1
|
Xin L, Kanghao N, Jiacheng L, Xiaodong Y, Juhan Y, Xinyang Z, Xiangdong L. Sodium aescinate protects renal ischemia-reperfusion and pyroptosis through AKT/NLRP3 signaling pathway. Ren Fail 2025; 47:2488140. [PMID: 40260531 DOI: 10.1080/0886022x.2025.2488140] [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: 07/07/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 04/23/2025] Open
Abstract
Renal ischemia-reperfusion injury (RIRI) is a common cause of acute renal injury. Studies have shown that sodium aescinate (SA) may serve as a potential therapeutic agent, although its exact mechanism remains unclear. This study first evaluated the efficacy of SA using a mouse renal ischemia-reperfusion model. Subsequently, its mechanism was elucidated through systematic bioinformatics, and finally validated through in vitro and in vivo experiments. The results demonstrated that SA has a protective effect on renal function in mice with RIRI. Bioinformatic analysis indicated that the pyroptosis pathway is significantly activated during renal ischemia-reperfusion injury, and immunohistochemistry showed that the level of renal pyroptosis is upregulated during ischemia-reperfusion injury. Administration of SA was able to reduce the expression of pyroptosis-related proteins (GSDMD, NLRP3, IL-1β) in RIRI. In vitro and in vivo experiments further confirmed that SA exerts an anti-pyroptotic effect by inhibiting the AKT/NLRP3 signaling pathway. Ultimately, SA mitigates kidney injury in IRI mice by suppressing renal failure through inhibition of the AKT/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Liu Xin
- The First Affiliated Hospital of Hebei North University, Hebei Province, China
| | - Ning Kanghao
- Graduate School of Hebei North University, Hebei Province, China
| | - Li Jiacheng
- The First Affiliated Hospital of Hebei North University, Hebei Province, China
| | - Yan Xiaodong
- The First Affiliated Hospital of Hebei North University, Hebei Province, China
| | - Yan Juhan
- Graduate School of Hebei North University, Hebei Province, China
| | - Zhao Xinyang
- The First Affiliated Hospital of Hebei North University, Hebei Province, China
| | - Li Xiangdong
- The First Affiliated Hospital of Hebei North University, Hebei Province, China
| |
Collapse
|
2
|
Wang Y, Li Y, Gu Y, Ma W, Guan Y, Guo M, Shao Q, Ji X, Liu J. Decreased levels of phosphorylated synuclein in plasma are correlated with poststroke cognitive impairment. Neural Regen Res 2025; 20:2598-2610. [PMID: 38845216 PMCID: PMC11801306 DOI: 10.4103/nrr.nrr-d-23-01348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/30/2023] [Accepted: 02/29/2024] [Indexed: 11/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00022/figure1/v/2024-11-05T132919Z/r/image-tiff Poststroke cognitive impairment is a major secondary effect of ischemic stroke in many patients; however, few options are available for the early diagnosis and treatment of this condition. The aims of this study were to (1) determine the specific relationship between hypoxic and α-synuclein during the occur of poststroke cognitive impairment and (2) assess whether the serum phosphorylated α-synuclein level can be used as a biomarker for poststroke cognitive impairment. We found that the phosphorylated α-synuclein level was significantly increased and showed pathological aggregation around the cerebral infarct area in a mouse model of ischemic stroke. In addition, neuronal α-synuclein phosphorylation and aggregation were observed in the brain tissue of mice subjected to chronic hypoxia, suggesting that hypoxia is the underlying cause of α-synuclein-mediated pathology in the brains of mice with ischemic stroke. Serum phosphorylated α-synuclein levels in patients with ischemic stroke were significantly lower than those in healthy subjects, and were positively correlated with cognition levels in patients with ischemic stroke. Furthermore, a decrease in serum high-density lipoprotein levels in stroke patients was significantly correlated with a decrease in phosphorylated α-synuclein levels. Although ischemic stroke mice did not show significant cognitive impairment or disrupted lipid metabolism 14 days after injury, some of them exhibited decreased cognitive function and reduced phosphorylated α-synuclein levels. Taken together, our results suggest that serum phosphorylated α-synuclein is a potential biomarker for poststroke cognitive impairment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Clinical Laboratory, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Capital Medical University, Beijing, China
| | - Yuning Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuying Guan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Qianqian Shao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Chen Z, Lin Z, Wan H, Li C, Jin W, Wan H, He Y. Rapid and accurate metabolite identification of traditional Chinese medicine based on UPLC-Q-TOF-MS coupled with UNIFI analysis platform and quantitative structure-retention relationship: Danshen-Honghua herbal pair as an example. J Pharm Biomed Anal 2025; 257:116696. [PMID: 39879817 DOI: 10.1016/j.jpba.2025.116696] [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: 09/27/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
In recent years, metabolite identification of chemical constituents of traditional Chinese medicine (TCM) has been extensively studied. However, due to the intricacy of metabolic processes and the low concentration of metabolites, identifying metabolites of TCM in vivo is still a tough work. Meanwhile, credibility of metabolite identification through mass spectrum technology has been called into question by reason of the lack of metabolite standards. In this study, ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS) was used to detect biological samples including plasma, feces, urine, liver, kidney, brain of normal and middle cerebral artery occlusion (MCAO) rats orally administrated water extract of Danshen-Honghua herbal pair (DHHP). An analysis strategy which combined MS data analysis platform UNIFI with quantitative structure-retention relationship (QSRR) model was established. First, metabolites of DHHP were identified rapidly by utilizing UNIFI analysis platform to analyze acquired MS data. Then, quantitative structure-retention relationships model was built through BP neural network optimized by the ant colony algorithm. Finally, predicted retention times of identified metabolites were produced by QSRR model. Metabolites identified by UNIFI whose difference between predicted and experimental retention time was beyond 1 min were considered false positive and excluded to improve the credibility of identification. According to the established analysis strategy, 26 prototypes and 16 metabolites were identified. Established MS data analysis strategy which combined UNIFI analysis platform with QSRR model was proven to be a creditable method to identify the in vivo metabolites of TCM rapidly and accurately.
Collapse
Affiliation(s)
- Zhaoyu Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Ziyi Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Haofang Wan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Chang Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
4
|
Ge W, Cao L, Liu C, Wang H, Lu M, Chen Y, Wang Y. Identifying Pyroptosis-Hub Genes and Inflammation Cell Type-Related Genes in Ischemic Stroke. Mol Neurobiol 2025; 62:6228-6255. [PMID: 39798044 PMCID: PMC11953102 DOI: 10.1007/s12035-024-04647-x] [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: 08/18/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025]
Abstract
Stroke is the second-leading global cause of death. The damage attributed to the immune storm triggered by ischemia-reperfusion injury (IRI) post-stroke is substantial. However, data on the transcriptomic dynamics of pyroptosis in IRI are limited. This study aimed to analyze the expression of key pyroptosis genes in stroke and their correlation with immune infiltration. Pyroptosis-related genes were identified from the obtained middle cerebral artery occlusion (MCAO) datasets. Differential expression and functional analyses of pyroptosis-related genes were performed, and differences in functional enrichment between high-risk and low-risk groups were determined. An MCAO diagnostic model was constructed and validated using selected pyroptosis-related genes with differential expression. High- and low-risk MCAO groups were constructed for expression and immune cell correlation analysis with pyroptosis-related hub genes. A regulatory network between pyroptosis-related hub genes and miRNA was also constructed, and protein domains were predicted. The expression of key pyroptosis genes was validated using an MCAO rat model. Twenty-five pyroptosis genes showed differential expression, including four hub genes, namely WISP2, MELK, SDF2L1, and AURKB. Characteristic genes were verified using real-time quantitative PCR analyses. The high- and low-risk groups showed significant expression differences for WISP2, MELK, and SDF2L1. In immune infiltration analysis, 12 immune cells showed differences in expression in MCAO samples. Further analysis demonstrated significant positive correlations between the pyroptosis-related hub gene SDF2L1 and immune cell-activated dendritic cells in the high-risk group and immune cell natural killer cells in the low-risk group. This study identified four pyroptosis-related hub genes, with elevated WISP2, MELK, and SDF2L1 expression closely associated with the high-risk group. The analysis of inflammatory cell types in immune infiltration can predict ischemic stroke risk levels and help to facilitate treatment.
