1
|
Wang H, Fan K, Dong H, Zheng G. Do the Cil/Dex/H-Dot nanoparticles possess potential anti-inflammatory effects? Kidney Int 2025; 107:360. [PMID: 39848748 DOI: 10.1016/j.kint.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/11/2024] [Indexed: 01/25/2025]
Affiliation(s)
- Heng Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Keyi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
2
|
Ning J, Wang J, Sun X, Li H, Cheng F. TRIM44 alleviates renal ischemia-reperfusion injury by inhibiting pyroptosis through the NLRP3 pathway. Mol Immunol 2025; 178:20-31. [PMID: 39813853 DOI: 10.1016/j.molimm.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) is a prevailing manifestation of acute kidney injury (AKI) with limited treatment options. TRIM44 has emerged as a possible target for treatment due to its regulatory function in inflammatory pathways. METHODS In vivo and in vitro models were employed to ascertain the TRIM44 impact on renal IRI. In vivo, we induced IRI in mice and assessed histological changes, oxidative stress markers, and pyroptosis-related proteins. In vitro, we subjected renal cells to hypoxia/reoxygenation (H/R) and manipulated TRIM44 expression to evaluate its effects on cell viability and pyroptosis. RESULTS IRI significantly increased inflammation, oxidative stress, and pyroptosis in both animal and cell models, evidenced by elevated cleaved caspase-1, GSDMD-N, and IL-1β/-18 levels. IRI conditions experienced a mitigated TRIM44 expression. Overexpression of TRIM44 in renal cells reduced pyroptosis, as shown by decreased levels of pyroptosis-related proteins and inflammatory cytokines and improved cell viability. Mechanistically, TRIM44 inhibited the NLRP3 inflammasome, as evidenced with reduced NLRP3 and cleaved caspase-1 levels upon TRIM44 overexpression and NLRP3 inhibition. In vivo, intravenous administration of TRIM44-expressing adenovirus post-IRI ameliorated renal damage, as reported with mitigated serum creatinine and blood urea nitrogen levels. CONCLUSION TRIM44 protects against renal IRI by inhibiting pyroptosis via the NLRP3 pathway, suggesting its potential to be targeted therapeutically for treating AKI.
Collapse
Affiliation(s)
- Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University. Wuhan, Hubei Province, PR China.
| | - Jinrun Wang
- Department of Urology, Renmin Hospital of Wuhan University. Wuhan, Hubei Province, PR China.
| | - Xuan Sun
- College of Nursing, Bengbu Medical University, Bengbu, Anhui Province, PR China.
| | - Haoyong Li
- Department of Urology, Renmin Hospital of Wuhan University. Wuhan, Hubei Province, PR China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University. Wuhan, Hubei Province, PR China.
| |
Collapse
|
3
|
Jarzebska N, Rodionov RN, Voit-Bak K, Straube R, Mücke A, Tselmin S, Rettig R, Julius U, Siow R, Gräßler J, Passauer J, Kok Y, Mavberg P, Weiss N, Bornstein SR, Aswani A. Neutrophil Extracellular Traps (NETs) as a Potential Target for Anti-Aging: Role of Therapeutic Apheresis. Horm Metab Res 2025. [PMID: 39788160 DOI: 10.1055/a-2444-3422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Neutrophil extracellular traps (NETs) are large structures composed of chromatin, histones and granule-derived proteins released extracellularly by neutrophils. They are generally considered to be a part of the antimicrobial defense strategy, preventing the dissemination of pathogens. However, overproduction of NETs or their ineffective clearance can drive various pathologies, many of which are associated with advanced age and involve uncontrolled inflammation, oxidative, cardiovascular and neurodegenerative stress as underlying mechanisms. Targeting NETs in the elderly as an anti-aging therapy seems to be a very attractive therapeutic approach. Therapeutic apheresis with a specific filter to remove NETs could be a promising strategy worth considering.
Collapse
Affiliation(s)
- Natalia Jarzebska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Karin Voit-Bak
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Richard Straube
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Anna Mücke
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Sergey Tselmin
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ronny Rettig
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ulrich Julius
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Richard Siow
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Ageing Research at King's (ARK), King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Jürgen Gräßler
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jens Passauer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | | | - Philip Mavberg
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Norbert Weiss
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Aswani
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom of Great Britain and Northern Ireland
- Santersus AG, Zurich, Switzerland
| |
Collapse
|
4
|
Yu F, Chen J, Zhang X, Ma Z, Wang J, Wu Q. Role of Neutrophil Extracellular Traps in Hypertension and Their Impact on Target Organs. J Clin Hypertens (Greenwich) 2025; 27:e14942. [PMID: 39686847 DOI: 10.1111/jch.14942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 12/18/2024]
Abstract
Hypertension is the predominant cause of cardiovascular diseases (CVDs) globally, and essential hypertension (EH) represents a significant public health challenge due to its multifactorial etiology involving complex interactions between genetic and environmental factors. However, the pathogenesis of EH is still unclear. Hypertension is a dysregulation in the renin-angiotensin-aldosterone system and sympathetic nervous system, both regulating saline homeostasis and cardiovascular function. However, current therapeutic interventions targeting these systems have limited efficacy in approximately 40% of cases, suggesting the involvement of alternative mechanisms. Inflammation is associated with the occurrence and progression of hypertension, but the underlying mechanism remains elusive, while chronic inflammation leads to tissue damage, fibrosis, and irreversible organ dysfunction. The development and maintenance of EH are caused by endothelial dysfunction, oxidative stress, and chronic inflammation. Neutrophils are involved in both acute and chronic inflammation since they represent the primary line of defense against inflammatory insults once recruited to the inflamed site where they remove harmful impurities. The process involving the formation of neutrophil extracellular traps (NETs) is called NETosis are involved in the pathogenesis and progression of CVDs, including coronary artery disease, acute myocardial infarction, peripheral arterial disease, heart failure, and atrial fibrillation. Recent investigations demonstrated that NETs facilitate the development of hypertension; however, the precise role of NETs in hypertension remains largely elusive. Therefore, this review aims to provide an overview of the current understanding regarding the involvement of NETosis in hypertension and explore the potential therapies targeting NETs for future interventions.
Collapse
Affiliation(s)
- Fei Yu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianshu Chen
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaowei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhengke Ma
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jingtao Wang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiang Wu
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
5
|
Wang Y, Guo J, Zhang D, Shi C, Zhang X, Gong Z. IDH1/MDH1 deacetylation promotes NETosis by regulating OPA1 and autophagy. Int Immunopharmacol 2024; 143:113270. [PMID: 39353390 DOI: 10.1016/j.intimp.2024.113270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND As a heterogeneous and life-threatening disease, the pathogenesis of acute liver failure (ALF) is complex. Our previous study has shown that IDH1/MDH1 deacetylation promotes ALF by regulating NETosis (a novel mode of cell death). In this article, we explore the manners of IDH1/MDH1 deacetylation regulates NETosis. METHODS In vitro experiments, the formation of NETs was detected by immunofluorescence staining and Western blotting. LC3 fluorescence staining was used to detect autophagosome formation. To observe mitochondrial morphology, cells were stained by Mito-Tracker Red. Western blotting was used to detect the levels of autophagy protein and mitochondrial dynamin. In vivo experiments, the ALF model in mouse was established with LPS/D-gal, and the formation of NETs was detected by immunofluorescence staining and Western blotting. The autophagy levels were detected by Western blotting in liver samples. RESULTS In dHL-60 cells, Western blotting results showed that the expression of OPA1 was higher in the IDH1/MDH1 deacetylated group compared with the IDH1/MDH1 WT group. And histone deacetylase inhibitor 6 (HDAC6i, ACY1215) decreased the expression level of OPA1 in IDH1/MDH1 deacetylated group. IDH1/MDH1 deacetylation increased the expression levels of both LC3B-II and Beclin 1, while decreasing the expression level of P62. It was reversed by ACY1215. Combined with our previous experiments, IDH1/MDH1 deacetylation upregulated autophagy concomitant with the increased expression of the markers of NETs formation. In a mouse model of ALF, ACY1215 further decreased the expression levels of LC3B-II and Beclin 1, while increasing the expression level of P62 in IDH1/MDH1 deacetylated mice. CONCLUSIONS IDH1/MDH1 deacetylation promoted NETosis by regulating autophagy and OPA1 in vitro. The regulation of neutrophil autophagy on NETosis during IDH1/MDH1 deacetylation might be masked in mice. ACY1215 might attenuate NETosis by regulating neutrophil autophagy, which alleviated ALF aggravated by IDH1/MDH1 deacetylation.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xiaoya Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
6
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024; 20:789-805. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Goshovska Y, Pashevin D, Goncharov S, Lapikova-Bryhinska T, Lisovyi O, Nagibin V, Portnichenko G, Tumanovska L, Dosenko V. Quercetin is a potential therapy for post-infarction NETosis formation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03602-w. [PMID: 39607549 DOI: 10.1007/s00210-024-03602-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024]
Abstract
The surgical intervention during myocardial infarction (MI) is associated with the risk of reperfusion injury, infiltration of tissues with polymorphonuclear neutrophils, and neutrophil extracellular trap (NET) formation. We hypothesized that inhibition of NETs with the use of quercetin might be a promising cardioprotective strategy. Wistar rats underwent LAD occlusion (MI) for 40 min followed by 90 min of reperfusion. The MI + Q group received a water-soluble form of quercetin (50 mg/kg, "Corvitin," BCPP, Ukraine) into the tail vein 10 min before reperfusion. The post-MI administration of quercetin significantly alleviated cardiac dysfunction. End-systolic pressure, stroke volume, cardiac output, and stroke work were significantly improved in the MI + Q vs. MI group. NET formation (examined by fluorescence microscopy and Hoechst staining) as well as free DNA in blood plasma was reduced in the MI + Q group that might be one of the mechanisms of the cardioprotective effect of quercetin. Postconditioning with quercetin might be used as a therapeutic tool for the alleviation of reperfusion injury and NETosis inhibition in vivo.
Collapse
Affiliation(s)
- Y Goshovska
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine.
| | - D Pashevin
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - S Goncharov
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - T Lapikova-Bryhinska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - O Lisovyi
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine
| | - V Nagibin
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - G Portnichenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - L Tumanovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - V Dosenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
8
|
Ge G, Zhu B, Zhu X, Yu Z, Zhu K, Cheng M. Mitochondrial DNA (mtDNA) accelerates oxygen-glucose deprivation-induced injury of proximal tubule epithelia cell via inhibiting NLRC5. Mitochondrion 2024:101989. [PMID: 39586387 DOI: 10.1016/j.mito.2024.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
The high morbidity and mortality associated with acute kidney injury (AKI) are global health concerns. AKI is commonly attributed to ischemia/reperfusion injury (IRI), a condition characterized by activation of inflammatory responses and mitochondrial dysfunction. Nonetheless, mitochondrial DNA (mtDNA) has the potential to induce renal IRI. This study aimed to elucidate the mechanism and function of mtDNA in HK-2 cells that had been exposed to oxygen-glucose deprivation/reperfusion (OGD/R) and in renal IRI mice. OGD/R was discovered to induce an increase in the amount of mtDNA in HK-2 cells. Moreover, our study demonstrated that mtDNA facilitated cellular apoptosis and inflammation in vivo and in vitro. Given the potential role of inflammation in OGD/R, we investigated the effect of mtDNA on various signaling pathways associated with inflammation. Western blot analysis demonstrated that mtDNA significantly upregulated NLRC5/TAP1 signaling. Furthermore, the upregulation of NLRC5 and TAP1 expression induced by mtDNA was reversed when NLRC5 was inhibited. It is worth mentioning that the loss of NLRC5 effectively nullified the beneficial effects of mtDNA on inflammation and cell apoptosis induced by OGD/R. In addition, in renal IRI mice, mtDNA treatment also aggravated inflammation and kidney damage, and increased the NLRC5 levels in kidney tissues. These results suggested that NLRC5 acts as an intermediary between mtDNA and the pathogenicity of renal IRI. In summary, this study provides evidence that mtDNA promotes apoptosis and inflammation in OGD/R treated HK-2 cells and renal IRI mice through upregulating NLRC5 levels.
