
Collapse
|
22
|
Thrombomodulin-mediated Inhibition of Neutrophil Extracellular Trap Formation Alleviates Hepatic Ischemia/Reperfusion Injury by Blocking TLR4 in Rats Subjected to Liver Transplantation. Transplantation 2021; 106:e126-e140. [PMID: 34534191 DOI: 10.1097/tp.0000000000003954] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hepatic ischemia/reperfusion injury (IRI) is an unavoidable outcome of liver transplantation, during which neutrophil extracellular traps (NETs) may play a critical role in the IRI-induced immune response to inflammation. The purpose of this study was to identify the function of recombinant human thrombomodulin (rTM) in the remission of hepatic IRI after liver transplantation and elucidate the specific mechanism. METHODS NET formation was detected in the serum of liver transplantation patients and rats following liver transplantation. Hematoxylin-eosin (HE) staining, terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) staining, immunohistochemistry and immunofluorescence were used to assess the effect of rTM on NET formation in vitro and in vivo. RESULTS We found that rTM markedly inhibited neutrophil formation in NETs, reduced apoptosis in hepatocytes, alleviated rat hepatic IRI and improved liver function. In vitro, rTM inhibited neutrophil formation in NETs, and lipopolysaccharide (LPS) (a Toll-like receptor (TLR)-4 agonist) reversed the inhibitory effect of rTM on NET formation. rTM blocked TLR-4 and the downstream extracellular signal-regulated kinase (ERK)/c-Jun NH2 terminal kinase (JNK) and nicotinamide adenine dinucleotide phosphate (NADPH)/ROS/peptidylarginine deiminase 4 (PAD4) signaling pathways to protect against hepatic IRI and inhibit NET formation. In addition, we demonstrated that combined treatment with rTM and an NADPH oxidative inhibitor had a better effect than either treatment alone. CONCLUSIONS NETs are a potential therapeutic target in hepatic IRI, and rTM could be used to prevent IR-induced hepatic injury. In addition, cotargeting NETosis-related signaling pathways might be a novel therapeutic strategy for hepatic IRI treatment.
Collapse
|
23
|
Liu Y, Qin X, Lei Z, Chai H, Wu Z. Diphenyleneiodonium ameliorates acute liver rejection during transplantation by inhibiting neutrophil extracellular traps formation in vivo. Transpl Immunol 2021; 68:101434. [PMID: 34216758 DOI: 10.1016/j.trim.2021.101434] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
Neutrophil extracellular traps (NETs) play critical roles in hepatic ischemic reperfusion injury (IRI) induced immune responses to inflammation. Diphenyleneiodonium (DPI) is an NADPH oxidative inhibitor that has been implicated in the regulation of NETs formation. However, the effects of NETs and their underlying mechanisms during DPI treatment of acute rejection (AR) after liver transplantation have not been elucidated. This study tested the hypothesis that blocking NETs formation by DPI treatment could be a potential therapeutic target against AR after liver transplantation. NETs were found to be excessively formed within the livers and serum of transplantation models, which could be an independent risk factor for AR. DPI was shown to alleviate hepatic injury and maintain liver functions by inhibiting NETs formation through the nicotinamide adenine dinucleotide phosphate (NADPH)/ROS/peptidylarginine deiminase 4 (PAD4) signaling pathway. NETs are highly involved in AR after liver transplantation. By inhibiting NETs formation, DPI suppresses activation of the NADPH/ROS/PAD4 signaling pathway which acts against AR after liver transplantation. Therefore, DPI is a potential candidate for the therapeutic management of AR after liver transplantation. Combination treatment containing both DPI and tacrolimus revealed a better antidamage efficacy than adjusting either treatment alone, suggesting that the joint therapy might be a promising solution in AR after liver transplantation.