Collapse
Affiliation(s)
- Wei Ge
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China
| | - Liangbin Cao
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China
| | - Can Liu
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China
| | - Hao Wang
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China
| | - Meijing Lu
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China
| | - Yongquan Chen
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China.
| | - Ye Wang
- Department of Anesthesiology, Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241004, China.
| |
Collapse
|
5
|
Patrick R, Pando BD, Yang C, Aponte A, Wang F, Ewing T, Ma Y, Yuan SY, Wu MH. Focal adhesion kinase mediates microvascular leakage and endothelial barrier dysfunction in ischemia-reperfusion injury. Microvasc Res 2025; 159:104791. [PMID: 39884384 DOI: 10.1016/j.mvr.2025.104791] [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: 08/02/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Intestinal ischemia-reperfusion (I/R) injury occurs under various surgical or disease conditions, where tissue hypoxia followed by reoxygenation results in the production of oxygen radicals and inflammatory mediators. These substances can target the endothelial barrier, leading to microvascular leakage. In this study, we induced intestinal I/R injury in mice by occluding the superior mesenteric artery, followed by removing the clamp to resume blood circulation. We assessed microvascular permeability to plasma proteins in vivo using intravital microscopy, measuring the time-dependent tracer distribution in the intravascular versus extravascular space in the mouse mesentery. Additionally, we examined endothelial cell-cell adhesive barrier resistance and junction morphology in cultured endothelial cell monolayers. At the molecular level, FAK inhibition similarly inhibited endothelial junction opening and barrier dysfunction in response to hydrogen peroxide-induced oxidative stress. To further investigate FAK's role with tissue/cell specificity, we developed an endothelial-specific inducible FAK knockout mouse model by crossbreeding FAK-floxed (FAKfl/fl) mice with Tie-2-CreERT2 transgenic mice. Compared to their wild-type controls, endothelial-specific FAK-deficient mice showed a blunted microvascular hyperpermeability response following I/R injury in the gut. Overall, our study demonstrates that FAK plays a significant signaling role in mediating endothelial barrier dysfunction and microvascular leakage during ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Rebecca Patrick
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America
| | - Briana D Pando
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America.
| | - Clement Yang
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America
| | - Alexandra Aponte
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America.
| | - Fang Wang
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America
| | - Tom Ewing
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America.
| | - Yonggang Ma
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America
| | - Sarah Y Yuan
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America.
| | - Mack H Wu
- University of South Florida, Morsani College of Medicine, James A Haley Veterans' Hospital, United States of America.
| |
Collapse
|
6
|
Eggenhofer E, Proneth B. Ferroptosis Inhibition: A Key Opportunity for the Treatment of Ischemia/Reperfusion Injury in Liver Transplantation. Transplantation 2025; 109:e228-e236. [PMID: 39294870 DOI: 10.1097/tp.0000000000005199] [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] [Indexed: 09/21/2024]
Abstract
The outcome after liver transplantation has improved in recent years, which can be attributed to superior storage and transportation conditions of the organs, as well as better peri- and postoperative management and advancements in surgical techniques. Nevertheless, there is an increasing discrepancy between the need for organs and their availability. Consequently, the mortality rate on the waiting list is high and continues to rise. One way of counteracting this trend is to increase the use of "expanded criteria donors." This means that more and more donors will be included, especially those who are older and having additional comorbidities (eg, steatosis). A major complication of any transplantation is the occurrence of ischemia/reperfusion injury (IRI), which often leads to liver dysfunction and failure. However, there have been various promising approaches to minimize IRI in recent years, but an effective and clinically applicable method to achieve a better outcome for patients after liver transplantation is still missing. Thereby, the so-called marginal organs are predominantly affected by IRI; thus, it is crucial to develop suitable and effective treatment options for patients. Recently, regulated cell death mechanisms, particularly ferroptosis, have been implicated to play a major role in IRI, including the liver. Therefore, inhibiting this kind of cell death modality presents a promising therapeutic approach for the management of this yet untreatable condition. Thus, this review provides an overview of the role of ferroptosis in liver IRI and transplantation and discusses possible therapeutic solutions based on ferroptosis inhibition to restrain IRI in marginal organs (especially steatosis and donation after circulatory death organs).
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Munich, Neuherberg, Germany
| |
Collapse
|
7
|
Hurd RE, Gu M, Okamura K, Shibata M, Ono Y, Haidar M, Riemer RK, Hanley FL, Spielman DM. Detection of elevated succinate in brain during circulatory arrest in a piglet model: A 3T 1H MR spectroscopy study. Magn Reson Med 2025; 93:1874-1885. [PMID: 39737693 DOI: 10.1002/mrm.30414] [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: 06/14/2024] [Revised: 11/17/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025]
Abstract
PURPOSE To measure and validate elevated succinate in brain during circulatory arrest in a piglet model of cardiopulmonary bypass. METHODS Using data from an archive of 3T 1H MR spectra acquired in previous in-magnet studies, dynamic plots of succinate, spectral simulations and difference spectra were generated for analysis and validation. RESULTS Elevation of succinate during circulatory arrest was observed and validated. Fitting bias was evaluated as a function of the line-widths and signal-to-noise ratios of the archived data. Succinate increases were independent of bypass temperature. Succinate elevation was also not observed with antegrade cerebral perfusion. CONCLUSION Although spectrally overlapped and at sub-millimolar levels, elevated brain succinate can be reliably measured by dynamic MR spectroscopy at 3T. Noise dependent bias of the stronger overlapping signals did not impact the succinate measurement. Elevated succinate during circulatory arrest and its recovery after reperfusion was observed. This finding is consistent with earlier reports that correlate elevated succinate with ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Ralph E Hurd
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | - Meng Gu
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | - Kenichi Okamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masafumi Shibata
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yoshikazu Ono
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Moussa Haidar
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - R Kirk Riemer
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Frank L Hanley
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel M Spielman
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Bongoni AK, Kiss B, McRae JL, Salvaris EJ, Fisicaro N, Muntz F, Németh BZ, Nagy ZA, Kocsis A, Gál P, Cowan PJ, Pál G. Targeting the complement lectin pathway with a highly specific MASP-2 inhibitor protects against renal ischemia-reperfusion injury. Proc Natl Acad Sci U S A 2025; 122:e2424754122. [PMID: 40228118 DOI: 10.1073/pnas.2424754122] [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: 11/26/2024] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a common complication in several clinical scenarios including kidney transplantation. Mannan-binding lectin-associated serine proteinase (MASP)-2 is essential for activation of the complement lectin pathway, which has been implicated in the pathogenesis of renal IRI and therefore represents a potential therapeutic target. We developed a new, affinity-enhanced MASP-2 inhibitor, EVO24, by directed evolution of the D2 domain of human tissue factor pathway inhibitor. EVO24 was fused with a human IgG1-Fc to create the homodimer EVO24L, which potently and selectively inhibited the lectin pathway in human and mouse serum in vitro. EVO24L was tested in a mouse model of unilateral warm renal IRI. EVO24L administered before and after ischemia significantly protected against IRI, with improved renal function as well as reduced tubular injury and inflammatory cell infiltration at 24 h compared to vehicle-treated mice. Immunofluorescence analyses showed reduced deposition of complement components (C3d, C4d, and C9) and reduced expression of VCAM-1, indicating a decrease in complement activation and endothelial cell activation. Additionally, EVO24L treatment lowered plasma levels of complement C5a, hyaluronan (a marker of endothelial glycocalyx shedding), and the proinflammatory cytokines IL-6 and TNF-α. Our findings indicate that EVO24L inhibits acute inflammatory responses in renal IRI by blocking the lectin pathway, confirming the important role of this pathway in acute ischemic kidney injury and warranting further investigation of EVO24L in clinical settings.