Collapse
Affiliation(s)
- Guojun Ge
- The 903 RD Hospital of PLA, No. 14 Lingyin Road, Xihu District, Hangzhou, Zhejiang 310013, China
| | - Bocheng Zhu
- The 903 RD Hospital of PLA, No. 14 Lingyin Road, Xihu District, Hangzhou, Zhejiang 310013, China
| | - Xiaofeng Zhu
- The 903 RD Hospital of PLA, No. 14 Lingyin Road, Xihu District, Hangzhou, Zhejiang 310013, China
| | - Zhenfei Yu
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No.453 Stadium Road, Hangzhou, Zhejiang 310007, China
| | - Keqing Zhu
- The 903 RD Hospital of PLA, No. 14 Lingyin Road, Xihu District, Hangzhou, Zhejiang 310013, China
| | - Mengshi Cheng
- Department of Intensive Care Unit, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No.453 Stadium Road, Hangzhou, Zhejiang 310007, China.
| |
Collapse
|
9
|
He J, Zheng F, Qiu L, Wang Y, Zhang J, Ye H, Zhang Q. Plasma neutrophil extracellular traps in patients with sepsis-induced acute kidney injury serve as a new biomarker to predict 28-day survival outcomes of disease. Front Med (Lausanne) 2024; 11:1496966. [PMID: 39629231 PMCID: PMC11611547 DOI: 10.3389/fmed.2024.1496966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Background There is currently no accurate, readily available, or validated biomarker for assessing disease severity and survival outcomes in sepsis-induced acute kidney injury (SAKI), which limits the ability to conduct effective therapeutic interventions. The neutrophil extracellular traps (NETs) may be involved in the pathophysiology of SAKI. The present study investigated the predictive value of plasma NETs for the survival outcome of patients with SAKI. Methods This observational study included 136 SAKI patients, all of whom underwent a 28-day follow-up. According to the follow-up records, SAKI patients were divided into two groups: the non-survivor group (60 subjects) and the survivor group (76 subjects). Blood samples were collected after the diagnosis of AKI to measure three NET markers and 12 inflammatory indices. Correlation analysis, logistic regression analysis, receiver operating characteristic curve analysis, and survival analysis were performed. Results Compared to survivors, non-survivors among SAKI patients exhibited significantly higher levels of three plasma NET markers (all p < 0.001). Meanwhile, in SAKI patients, plasma levels of NET markers were significantly associated with serum levels of inflammatory indices. Additionally, serum interleukin (IL)-2, IL-8, IL-10, and tumor necrosis factor-alpha showed an interactive effect with plasma NET markers on the survival of SAKI patients. Furthermore, the combination of three plasma NET markers could identify SAKI patients with a poor 28-day survival with better accuracy (area under the curve = 0.857). Finally, plasma NET markers may independently predict the 28-day survival in SAKI patients. Conclusion Plasma NET markers were elevated in SAKI patients with poor outcomes and served as biomarkers for predicting 28-day survival in SAKI patients.
Collapse
Affiliation(s)
| | | | | | | | - Jing Zhang
- Department of Intensive Care Unit, First People’s Hospital of Changshu City, Changshu Hospital Affiliated to Soochow University, Changshu, Jiangsu, China
| | - Hongwei Ye
- Department of Intensive Care Unit, First People’s Hospital of Changshu City, Changshu Hospital Affiliated to Soochow University, Changshu, Jiangsu, China
| | - Qian Zhang
- Department of Intensive Care Unit, First People’s Hospital of Changshu City, Changshu Hospital Affiliated to Soochow University, Changshu, Jiangsu, China
| |
Collapse
|
10
|
Xia K, Jin Z, Qiu Q, Zhou Y, Lu Y, Qiu T, Zhou J, Chen Z. Ligustilide alleviates oxidative stress during renal ischemia-reperfusion injury through maintaining Sirt3-dependent mitochondrial homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155975. [PMID: 39216302 DOI: 10.1016/j.phymed.2024.155975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion (I/R) injury is an inevitable complication during renal transplantation and is closely related to patient prognosis. Mitochondrial damage induced oxidative stress is the core link of renal I/R injury. Ligustilide (LIG), a natural compound extracted from ligusticum chuanxiong hort and angelica sinensis, has exhibited the potential to protect mitochondrial function. However, whether LIG can ameliorate renal I/R injury requires further investigation. Delving deeper into the precise targets and mechanisms of LIG's effect on renal I/R injury is crucial. PURPOSE This study aimed to elucidate the specific mechanism of LIG's protective effect on renal I/R injury. METHODS In this study, an in vivo model of renal ischemia-reperfusion (I/R) injury was developed in mice, along with an in vitro model of hypoxia-reoxygenation (H/R) using human proximal renal tubular epithelial cells (HK-2). To assess the impact of LIG on renal injury, various methods were employed, including serum creatinine (Cr) and blood urea nitrogen (BUN) testing, hematoxylin and eosin (HE) staining, and immunohistochemistry (IHC) for kidney injury molecule-1 (KIM-1). The effects of LIG on oxidative stress were examined using fluorescent probes dihydroethidium (DHE) and dichlorodihydrofluorescein diacetate (DCFH-DA), TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and flow cytometry. Additionally, the influence of LIG on mitochondrial morphology and function was evaluated through transmission electron microscopy (TEM), Mito Tracker Red CMXRos staining, adenosine triphosphate (ATP) concentration assays, and JC-1 staining. The potential mechanism involving LIG and Sirt3 was explored by manipulating Sirt3 expression through cell transfection. RESULTS The results showed that LIG could provide protective function for mitochondria to alleviate oxidative stress induced by renal I/R. Further mechanistic studies indicated that LIG maintained mitochondrial homeostasis by targeting Sirt3. CONCLUSION Our findings demonstrated that LIG alleviated oxidative stress during renal I/R injury through maintaining Sirt3-dependent mitochondrial homeostasis. Overall, our data raised the possibility of LIG as a novel therapy for renal I/R injury.
Collapse
Affiliation(s)
- Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zeya Jin
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiangmin Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yujie Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yifan Lu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Liang W, Bai Y, Zhang H, Mo Y, Li X, Huang J, Lei Y, Gao F, Dong M, Li S, Liang J. Identification and Analysis of Potential Biomarkers Associated with Neutrophil Extracellular Traps in Cervicitis. Biochem Genet 2024:10.1007/s10528-024-10919-x. [PMID: 39419909 DOI: 10.1007/s10528-024-10919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/14/2024] [Indexed: 10/19/2024]
Abstract
Early diagnosis of cervicitis is important. Previous studies have found that neutrophil extracellular traps (NETs) play pro-inflammatory and anti-inflammatory roles in many diseases, suggesting that they may be involved in the inflammation of the uterine cervix and NETs-related genes may serve as biomarkers of cervicitis. However, what NETs-related genes are associated with cervicitis remains to be determined. Transcriptome analysis was performed using samples of exfoliated cervical cells from 15 patients with cervicitis and 15 patients without cervicitis as the control group. First, the intersection of differentially expressed genes (DEGs) and neutrophil extracellular trap-related genes (NETRGs) were taken to obtain genes, followed by functional enrichment analysis. We obtained hub genes through two machine learning algorithms. We then performed Artificial Neural Network (ANN) and nomogram construction, confusion matrix, receiver operating characteristic (ROC), gene set enrichment analysis (GSEA), and immune cell infiltration analysis. Moreover, we constructed ceRNA network, mRNA-transcription factor (TF) network, and hub genes-drug network. We obtained 19 intersecting genes by intersecting 1398 DEGs and 136 NETRGs. 5 hub genes were obtained through 2 machine learning algorithms, namely PKM, ATG7, CTSG, RIPK3, and ENO1. Confusion matrix and ROC curve evaluation ANN model showed high accuracy and stability. A nomogram containing the 5 hub genes was established to assess the disease rate in patients. The correlation analysis revealed that the expression of ATG7 was synergistic with RIPK3. The GSEA showed that most of the hub genes were related to ECM receptor interactions. It was predicted that the ceRNA network contained 2 hub genes, 3 targeted miRNAs, and 27 targeted lnRNAs, and that 5 mRNAs were regulated by 28 TFs. In addition, 36 small molecule drugs that target hub genes may improve the treatment of cervicitis. In this study, five hub genes (PKM, ATG7, CTSG, RIPK3, ENO1) provided new directions for the diagnosis and treatment of patients with cervicitis.
Collapse
Affiliation(s)
- Wantao Liang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Yanyuan Bai
- Guangxi University of Chinese Medicine, Nanning, 530001, Guangxi, China
| | - Hua Zhang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Yan Mo
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Xiufang Li
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Junming Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Yangliu Lei
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Fangping Gao
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Mengmeng Dong
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Shan Li
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Juan Liang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China.
| |
Collapse
|
12
|
Jiang ZH, Wang JS, Wang JL, Zheng JF, Li XL, Yang ZC, Xu MQ, Zhang YL, Wang Y. Hydrogen attenuates ischaemia-reperfusion injury in skeletal muscles post-limb replantation by activating the NRF2/HO-1 signalling pathway to reduce BAX expression. Heliyon 2024; 10:e37018. [PMID: 39309900 PMCID: PMC11414507 DOI: 10.1016/j.heliyon.2024.e37018] [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: 04/24/2024] [Revised: 07/18/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Background Ischaemia-reperfusion injury (IRI) is a critical complication post-limb replantation. The oxidative stress and cellular apoptosis due to IRI considerably hinder the healing process. This study aimed to investigate the modulatory effects of pre-perfusion with hydrogen-rich heparin sodium on the nuclear factor erythroid 2-related factor 2 (NRF2)/haeme oxygenase-1 (HO-1) pathway and its potential mechanisms in mitigating skeletal muscle IRI post-limb replantation. Methods Forty healthy Sprague-Dawley rats (250-300 g) were classified into five groups (n = 8 each): normal control, IRI + heparin sodium pre-perfusion (heparin group), IRI + hydrogen-rich heparin sodium pre-perfusion (hydrogen-rich heparin group), IRI + hydrogen-rich heparin sodium pre-perfusion + NRF2 inhibitor (hydrogen-rich heparin + all-trans retinoic acid [ATRA] group), and IRI + heparin sodium pre-perfusion + NRF2 inhibitor (heparin + ATRA group). The activation of the NRF2/HO-1 pathway in skeletal muscle IRI was evaluated based on HO-1 expression using western blotting and immunofluorescence. Furthermore, haematoxylin and eosin staining and transmission electron microscopy were employed to determine the histopathological characteristics. Additionally, superoxide dismutase and malondialdehyde levels in skeletal muscle tissue were measured to assess antioxidant capacity and the degree of oxidative stress damage. Tissue hypoxia was assessed based on hypoxia-inducible factor 1-alpha expression, whereas apoptosis markers BCL-2-associated X protein (BAX) and Caspase-3 in skeletal muscle tissues were analysed using western blotting with terminal deoxynucleotidyl transferase dUTP nick end labelling staining to quantify cell apoptosis. Results Compared with the control group, the heparin group exhibited significant pathological changes, including inflammatory infiltration and cellular hypertrophy, with increased apoptosis and oxidative stress. Notably, NRF2 suppression aggravated these effects. However, hydrogen-rich heparin sodium prominently activated the NRF2/HO-1 pathway, enhancing antioxidant defence and reducing BAX/Caspase-3-mediated apoptosis, thereby mitigating IRI-induced damage. The use of an NRF2 inhibitor to inhibit NRF2 excitation by hydrogen-rich heparin sodium notably weakened NRF2 activation and the antioxidant response, resulting in a substantial increase in cellular apoptosis. Conclusion Pre-perfusion with hydrogen-rich heparin sodium markedly diminishes the BAX/Caspase-3-mediated apoptotic pathway in skeletal muscle tissues with IRI through the excitation of the NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- zi-hao Jiang
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - jun-sheng Wang
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - jin-ling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - jiang-fan Zheng
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - xiao-ling Li
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - zhi-cheng Yang
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - meng-qiu Xu
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - yong-li Zhang
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - yu Wang
- Department of Emergency, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
13
|
Maier-Begandt D, Alonso-Gonzalez N, Klotz L, Erpenbeck L, Jablonska J, Immler R, Hasenberg A, Mueller TT, Herrero-Cervera A, Aranda-Pardos I, Flora K, Zarbock A, Brandau S, Schulz C, Soehnlein O, Steiger S. Neutrophils-biology and diversity. Nephrol Dial Transplant 2024; 39:1551-1564. [PMID: 38115607 PMCID: PMC11427074 DOI: 10.1093/ndt/gfad266] [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: 08/31/2023] [Indexed: 12/21/2023] Open
Abstract
Neutrophils, the most abundant white blood cells in the human circulation, play crucial roles in various diseases, including kidney disease. Traditionally viewed as short-lived pro-inflammatory phagocytes that release reactive oxygen species, cytokines and neutrophil extracellular traps, recent studies have revealed their complexity and heterogeneity, thereby challenging this perception. Neutrophils are now recognized as transcriptionally active cells capable of proliferation and reverse migration, displaying phenotypic and functional heterogeneity. They respond to a wide range of signals and deploy various cargo to influence the activity of other cells in the circulation and in tissues. They can regulate the behavior of multiple immune cell types, exhibit innate immune memory, and contribute to both acute and chronic inflammatory responses while also promoting inflammation resolution in a context-dependent manner. Here, we explore the origin and heterogeneity of neutrophils, their functional diversity, and the cues that regulate their effector functions. We also examine their emerging role in infectious and non-infectious diseases with a particular emphasis on kidney disease. Understanding the complex behavior of neutrophils during tissue injury and inflammation may provide novel insights, thereby paving the way for potential therapeutic strategies to manage acute and chronic conditions. By deciphering their multifaceted role, targeted interventions can be developed to address the intricacies of neutrophil-mediated immune responses and improve disease outcomes.