Collapse
Affiliation(s)
- Yanyao Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Qin
- Department of General Surgery of Yuzhong District, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Children Health and Disorders, China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zilun Lei
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Chai
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
24
|
Maeda A, Hayase N, Doi K. Acute Kidney Injury Induces Innate Immune Response and Neutrophil Activation in the Lung. Front Med (Lausanne) 2020; 7:565010. [PMID: 33330525 PMCID: PMC7718030 DOI: 10.3389/fmed.2020.565010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/12/2020] [Indexed: 01/08/2023] Open
Abstract
Complication in acute kidney injury (AKI) is significantly associated with developing acute respiratory failure (ARF), while ARF is one of the most important risks for AKI. These data suggest AKI and ARF may synergistically worsen the outcomes of critically ill patients and these organ injuries may not occur independently. Organ crosstalk between the kidney and the lung has been investigated by using animal models so far. This review will focus on innate immune response and neutrophil activation among the mechanisms that contribute to this organ crosstalk. AKI increased the blood level of an inflammatory mediator in high-mobility group box 1, which induces an innate immune reaction via toll-like receptor 4. The remarkable infiltration of neutrophils to the lung was observed in animal AKI models. IL-6 and IL-8 have been demonstrated to contribute to pulmonary neutrophil activation in AKI. In addition, the formation of a neutrophil extracellular trap was also observed in the lung after the exposure of renal ischemia reperfusion in the animal model. Further investigation is necessary to determine whether targeting innate immune response and neutrophil activation will be useful for developing new therapeutics that could improve multiple organ failure in critically ill patients.
Collapse
Affiliation(s)
- Akinori Maeda
- Department of Acute Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Naoki Hayase
- Department of Acute Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Kent Doi
- Department of Acute Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
25
|
Li Y, Jiao HL, Bai YK, Wang P. Effect of manipulative reduction combined with air enema on intestinal mucosal immune function in children with intussusception. Pak J Med Sci 2020; 36:1640-1644. [PMID: 33235589 PMCID: PMC7674862 DOI: 10.12669/pjms.36.7.3105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective: To explore the effect of manipulative reduction combined with air enema on intestinal mucosal immune function in children with intussusception. Methods: This is a prospective randomized controlled study in which 60 children with primary intussusception admitted to Hebei Children’s Hospital from October 2018 to October 2019 were selected for this study. They were randomly divided into two groups. The 30 patients in the experimental group underwent manipulative reduction and air enema reduction, and 30 patients in the control group underwent only air enema reduction. Pain scores and pressure during enema were recorded and analyzed. Fasting blood of children in the experimental group were drawn to test the serum T lymphocyte subsets CD3+, CD4+, CD8+ levels, B lymphocyte subsets CD19+ level, and NK cell subsets CD56+ levels before reduction. Among them, fasting blood of 28 children with successful reduction were drawn again in the morning after reduction, and the indicators of each immune cell subgroup before and after reduction were analyzed. Two children with unsuccessful reduction were no longer tested for these indicators. Results: Twenty-Eight children in the experimental group had successful reduction, and two children with unsuccessful reduction were changed to open surgery (28/30). Twenty five Children in the control group had successful reduction, and five were changed to open surgery (25/30). There was no significant difference in the success rate of reduction between two groups (p>0.05). Close observation for 12~24h after reduction found that none of the children had signs of peritonitis. The pain score and reduction pressure of the observation group were lower than those of the control group, and the difference was statistically significant (p<0.05). The levels of serum CD3+, CD4+, and CD8+ after reduction in the experimental group were significantly higher than before reduction, and the difference was statistically significant (p<0.05). CD19+ level was significantly lower than before reduction, and the difference was statistically significant (p<0.05). There was no significant difference in changes of other indicators. Conclusions: Manipulative reduction combined with air enema reduction can relieve pain and air injection pressure during enema, reduce reperfusion injury caused by intestinal ischemia, and protect intestinal mucosal immune function, which is a favored treatment.
Collapse
Affiliation(s)
- Yang Li
- Yang Li, Department of Minimally Invasive Surgery, Hebei Children's Hospital, Shijiazhuang, 050031, Hebei, P.R. China
| | - Han-Liang Jiao
- Han-liang Jiao, Department of Pediatrics, Hebei Children's Hospital, Shijiazhuang, 050031, Hebei, P.R. China
| | - Yu-Kun Bai
- Yu-kun Bai, Department of Pediatrics, Hebei Children's Hospital, Shijiazhuang, 050031, Hebei, P.R. China
| | - Ping Wang
- Ping Wang, Department of Pediatrics, Hebei Children's Hospital, Shijiazhuang, 050031, Hebei, P.R. China
| |
Collapse
|
26
|
Abstract
Acute kidney injury (AKI) is a threatening medical condition associated with poor outcomes at different settings. The development of standardized diagnostic criteria and new biomarkers addressed significant clinical impacts of AKI and the need for an early AKI detection, respectively. There have been some breakthroughs in understanding the pathogenesis of AKI through basic research; however, treatments against AKI aside from renal replacement therapy (RRT) have not shown adequate successful results. Biomarkers that could identify good responders to certain treatment are expected to facilitate translation of basic research findings. Most patients with severe AKI treated with RRT died due to multiple-organ failure, not renal dysfunction. Hence, it is essential to identify other organ dysfunctions induced by AKI as organ crosstalk. Also, a multidisciplinary approach of critical care nephrology is needed to evaluate a complex organ crosstalk in AKI. For disruptive innovation for AKI, we further explore these new aspects of AKI, which previously were considered outside the scope of nephrology.