Collapse
Affiliation(s)
- Anjan K Bongoni
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Bence Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
| | - Jennifer L McRae
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Evelyn J Salvaris
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Nella Fisicaro
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Fenella Muntz
- Bioresources Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Bálint Zoltán Németh
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
| | - Zoltán Attila Nagy
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
| | - Andrea Kocsis
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
- Institute of Molecular Life Sciences, Hungarian Research Network, Research Centre for Natural Sciences, Budapest H-1117, Hungary
| | - Péter Gál
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
- Institute of Molecular Life Sciences, Hungarian Research Network, Research Centre for Natural Sciences, Budapest H-1117, Hungary
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3052, Australia
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
- EvolVeritas Biotechnology Ltd., Budapest H-1117, Hungary
| |
Collapse
|
9
|
Xu G, Sun X, An J, Sun F, Zhang C, Williams JP. Ozone protects from myocardial ischemia-reperfusion injury via inhibition of the NLRP3 inflammasome. Eur J Pharmacol 2025; 997:177631. [PMID: 40246138 DOI: 10.1016/j.ejphar.2025.177631] [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: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Ischemic heart disease (IHD) is a leading cause of morbidity and mortality worldwide. Myocardial ischemia/reperfusion injury (MIRI) is the primary cause of myocardial injury triggered by post-myocardial infarction reperfusion therapy. Its pathogenesis involves Ca2+ overload, the production of large amounts of oxygen-free radicals, inflammation, and cell necrosis. Growing evidence suggests that the NLRP3 inflammasome significantly contributes to the sterile inflammatory response and pyroptosis in MIRI, linking damage sensing to the initiation and amplification of the inflammatory response. Reportedly, ozone exerts anti-inflammatory and anti-infection effects by activating the antioxidant system. Additional evidence suggests that ozone inhibits NLRP3 inflammasome expression to relieve ischemic injury. In this study, we aimed to explore whether pretreating the myocardium with ozone protects it from MIRI by inhibiting the NLRP3 inflammasome. Rats were subjected to rectal infusion of ozone for 5 consecutive days, followed by ligation of the left anterior descending coronary artery for 30 min and reperfusion for 120 min to induce MIRI. Experimental results were obtained using echocardiography, triphenyltetrazolium chloride and hematoxylin and eosin staining, western blotting, and enzyme-linked immunosorbent assay. The results showed that ozone significantly improved the diastolic function of the heart, reduced the area of myocardial infarction, and decreased the expression levels of NLRP3, pro-caspase-1, ASC, and the secretion of caspase-1, interleukin (IL)-1β, and IL-18. In summary, these findings reveal that ozone pretreatment can alleviate the damage that occurs during MIRI by inhibiting the NLRP3 Inflammasome.
Collapse
Affiliation(s)
- Guohao Xu
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaotong Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Jianxiong An
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China; Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Brain Disease Institute & Department of Anesthesiology for the Integrated Traditional Chinese and Western Medicine Hospital, Southern Medical University, Guangzhou, China.
| | - Fan Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Chengming Zhang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China.
| | - John P Williams
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Qiu YH, Zhang YH, Wu ZC, Huang JY, Chen BC, Xiao J, Chen FF. 3,4-Dimethoxychalcone alleviates limb ischemia/reperfusion injury by TFEB-mediated autophagy enhancement and antioxidative response. FASEB J 2025; 39:e70496. [PMID: 40162605 DOI: 10.1096/fj.202402609rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Caloric restriction mimetics (CRMs) replicate the positive effects of caloric restriction (CR) and have demonstrated therapeutic benefits in neuroinflammatory and cardiovascular diseases. However, it remains uncertain whether CRMs enhance functional recovery following ischemia/reperfusion (I/R) injury, as well as the specific mechanisms involved in this process. This study examines the therapeutic potential of the CRM 3,4-dimethoxychalcone (3,4-DC) in limb I/R injury. Histology, tissue swelling analysis, and laser doppler imaging (LDI) were used to assess the viability of the limbs. Western blotting and immunofluorescence were utilized to examine apoptosis levels, oxidative stress (OS), autophagy, transcription factor EB (TFEB) activity, and mucolipin 1 (MCOLN1)-calcineurin signaling pathway. The administration of 3,4-DC notably alleviated hypoperfusion, tissue swelling, skeletal muscle fiber damage, and cellular injury in the limb caused by I/R. The pharmacological blockade of autophagy negated the antioxidant and antiapoptotic effects of 3,4-DC. Moreover, RNA interference-mediated TFEB silencing eliminated the 3,4-DC-induced restoration of autophagy, antioxidant response, and antiapoptotic effects. Additionally, our findings revealed that 3,4-DC modulates TFEB activity via the MCOLN1-calcineurin signaling pathway. 3,4-DC facilitates functional recovery by enhancing TFEB-driven autophagy, while simultaneously suppressing oxidative stress and apoptosis following I/R injury, suggesting its potential value in clinical applications.
Collapse
Affiliation(s)
- Yi-Hui Qiu
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yin-He Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Zi-Chang Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jing-Yong Huang
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bi-Cheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Fan-Feng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Akıllıoğlu K, Köse Korkmaz S, Dönmez Kutlu M. The effect of caffeine in a model of schizophrenia-like behavior induced by MK-801 in mice. Behav Brain Res 2025; 483:115468. [PMID: 39922384 DOI: 10.1016/j.bbr.2025.115468] [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: 12/16/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVE The blockade of NMDA receptors during early developmental stages is accepted as a model for schizophrenia-like behavior. This study aimed to investigate the effects of caffeine on adult behaviors in mice subjected to tests of schizophrenia-like behaviors induced by the NMDA receptor antagonist MK-801. MATERIALS AND METHODS MK-801 (0.25 mg/kg, twice daily, 0.1 ml/10 g body weight, intraperitoneally) was administered to Balb/c mice during PND 7-10 to establish a schizophrenia-like behavior model. In one group, caffeine (10 mg/kg, twice daily, 0.1 ml/10 g body weight, intraperitoneally) was given 30 min after MK-801 administration. In another group, MK-801 was administered 30 min after caffeine injection. At 8-10 weeks of age, behavioral tests were performed sequentially: open field test (OFT), elevated plus maze test (EPM), Morris water maze test (MWM), and social interaction test. RESULTS MK-801 administration significantly increased anxiety-like behaviors and decreased exploratory behavior in the OFT by reducing the time spent in the center, the frequency of center entries, and rearing frequency, while increasing the latency to the first center entry. In the EPM, MK-801 significantly decreased the time spent in the open arms, the frequency of open arm entries, and the head-dipping behavior of the open arm while increasing the time spent in the closed arms and the latency to the first open arm entry. In the MWM, MK-801 impaired learning and memory performance. MK-801 reduced social interaction. Caffeine reversed the anxiety, social interaction, learning, and memory impairments caused by MK-801. CONCLUSION MK-801 administration during the neonatal period induces schizophrenia-like behaviors in adulthood, whereas low-dose caffeine can mitigate these effects.
Collapse
Affiliation(s)
- Kübra Akıllıoğlu
- Çukurova University Faculty of Medicine. Department of Physiology, Department of Neurophysiology, Adana 01330, Turkey
| | - Seda Köse Korkmaz
- Çukurova University Faculty of Medicine. Department of Physiology, Department of Neurophysiology, Adana 01330, Turkey
| | - Meltem Dönmez Kutlu
- Çukurova University Faculty of Medicine. Department of Physiology, Department of Neurophysiology, Adana 01330, Turkey.
| |
Collapse
|
12
|
Wang D, Zhai Y, Wang Y, Fu X, Ji Y, Li R. Dual-color reversible fluorescent carbon dots designed for dynamic monitoring of cellular superoxide anion radicals. J Mater Chem B 2025. [PMID: 40205991 DOI: 10.1039/d5tb00099h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
The superoxide anion radical (O2˙-) represents the primary reactive oxygen species generated in biological systems. Real-time monitoring of its dynamic fluctuations provides valuable insights into disease progression and enables early diagnosis of hepatic ischemia-reperfusion injury (HIRI). In this work, we developed a novel dual-color fluorescent carbon dot (CD) probe through a one-step hydrothermal synthesis for reversible O2˙- detection. The CDs demonstrated excellent sensitivity, dynamically detecting O2˙- concentrations ranging from 0 to 60 μM with a detection limit of 0.56 μM. The probe exhibited remarkable reversibility, maintaining stable performance through at least three complete oxidation-reduction cycles following glutathione (GSH) treatment. In practical applications, the CDs achieved 95.2-104% recovery rates when detecting O2˙- in serum samples. Cellular imaging experiments confirmed the probe's effectiveness in normal hepatocytes (LO2), showing clear reversible responses to O2˙- fluctuations. Application in a HIRI cell model revealed significant elevation of O2˙- levels and provided new evidence for its role in HIRI-related signaling pathways. This study not only presents an effective dual-color fluorescent probe for dynamic O2˙- monitoring but also establishes a versatile synthetic strategy that could be adapted for imaging other biologically relevant molecules in living cells.