Collapse
Affiliation(s)
- Daniela Maier-Begandt
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Luisa Klotz
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Münster, Germany
| | - Luise Erpenbeck
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) partner site Düsseldorf/Essen, Essen, Germany
| | - Roland Immler
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anja Hasenberg
- Institute of Experimental Immunology and Imaging, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Tonina T Mueller
- Department of Medicine I, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andrea Herrero-Cervera
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Universität of Münster, Münster, Germany
| | | | - Kailey Flora
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Zarbock
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Schulz
- Department of Medicine I, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Universität of Münster, Münster, Germany
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
14
|
Li Y, Min X, Zhang X, Cao X, Kong Q, Mao Q, Cheng H, Gou L, Li Y, Li C, Liu L, Ding Z. HSPA12A promotes c-Myc lactylation-mediated proliferation of tubular epithelial cells to facilitate renal functional recovery from kidney ischemia/reperfusion injury. Cell Mol Life Sci 2024; 81:404. [PMID: 39277835 PMCID: PMC11402889 DOI: 10.1007/s00018-024-05427-5] [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: 05/29/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
Proliferation of renal tubular epithelial cells (TEC) is essential for restoring tubular integrity and thereby to support renal functional recovery from kidney ischemia/reperfusion (KI/R) injury. Activation of transcriptional factor c-Myc promotes TEC proliferation following KI/R; however, the mechanism regarding c-Myc activation in TEC is incompletely known. Heat shock protein A12A (HSPA12A) is an atypic member of HSP70 family. In this study, we found that KI/R decreased HSPA12A expression in mouse kidneys and TEC, while ablation of HSPA12A in mice impaired TEC proliferation and renal functional recovery following KI/R. Gain-of-functional studies demonstrated that HSPA12A promoted TEC proliferation upon hypoxia/reoxygenation (H/R) through directly interacting with c-Myc and enhancing its nuclear localization to upregulate expression of its target genes related to TEC proliferation. Notably, c-Myc was lactylated in TEC after H/R, and this lactylation was enhanced by HSPA12A overexpression. Importantly, inhibition of c-Myc lactylation attenuated the HSPA12A-induced increases of c-Myc nuclear localization, proliferation-related gene expression, and TEC proliferation. Further experiments revealed that HSPA12A promoted c-Myc lactylation via increasing the glycolysis-derived lactate generation in a Hif1α-dependent manner. The results unraveled a role of HSPA12A in promoting TEC proliferation and facilitating renal recovery following KI/R, and this role of HSPA12A was achieved through increasing lactylation-mediated c-Myc activation. Therefore, targeting HSPA12A in TEC might be a viable strategy to promote renal functional recovery from KI/R injury in patients.
Collapse
Affiliation(s)
- Yunfan Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinxu Min
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaofei Cao
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qiuyue Kong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qian Mao
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Cheng
- Department of Anesthesiology, The First Affiliated Hospital With Wannan Medical College, Wuhu, 241001, China
| | - Liming Gou
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
15
|
Zhang M, Li S, Ying J, Qu Y. Neutrophils: a key component in ECMO-related acute organ injury. Front Immunol 2024; 15:1432018. [PMID: 39346902 PMCID: PMC11427252 DOI: 10.3389/fimmu.2024.1432018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Extracorporeal membrane oxygenation (ECMO), as an extracorporeal life support technique, can save the lives of reversible critically ill patients when conventional treatments fail. However, ECMO-related acute organ injury is a common complication that increases the risk of death in critically ill patients, including acute kidney injury, acute brain injury, acute lung injury, and so on. In ECMO supported patients, an increasing number of studies have shown that activation of the inflammatory response plays an important role in the development of acute organ injury. Cross-cascade activation of the complement system, the contact system, and the coagulation system, as well as the mechanical forces of the circuitry are very important pathophysiological mechanisms, likely leading to neutrophil activation and the production of neutrophil extracellular traps (NETs). NETs may have the potential to cause organ damage, generating interest in their study as potential therapeutic targets for ECMO-related acute organ injury. Therefore, this article comprehensively summarized the mechanism of neutrophils activation and NETs formation following ECMO treatment and their actions on acute organ injury.
Collapse
Affiliation(s)
- Mingfu Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Zollet V, Arenas Hoyos I, Hirsiger S, Brahim BB, Petrucci MF, Casoni D, Wang J, Spirig R, Nettelbeck K, Garcia L, Fuest L, Vögelin E, Constantinescu M, Rieben R. Neutrophil extracellular traps and citrullinated fibrinogen contribute to injury in a porcine model of limb ischemia and reperfusion. Front Immunol 2024; 15:1436926. [PMID: 39315100 PMCID: PMC11416929 DOI: 10.3389/fimmu.2024.1436926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Background Ischemia/reperfusion injury (IRI) is a complex pathological process, triggered by the restoration of blood flow following an interrupted blood supply. While restoring the blood flow is the only option to salvage the ischemic tissue, reperfusion after a prolonged period of ischemia initiates IRI, triggering a cascade of inflammatory responses ultimately leading to neutrophil recruitment to the inflamed tissue, where they release neutrophil extracellular traps (NETs). NETs are web-like structures of decondensed chromatin and neutrophilic proteins, including peptidyl-arginine deiminase 2 and 4 (PAD2, PAD4), that, once outside, can citrullinate plasma proteins, irreversibly changing their conformation and potentially their function. While the involvement of NETs in IRI is known mainly from rodent models, we aimed to determine the effect of NET formation and especially PADs-mediated extracellular protein citrullination in a porcine model of limb IRI. Methods We conducted our study on amputated pig forelimbs exposed to 1 h or 9 h of ischemia and then reperfused in vivo for 12 h. Limb weight, edema formation, compartmental pressure were measured, and skeletal muscle was analyzed by immunofluorescence (TUNEL assay and dystrophin staining) to evaluate tissue damage. Fibrin tissue deposition, complement deposition and NETs were investigated by immunofluorescence. Citrullinated plasma proteins were immunoprecipitated and citrullinated fibrinogen was identified in the plasma by Western blot and in the tissue by immunofluorescence and Western blot. Results Our data consolidate the involvement of NETs in a porcine model of limb IRI, correlating their contribution to damage extension with the duration of the ischemic time. We found a massive infiltration of NETs in the group subjected to 9 h ischemia compared to the 1 h and citrullinated fibrinogen levels, in plasma and tissue, were higher in 9 h ischemia group. We propose fibrinogen citrullination as one of the mechanisms contributing to the worsening of IRI. NETs and protein citrullination represent a potential therapeutic target, but approaches are still a matter of debate. Here we introduce the idea of therapeutic approaches against citrullination to specifically inhibit PADs extracellularly, avoiding the downstream effects of hypercitrullination and keeping PADs' and NETs' intracellular regulatory functions.
Collapse
Affiliation(s)
- Valentina Zollet
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Isabel Arenas Hoyos
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Department of Plastic and Hand Surgery, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Stefanie Hirsiger
- Department of Plastic and Hand Surgery, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Bilal Ben Brahim
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Maria Francesca Petrucci
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Daniela Casoni
- Experimental Surgery Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Junhua Wang
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Rolf Spirig
- Commonwealth Serum Laboratories (CSL) Behring, Research, Commonwealth Serum Laboratories (CSL) Behring Biologics Research Center, Bern, Switzerland
| | - Kay Nettelbeck
- Experimental Surgery Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Luisana Garcia
- Experimental Surgery Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Lena Fuest
- Department of Plastic and Hand Surgery, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Esther Vögelin
- Department of Plastic and Hand Surgery, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Mihai Constantinescu
- Department of Plastic and Hand Surgery, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
van Zyl M, Cramer E, Sanders JSF, Leuvenink HGD, Lisman T, van Rooy MJ, Hillebrands JL. The role of neutrophil extracellular trap formation in kidney transplantation: Implications from donors to the recipient. Am J Transplant 2024; 24:1547-1557. [PMID: 38719094 DOI: 10.1016/j.ajt.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/19/2024] [Accepted: 04/05/2024] [Indexed: 05/23/2024]
Abstract
Kidney transplantation remains the gold standard for patients with end-stage renal disease, but severe donor organ shortage has led to long waiting lists. The utilization of expanded criteria donor kidneys within the category of deceased donors has enlarged the pool of available kidneys for transplantation; however, these grafts often have an increased risk for delayed graft function or reduced graft survival following transplantation. During brain or circulatory death, neutrophils are recruited to the vascular beds of kidneys where a proinflammatory microenvironment might prime the formation of neutrophil extracellular traps (NETs), web-like structures, containing proteolytic enzymes, DNA, and histones. NETs are known to cause tissue damage and specifically endothelial damage while activating other systems such as coagulation and complement, contributing to tissue injury and an unfavorable prognosis in various diseases. In lung transplantation and kidney transplantation studies, NETs have also been associated with primary graft dysfunction or rejection. In this review, the role that NETs might play across the different phases of transplantation, already initiated in the donor, during preservation, and in the recipient, will be discussed. Based on current knowledge, NETs might be a promising therapeutic target to improve graft outcomes.
Collapse
Affiliation(s)
- Maryna van Zyl
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands; Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Elodie Cramer
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Ton Lisman
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Mia-Jeanne van Rooy
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
18
|
Kırgız Ö, Altuğ M, Özkan H, Han M, Akçakavak G, Özarslan A, Yücel S. 45S5 Bioactive Glass-Ointment Positively Effects on Wound Healing in Rats by Regulating TNFα, Il-10, VEGF, and TGFβ. J Clin Lab Anal 2024; 38:e25094. [PMID: 39235180 PMCID: PMC11484740 DOI: 10.1002/jcla.25094] [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: 04/07/2024] [Revised: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
AIM This study aimed to investigate the effects of 45S5 bioactive glass-ointment (BG) on cutaneous wound healing in rats at the molecular, biochemical, and histopathological levels. MATERIALS AND METHODS Thirty-two rats were divided into four groups (n = 8): Control, Sham, BG, and DEX (Dexpanthenol). While no wound treatment was applied to the CONTROL, a wound model was created in the Sham, and no treatment was applied. A wound model was created for other groups, and BG and DEX were applied locally for 21 days. During the 21-day experiment period, feed and water consumption and weight changes were observed. Wound areas were calculated on days 0, 3, 7, 4, and 21. Following treatment, the rats were euthanized and tissues from the wound area and blood samples were collected. While the expression levels of tumor necrosis factor-alpha (TNFα), Interleukin 6 (IL6), Interleukin 10 (IL10), transforming growth factor-beta (TGFβ), and vascular endothelial growth factor (VEGF) genes were determined by qPCR, the levels of TNFα, IL6, and IL10 proteins were measured by ELISA. RESULTS It was observed that the BG group showed anti-inflammatory activity by suppressing TNFα levels and stimulating IL-10. In addition, it was determined that BG increased fibroblast activity and vascularization. CONCLUSION Current findings showed that topical application of BG has anti-inflammatory effects, while also accelerating healing by increasing vascularity and making positive contributions to tissue healing.