Collapse
Affiliation(s)
- Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan.
| |
Collapse
|
27
|
Li T, Jiang H, Liu H, Cooper DKC, Wang Y. Extracellular histones and xenotransplantation. Xenotransplantation 2020; 27:e12618. [PMID: 32940936 DOI: 10.1111/xen.12618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Tao Li
- Department of Organ Transplantation The Second Affiliated Hospital of Hainan Medical University Haikou Hainan China
- The Transplantation Insititute of Hainan Medical University Haikou Hainan China
| | - Hongtao Jiang
- Department of Organ Transplantation The Second Affiliated Hospital of Hainan Medical University Haikou Hainan China
- The Transplantation Insititute of Hainan Medical University Haikou Hainan China
| | - Houqin Liu
- Department of Organ Transplantation The Second Affiliated Hospital of Hainan Medical University Haikou Hainan China
- The Transplantation Insititute of Hainan Medical University Haikou Hainan China
| | - David K. C. Cooper
- Xenotransplantation Program Department of Surgery University of Alabama at Birmingham Birmingham AL USA
| | - Yi Wang
- Department of Organ Transplantation The Second Affiliated Hospital of Hainan Medical University Haikou Hainan China
- The Transplantation Insititute of Hainan Medical University Haikou Hainan China
| |
Collapse
|
28
|
Watanabe-Kusunoki K, Nakazawa D, Ishizu A, Atsumi T. Thrombomodulin as a Physiological Modulator of Intravascular Injury. Front Immunol 2020; 11:575890. [PMID: 33042158 PMCID: PMC7525002 DOI: 10.3389/fimmu.2020.575890] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
Thrombomodulin (TM), which is predominantly expressed on the endothelium, plays an important role in maintaining vascular homeostasis by regulating the coagulation system. Intravascular injury and inflammation are complicated physiological processes that are induced by injured endothelium-mediated pro-coagulant signaling, necrotic endothelial- and blood cell-derived damage-associated molecular patterns (DAMPs), and DAMP-mediated inflammation. During the hypercoagulable state after endothelial injury, TM is released into the intravascular space by proteolytic cleavage of the endothelium component. Recombinant TM (rTM) is clinically applied to patients with disseminated intravascular coagulation, resulting in protection from tissue injury. Recent studies have revealed that rTM functions as an inflammatory regulator beyond hemostasis through various molecular mechanisms. More specifically, rTM neutralizes DAMPs, including histones and high mobility group box 1 (HMGB1), suppresses excessive activation of the complement system, physiologically protects the endothelium, and influences both innate and acquired immunity. Neutrophil extracellular traps (NETs) promote immunothrombosis by orchestrating platelets to enclose infectious invaders as part of the innate immune system, but excessive immunothrombosis can cause intravascular injury. However, rTM can directly and indirectly regulate NET formation. Furthermore, rTM interacts with mediators of acquired immunity to resolve vascular inflammation. So far, rTM has shown good efficacy in suppressing inflammation in various experimental models, including thrombotic microangiopathy, sterile inflammatory disorders, autoimmune diseases, and sepsis. Thus, rTM has the potential to become a novel tool to regulate intravascular injury via pleiotropic effects.
Collapse
Affiliation(s)
- Kanako Watanabe-Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
29
|
Cai Z, Zhang M, Boafo Kwantwi L, Bi X, Zhang C, Cheng Z, Ding X, Su T, Wang H, Wu Q. Breast cancer cells promote self-migration by secreting interleukin 8 to induce NET formation. Gene 2020; 754:144902. [PMID: 32544496 DOI: 10.1016/j.gene.2020.144902] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Zeyu Cai
- Department of Pathology, Anhui Medical University, Hefei 230032, PR China
| | - Mingxun Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Center for Diagnostic Pathology, Hefei 230001, PR China
| | | | - Xiaomin Bi
- Department of Pathology, Anhui Medical University, Hefei 230032, PR China
| | - Chenchen Zhang
- The Second Affiliated Hospital of Anhui Medical University, Department of Pathology, Hefei 230601, PR China
| | - Zhongle Cheng
- The First Affiliated Hospital of Anhui Medical University, Department of Clinical Laboratory, Hefei 230032, PR China
| | - Xiaojuan Ding
- Department of Microbiology, Anhui Medical University, Hefei, 230032, PR China
| | - Tianhong Su
- Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, NY 10032, USA
| | - Hua Wang
- The First Affiliated Hospital of Anhui Medical University, Department of Oncology, Hefei 230032, PR China
| | - Qiang Wu
- Department of Pathology, Anhui Medical University, Hefei 230032, PR China.