Collapse
Affiliation(s)
- Dan Wang
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yanke Zhai
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Nanjing 210009, China.
| | - Yun Wang
- Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Gansu, Lanzhou, 730000, China.
| | - Xu Fu
- Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Gansu, Lanzhou, 730000, China.
| | - Yibing Ji
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Nanjing 210009, China.
| | - Ruijun Li
- Department of Analytical Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Nanjing 210009, China.
| |
Collapse
|
13
|
Gyöngyösi M, Guthrie J, Hasimbegovic E, Han E, Riesenhuber M, Hamzaraj K, Bergler-Klein J, Traxler D, Emmert MY, Hackl M, Derdak S, Lukovic D. Critical analysis of descriptive microRNA data in the translational research on cardioprotection and cardiac repair: lost in the complexity of bioinformatics. Basic Res Cardiol 2025:10.1007/s00395-025-01104-1. [PMID: 40205177 DOI: 10.1007/s00395-025-01104-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
The unsuccessful translation of cardiac regeneration and cardioprotection from animal experiments to clinical applications in humans has raised the question of whether microRNA bioinformatics can narrow the gap between animal and human research outputs. We reviewed the literature for the period between 2000 and 2024 and found 178 microRNAs involved in cardioprotection and cardiac regeneration. On analyzing the orthologs and annotations, as well as downstream regulation, we observed species-specific differences in the diverse regulation of the microRNAs and related genes and transcriptomes, the influence of the experimental setting on the microRNA-guided biological responses, and database-specific bioinformatics results. We concluded that, in addition to reducing the number of in vivo experiments, following the 3R animal experiment rules, the bioinformatics approach allows the prediction of several currently unknown interactions between pathways, coding and non-coding genes, proteins, and downstream regulatory elements. However, a comprehensive analysis of the miRNA-mRNA-protein networks needs a profound bioinformatics and mathematical education and training to appropriately design an experimental study, select the right bioinformatics tool with programming language skills and understand and display the bioinformatics output of the results to translate the research data into clinical practice. In addition, using in-silico approaches, a risk of deviating from the in vivo processes exists, with adverse consequences on the translational research.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| | - Julia Guthrie
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Ena Hasimbegovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Emilie Han
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Martin Riesenhuber
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Kevin Hamzaraj
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jutta Bergler-Klein
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charite (DHZC), Berlin, Germany
| | | | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Wang Y, Li S. Continuous synthesis of PEGylated MIL-101(Cr) nanoparticles for neuroprotection. RSC Adv 2025; 15:12020-12027. [PMID: 40242494 PMCID: PMC12002162 DOI: 10.1039/d4ra09107h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
The application of metal organic frameworks (MOFs) in targeted drug delivery for ischemic stroke therapy has emerged as a hot issue recently. Although significant progress has been made in immobilizing neuroprotective agents on MOFs, environmentally friendly large-scale preparation of nano-drug-loaded MOFs with controlled size, morphology, purity and therapeutic effect remains challenging. PEGylation of MIL-101(Cr) nanoparticles with dual ligands that have the 2,2-dimethylthiazolidine (DMTD) structure was developed in this work to mitigate nervous system injury induced by ischemia/reperfusion (IR) during a stroke. A green ultrasound-assisted continuous-flow system was established for efficient production of the versatile MOF nanoparticles. Unified nanoparticles (diameter: ∼250-350 nm) were obtained with both high quality and high space-time yield (5664 kg m-3 d-1). The MOF exhibited protective activity in SH-SY5Y cells against oxygen and glucose deprivation and H2O2 insults, and prevented reactive oxygen species accumulation. The cellular uptake of the PEGylated MOFs by brain capillary endothelial cells was investigated, showing targeting capability in vitro, which proposes the biomaterial as a promising therapeutic candidate for reducing IR-induced nervous system injury.
Collapse
Affiliation(s)
- Yuheng Wang
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Shuirong Li
- College of Energy, Xiamen University Xiamen 361102 China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM) Xiamen 361005 China
| |
Collapse
|
15
|
Davari M, Khansari M, Hosseini S, Morovatshoar R, Azani A, Mirzohreh ST, Mahabadi MA, Ghasemi M, Meigoli MSS, Nematollahi SF, Pourranjbar S, Behfar Q, Baghdadi M, Hosseini AM. The Impact of Opioids on Epigenetic Modulation in Myocardial Ischemia and Reperfusion Injury: Focus on Non-coding RNAs. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10609-y. [PMID: 40198537 DOI: 10.1007/s12265-025-10609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
Myocardial ischemia-reperfusion injury (IRI) is a major issue in cardiovascular medicine, marked by tissue damage from the restoration of blood flow after ischemia. Opioids, known for their pain-relieving properties, have emerged as potential cardioprotective agents in IRI. Recent research suggests opioids influence epigenetic mechanisms-such as histone modifications and non-coding RNAs (ncRNAs)-which are essential for regulating gene expression and cellular responses during myocardial IRI. This review delves into how opioids like remifentanil affect histone modifications, DNA methylation, and ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Remifentanil postconditioning (RPC) reduces apoptosis in cardiomyocytes through histone deacetylation, specifically downregulating histone deacetylase 3 (HDAC3). Similarly, opioids impact miRNAs such as miR- 206 - 3p and miR- 320 - 3p, and lncRNAs like TINCR and UCA1, which influence apoptosis, inflammation, and oxidative stress. Understanding these interactions highlights the potential for opioid-based therapies in mitigating IRI-induced myocardial damage.
Collapse
Affiliation(s)
- Mohsen Davari
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Mahmoud Khansari
- General Surgery Department, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Sahar Hosseini
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Reza Morovatshoar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Alireza Azani
- Department of Medical Genetic, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Seyedeh Tarlan Mirzohreh
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Mohammadjavad Ashrafi Mahabadi
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Moein Ghasemi
- Faculty of Medicine, Tehran University of Medical Science, Tehran, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Mohammad Saeed Soleimani Meigoli
- School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Sima Foroughi Nematollahi
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Sina Pourranjbar
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran
| | - Qumars Behfar
- National Institute for Health Research, Tehran University of Medical Sciences, Tehran, Iran.
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran.
| | - Mandana Baghdadi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran.
| | - Ahmad Mir Hosseini
- Mashhad University of Medical Sciences, Mashhad, Iran.
- Cardiology Department, Mentoring Program in Medical Sciences, MSA Research Group, Tehran, Iran.
| |
Collapse
|
16
|
Tang L, Zhang W, Liao Y, Wang W, Deng X, Wang C, Shi W. Autophagy: a double-edged sword in ischemia-reperfusion injury. Cell Mol Biol Lett 2025; 30:42. [PMID: 40197222 PMCID: PMC11978130 DOI: 10.1186/s11658-025-00713-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Ischemia-reperfusion (I/R) injury describes the pathological process wherein tissue damage, initially caused by insufficient blood supply (ischemia), is exacerbated upon the restoration of blood flow (reperfusion). This phenomenon can lead to irreversible tissue damage and is commonly observed in contexts such as cardiac surgery and stroke, where blood supply is temporarily obstructed. During ischemic conditions, the anaerobic metabolism of tissues and organs results in compromised enzyme activity. Subsequent reperfusion exacerbates mitochondrial dysfunction, leading to increased oxidative stress and the accumulation of reactive oxygen species (ROS). This cascade ultimately triggers cell death through mechanisms such as autophagy and mitophagy. Autophagy constitutes a crucial catabolic mechanism within eukaryotic cells, facilitating the degradation and recycling of damaged, aged, or superfluous organelles and proteins via the lysosomal pathway. This process is essential for maintaining cellular homeostasis and adapting to diverse stress conditions. As a cellular self-degradation and clearance mechanism, autophagy exhibits a dualistic function: it can confer protection during the initial phases of cellular injury, yet potentially exacerbate damage in the later stages. This paper aims to elucidate the fundamental mechanisms of autophagy in I/R injury, highlighting its dual role in regulation and its effects on both organ-specific and systemic responses. By comprehending the dual mechanisms of autophagy and their implications for organ function, this study seeks to explore the potential for therapeutic interventions through the modulation of autophagy within clinical settings.