Collapse
Affiliation(s)
- Ömer Kırgız
- Department of Surgery, Faculty of Veterinary MedicineHatay Mustafa Kemal UniversityHatayTurkey
| | - Muhammed Enes Altuğ
- Department of Surgery, Faculty of Veterinary MedicineHatay Mustafa Kemal UniversityHatayTurkey
| | - Hüseyin Özkan
- Department of Genetics, Faculty of Veterinary MedicineHatay Mustafa Kemal UniversityHatayTurkey
| | - Mehmet Cengiz Han
- Department of SurgeryFirat University Faculty of Veterinary MedicineElazığTurkey
| | - Gökhan Akçakavak
- Department of PathologyAksaray University Faculty of Veterinary MedicineAksarayTurkey
| | - Ali Can Özarslan
- Department of Metallurgical and Materials Engineering, Faculty of EngineeringIstanbul University‐CerrahpasaIstanbulTurkey
- Health Biotechnology Joint Research and Application Center of ExcellenceIstanbulTurkey
| | - Sevil Yücel
- Health Biotechnology Joint Research and Application Center of ExcellenceIstanbulTurkey
- Department of Bioengineering, Faculty of Chemical and Metallurgical EngineeringYildiz Technical UniversityIstanbulTurkey
| |
Collapse
|
19
|
Tao L, Zhang Z, Li C, Huang M, Chang P. The therapeutic targets and signaling mechanisms of ondansetron in the treatment of critical illness in the ICU. Front Pharmacol 2024; 15:1443169. [PMID: 39234104 PMCID: PMC11372243 DOI: 10.3389/fphar.2024.1443169] [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: 06/03/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024] Open
Abstract
Background There is accumulating evidence regarding the benefits of the 5-HT3 receptor antagonist ondansetron for the treatment of critical illness due to its potential anti-inflammatory effect. This study attempted to determine the potential targets and molecular mechanisms of ondansetron's action against critical illnesses. Methods A bioinformatics analysis of network pharmacology was conducted to demonstrate screening targets and the signaling pathways of ondansetron action against the most common critical illnesses such as acute kidney injury (AKI), sepsis, and acute respiratory distress syndrome (ARDS). Experiments of LPS-stimulated rat neutrophils with ondansetron treatment were conducted to further validate the relevant hypothesis. Results A total of 198, 111, and 26 primary causal targets were identified from the data for the action of ondansetron against AKI, sepsis, and ARDS respectively. We found that the pathway of neutrophil extracellular traps (NETs) formation is statistically significantly involved in the action of ondansetron against these three critical illnesses. In the pathway of NETs formation, the common drug-disease intersection targets in these three critical illnesses were toll-like receptor 8 (TLR8), mitogen-activated protein kinase-14 (MAPK14), nuclear factor kappa-B1 (NFKB1), neutrophil elastase (NE), and myeloperoxidase (MPO). Considering these bioinformatics findings, we concluded that ondansetron anti-critical illness effects are mechanistically and pharmacologically implicated with suppression of neutrophils-associated inflammatory processes. It was also showed that after treatment of LPS-stimulated rat neutrophils with ondansetron, the key proteins NE, MPO, and Peptide Arginine Deaminase 4 (PAD4) in the NETs formation were significantly reduced, and the inflammatory factors IL-6, IL-1β, TNF-α, and chemokine receptor (CXCR4) were also significantly decreased. Conclusion The excessive formation of NETs may have important research value in the development and progression of critical illness. Ondansetron may reduce excessive inflammatory injury in critical diseases by reducing the formation of NETs via influencing the five targets: TLR8, NFKB1, MAPK14, NE, and MPO. Ondansetron and these primary predictive biotargets may potentially be used to treat critical illness in future clinical practice.
Collapse
Affiliation(s)
- Lili Tao
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhenhui Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chuang Li
- Department of Emergency Department, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Minxuan Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Yang T, Peng J, Zhang Z, Chen Y, Liu Z, Jiang L, Jin L, Han M, Su B, Li Y. Emerging therapeutic strategies targeting extracellular histones for critical and inflammatory diseases: an updated narrative review. Front Immunol 2024; 15:1438984. [PMID: 39206200 PMCID: PMC11349558 DOI: 10.3389/fimmu.2024.1438984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular histones are crucial damage-associated molecular patterns involved in the development and progression of multiple critical and inflammatory diseases, such as sepsis, pancreatitis, trauma, acute liver failure, acute respiratory distress syndrome, vasculitis and arthritis. During the past decade, the physiopathologic mechanisms of histone-mediated hyperinflammation, endothelial dysfunction, coagulation activation, neuroimmune injury and organ dysfunction in diseases have been systematically elucidated. Emerging preclinical evidence further shows that anti-histone strategies with either their neutralizers (heparin, heparinoids, nature plasma proteins, small anion molecules and nanomedicines, etc.) or extracorporeal blood purification techniques can significantly alleviate histone-induced deleterious effects, and thus improve the outcomes of histone-related critical and inflammatory animal models. However, a systemic evaluation of the efficacy and safety of these histone-targeting therapeutic strategies is currently lacking. In this review, we first update our latest understanding of the underlying molecular mechanisms of histone-induced hyperinflammation, endothelial dysfunction, coagulopathy, and organ dysfunction. Then, we summarize the latest advances in histone-targeting therapy strategies with heparin, anti-histone antibodies, histone-binding proteins or molecules, and histone-affinity hemoadsorption in pre-clinical studies. Finally, challenges and future perspectives for improving the clinical translation of histone-targeting therapeutic strategies are also discussed to promote better management of patients with histone-related diseases.
Collapse
Affiliation(s)
- Tinghang Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Peng
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Luojia Jiang
- Jiujiang City Key Laboratory of Cell Therapy, Department of Nephrology, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Lunqiang Jin
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Mei Han
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Med+ Biomaterial Institute of West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
- Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Luo G, Gao Y, Zheng Z, Gao B, Wang L, Tang X, Lei C. Exploring the role of ferroptosis-related genes as biomarkers in acute kidney injury. PLoS One 2024; 19:e0307472. [PMID: 39042632 PMCID: PMC11265698 DOI: 10.1371/journal.pone.0307472] [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: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is a severe condition with high morbidity and mortality. Innovative biomarkers and treatments are essential for improving patient outcomes. This study aims to investigate the role of ferroptosis-related genes (FRGs) in AKI for identifying potential biomarkers and therapeutic targets. METHODS We analyzed mRNA expression profiles from the Gene Expression Omnibus (GEO: GSE139061) dataset, comparing 36 AKI samples with 9 normal samples. Differentially expressed genes (DEGs) were identified using the R software package limma. Functional enrichment analyses were conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Key biomarkers were validated through area under the curve (AUC) values, and immune cell infiltration was analyzed using CIBERSORT. RESULTS We identified 78 differentially expressed FRGs, with 27 up-regulated and 51 down-regulated genes. Key signaling pathways included MAPK, ferroptosis, and p53. Five genes-NR4A1, GLRX5, USP35, AEBP2, and MDM4-were identified as potential biomarkers, each demonstrating AUC values greater than 0.800. Specifically, MDM4 showed significant potential by promoting the phosphorylation of p53 at Ser46, enhancing mitochondrial apoptotic activity. Immune analysis revealed a significant elevation of M0 macrophages in AKI samples compared to normal samples (P < 0.01). CONCLUSION Our findings highlight the critical role of ferroptosis-related genes in AKI, identifying NR4A1, GLRX5, USP35, AEBP2, and MDM4 as key biomarkers with high diagnostic potential. These results provide novel insights into the molecular mechanisms of AKI.
Collapse
Affiliation(s)
- Gang Luo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Yi Gao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Ziyu Zheng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Baobao Gao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Lini Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Xuemiao Tang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| | - Chong Lei
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Shaanxi, China
| |
Collapse
|
22
|
陶 怀, 骆 金, 闻 志, 虞 亘, 苏 萧, 王 鑫, 关 翰, 陈 志. [High STING expression exacerbates renal ischemia-reperfusion injury in mice by regulating the TLR4/NF-κB/NLRP3 pathway and promoting inflammation and apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1345-1354. [PMID: 39051080 PMCID: PMC11270667 DOI: 10.12122/j.issn.1673-4254.2024.07.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Indexed: 07/27/2024]
Abstract
OBJECTIVE To investigate renal expression level of STING in mice with renal ischemia-reperfusion injury (IRI) and its regulatory role in IRI. METHODS C57BL/6 mice were divided into sham operation group, IRI (induced by clamping the renal artery) model group, IRI+DMSO treatment group, and IRI+SN-011 treatment group. Serum creatinine and blood urea nitrogen of the mice were analyzed, and pathological changes in the renal tissue were assessed with PAS staining. RT-qPCR, ELISA, Western blotting, and immunohistochemistry were used to detect the expression levels of STING, KIM-1, Bcl-2, Bax, caspase-3, TLR4, P65, NLRP3, caspase-1, CD68, MPO, IL-1β, IL-6, and TNF-α in the renal tissues. In the cell experiment, HK-2 cells exposed to hypoxia-reoxygenation (H/R) were treated with DMSO or SN-011, and cellular STING expression levels and cell apoptosis were analyzed using RT-qPCR, Western blotting or flow cytometry. RESULTS In C57BL/6 mice, renal IRI induced obvious renal tissue damage, elevation of serum creatinine and blood urea nitrogen levels and renal expression levels of KIM-1, STING, TLR4, P65, NLRP3, caspase-1, caspase-3, Bax, CD68, MPO, IL-1β, IL-6, and TNF-α, and reduction of Bcl-2 expression level. Treatment of the mouse models with SN-011 for inhibiting STING expression significantly alleviated these changes. In HK-2 cells, H/R exposure caused significant elevation of cellular STING expression and obviously increased cell apoptosis rate, which was significantly lowered by treatment with SN-011. CONCLUSION Renal STING expression is elevated in mice with renal IRI to exacerbate renal injury by regulating the TLR4/NF-κB/NLRP3 pathway and promoting inflammation and apoptosis in the renal tissues.
Collapse
|
23
|
Mousset A, Bellone L, Gaggioli C, Albrengues J. NETscape or NEThance: tailoring anti-cancer therapy. Trends Cancer 2024; 10:655-667. [PMID: 38664080 DOI: 10.1016/j.trecan.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 07/12/2024]
Abstract
Neutrophils, major regulators of innate immunity, have recently emerged as key components of the tumor microenvironment. The role of neutrophils in cancer has been linked to their ability to form neutrophil extracellular traps (NETs), structures composed of decondensed DNA decorated with enzymes that are released into the extracellular space. Here, we discuss the pivotal roles of NETs in influencing responses to anticancer therapies such as chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Highlighting recent insights, we delve into the dual nature of NETs in the context of anticancer treatments, examining their potential to either counteract or enhance treatment outcomes. Strategic targeting of NETs may be a promising avenue for crafting combination therapies to counteract resistance or enhance anticancer treatments' efficacy.
Collapse
Affiliation(s)
- Alexandra Mousset
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Lola Bellone
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Cedric Gaggioli
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Jean Albrengues
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France.
| |
Collapse
|
24
|
Mousset A, Albrengues J. Neutrophil extracellular traps modulate chemotherapy efficacy and its adverse side effects. Biol Cell 2024; 116:e2400031. [PMID: 38724262 DOI: 10.1111/boc.202400031] [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: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 07/13/2024]
Abstract
Neutrophils, major regulator of innate immunity have recently emerged as key components of the tumor microenvironment. The role of neutrophils in cancer has been linked to their ability to form neutrophil extracellular traps (NETs), structures composed of decondensed DNA decorated with enzymes that are released into the extracellular space. Here, we discuss the pivotal roles of NETs, in influencing responses to chemotherapy and its severe adverse effect. Highlighting recent insights, we discuss the dual nature of NETs in the context of chemotherapy treatment, examining their potential to either counteract or enhance treatment outcomes. Strategic targeting of NETs emerges as a promising avenue for determining combination therapies that could help counteracting resistance or enhancing chemotherapy efficacy as well as limiting complications due to this type of treatment.
Collapse
Affiliation(s)
- Alexandra Mousset
- Institute for Research on Cancer and Aging, University Côte d'Azur, Nice, France
| | - Jean Albrengues
- Institute for Research on Cancer and Aging, University Côte d'Azur, Nice, France
| |
Collapse
|
25
|
Sheng XH, Han LC, Gong A, Meng XS, Wang XH, Teng LS, Sun XH, Xu KC, Liu ZH, Wang T, Ma JP, Zhang L. Discovery of Novel Ortho-Aminophenol Derivatives Targeting Lipid Peroxidation with Potent Antiferroptotic Activities. J Med Chem 2024; 67:9536-9551. [PMID: 38822802 DOI: 10.1021/acs.jmedchem.4c00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
The concept of ferroptosis inhibition has gained growing recognition as a promising therapeutic strategy for addressing a wide range of diseases. Here, we present the discovery of four series of ortho-aminophenol derivatives as potential ferroptosis inhibitors beginning with the endogenous substance 3-hydroxyanthranilic acid (3-HA) by employing quantum chemistry techniques, in vitro and in vivo assays. Our findings reveal that these ortho-aminophenol derivatives exhibit unique intra-H bond interactions, compelling ortho-amines to achieve enhanced alignment with the aromatic π-system, thereby expanding their activity. Notably, compounds from all four series display remarkable activity against RSL3-induced ferroptosis, showcasing an activity 100 times more than that of 3-HA. Furthermore, these compounds also demonstrate robust in vivo efficacy in protecting mice from kidney ischemia-reperfusion injury and acetaminophen-induced hepatotoxicity. In summary, we provide four distinct series of active scaffolds that significantly expand the chemical space of ferroptosis inhibitors, serving as valuable insights for future structural modifications.