| |
Collapse
|
30
|
Jin Y, Sun M, Lv X, Wang X, Jiang G, Chen C, Wen Z. Extracellular histones play a pathogenic role in primary graft dysfunction after human lung transplantation. RSC Adv 2020; 10:12485-12491. [PMID: 35497627 PMCID: PMC9051052 DOI: 10.1039/d0ra00127a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/22/2020] [Indexed: 11/21/2022] Open
Abstract
Primary graft dysfunction (PGD) causes early mortality and late graft failure after lung transplantation. The mechanisms of PGD are not fully understood but ischemia/reperfusion (I/R) injury may be involved. Extracellular histones have recently been identified as major contributors to I/R injury. Hence, we investigated whether extracellular histones are associated with PGD after lung transplantation. In total, 65 lung transplant patients were enrolled into this study. Blood samples were collected from patients before and serially after transplantation (24 h, 48 h, and 72 h) and measured for extracellular histones, myeloperoxidase (MPO), lactate dehydrogenase (LDH), and multiple cytokines. Besides, the patients' sera were cultured with human pulmonary artery endothelial cells (HPAEC) and human monocyte cell line (THP1) cells, respectively, and cellular viability and cytokine production were determined. Heparin or anti-histone antibody were used to study the effects of histone-neutralized interventions. The results showed that extracellular histones increased markedly after lung transplantation, peaked by 24 h and tended to decrease thereafter, but still retained high levels up to 72 h. Extracellular histones were more abundant in patients with PGD (n = 8) than patients without PGD (n = 57) and linearly correlated with MPO, LDH, and most detected cytokines. Ex vivo studies showed that the patients' sera collected within 24 h after transplantation were very damaging to HPAEC cells and promoted cytokine production in cultured THP1 cells, which could be largely prevented by heparin or anti-histone antibodies. These data suggested a pathogenic role for extracellular histones in PGD after lung transplantation. Targeting extracellular histones may serve as a preventive and therapeutic strategy for PGD following lung transplantation.
Collapse
Affiliation(s)
- Yang Jin
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Meng Sun
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine Pittsburgh PA 15213 USA
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| |
Collapse
|
31
|
Shrestha B, Ito T, Kakuuchi M, Totoki T, Nagasato T, Yamamoto M, Maruyama I. Recombinant Thrombomodulin Suppresses Histone-Induced Neutrophil Extracellular Trap Formation. Front Immunol 2019; 10:2535. [PMID: 31736962 PMCID: PMC6828967 DOI: 10.3389/fimmu.2019.02535] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022] Open
Abstract
Histones, the major protein components of chromatin, are released into the extracellular space during sepsis, trauma, and ischemia-reperfusion injury, and subsequently mediate organ failure. Extracellular histones can promote endothelial damage and platelet aggregation, which can be suppressed by administration of recombinant thrombomodulin (rTM). The present study aimed to clarify whether histones can activate neutrophils to induce NET formation and whether rTM can prevent histone-induced NET formation. NET formation was analyzed in vitro by stimulating human neutrophils with histones in the absence or presence of rTM. NET formation was further analyzed in vivo by intravenous infusion of histones into rats with or without rTM. Histones induced NET release in a dose-dependent manner in vitro and NET release was induced as early as 1 h after stimulation. Histone-induced NET release was independent of NADPH oxidase. rTM suppressed histone-induced NET release in vitro as well as in vivo. The suppression might be mediated by rTM binding to histones, as suggested by analysis using a quartz crystal microbalance system. The present findings suggest that histones can activate neutrophils to form NETs and that rTM can inhibit histone-induced NET formation.
Collapse
Affiliation(s)
- Binita Shrestha
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Medicinal Chemistry, Rogel Cancer Center, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Midori Kakuuchi
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takaaki Totoki
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomoka Nagasato
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mika Yamamoto
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|