Collapse
Affiliation(s)
- Lingxuan Tang
- Basic Medical University, Naval Medical University, Shanghai, 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Weijie Wang
- Basic Medical University, Naval Medical University, Shanghai, 200433, China
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Changli Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Wenwen Shi
- School of Nursing, Navy Military Medical University, Shanghai, China.
| |
Collapse
|
17
|
Grignano MA, Pisani S, Gregorini M, Rainaudo G, Avanzini MA, Croce S, Valsecchi C, Ceccarelli G, Islami T, Margiotta E, Portalupi V, De Mauri A, Stea ED, Pattonieri EF, Iadarola P, Viglio S, Conti B, Rampino T. Engineered ATP-Loaded Extracellular Vesicles Derived from Mesenchymal Stromal Cells: A Novel Strategy to Counteract Cell ATP Depletion in an In Vitro Model. Int J Mol Sci 2025; 26:3424. [PMID: 40244293 PMCID: PMC11990007 DOI: 10.3390/ijms26073424] [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: 03/03/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
The use of adenosine triphosphate (ATP) has shown promising effects in alleviating ischemic damage across various tissues. However, the penetration of ATP into kidney tubular cells presents a challenge due to their unique anatomical and physiological properties. In this study, we introduce a novel bioinspired drug delivery system utilizing extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) and engineered to carry ATP. ATP-loaded liposomes (ATP-LPs) and ATP-loaded EVs (ATP-EVs) were prepared using microfluidic technology, followed by characterization of their morphology (DLS, NTA, SEM, TEM), ATP content, and release rate at 37 °C (pH 7.4). Additionally, the delivery efficacy of ATP-LPs and ATP-EVs was evaluated in vitro on renal cells (HK2 cells) under chemically induced ischemia. The results indicated successful ATP enrichment in EVs, with ATP-EVs showing no significant changes in morphology or size compared to naïve EVs. Notably, ATP-EVs demonstrated superior ATP retention compared to ATP-LPs, protecting the ATP from degradation in the extracellular environment. In an ATP-depleted HK2 cell model, only ATP-EVs effectively restored ATP levels, preserving cell viability and reducing apoptotic gene expression (BCL2-BAX). This study is the first to successfully demonstrate the direct delivery of ATP into renal tubular cells in vitro using EVs as carriers.
Collapse
Affiliation(s)
- Maria Antonietta Grignano
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, Viale Torquato Taramelli 12, 27100 Pavia, Italy;
| | - Marilena Gregorini
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Giorgia Rainaudo
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Maria Antonietta Avanzini
- Pediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.A.); (C.V.)
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Stefania Croce
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Chiara Valsecchi
- Pediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.A.); (C.V.)
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gabriele Ceccarelli
- Human Anatomy Unit, Department of Public Health, Experimental Medicine and Forensic, University of Pavia, 27100 Pavia, Italy;
- Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy
| | - Tefik Islami
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Elisabetta Margiotta
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Valentina Portalupi
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Andreana De Mauri
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Emma Diletta Stea
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Eleonora Francesca Pattonieri
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Simona Viglio
- Lung Transplantation Unit, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Viale Torquato Taramelli 12, 27100 Pavia, Italy;
| | - Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
18
|
Schneider S, Biggerstaff D, Barber TM. Dietary Guidelines Post Kidney Transplant: Is This the Missing Link in Recovery and Graft Survival? Transpl Int 2025; 38:14288. [PMID: 40248508 PMCID: PMC12004285 DOI: 10.3389/ti.2025.14288] [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: 01/02/2025] [Accepted: 03/11/2025] [Indexed: 04/19/2025]
Abstract
The physiology of a transplanted kidney is affected from the moment it is separated from the donor. The risk of complications arising from surgery are highly associated with ischemic-reperfusion injury (IRI) due to the effects of hypoxia and oxidative stress during the procurement, preservation and reperfusion procedures. Hypoxia promotes the formation of reactive oxygen species (ROS) and it seems apparent that finding ways of optimising the metabolic milieu for the transplanted kidney would improve recovery and graft survival. Studies have demonstrated the benefits of nutrition and antioxidant compounds in mitigating the disturbance of energy supply to cells post-transplant and at improving long-term graft survival. Particularly in patients who may be nutritionally deficient following long-term dialysis. Despite the high incidence of allograft failure, a search of the literature and grey literature reveals no medical nutriti on therapy guidelines on beneficial nutrient intake to aid transplant recovery and survival. This narrative review aims to summarise current knowledge of specific macro and micronutrients and their effect on allograft recovery and survival in the perioperative period, up to 1-year post transplant, to optimise the metabolic environment and mitigate risk to graft injury.
Collapse
Affiliation(s)
- Suzanne Schneider
- Directorate Applied Health, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Deborah Biggerstaff
- Directorate Applied Health, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Thomas M. Barber
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| |
Collapse
|
19
|
He J, Yao Y, Wang R, Liu Y, Wan X, Wang H, Zhou Y, Wang W, Ma Y, Lv X. Enhanced renal ischemia/reperfusion injury repair potential of exosomes derived from B7-H1 high mesenchymal stem cells. Front Genet 2025; 16:1516626. [PMID: 40242472 PMCID: PMC12000007 DOI: 10.3389/fgene.2025.1516626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
Two subgroups with high expression of B7-H1 and low expression of B7-H1 were successfully isolated from primitive human umbilical cord mesenchymal stem cells. And exosomes with high B7-H1 expression and low B7-H1 expression were successfully isolated. In comparison to the sham-operated group, mice in the IRI group demonstrated elevated serum levels of blood urea nitrogen (BUN) and serum creatinine (Scr), accompanied by a more pronounced degree of renal tissue damage. The administration of exosomes via the tail vein markedly accelerated the recovery of renal function in IRI mice, with the therapeutic effect beingmore pronounced in those treated with B7-H1high-Exo. Moreover RNA sequencing of mouse kidney treated with B7-H1high-Exo and B7-H1low-Exo showed that eight genes (C3, IRF7, AREG, CXCL10, Aldh1l2, Fnip2, Vcam1, St6galnac3) were involved in the pathophysiological process of ischemia-reperfusion injury. The in vitro and in vivo experiments showed that the expression level of C3 protein was significantly decreased, which indicated that B7-H1high-Exo played a therapeutic role by down-regulating C3.