Collapse
Affiliation(s)
- Xie-Huang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Li-Cong Han
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Ao Gong
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250001, China
| | - Xiang-Shuai Meng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Xin-Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Lin-Song Teng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| | - Xiao-Han Sun
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Kuo-Chen Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| | - Zhao-Hua Liu
- The Model Animal Research Center, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Ting Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Jian-Ping Ma
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, China
| | - Lei Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan 250014, China
| |
Collapse
|
26
|
Yuan X, Sheng L, Shi G, Jiang L, Lian C. Colchicine Alleviates Rosacea by Inhibiting Neutrophil Inflammation Activated by the TLR2 Pathway. Inflammation 2024; 47:1002-1014. [PMID: 38279067 DOI: 10.1007/s10753-023-01956-6] [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: 12/11/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/28/2024]
Abstract
Rosacea is a chronic facial inflammatory skin disease that occurs with dysfunction of the immune system. Colchicine was reported to have anti-inflammatory properties. However, the impact of colchicine on rosacea remains unclear. In the present study, the phenotype of rosacea lesions was evaluated by the redness score, inflammatory biomarkers were analyzed by reverse transcription PCR (RT‒PCR), and the infiltration of inflammatory cells was assessed by IHC analysis and immunofluorescence in a rosacea-like mouse model. In vitro, RT‒PCR was used to identify the inflammatory factors that Toll-like receptor 2 (TLR2) agonist caused neutrophils to produce, and immunofluorescence and coimmunoprecipitation were used to identify putative signalling pathways. We found that skin erythema and histopathological alterations, as well as elevated proinflammatory factors (IL-1β, IL-6, TNFα, CXCL2) and CAMP, were significantly ameliorated by colchicine treatment in LL37-induced rosacea-like mice. In addition, colchicine reduced the colocalization of TLR2 and neutrophils and the formation of neutrophil extracellular trap networks (NET) in mouse lesions. In neutrophils, colchicine markedly reduced TLR2 agonist-induced inflammatory biomarker expression, NET formation, and ROS production. Moreover, we found that LL37 could bind to TLR2 upon activation of TLR2 in neutrophils. Importantly, colchicine could repress the combination of TLR2 and LL37 in vivo. Finally, bioinformatics methods further validated the key molecules of neutrophil-related inflammation in rosacea, which is consistent with our experimental findings. Collectively, colchicine ameliorated rosacea-like dermatitis by regulating the neutrophil immune response activated by the TLR2 pathway, indicating that it could be an effective therapeutic option for patients with rosacea.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Dermatology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Liang Sheng
- Department of Dermatology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Guang Shi
- Department of Dermatology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Leiwei Jiang
- Department of Dermatology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China.
- GuiYang First People Hospital, Guiyang, Guizhou, China.
| | - Chengxiang Lian
- Department of Dermatology, The First Affiliated Hospital of Jinan University & Jinan University Institute of Dermatology, Guangzhou, 510632, China.
| |
Collapse
|
27
|
Zheng Q, Li X, Xu X, Tang X, Hammad B, Xing J, Zhang D. The mmu_circ_003062, hsa_circ_0075663/miR-490-3p/CACNA1H axis mediates apoptosis in renal tubular cells in association with endoplasmic reticulum stress following ischemic acute kidney injury. Int Immunopharmacol 2024; 132:111956. [PMID: 38554447 DOI: 10.1016/j.intimp.2024.111956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND While recent studies have suggested a potential involvement of circRNAs in acute kidney injury (AKI) after ischemia, mmu_circ_003062 role is undetermined. METHODS The levels of mmu_circ_003062, miR-490-3p, CACNA1H, GRP78, CHOP and hsa_circ_0075663 were detected by Relative qPCR in Boston University mouse proximal tubule (BUMPT) cells, mouse kidneys, and human renal tubular epithelial (HK-2) cells. Moreover, the levels of hsa_circ_0075663 in serum and urine of patients with AKI following cardiopulmonary resuscitation (CPR) were detected by absolute quantitative PCR. Western blot was used to detect the relative expression of the protein. The function and regulatory mechanism of mmu_circ_003062 and hsa_circ_0075663 were investigated through a series of in vitro and in vivo experiments, including bioinformatic prediction, luciferase reporter assays, FISH, FCM, TUNEL staining, and H&E staining. RESULTS It was found that mmu_circ_003062, hsa_circ_0075663 mediated apoptosis after ischemia/reperfusion (I/R) by interaction with miR-490-3p to enhance CACNA1H expression, thereby leading to the upregulation of endoplasmic reticulum stress (ERS)-relevant proteins GRP78 and CHOP. Ultimately, mmu_circ_003062 downregulation significantly ameliorated ischemic AKI by modulating the miR-490-3p/CACNA1H/GRP78 and CHOP pathway. Furthermore, the plasma and urinary levels of hsa_circ_0075663 in patients with AKI following CPR were significantly higher than non-AKI patients, exhibited a strongly correlation with serum creatinine. CONCLUSION The involvement of mmu_circ_003062, hsa_circ_0075663/miR-490-3p/CACNA1H/GRP78 and CHOP axis is significant in the development of ischemic AKI. Moreover, hsa_circ_0075663 has potential as an early diagnostic biomarker.
Collapse
Affiliation(s)
- Qiang Zheng
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaozhou Li
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xuan Xu
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xianming Tang
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bacha Hammad
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jihong Xing
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Dongshan Zhang
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
Li Z, Xing J. Potential therapeutic applications of circular RNA in acute kidney injury. Biomed Pharmacother 2024; 174:116502. [PMID: 38569273 DOI: 10.1016/j.biopha.2024.116502] [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: 12/27/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
Acute kidney injury (AKI) is a common clinical syndrome characterized by a rapid deterioration in renal function, manifested by a significant increase in creatinine and a sharp decrease in urine output. The incidence of morbidity and mortality associated with AKI is on the rise, with most patients progressing to chronic kidney disease or end-stage renal disease. Treatment options for patients with AKI remain limited. Circular RNA (circRNA) is a wide and diverse class of non-coding RNAs that are present in a variety of organisms and are involved in gene expression regulation. Studies have shown that circRNA acts as a competing RNA, is involved in disease occurrence and development, and has potential as a disease diagnostic and prognostic marker. CircRNA is involved in the regulation of important biological processes, including apoptosis, oxidative stress, and inflammation. This study reviews the current status and progress of circRNA research in the context of AKI.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
29
|
Li C, Wu C, Li F, Xu W, Zhang X, Huang Y, Xia D. Targeting Neutrophil Extracellular Traps in Gouty Arthritis: Insights into Pathogenesis and Therapeutic Potential. J Inflamm Res 2024; 17:1735-1763. [PMID: 38523684 PMCID: PMC10960513 DOI: 10.2147/jir.s460333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Gouty arthritis (GA) is an immune-mediated disorder characterized by severe inflammation due to the deposition of monosodium urate (MSU) crystals in the joints. The pathophysiological mechanisms of GA are not yet fully understood, and therefore, the identification of effective therapeutic targets is of paramount importance. Neutrophil extracellular traps (NETs), an intricate structure of DNA scaffold, encompassing myeloperoxidase, histones, and elastases - have gained significant attention as a prospective therapeutic target for gouty arthritis, due to their innate antimicrobial and immunomodulatory properties. Hence, exploring the therapeutic potential of NETs in gouty arthritis remains an enticing avenue for further investigation. During the process of gouty arthritis, the formation of NETs triggers the release of inflammatory cytokines, thereby contributing to the inflammatory response, while MSU crystals and cytokines are sequestered and degraded by the aggregation of NETs. Here, we provide a concise summary of the inflammatory processes underlying the initiation and resolution of gouty arthritis mediated by NETs. Furthermore, this review presents an overview of the current pharmacological approaches for treating gouty arthritis and summarizes the potential of natural and synthetic product-based inhibitors that target NET formation as novel therapeutic options, alongside elucidating the intrinsic challenges of these inhibitors in NETs research. Lastly, the limitations of HL-60 cell as a suitable substitute of neutrophils in NETs research are summarized and discussed. Series of recommendations are provided, strategically oriented towards guiding future investigations to effectively address these concerns. These findings will contribute to an enhanced comprehension of the interplay between NETs and GA, facilitating the proposition of innovative therapeutic strategies and novel approaches for the management of GA.
Collapse
Affiliation(s)
- Cantao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Wenjing Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoxi Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
30
|
Chen L, Lu S, Wu Z, Zhang E, Cai Q, Zhang X. Innate immunity in diabetic nephropathy: Pathogenic mechanisms and therapeutic targets. MEDCOMM – FUTURE MEDICINE 2024; 3. [DOI: 10.1002/mef2.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/18/2024] [Indexed: 01/02/2025]
Abstract
AbstractDiabetic nephropathy (DN) represents a prevalent chronic microvascular complication of diabetes mellitus (DM) and is a major cause of end‐stage renal disease. The anfractuous surrounding of DN pathogenesis and the intricate nature of this metabolic disorder often pose challenges in both the diagnosis and treatment of DN compared to other kidney diseases. Hyperglycaemia in DM predispose vulnerable renal cells into microenvironmental disequilibrium and thereby results in innate immunocytes infiltration including neutrophils, macrophages, myeloid‐derived suppressor cells, dendritic cells, and so forth. These immune cells play dual roles in kidney injury and closely correlated with the degree of proteinuria in DN patients. Additionally, innate immune signaling cascades, initiated by altered metabolic and hemodynamic in diabetic context, are crucial in instigating and perpetuating renal inflammation, which detrimentally contribute to DN pathogenesis. As such, anti‐inflammatory therapies, particularly those targeting innate immunity, hold renoprotective promise in DN. In this article, we reviewed the origin and feature of the above four prominent kidney innate immune cells, analyze their pathogenic role in DN, and discuss potential targeted‐therapeutic strategies, aiming to enhance the current understanding of renal innate immunity and hence help to discover promising therapeutic approaches for DN.
Collapse
Affiliation(s)
- Le‐Xin Chen
- School of Pharmaceutical Science Guangzhou University of Chinese Medicine Guangzhou PR China
| | - Shu‐Ru Lu
- School of Pharmaceutical Science Guangzhou University of Chinese Medicine Guangzhou PR China
| | - Zhi‐Hao Wu
- School of Pharmaceutical Science Guangzhou University of Chinese Medicine Guangzhou PR China
| | - En‐Xin Zhang
- Shenzhen Bao'an Authentic TCM Therapy Hospital Shenzhen PR China
| | - Qing‐Qun Cai
- The First Affiliated Hospital Guangzhou University of Chinese Medicine Guangzhou PR China
| | - Xiao‐Jun Zhang
- School of Pharmaceutical Science Guangzhou University of Chinese Medicine Guangzhou PR China
| |
Collapse
|
31
|
Du C, Xu C, Jia P, Cai N, Zhang Z, Meng W, Chen L, Zhou Z, Wang Q, Feng R, Li J, Meng X, Huang C, Ma T. PSTPIP2 ameliorates aristolochic acid nephropathy by suppressing interleukin-19-mediated neutrophil extracellular trap formation. eLife 2024; 13:e89740. [PMID: 38314821 PMCID: PMC10906995 DOI: 10.7554/elife.89740] [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: 05/29/2023] [Accepted: 02/04/2024] [Indexed: 02/07/2024] Open
Abstract
Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by herbal medicines. Proline-serine-threonine phosphatase-interacting protein 2 (PSTPIP2) and neutrophil extracellular traps (NETs) play important roles in kidney injury and immune defense, respectively, but the mechanism underlying AAN regulation by PSTPIP2 and NETs remains unclear. We found that renal tubular epithelial cell (RTEC) apoptosis, neutrophil infiltration, inflammatory factor, and NET production were increased in a mouse model of AAN, while PSTPIP2 expression was low. Conditional knock-in of Pstpip2 in mouse kidneys inhibited cell apoptosis, reduced neutrophil infiltration, suppressed the production of inflammatory factors and NETs, and ameliorated renal dysfunction. Conversely, downregulation of Pstpip2 expression promoted kidney injury. In vivo, the use of Ly6G-neutralizing antibody to remove neutrophils and peptidyl arginine deiminase 4 (PAD4) inhibitors to prevent NET formation reduced apoptosis, alleviating kidney injury. In vitro, damaged RTECs released interleukin-19 (IL-19) via the PSTPIP2/nuclear factor (NF)-κB pathway and induced NET formation via the IL-20Rβ receptor. Concurrently, NETs promoted apoptosis of damaged RTECs. PSTPIP2 affected NET formation by regulating IL-19 expression via inhibition of NF-κB pathway activation in RTECs, inhibiting RTEC apoptosis, and reducing kidney damage. Our findings indicated that neutrophils and NETs play a key role in AAN and therapeutic targeting of PSTPIP2/NF-κB/IL-19/IL-20Rβ might extend novel strategies to minimize Aristolochic acid I-mediated acute kidney injury and apoptosis.
Collapse
Affiliation(s)
- Changlin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Chuanting Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Pengcheng Jia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Na Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Zhenming Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Wenna Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Lu Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Zhongnan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Qi Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Rui Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical UniversityHefeiChina
| |
Collapse
|
32
|
Sheng XH, Han LC, Ma XJ, Gong A, Hao MZ, Zhu H, Meng XS, Wang T, Sun CH, Ma JP, Zhang L. Design, synthesis, and biological evaluation of 2-amino-6-methyl-phenol derivatives targeting lipid peroxidation with potent anti-ferroptotic activities. Eur J Med Chem 2024; 264:115997. [PMID: 38056303 DOI: 10.1016/j.ejmech.2023.115997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
The suppression of ferroptosis is emerging as a promising therapeutic strategy for effectively treating a wide range of diseases, including neurodegenerative disorders, organ ischemia-reperfusion injury, and inflammatory conditions. However, the clinical utility of ferroptosis inhibitors is significantly impeded by the limited availability of rational drug designs. In our previous study, we successfully unraveled the efficacy of ferrostatin-1 (Fer-1) attributed to the synergistic effect of its ortho-diamine (-NH) moiety. In this study, we present the discovery of the ortho-hydroxyl-amino moiety as a novel scaffold for ferroptosis inhibitors, employing quantum chemistry as well as in vitro and in vivo assays. 2-amino-6-methylphenol derivatives demonstrated remarkable inhibition of RSL3-induced ferroptosis, exhibiting EC50 values ranging from 25 nM to 207 nM. These compounds do not appear to modulate iron homeostasis or lipid reactive oxygen species (ROS) generation pathways. Nevertheless, they effectively prevent the accumulation of lipid peroxides in living cells. Furthermore, compound 13 exhibits good in vivo activities as it effectively protect mice from kidney ischemia-reperfusion injury. In summary, compound 13 has been identified as a potent ferroptosis inhibitor, warranting further investigation as a promising lead compound.
Collapse
Affiliation(s)
- Xie-Huang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China
| | - Li-Cong Han
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China
| | - Xiao-Jie Ma
- Department of Rheumatology and Immunology, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Ao Gong
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China
| | - Meng-Zhu Hao
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China
| | - Hao Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China
| | - Xiang-Shuai Meng
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China
| | - Ting Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Chang-Hua Sun
- Shandong Center for Disease Control and Prevention, Jinan, 250014, China
| | - Jian-Ping Ma
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, China.
| | - Lei Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, 250014, China.
| |
Collapse
|
33
|
Jones BA, Myakala K, Guha M, Davidson S, Adapa S, Lopez Santiago I, Schaffer I, Yue Y, Allegood JC, Cowart LA, Wang XX, Rosenberg AZ, Levi M. Farnesoid X receptor prevents neutrophil extracellular traps via reduced sphingosine-1-phosphate in chronic kidney disease. Am J Physiol Renal Physiol 2023; 325:F792-F810. [PMID: 37823198 PMCID: PMC10894665 DOI: 10.1152/ajprenal.00292.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023] Open
Abstract
Farnesoid X receptor (FXR) activation reduces renal inflammation, but the underlying mechanisms remain elusive. Neutrophil extracellular traps (NETs) are webs of DNA formed when neutrophils undergo specialized programmed cell death (NETosis). The signaling lipid sphingosine-1-phosphate (S1P) stimulates NETosis via its receptor on neutrophils. Here, we identify FXR as a negative regulator of NETosis via repressing S1P signaling. We determined the effects of the FXR agonist obeticholic acid (OCA) in mouse models of adenosine phosphoribosyltransferase (APRT) deficiency and Alport syndrome, both genetic disorders that cause chronic kidney disease. Renal FXR activity is greatly reduced in both models, and FXR agonism reduces disease severity. Renal NETosis and sphingosine kinase 1 (Sphk1) expression are increased in diseased mice, and they are reduced by OCA in both models. Genetic deletion of FXR increases Sphk1 expression, and Sphk1 expression correlates with NETosis. Importantly, kidney S1P levels in Alport mice are two-fold higher than controls, and FXR agonism restores them back to baseline. Short-term inhibition of sphingosine synthesis in Alport mice with severe kidney disease reverses NETosis, establishing a causal relationship between S1P signaling and renal NETosis. Finally, extensive NETosis is present in human Alport kidney biopsies (six male, nine female), and NETosis severity correlates with clinical markers of kidney disease. This suggests the potential clinical relevance of the newly identified FXR-S1P-NETosis pathway. In summary, FXR agonism represses kidney Sphk1 expression. This inhibits renal S1P signaling, thereby reducing neutrophilic inflammation and NETosis.NEW & NOTEWORTHY Many preclinical studies have shown that the farnesoid X receptor (FXR) reduces renal inflammation, but the mechanism is poorly understood. This report identifies FXR as a novel regulator of neutrophilic inflammation and NETosis via the inhibition of sphingosine-1-phosphate signaling. Additionally, NETosis severity in human Alport kidney biopsies correlates with clinical markers of kidney disease. A better understanding of this signaling axis may lead to novel treatments that prevent renal inflammation and chronic kidney disease.
Collapse
Affiliation(s)
- Bryce A Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, District of Columbia, United States
| | - Komuraiah Myakala
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Mahilan Guha
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Shania Davidson
- Department of Biology, Howard University, Washington, District of Columbia, United States
| | - Sharmila Adapa
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Isabel Lopez Santiago
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Isabel Schaffer
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Yang Yue
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Jeremy C Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| |
Collapse
|
34
|
Wang H, Gao T, Zhang R, Hu J, Gao S, Wang Y, Qi X, Zhou Y, Zheng G, Dong H. Neutrophil Extracellular Traps Aggravate Contrast-Induced Acute Kidney Injury by Damaging Glomeruli and Peritubular Capillaries. J Inflamm Res 2023; 16:5629-5646. [PMID: 38046404 PMCID: PMC10693253 DOI: 10.2147/jir.s433110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023] Open
Abstract
Background Contrast-induced acute kidney injury (CI-AKI) is considered to be the third leading cause of hospital-acquired kidney injury. Current studies mostly suggest that contrast agents mainly harm renal tubular epithelial cells, but we hypothesized that the development of CI-AKI should be the result of the interaction of renal vascular and tubular injury. Methods First we constructed a CI-AKI mouse model and verified the success of the model by pathological injury and serum creatinine level. Immunohistochemistry, protein quantification and qRT-PCR were used to detect the location and level of expression of neutrophil extracellular traps (NETs) in the kidney. Then, we blocked the in vivo accumulation of NETs using GSK484 and DNase I and detected the expression of NETs and the damage of glomerular and peritubular capillaries. Results We first identified the presence of NETs in CI-AKI mice, and NETs were mainly accumulated in glomeruli and peritubular capillaries. The expression of NETs was positively correlated with the severity of CI-AKI kidney. After inhibition of NETs release or promotion of NETs degradation by drugs, renal vascular endothelial cell injury was reduced and renal pathological changes and creatinine levels were reversed in CI-AKI mice. In addition, inhibition of NETs reduced apoptosis and pyroptosis of renal cells and attenuated inflammation in vivo. Conclusion These findings suggest that NETs are involved in the development of CI-AKI by damaging glomerular and peritubular capillary endothelial cells. This study will provide a new strategy for clinical prevention and treatment of CI-AKI.
Collapse
Affiliation(s)
- Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jie Hu
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yuwen Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xiaotong Qi
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yun Zhou
- Shanxi Provincial Integrated TCM and WM Hospital, Taiyuan, People’s Republic of China
| | - Guoping Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
35
|
虞 亘, 王 鑫, 骆 金, 苏 萧, 陶 怀, 闻 志, 关 翰. [Role of SPP1 in acute kidney injury induced by renal ischemia-reperfusion in rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1947-1954. [PMID: 38081614 PMCID: PMC10713472 DOI: 10.12122/j.issn.1673-4254.2023.11.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Indexed: 12/18/2023]
Abstract
OBJECTIVE To investigate the role of SPP1 gene in acute kidney injury induced by renal ischemia-reperfusion injury (IRI). METHODS Twelve Sprague-Dawley rats were randomly divided into sham group and IRI group (n=6) and subjected to sham operation and renal ischemia for 30 min induced by penal pedicle clamping using non-traumatic microvascular clamps, respectively.Serum creatinine and blood urea nitrogen levels were detected, and PAS staining was used for pathological examination of the kidneys in the two groups.The renal expressions of SPP1, α-SMA and caspase-3 were detected using immunohistochemistry and immunofluorescent staining.In cultured renal tubular epithelial cells (HK-2 cells), Western blotting was performed to detect the changes in expressions of SPP1, caspase-3, and Kim-1 proteins following hypoxiareoxygenation (H/R) and transfection with si-NC or si-SPP1;flow cytometry was employed to analyze apoptosis of the treated cells. RESULTS Renal IRI caused significant elevations of serum creatinine and blood urea nitrogen levels (P<0.05) and induced severe shedding and necrosis of the renal tubular epithelial cells in the rats, resulting also in significantly up-regulated renal expressions of SPP1, α-SMA and caspase-3(P<0.05).In HK-2 cells, H/R significantly increased the protein expression levels of SPP1, caspase-3, and Kim-1(P<0.05), and compared si-NC transfection, transfection with SPP1 obviously reduced caspase-3 and Kim-1 expressions and lowered apoptosis rate of the cells with H/R exposure (P<0.05). CONCLUSION SPP1 is up-regulated in the kidneys of rats with renal IRI, and down-regulation of SPP1 expression can inhibit H/R-induced apoptosis of renal tubular epithelial cells.
Collapse
Affiliation(s)
- 亘明 虞
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 鑫玮 王
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 金光 骆
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 萧 苏
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 怀祥 陶
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| | - 志远 闻
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 翰 关
- 蚌埠医学院第一附属医院泌尿外科,安徽 蚌埠 233004Department of Urology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
- 蚌埠医学院慢性疾病免疫学基础与临床安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Immunology in Chronic Disease, Bengbu Medical College, Bengbu 233030, China
| |
Collapse
|
36
|
Yang W, Zhang M, Li J, Qu S, Zhou F, Liu M, Li L, Liu Z, Zen K. YTHDF1 mitigates acute kidney injury via safeguarding m 6A-methylated mRNAs in stress granules of renal tubules. Redox Biol 2023; 67:102921. [PMID: 37857002 PMCID: PMC10587769 DOI: 10.1016/j.redox.2023.102921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 10/21/2023] Open
Abstract
Acute kidney injury (AKI) presents a daunting challenge with limited therapeutic options. To explore the contribution of N6-methyladenosine (m6A) in AKI development, we have investigated m6A-modified mRNAs within renal tubular cells subjected to injuries induced by diverse stressors. Notably, while the overall level of m6A-modified RNA remains unaltered in renal tubular cells facing stress, a distinct phenomenon emerges-mRNAs bearing m6A methylation exhibit a pronounced tendency to accumulate within stress granules (SGs), structures induced in response to these challenges. Cumulation of m6A-modified mRNA in SGs is orchestrated by YTHDF1, a m6A 'reader' closely associated with SGs. Strikingly, AKI patients and various mouse AKI models showcase elevated levels of renal tubular YTHDF1. Depleting YTHDF1 within renal tubular cells leads to a marked reduction in m6A-modified mRNA accumulation within SGs, accompanied by an escalation in cell apoptosis under stress challenges. The significance of YTHDF1's protective role is further underscored by findings in AKI mouse models triggered by cisplatin or renal ischemia-reperfusion treatments. In particular, renal tubular-specific YTHDF1 knockout mice exhibit heightened AKI severity when contrasted with their wild-type counterparts. Mechanistic insights reveal that YTHDF1 fulfills a crucial function by safeguarding m6A-modified mRNAs that favor cell survival-exemplified by SHPK1-within SGs amid stress-challenged renal tubular cells. Our findings collectively shed light on the pivotal role of YTHDF1 in shielding renal tubules against AKI, through its adeptness in recruiting and preserving m6A-modified mRNAs within stress-induced SGs.
Collapse
Affiliation(s)
- Wenwen Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, Jiangsu, 210046, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China
| | - Jiacheng Li
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, Jiangsu, 210046, China
| | - Shuang Qu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fenglian Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, Jiangsu, 210046, China
| | - Minghui Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Limin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, Jiangsu, 210046, China.
| |
Collapse
|
37
|
Geng H, An Q, Zhang Y, Huang Y, Wang L, Wang Y. Role of Peptidylarginine Deiminase 4 in Central Nervous System Diseases. Mol Neurobiol 2023; 60:6748-6756. [PMID: 37480499 DOI: 10.1007/s12035-023-03489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/05/2023] [Indexed: 07/24/2023]
Abstract
The deimination or citrullination of arginine residues in the polypeptide chain by peptidylarginine deiminase 4 alters the charge state of the polypeptide chain and affects the function of proteins. It is one of the main ways of protein post-translational modifications to regulate its function. Peptidylarginine deiminase 4 is widely expressed in multiple tissues and organs of the body, especially the central nervous system, and regulates the normal development of organisms. The abnormal expression and activation of peptidylarginine deiminase 4 is an important pathological mechanism for the occurrence and development of central nervous system diseases such as multiple sclerosis, Alzheimer's disease, cerebral ischemia reperfusion injury, and glioblastoma.
Collapse
Affiliation(s)
- Huixia Geng
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health Sciences, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China
| | - Qihang An
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health Sciences, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China
| | - Yanshuo Zhang
- School of Life Science, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China
| | - Yunhang Huang
- School of Life Science, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China
| | - Lai Wang
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health Sciences, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China.