Collapse
Affiliation(s)
- Jiahui He
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yawei Yao
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruiyan Wang
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yujia Liu
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xingyu Wan
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Hao Wang
- Department of Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuqiang Zhou
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Wenjing Wang
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yan Ma
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xinghua Lv
- Department of Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
20
|
Zhao Y, Xu Y, Xu Q, He N, Zhao J, Liu Y. p23 protects against ferroptosis of brain microvascular endothelial cells in ischemic stroke. Int J Mol Med 2025; 55:64. [PMID: 39981897 PMCID: PMC11878478 DOI: 10.3892/ijmm.2025.5505] [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: 09/14/2024] [Accepted: 01/03/2025] [Indexed: 02/22/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent regulated cell death that differs from apoptosis, autophagy or necrosis. p23 serves as a co‑chaperone and performs a unique biological function in various diseases by binding to client proteins to modulate their biological functions; however, its effect on ferroptosis remains largely unknown. In the present study, the effects of cerebral ischemia/reperfusion (I/R) injury (CIRI) or oxygen‑glucose deprivation/reoxygenation on the blood‑brain barrier (BBB) and ferroptosis in brain microvascular endothelial cells (BMECs), as well as the expression of p23, were examined. Subsequently, the effects of p23 on CIRI‑induced BBB dysfunction and BMEC ferroptosis were determined. Finally, the role of glutathione peroxidase 4 (GPX4) in the regulatory effects of p23 on ferroptosis was detected. The results revealed that p23 protected against BBB injury caused by CIRI by inhibiting ferroptosis in BMECs. The effect of p23 on ferroptosis was then explored, and it was found that the expression of GPX4, a major regulator of ferroptosis, was promoted by p23. Furthermore, molecular docking and co‑immunoprecipitation experiments revealed that p23 could bind to GPX4 through its N‑terminal domain (1‑90aa), enhance the stability of GPX4 and inhibit the degradation of GPX4 by cycloheximide. Finally, a cerebral I/R animal model was established using GPX4 conditional knockout mice (GPX4 FosCreERT2/+), and it was revealed that the protective effect of p23 overexpression on the BBB in GPX4 FosCreERT2/+ mice was attenuated compared with that in GPX4 FosCreERT2/‑ mice. In conclusion, p23 may serve a protective role against cerebral I/R‑induced BBB injury by inhibiting ferroptosis in BMECs through enhancing the stability of GPX4, providing a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yao Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Yunfei Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, P.R. China
- Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan 410013, P.R. China
- National Medicine Functional Experimental Teaching Center, Changsha, Hunan 410013, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
21
|
Dingfelder J, Kollmann D, Rauter L, Pereyra D, Kacar S, Weijler AM, Saffarian Zadeh T, Tortopis C, Silberhumer G, Salat A, Soliman T, Berlakovich G, Györi GP. Validation of mitochondrial FMN as a predictor for early allograft dysfunction and patient survival measured during hypothermic oxygenated perfusion. Liver Transpl 2025; 31:476-488. [PMID: 39787526 PMCID: PMC11895825 DOI: 10.1097/lvt.0000000000000512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/21/2024] [Indexed: 01/12/2025]
Abstract
Hypothermic oxygenated machine perfusion (HOPE) preconditions liver grafts before transplantation. While beneficial effects on patient outcomes were demonstrated, biomarkers for viability assessment during HOPE are scarce and lack validation. This study aims to validate the predictive potential of perfusate flavin mononucleotide (FMN) during HOPE to enable the implementation of FMN-based assessment into clinical routine and to identify safe organ acceptance thresholds. FMN was measured in perfusate samples of 50 liver grafts at multiple time points. After transplantation, patients were followed up for development of early allograft dysfunction (EAD), transplantation, and 1-year survival. FMN concentrations were significantly higher for grafts that developed EAD at 5 and 60 minutes into HOPE ( p = 0.008, p = 0.026). The strongest predictive potential of FMN for EAD was observed at 5 minutes of HOPE with an AUC of 0.744. Similarly, 5-minute FMN was predictive for 1-year mortality ( p < 0.001), reaching a remarkable AUC of 0.890. Cutoffs for prediction of EAD (10.6 ng/mL) and early mortality (23.5 ng/mL) were determined and allowed risk stratification of grafts. Particularly, patients receiving low-risk grafts developed EAD in 9% of cases, while all patients survived the first postoperative year. In contrast, high-risk organs developed an incidence of EAD at 62%, accompanied by the necessity of retransplantation in 38% of cases. One-year mortality in the high-risk cohort was 62%. Evaluation of FMN as early as 5 minutes during HOPE allows for risk stratification of liver grafts. Low-risk grafts, according to FMN, display a negligible risk for patients. Yet, high-risk grafts are associated with increased risk for EAD, transplantation, and early mortality and should not be used for transplantation without further assessment.
Collapse
Affiliation(s)
- Jule Dingfelder
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Dagmar Kollmann
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Laurin Rauter
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - David Pereyra
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Sertac Kacar
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Anna M. Weijler
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Tina Saffarian Zadeh
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Chiara Tortopis
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Gerd Silberhumer
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Salat
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Thomas Soliman
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Gabriela Berlakovich
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Georg P. Györi
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Xu H, Lu X, Qin R, Shao L, Chen L. The Evolution of Ischemia-Reperfusion Injury Research in Ischemic Stroke: Insights From a Two-Decade Bibliometric Analysis. Brain Behav 2025; 15:e70445. [PMID: 40165524 PMCID: PMC11959154 DOI: 10.1002/brb3.70445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Ischemic stroke is a complex disease with high mortality and disability rates. Ischemia-reperfusion injury is a common aftermath. There have been significant advancements in understanding ischemia-reperfusion injury in ischemic stroke over the past two decades. This study aims to evaluate the current state of ischemia-reperfusion injury in ischemic stroke through bibliometric analysis, identifying key research areas and emerging trends. METHODS Relevant documents in the Web of Science Core Collection, SCI-Expanded from January 1, 2003, to December 31, 2023, were downloaded on July 10, 2024. Bibliometric analysis was performed using HistCite, VOSviewer, CiteSpace, and Bibliometrics online analysis platform. RESULTS A total of 2179 research papers from 611 journals in 66 countries were included in this study. Among these papers, China emerged as the leading contributor of ischemia-reperfusion injury in ischemic stroke publications, with Capital Medical University standing out as the institution with the highest number of publications in this area. Y. Zhang was identified as the author with the most publications during the study period. Brain Research was found to be the most prolific journal for this research. The keywords "ferroptosis", "circular RNA", "polarization", and "fatty acid binding protein" represent the current hot spots of ischemia-reperfusion injury in ischemic stroke research. CONCLUSION This bibliometric analysis offers the first thorough overview of hot spots and research trends in ischemia-reperfusion injury in ischemic stroke over the previous 21 years, providing researchers with new ideas in the field. "ferroptosis", "circular RNA", "polarization", and "fatty acid binding protein" may be the focus of future studies.
Collapse
Affiliation(s)
- Hongyu Xu
- Department of Neurologythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xinglin Lu
- Department of Critical Care MedicineAffiliated Minzu Hospital of Guangxi Medical UniversityNanningChina
| | - Rongxing Qin
- Department of Neurologythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Lingduo Shao
- Department of Neurologythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Li Chen
- Department of Neurologythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
23
|
Xu Y, Yu Y, Guo Z. Hydrogels in cardiac tissue engineering: application and challenges. Mol Cell Biochem 2025; 480:2201-2222. [PMID: 39495368 DOI: 10.1007/s11010-024-05145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Cardiovascular disease remains the leading cause of global mortality. Current stem cell therapy and heart transplant therapy have limited long-term stability in cardiac function. Cardiac tissue engineering may be one of the key methods for regenerating damaged myocardial tissue. As an ideal scaffold material, hydrogel has become a viable tissue engineering therapy for the heart. Hydrogel can not only provide mechanical support for infarcted myocardium but also serve as a carrier for various drugs, bioactive factors, and cells to increase myocardial contractility and improve the cell microenvironment in the infarcted area, thereby improving cardiac function. This paper reviews the applications of hydrogels and biomedical mechanisms in cardiac tissue engineering and discusses the challenge of clinical transformation of hydrogel in cardiac tissue engineering, providing new strategies for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yaping Xu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Yuexin Yu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China.
| |
Collapse
|
24
|
Zhao Y, Cao Y, Su Y, Chen J, Wang X, Ding P, Hu W, Zhu T, Hu C. Identification of c-Jun phosphorylation as a crucial mediator of complement activation in renal ischemia-reperfusion injury revealed by phosphoproteomics and functional validation. Mol Biol Rep 2025; 52:345. [PMID: 40146438 DOI: 10.1007/s11033-025-10414-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Ischemia reperfusion injury (IRI) is an unavoidable condition that primarily affects graft function in renal transplantation. Blockage of complement activation by complement receptor immunoglobulin/ factor H (CRIg/FH), a novel complement inhibitor, shows great potency to ameliorate renal IRI. Sublytic membrane attack complex (MAC) disrupts cellular functions via the activation of different protein kinases and phosphorylation of critical signal transduction factors. We aimed to investigate whether complement activation triggered shift in phosphorylation status in IRI. METHODS AND RESULTS We performed a LC-MS/MS-based quantitative phosphoproteomic analysis of CRIg/FH-IRI, PBS-IRI and Sham mice, depicting a thorough protein phosphorylation profile. C3d and MAC staining were conducted to study the complement activation status. In vitro model mimicking complement mediated IRI tubular injury was achieved by applying normal human serum (NHS) to TCMK cells. By hierarchical clustering, we observed that CRIg/FH treatment reversed the hyperphosphorylation status triggered by IRI. Differentially expressed phosphoproteins (DEPs) were associated with focal adhesion, integrin activation, actin cytoskeleton organization and cell junction. We identified c-Jun as the most differentially phosphorylated transcriptional factor regulated by complement activation, the S63 phosphorylation of which was verified both in vitro and in vivo and screened for its downstream targets. JNK inhibitor reduced the phosphorylation of c-Jun and attenuated accumulation of the C3d on the tubular epithelial cells. CONCLUSION We proposed a crucial role of c-Jun phosphorylation in complement activation induced by renal IRI by combining phosphoproteomic approaches and protein validation, which hopefully could provide novel insights into the pathological mechanisms of IRI.
Collapse
Affiliation(s)
- Yufeng Zhao
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yirui Cao
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ying Su
- Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juntao Chen
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Xuanchuan Wang
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Collaborative Innovation Center of Cancer Medicine, Fudan University, Shanghai, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Collaborative Innovation Center of Cancer Medicine, Fudan University, Shanghai, China.