- School of Life Science, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China.
| | - Yanming Wang
- School of Life Science, Henan University, Henan Province, Kaifeng, 475004, People's Republic of China.
| |
Collapse
|
38
|
Liu X, Qian N, Zhu L, Fan L, Fu G, Ma M, Bao J, Cao C, Liang X. Geniposide ameliorates acute kidney injury via enhancing the phagocytic ability of macrophages towards neutrophil extracellular traps. Eur J Pharmacol 2023; 957:176018. [PMID: 37634840 DOI: 10.1016/j.ejphar.2023.176018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Acute kidney injury (AKI) is a clinically serious disorder associated with high mortality rates and an increased risk of progression to end-stage renal disease. As an essential supportive treatment for patients with respiratory failure, mechanical ventilation not only save many critically ill patients, but also affect glomerular filtration function by changing renal hemodynamics, neurohumoral and positive end-expiratory pressure, eventually leading to AKI. AMP-activated protein kinase (AMPK), a crucial energy homeostasis regulator, could enhance macrophage phagocytic ability and inhibit inflammation, but whether it can engulf neutrophil extracellular traps (NETs) and alleviate mechanical ventilation-associated AKI is still unclear. In this study, we found that geniposide significantly ameliorated histopathological damage, reduced serum Cre and BUN levels. Besides, geniposide can also induce AMPK activation and enhance macrophage phagocytic ability toward NETs. Moreover, geniposide can markedly reduce the levels of high mobility group box 1 (HMGB1), and these effects were dependent on AMPK-PI3K/Akt signaling. Altogether, these results indicated that geniposide promoted macrophage efferocytosis by inducing AMPK-PI3K/Akt signaling activation, clearing NETs and ameliorating AKI.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China; The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Na Qian
- The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Li Zhu
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Li Fan
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China; Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Guanghao Fu
- The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Mengqing Ma
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Jiaxin Bao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Changchun Cao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China.
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
39
|
Wu X, You D, Pan M, Weng M, Xie Q, Guan Y, Zheng J, Lin S, Zhang X, Hao C, Wan J. Knockout of the C3a receptor protects against renal ischemia reperfusion injury by reduction of NETs formation. Cell Mol Life Sci 2023; 80:322. [PMID: 37816851 PMCID: PMC11072185 DOI: 10.1007/s00018-023-04967-6] [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: 04/26/2023] [Revised: 08/20/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023]
Abstract
Renal ischemia/reperfusion (I/R) injury is a local sterile inflammatory response driven by innate immunity. Emerging data have revealed that complement and neutrophils contribute to hyperinflammation and oxidative stress in I/R induced acute kidney injury (AKI). However, the interplay between the C3a/C3aR axis and neutrophil extracellular traps (NETs) is imcompletelyunderstood. Here, we utilize genetically engineered mouse models and pharmacological inhibitors to investigate this association. The C3a/C3aR axis is found to promote neutrophil recruitment and NETs formation, thereby accelerating renal damage and dysfunction. Knockout of C3aR restores NETs release and improves renal function after I/R injury. Antibody-mediated blockade of NETs can also significantly ameliorate renal tubular injury and inflammation. Consistently, under stimulation by C3a, neutrophils are activated to promote NETs formation and subsequent renal tubular epithelial cell damage, and blocking C3aR rescued the injury. Interfering with reactive oxygen species (ROS) accumulation in neutrophils by antioxidant treatment significantly attenuates NETs formation. Our findings demonstrate that the C3a/C3aR-ROS-NETs axis constitutes a promising target for prevention or treatment of renal I/R injury.
Collapse
Affiliation(s)
- Xiaoting Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Danyu You
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Maoen Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mengjie Weng
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qionghong Xie
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yi Guan
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Zheng
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Songhua Lin
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiaohong Zhang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Chuanming Hao
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
40
|
Zhao C, Liang F, Ye M, Wu S, Qin Y, Zhao L, Zhang L, He J, Cen L, Lin F. GSDMD promotes neutrophil extracellular traps via mtDNA-cGAS-STING pathway during lung ischemia/reperfusion. Cell Death Discov 2023; 9:368. [PMID: 37794018 PMCID: PMC10551007 DOI: 10.1038/s41420-023-01663-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
Lung ischemia/reperfusion injury (LIRI) is a complex pathophysiological process, with the histopathological hallmark of neutrophils migrating into the lungs. Neutrophil extracellular traps (NETs) have been suggested to exert a critical role in the pathogenesis of inflammation and infection in humans and animals, while the exact functions and underlying mechanisms of NETs in LIRI remain insufficiently elucidated. In this study, we investigated the role of pore-forming protein gasdermin D (GSDMD) on NETs release in LIRI induced by lung ischemia/reperfusion (I/R). We found that disulfiram, a GSDMD inhibitor, dramatically reduced NETs release and pathological injury in lung I/R in vivo and in vitro. Additionally, GSDMD caused mitochondrial DNA (mtDNA) leaking into the neutrophil cytosol, and then the cytoplasmic mtDNA activated the cGAS-STING signaling pathway and stimulated NETs formation in lung I/R. Furthermore, inhibition of cGAS/STING pathway could inhibit cytosol mtDNA mediated NETs formation.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Fangte Liang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Mengling Ye
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siyi Wu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Yi Qin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Lu Zhao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Lu Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Jing He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Liming Cen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China.
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China.
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China.
| |
Collapse
|
41
|
Kumar M, Kenwar DB, Sekar A, Singh J, Nada R, Saikia B, Sharma A, Kohli HS, Anand S, Minz RW. Circulating "Neutrophils extra-cellular traps" during the early post-renal transplant period and correlation with graft dysfunction and rejection. Transpl Immunol 2023; 80:101898. [PMID: 37437666 DOI: 10.1016/j.trim.2023.101898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) have a role in infection, autoimmunity, autoinflammation, thrombosis, ischemia-reperfusion injury (IRI), epithelial-mesenchymal transition, vasculitis, and metabolic diseases. However, its role in early graft injury and graft outcome has not been elucidated till now. We evaluated the circulating NETs during early post-transplant periods and their correlation with graft outcome and IRI. METHODS Prospectively, thirty kidney transplants recipient (KTR) were recruited and grouped into non-dysfunction (Group-A) and dysfunction groups (Group-B). Serum levels of circulating NETs were estimated by measuring myeloperoxidase-DNA complex at three-time points: pre-transplant, 8 h post-transplant, and 18 h post-transplant; and correlated with early graft outcome. Malondialdehyde (MDA), a marker of oxidative stress or IRI, was also measured to assess its relation with NETs and early graft outcome. RESULTS Circulating NETs were significantly increased in both non-dysfunctional [Median OD: 0.11 (0.01-0.19) to 0.51 (0.22-0.91); p = 0.001] and dysfunctional [Median OD: 0.16 (0.12-0.27) to 0.38 (0.19-0.68); p = 0.047] KTR during first 8 h of transplant followed by fall at 18 h post-transplant [0.25 (0.18-0.72) and 0.35 (0.26-0.36) respectively]; however, no significant difference were observed between two groups at any time points. Isolated biopsy-proven graft rejection KTR also had higher circulating NETs during the early post-transplant period [Median OD: 0.16 (0.13-0.31) to 0.38 (0.28-1.5); p > 0.05] but no significant difference compared to non-dysfunctional KTR. MDA also displayed similar trends with an early significant rise [9.30 (7.74-12.56) μM to 17.37 (9.11-22.25) μM; p = 0.03 in group-A, and 8.7 (6.04-10.30) μM to 14.66 (13.39-21.63) μM; p = 0.01in group-B] followed by fall at 18 h in both groups [10.21 (7.64-13.90) μM and 11.11 (9.15-17.54) μM respectively]. Despite similar trends of both NETs and MDA, there was no significant correlation between these; however, creatinine exhibits a significant inverse correlation with NETs and MDA both. CONCLUSION Circulating NETs are significantly increased during the early post-transplant period in KTR irrespective of early graft outcome. Similar dynamics of MDA indicate that the early rise of NETs might be a part of IRI. However, molecular studies with large sample sizes and longer follow up are required to reach more defined conclusions.
Collapse
Affiliation(s)
- Mahendra Kumar
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Deepesh B Kenwar
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Aravind Sekar
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jagdeep Singh
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritambhra Nada
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Biman Saikia
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashish Sharma
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harbir Singh Kohli
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shashi Anand
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ranjana W Minz
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
42
|
Xia K, Qiu T, Jian Y, Liu H, Chen H, Liu X, Chen Z, Wang L. Degradation of histone deacetylase 6 alleviates ROS-mediated apoptosis in renal ischemia-reperfusion injury. Biomed Pharmacother 2023; 165:115128. [PMID: 37429230 DOI: 10.1016/j.biopha.2023.115128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/26/2023] [Accepted: 07/02/2023] [Indexed: 07/12/2023] Open
Abstract
Renal ischemia reperfusion injury (RIRI) is an inevitable complication during renal surgery. Histone deacetylase 6 (HDAC6), a key member of the histone deacetylase family, is associated with multiple pathologies, including renal diseases. However, whether HDAC6 could become a potential therapeutic target for clinical application of RIRI remained to be proven. Here, we found that HDAC6 expression was abnormally enhanced by the transcription factor OSR2 in RIRI. Moreover, we were the first to validate that a selective HDAC6 degrader, proteolysis-targeting chimeras (PROTAC) NP8, could significantly improve RIRI. Further in vivo and in vitro mechanism studies have found that the reduction of HDAC6 alleviated RIRI by inhibiting ROS mediated apoptosis. Remarkably, a renal protective protein, Klotho, has been proven to be a target of HDAC6, and the degradation of HDAC6 restored KL expression, thereby ameliorating ROS mediated apoptosis. Overall, our results illustrated that the degradation of HDAC6 restrained ROS mediated apoptosis by restoring Klotho expression during RIRI. PROTAC-NP8 might be a potential therapeutic strategy for clinical prevention of RIRI.
Collapse
Affiliation(s)
- Kang Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Wuhan University Institute of Urological Disease, Wuhan, Hubei, China
| | - Tao Qiu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yonghong Jian
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hao Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Wuhan University Institute of Urological Disease, Wuhan, Hubei, China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Wuhan University Institute of Urological Disease, Wuhan, Hubei, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Wuhan University Institute of Urological Disease, Wuhan, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Wuhan University Institute of Urological Disease, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Qu J, Jin J, Zhang M, Ng LG. Neutrophil diversity and plasticity: Implications for organ transplantation. Cell Mol Immunol 2023; 20:993-1001. [PMID: 37386174 PMCID: PMC10468536 DOI: 10.1038/s41423-023-01058-1] [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: 04/02/2023] [Accepted: 06/11/2023] [Indexed: 07/01/2023] Open
Abstract
Neutrophils, as the first defenders against external microbes and stimuli, are highly active and finely regulated innate immune cells. Emerging evidence has challenged the conventional dogma that neutrophils are a homogeneous population with a short lifespan that promotes tissue damage. Recent findings on neutrophil diversity and plasticity in homeostatic and disease states have centered on neutrophils in the circulation. In contrast, a comprehensive understanding of tissue-specialized neutrophils in health and disease is still lacking. This article will first discuss how multiomics advances have contributed to our understanding of neutrophil heterogeneity and diversification in resting and pathological settings. This discussion will be followed by a focus on the heterogeneity and role of neutrophils in solid organ transplantation and how neutrophils may contribute to transplant-related complications. The goal of this article is to provide an overview of the research on the involvement of neutrophils in transplantation, with the aim that this may draw attention to an underappreciated area of neutrophil research.
Collapse
Affiliation(s)
- Junwen Qu
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jingsi Jin
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ming Zhang
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
44
|
Xu Z, Zhu Y, Xie M, Liu K, Cai L, Wang H, Li D, Chen H, Gao L. Mackinawite nanozymes as reactive oxygen species scavengers for acute kidney injury alleviation. J Nanobiotechnology 2023; 21:281. [PMID: 37598162 PMCID: PMC10439570 DOI: 10.1186/s12951-023-02034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Iron sulfide nanomaterials have been successfully employed as therapeutic agents for bacterial infection therapy and catalytic-ferroptosis synergistic tumor therapy due to their unique structures, physiochemical properties, and biocompatibility. However, biomedical research and understanding of the biological functions of iron sulfides are insufficient, and how iron sulfide nanomaterials affect reactive oxygen species (ROS) in diseases remains unknown. Acute kidney injury (AKI) is associated with high levels of ROS, and therefore nanomedicine-mediated antioxidant therapy has emerged as a novel strategy for its alleviation. RESULTS Here, mackinawite nanozymes were synthesized from glutathione (GSH) and iron ions (Fe3+) (denoted as GFeSNs) using a hydrothermal method, and then evaluated as ROS scavengers for ROS-related AKI treatment. GFeSNs showed broad-spectrum ROS scavenging ability through synergistic interactions of multiple enzymes-like and hydrogen polysulfide-releasing properties. Furthermore, both in vitro and in vivo experiments demonstrated that GFeSNs exhibited outstanding cytoprotective effects against ROS-induced damage at extremely low doses and significantly improved treatment outcomes in AKI. CONCLUSIONS Given the synergetic antioxidant properties and high biocompatibility, GFeSNs exhibit great potential for the treatment of AKI and other ROS-associated diseases.