- Department of Oncology, Fudan University, Shanghai, China.
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.
| | - Tongyu Zhu
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.
| | - Chao Hu
- Department of Kidney Transplantation, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Second Yongjia Road, Huangpu District, Shanghai, China.
| |
Collapse
|
25
|
Ran Q, Zhang J, Zhong J, Lin J, Zhang S, Li G, You B. Organ preservation: current limitations and optimization approaches. Front Med (Lausanne) 2025; 12:1566080. [PMID: 40206471 PMCID: PMC11980443 DOI: 10.3389/fmed.2025.1566080] [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: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Despite the annual rise in patients with end-stage diseases necessitating organ transplantation, the scarcity of high-quality grafts constrains the further development of transplantation. The primary causes of the graft shortage are the scarcity of standard criteria donors, unsatisfactory organ preservation strategies, and mismatching issues. Organ preservation strategies are intimately related to pre-transplant graft viability and the incidence of adverse clinical outcomes. Static cold storage (SCS) is the current standard practice of organ preservation, characterized by its cost-effectiveness, ease of transport, and excellent clinical outcomes. However, cold-induced injury during static cold preservation, toxicity of organ preservation solution components, and post-transplantation reperfusion injury could further exacerbate graft damage. Long-term ex vivo dynamic machine perfusion (MP) preserves grafts in a near-physiological condition, evaluates graft viability, and cures damage to grafts, hence enhancing the usage and survival rates of marginal organs. With the increased use of extended criteria donors (ECD) and advancements in machine perfusion technology, static cold storage is being gradually replaced by machine perfusion. This review encapsulates the latest developments in cryopreservation, subzero non-freezing storage, static cold storage, and machine perfusion. The emphasis is on the injury mechanisms linked to static cold storage and optimization strategies, which may serve as references for the optimization of machine perfusion techniques.
Collapse
Affiliation(s)
- Qiulin Ran
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jisheng Zhong
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ji Lin
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shuai Zhang
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guang Li
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bin You
- Department of Cardiovascular Surgery, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Allois R, Pertusio R, Pagliaro P, Roatta S. Ischemic preconditioning: exploring local ergogenic mechanisms in non-fatiguing voluntary contractions. Front Physiol 2025; 16:1542394. [PMID: 40200986 PMCID: PMC11975927 DOI: 10.3389/fphys.2025.1542394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Background IPC has been suggested to boost skeletal muscle performance, though its effectiveness remains controversial. This study evaluates whether IPC influences local hemodynamic responses and surface electromyographic (sEMG) activity during non-fatiguing voluntary sustained and intermittent contractions. Methods Ten male participants were subjected to IPC (3 cycles, 5-min ON/5-min OFF right arm ischemia, cuff pressure: 250 mmHg) and SHAM (same protocol at 20 mmHg) in two different sessions. Near-infrared spectroscopy was used to monitor tissue oxygenation (TOI) and deoxy-hemoglobin (HHb) in extensor and flexor forearm muscles. sEMG was also recorded. Measurements were taken during sustained (20-s duration) and intermittent (5 s ON/5 s OFF) isometric contractions at 20, 30, and 40% of the maximal voluntary contraction. These non-fatiguing exercise tasks were performed before and 30 min after the IPC/SHAM intervention. Results sEMG exhibited a significant increase post vs. pre-treatment in both IPC and SHAM in extensors. A significant decrease in TOI at rest was noted pre vs. post-treatment for both IPC and SHAM (p < 0.01). In general, no main effect of treatment was observed, except for HHb changes during contraction in extensor muscles, associated with no effect of time and no time-treatment interaction. All variables exhibited a main effect of force level (p < 0.05), with no interaction with treatment or time. Conclusion IPC had no effect on hemodynamic and electromyographic variables during sustained and intermittent handgrip. These results do not support IPC-related ergogenic effects at the muscle level, aligning with previous findings on electrically stimulated contractions.
Collapse
Affiliation(s)
- Ruben Allois
- Department of Neuroscience, University of Torino, Torino, Italy
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | |
Collapse
|
27
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [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: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Cenik I, Van Slambrouck J, Barbarossa A, Vanluyten C, Jin X, Prisciandaro E, Provoost AL, Vandervelde CM, Novysedlák R, Serçik Ö, De Leyn P, Van Veer H, Depypere L, Jansen Y, Pirenne J, Van Raemdonck DE, Ceulemans LJ. Temperature dynamics of donor lungs from procurement to reperfusion: Static ice versus controlled hypothermic storage. J Heart Lung Transplant 2025:S1053-2498(25)01777-2. [PMID: 40118306 DOI: 10.1016/j.healun.2025.02.1695] [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: 11/27/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) remains an important challenge in lung transplantation (LTx). Ischemia can be divided into 3 phases: cooling during procurement, preservation, and rewarming during implantation. Temperature fluctuations influence metabolic processes, exacerbating IRI. However, actual lung temperatures have not been previously studied. Therefore, we aimed to characterize lung temperature dynamics in clinical LTx for static ice storage (SIS) and controlled hypothermic storage (CHS). METHODS From December 2022 to February 2024, we included 35 SIS and 19 CHS bilateral LTx cases at a single center, resulting in 70 SIS and 38 CHS lungs. Surface temperature (surfaceT°) was measured with a thermography camera. Preservation temperature (preservationT°) was remotely recorded for 6 SIS and 6 CHS. Core temperature (coreT°) was measured with a flexible probe in the lower lobe bronchus after unpacking and every 10 minutes during implantation. RESULTS Regarding SIS, mean ± standard deviation (SD) surfaceT° was 30°C ± 3.3°C before flushing, 17°C ± 4.1 °C after extraction, 8.6°C ± 3.3°C before packing. PreservationT° reached 4°C after 98 minutes and 0°C after 266 minutes. After unpacking, surfaceT° was 1.8°C ± 2.3°C, coreT° was 1.6°C ± 1.2°C. At 30 minutes implantation, surfaceT° was 25.0°C ± 2.9°C, coreT° was 22.0°C ± 4.4°C. CHS surfaceT° was 30.0°C ± 2.5°C before flushing, 17°C ± 3.8°C after extraction, and 11°C ± 3.5°C before packing. After unpacking, surfaceT° was 7.9°C ± 2.0°C, and coreT° was 7.1°C ± 1.1°C. At 30 minutes of implantation, surfaceT° was 26.0°C ± 1.7°C, and coreT° was 24.0°C ± 3.6°C. Postoperative outcome was comparable between both groups. CONCLUSIONS We characterized temperature dynamics in clinical LTx, revealing a rapid temperature drop with pulmonary flushing, potential freezing injury with SIS, and rapid rewarming during implantation.
Collapse
Affiliation(s)
- Ismail Cenik
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Annalisa Barbarossa
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Cedric Vanluyten
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Xin Jin
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Elena Prisciandaro
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - An-Lies Provoost
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Christelle M Vandervelde
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - René Novysedlák
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Ömer Serçik
- Data Science Institute, Interuniversity Institute for Biostatistics and statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium
| | - Paul De Leyn
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Hans Van Veer
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Lieven Depypere
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Yanina Jansen
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Wu L, Li Z, Wang K, Groleau RR, Rong X, Liu X, Liu C, Lewis SE, Zhu B, James TD. Advances in Organic Small Molecule-Based Fluorescent Probes for Precision Detection of Liver Diseases: A Perspective on Emerging Trends and Challenges. J Am Chem Soc 2025; 147:9001-9018. [PMID: 40036086 PMCID: PMC11926879 DOI: 10.1021/jacs.4c17092] [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: 03/06/2025]
Abstract
Liver disease poses a significant challenge to global health, and its early diagnosis is crucial for improving treatment outcomes and patient prognosis. Since fluctuation of key biomarkers during the onset and progression of liver diseases can directly reflect liver health and normal/abnormal function, biomarker-based assays are vital tools for the early detection of liver disease. In this context, small molecule fluorescent probes have undeniably emerged as indispensable tools for diagnosis and analysis, with an ever-growing number of small molecule-based fluorescent probes being developed over recent years, with the sole aim of monitoring relevant biomarkers of liver disease. This perspective will focus on the development and application of probes developed primarily over the last 10 years for diagnosing a range liver disease-related processes. It will outline the foundational design strategies for developing promising probes, their optical response to key biomarkers, and how they have been demonstrated in proof-of-concept imaging applications. Current challenges and new developments in the field will be discussed, with the aim of providing insights and highlighting opportunities in the field.