Collapse
Affiliation(s)
- Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Yufei Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Mengke Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Kankan Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Huihui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Hao Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
45
|
Wu K, Lu X, Li Y, Wang Y, Liu M, Li H, Li H, Liu Q, Shao D, Chen W, Zhou Y, Tu Z, Mao H. Polyglycerol-Amine Covered Nanosheets Target Cell-Free DNA to Attenuate Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300604. [PMID: 37276385 PMCID: PMC10427348 DOI: 10.1002/advs.202300604] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/08/2023] [Indexed: 06/07/2023]
Abstract
Increased levels of circulating cell-free DNA (cfDNA) are associated with poor clinical outcomes in patients with acute kidney injury (AKI). Scavenging cfDNA by nanomaterials is regarded as a promising remedy for cfDNA-associated diseases, but a nanomaterial-based cfDNA scavenging strategy has not yet been reported for AKI treatment. Herein, polyglycerol-amine (PGA)-covered MoS2 nanosheets with suitable size are synthesized to bind negatively charged cfDNA in vitro, in vivo and ex vivo models. The nanosheets exhibit higher cfDNA binding capacity than polymer PGA and PGA-based nanospheres owing to the flexibility and crimpability of their 2D backbone. Moreover, with low cytotoxicity and mild protein adsorption, the nanosheets effectively reduced serum cfDNA levels and predominantly accumulated in the kidneys to inhibit the formation of neutrophil extracellular traps and renal inflammation, thereby alleviating both lipopolysaccharide and ischemia-reperfusion induced AKI in mice. Further, they decreased the serum cfDNA levels in samples from AKI patients. Thus, PGA-covered MoS2 nanosheets can serve as a potent cfDNA scavenger for treating AKI and other cfDNA-associated diseases. In addition, this work demonstrates the pivotal feature of a 2D sheet-like structure in the development of the cfDNA scavenger, which can provide a new insight into the future design of nanoplatforms for modulating inflammation.
Collapse
Affiliation(s)
- Kefei Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Xiaohui Lu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yi Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yating Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Ming Liu
- Department of Otolaryngologythe Sixth Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Hongyu Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Huiyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Qinghua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Dan Shao
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong511442China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Zhaoxu Tu
- Department of Otolaryngologythe Sixth Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| |
Collapse
|
46
|
Wang J, Liu X, Gu Y, Gao Y, Jankowski V, Was N, Leitz A, Reiss LK, Shi Y, Cai J, Fang Y, Song N, Zhao S, Floege J, Ostendorf T, Ding X, Raffetseder U. DNA binding protein YB-1 is a part of the neutrophil extracellular trap mediation of kidney damage and cross-organ effects. Kidney Int 2023; 104:124-138. [PMID: 36963487 DOI: 10.1016/j.kint.2023.02.032] [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: 11/09/2022] [Revised: 02/10/2023] [Accepted: 02/27/2023] [Indexed: 03/26/2023]
Abstract
Open-heart surgery is associated with high morbidity, with acute kidney injury (AKI) being one of the most commonly observed postoperative complications. Following open-heart surgery, in an observational study we found significantly higher numbers of blood neutrophils in a group of 13 patients with AKI compared to 25 patients without AKI (AKI: 12.9±5.4 ×109 cells/L; non-AKI: 10.1±2. 9 ×109 cells/L). Elevated serum levels of neutrophil extracellular trap (NETs) components, such as dsDNA, histone 3, and DNA binding protein Y-box protein (YB)-1, were found within the first 24 hours in patients who later developed AKI. We could demonstrate that NET formation and hypoxia triggered the release of YB-1, which was subsequently shown to act as a mediator of kidney tubular damage. Experimentally, in two models of AKI mimicking kidney hypoperfusion during cardiac surgery (bilateral ischemia/reperfusion (I/R) and systemic lipopolysaccharide (LPS) administration), a neutralizing YB-1 antibody was administered to mice. In both models, prophylactic YB-1 antibody administration significantly reduced the tubular damage (damage score range 1-4, the LPS model: non-specific IgG control, 0.92±0.23; anti-YB-1 0.65±0.18; and in the I/R model: non-specific IgG control 2.42±0.23; anti-YB-1 1.86±0.44). Even in a therapeutic, delayed treatment model, antagonism of YB-1 ameliorated AKI (damage score, non-specific IgG control 3.03±0.31; anti-YB-1 2.58±0.18). Thus, blocking extracellular YB-1 reduced the effects induced by hypoxia and NET formation in the kidney and significantly limited AKI, suggesting that YB-1 is part of the NET formation process and an integral mediator of cross-organ effects.
Collapse
Affiliation(s)
- Jialin Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiyang Liu
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany
| | - Yulu Gu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yingying Gao
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research, RWTH, Aachen University, Aachen, Germany
| | - Nina Was
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, Wuerzburg, Germany
| | - Anna Leitz
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany
| | - Lucy K Reiss
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH, Aachen University, Germany
| | - Yiqin Shi
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jieru Cai
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jürgen Floege
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany
| | - Tammo Ostendorf
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Ute Raffetseder
- Department of Nephrology and Clinical Immunology, University Hospital, Rhine-Westphalia Technical University (RWTH)-Aachen, Aachen, Germany.
| |
Collapse
|
47
|
Wei JY, Hu MY, Chen XQ, Wei JS, Chen J, Qin XK, Lei FY, Zou JS, Zhu SQ, Qin YH. Hypobaric Hypoxia Aggravates Renal Injury by Inducing the Formation of Neutrophil Extracellular Traps through the NF-κB Signaling Pathway. Curr Med Sci 2023:10.1007/s11596-023-2744-3. [PMID: 37264195 DOI: 10.1007/s11596-023-2744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/24/2022] [Indexed: 06/03/2023]
Abstract
OBJECTIVE The hypersensitivity of the kidney makes it susceptible to hypoxia injury. The involvement of neutrophil extracellular traps (NETs) in renal injury resulting from hypobaric hypoxia (HH) has not been reported. In this study, we aimed to investigate the expression of NETs in renal injury induced by HH and the possible underlying mechanism. METHODS A total of 24 SD male rats were divided into three groups (n=8 each): normal control group, hypoxia group and hypoxia+pyrrolidine dithiocarbamate (PDTC) group. Rats in hypoxia group and hypoxia+PDTC group were placed in animal chambers with HH which was caused by simulating the altitude at 7000 meters (oxygen partial pressure about 6.9 kPa) for 7 days. PDTC was administered at a dose of 100 mg/kg intraperitoneally once daily for 7 days. Pathological changes of the rat renal tissues were observed under a light microscope; the levels of serum creatinine (SCr), blood urea nitrogen (BUN), cell-free DNA (cf-DNA) and reactive oxygen species (ROS) were measured; the expression levels of myeloperoxidase (MPO), citrullinated histone H3 (cit-H3), B-cell lymphoma 2 (Bcl-2), Bax, nuclear factor kappa B (NF-κB) p65 and phospho-NF-κB p65 (p-NF-κB p65) in rat renal tissues were detected by qRT-qPCR and Western blotting; the localization of NF-κB p65 expression in rat renal tissues was observed by immunofluorescence staining and the expression changes of NETs in rat renal tissues were detected by multiplex fluorescence immunohistochemical staining. RESULTS After hypoxia, the expression of NF-κB protein in renal tissues was significantly increased, the levels of SCr, BUN, cf-DNA and ROS in serum were significantly increased, the formation of NETs in renal tissues was significantly increased, and a large number of tubular dilatation and lymphocyte infiltration were observed in renal tissues. When PDTC was used to inhibit NF-κB activation, NETs formation in renal tissue was significantly decreased, the expression level of Bcl-2 in renal tissues was significantly increased, the expression level of Bax was significantly decreased, and renal injury was significantly alleviated. CONCLUSION HH induces the formation of NETs through the NF-κB signaling pathway, and it promotes apoptosis and aggravates renal injury by decreasing Bcl-2 and increasing Bax expression.
Collapse
Affiliation(s)
- Jun-Yu Wei
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Miao-Yue Hu
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xiu-Qi Chen
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Jin-Shuang Wei
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Jie Chen
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xuan-Kai Qin
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Feng-Ying Lei
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Jia-Sen Zou
- Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Shi-Qun Zhu
- Shenzhen Children's Hospital, Shenzhen, 518034, China
| | - Yuan-Han Qin
- Department of Pediatrics, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
48
|
Sharland AF, Hill AE, Son ET, Scull KE, Mifsud NA, Purcell AW. Are Induced/altered Self-peptide Antigens Responsible for De Novo Autoreactivity in Transplantation? Transplantation 2023; 107:1232-1236. [PMID: 36706066 PMCID: PMC10205114 DOI: 10.1097/tp.0000000000004499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/10/2022] [Accepted: 11/02/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Alexandra F. Sharland
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra E. Hill
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Eric T. Son
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Katherine E. Scull
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Nicole A. Mifsud
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Anthony W. Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
49
|
Alaygut D, Ozturk I, Ulu S, Gungor O. NETosis and kidney disease: what do we know? Int Urol Nephrol 2023:10.1007/s11255-023-03527-y. [PMID: 36840801 DOI: 10.1007/s11255-023-03527-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Neutrophils are the most abundant leukocytes in the blood. They are rapidly mobilized from the circulation to sites of inflammation and/or infection. In affected tissues, neutrophils exhibit some dramatic antimicrobial functions, including degranulation, reactive oxygen species (ROS) production, phagocytosis, and formation of neutrophil extracellular traps (NETs). Like other cells of the immune system, after fulfilling their biological duties, they enter the path of death. Depending on the conditions, they may undergo different types of cell death (apoptosis, necrosis, necroptosis, autophagy, NETosis, and pyroptosis) that require the participation of multiple signaling pathways. NETosis is a unique neutrophil cell death mechanism that gives rise to different inflammatory and autoimmune pathological conditions. Recent studies have shown that NETosis also plays a role in the formation and/or progression of kidney diseases. This review discusses the underlying mechanism of NETosis and its relationship with some major kidney diseases in light of the current knowledge.
Collapse
Affiliation(s)
- Demet Alaygut
- Department of Pediatric Nephrology, University of Health Sciences, Izmir Faculty of Medicine, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ilyas Ozturk
- Department of Internal Medicine, Division of Nephrology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey.
| | - Sena Ulu
- Department of Internal Medicine, Division of Nephrology, Bahcesehir University Faculty of Medicine, Istanbul, Turkey
| | - Ozkan Gungor
- Department of Internal Medicine, Division of Nephrology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| |
Collapse
|
50
|
Gajendran C, Fukui S, Sadhu NM, Zainuddin M, Rajagopal S, Gosu R, Gutch S, Fukui S, Sheehy CE, Chu L, Vishwakarma S, Jeyaraj DA, Hallur G, Wagner DD, Sivanandhan D. Alleviation of arthritis through prevention of neutrophil extracellular traps by an orally available inhibitor of protein arginine deiminase 4. Sci Rep 2023; 13:3189. [PMID: 36823444 PMCID: PMC9950073 DOI: 10.1038/s41598-023-30246-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Protein arginine deiminases (PAD) 4 is an enzyme that catalyzes citrullination of protein and its role in autoimmune diseases has been established through clinical genetics and gene knock out studies in mice. Further, studies with PAD4 - deficient mice have shown that PAD4 deficiency does not lead to increased infection or immune suppression, which makes PAD4 an attractive therapeutic target for auto-immune and inflammatory diseases. PAD4 has critical enzymatic role of promoting chromatin decondensation and neutrophil extracellular traps (NETs) formation that is associated with a number of immune-mediated pathological conditions. Here, we present a non-covalent PAD4 inhibitor JBI-589 with high PAD4 isoform selectivity and delineated its binding mode at 2.88 Å resolution by X-ray crystallography. We confirmed its effectiveness in inhibiting NET formation in vitro. Additionally, by using two mouse arthritis models for human rheumatoid arthritis (RA), the well-known disease associated with PAD4 clinically, we established its efficacy in vivo. These results suggest that JBI-589 would be beneficial for both PAD4 and NET-associated pathological conditions.
Collapse
Affiliation(s)
| | - Shoichi Fukui
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | | | | | | | | | - Sarah Gutch
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Saeko Fukui
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Casey E Sheehy
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Long Chu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | | | | | | | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02125, USA
| | | |
Collapse
|