Collapse
Affiliation(s)
- Luling Wu
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
| | - Zilu Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Kun Wang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Robin R Groleau
- Department of Life Sciences, University of Bath, Bath BA2 7AY, U.K
| | - Xiaodi Rong
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Xueting Liu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Caiyun Liu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Simon E Lewis
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
| | - Baocun Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, People's Republic of China
| |
Collapse
|
30
|
Ullah K, Ai L, Li Y, Liu L, Zhang Q, Pan K, Humayun Z, Piao L, Sitikov A, Zhao Q, Su Q, Sharp W, Fang Y, Wu D, Liao JK, Wu R. ARNT-dependent HIF-2α signaling protects cardiac microvascular barrier integrity and heart function post-myocardial infarction. Commun Biol 2025; 8:440. [PMID: 40089572 PMCID: PMC11910586 DOI: 10.1038/s42003-025-07753-1] [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: 05/23/2024] [Accepted: 02/15/2025] [Indexed: 03/17/2025] Open
Abstract
Myocardial infarction (MI) compromises the cardiac microvascular endothelial barrier, increasing leakage and inflammation. HIF2α, predominantly expressed in cardiac endothelial cells during ischemia, has an unclear role in barrier function during MI. Here, we show that inducible, adult endothelial-specific deletion of Hif2α in mice leads to increased mortality, cardiac leakage, inflammation, reduced heart function, and adverse remodeling after MI. In parallel, human cardiac microvascular endothelial cells (HCMVECs) lacking HIF2α display impaired barrier integrity, reduced tight-junction proteins, increased cell death, and elevated IL-6 levels, effects that are alleviated by overexpressing ARNT, a key partner of HIF2α under hypoxic conditions. Interestingly, ARNT, but not HIF2α, directly binds the IL-6 promoter to suppress its expression. These findings suggest the HIF2α/ARNT axis as a protective mechanism in heart failure post-MI and identify potential therapeutic targets to support cardiac function.
Collapse
Affiliation(s)
- Karim Ullah
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Lizhuo Ai
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Yan Li
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Lifeng Liu
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Qin Zhang
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Kaichao Pan
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Zainab Humayun
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Lin Piao
- Emergency Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Albert Sitikov
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Qiong Zhao
- Division of Cardiology, Department of Medicine, Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Qiaozhu Su
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - Willard Sharp
- Emergency Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Yun Fang
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - James K Liao
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Rongxue Wu
- Section of Cardiology,, Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
31
|
Liu J, Jin Y, Lv F, Yang Y, Li J, Zhang Y, Zhong L, Liu W. Identification of biomarkers associated with programmed cell death in liver ischemia-reperfusion injury: insights from machine learning frameworks and molecular docking in multiple cohorts. Front Med (Lausanne) 2025; 12:1501467. [PMID: 40160318 PMCID: PMC11949969 DOI: 10.3389/fmed.2025.1501467] [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: 09/26/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Liver ischemia-reperfusion injury (LIRI) is a major reason for liver injury that occurs during surgical procedures such as hepatectomy and liver transplantation and is a major cause of graft dysfunction after transplantation. Programmed cell death (PCD) has been found to correlate with the degree of LIRI injury and plays an important role in the treatment of LIRI. We aim to comprehensively explore the expression patterns and mechanism of action of PCD-related genes in LIRI and to find novel molecular targets for early prevention and treatment of LIRI. Methods We first compared the expression profiles, immune profiles, and biological function profiles of LIRI and control samples. Then, the potential mechanisms of PCD-related differentially expressed genes in LIRI were explored by functional enrichment analysis. The hub genes for LIRI were further screened by applying multiple machine learning methods and Cytoscape. GSEA, GSVA, immune correlation analysis, transcription factor prediction, ceRNA network analysis, and single-cell analysis further revealed the mechanisms and regulatory network of the hub gene in LIRI. Finally, potential therapeutic agents for LIRI were explored based on the CMap database and molecular docking technology. Results Forty-seven differentially expressed genes associated with PCD were identified in LIRI, and functional enrichment analysis showed that they were involved in the regulation of the TNF signaling pathway as well as the regulation of hydrolase activity. By utilizing machine learning methods, 11 model genes were identified. ROC curves and confusion matrix from the six cohorts illustrate the superior diagnostic value of our model. MYC was identified as a hub PCD-related target in LIRI by Cytoscape. Finally, BMS-536924 and PF-431396 were identified as potential therapeutic agents for LIRI. Conclusion This study comprehensively characterizes PCD in LIRI and identifies one core molecule, providing a new strategy for early prevention and treatment of LIRI.
Collapse
Affiliation(s)
- Jifeng Liu
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yeheng Jin
- Department of Second Clinical College, China Medical University, Shenyang, Liaoning, China
| | - Fengchen Lv
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yao Yang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Junchen Li
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yunshu Zhang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lei Zhong
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Wei Liu
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
32
|
Cicek I, Esenulku CM, Somuncu AM, Bulut S, Yucel N, Bal Tastan T, Coban TA, Suleyman H. Sunitinib's Effect on Bilateral Optic Nerve Damage in Rats Following the Unilateral Clamping and Unclamping of the Common Carotid Artery. Biomedicines 2025; 13:620. [PMID: 40149596 PMCID: PMC11940743 DOI: 10.3390/biomedicines13030620] [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: 01/16/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Background/objectives: Common carotid artery occlusion can cause oxidant and inflammatory damage to the optic nerve. In this study, the effect of sunitinib was investigated, the antioxidant and anti-inflammatory properties of which have been previously reported and shown to be protective in I/R injury and in preventing bilateral optic nerve ischemia-reperfusion (I/R) injuries after unilateral common carotid artery ligation in rats. Methods: In this study, 18 Albino Wistar male rats were divided into SG (sham-operated), CCU (clamping and unclamping), and SCCU (sunitinib + clamping and unclamping) groups. One hour before the surgical procedures, sunitinib (25 mg/kg, oral) was given to SCCU rats. Anesthesia was induced with ketamine (60 mg/kg, ip) and sevoflurane. The right common carotid arteries of all rats were accessed under anesthesia. While the skin opened in SG rats was closed with sutures, the right common carotid arteries of CCU and SCCU rats were clipped, and an ischemia period was created for 10 min. Then, reperfusion (6 h) was achieved by unclipping. After euthanasia with ketamine (120 mg/kg, intraperitoneally), the right and left optic nerves of the rats were removed and examined biochemically and histopathologically. Results: Malondialdehyde, tumor necrosis factor α, interleukin-1β, and interleukin-6 were increased, and total glutathione levels had decreased in both ipsilateral and contralateral optic nerves (p < 0.05). These changes were more prominent on the ipsilateral side. Similarly, histopathological damage was observed to be more on the ipsilateral side (p < 0.05). Biochemical and histopathological changes were significantly suppressed in rats receiving sunitinib treatment (p < 0.05). Conclusions: Sunitinib may protect optic nerve tissue against I/R injury by reducing oxidative stress and inflammation.
Collapse
Affiliation(s)
- Ibrahim Cicek
- Department of Ophtalmology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey;
| | - Cenap Mahmut Esenulku
- Department of Ophthalmology, Trabzon Kanuni Health Application and Research Center, Health Sciences University, Trabzon 61040, Turkey; (C.M.E.); (A.M.S.)
| | - Ahmet Mehmet Somuncu
- Department of Ophthalmology, Trabzon Kanuni Health Application and Research Center, Health Sciences University, Trabzon 61040, Turkey; (C.M.E.); (A.M.S.)
| | - Seval Bulut
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey;
| | - Nurinisa Yucel
- Pharmacy Services Program, Vocational School of Health Services, Erzincan Binali Yildirim University, Erzincan 24036, Turkey;
| | - Tugba Bal Tastan
- Department of Histology and Embryology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan 24100, Turkey;
| | - Taha Abdulkadir Coban
- Department of Biochemistry, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan 24100, Turkey;
| | - Halis Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey;
| |
Collapse
|
33
|
Yan J, Kim H, Kim B, Piao H, Jang JY, Kang TK, Lee W, Kim D, Jo S, Shin D, Abuzar SMD, Kim ML, Yang J, Jon S. Synthetic Bilirubin-Based Nanomedicine Protects Against Renal Ischemia/Reperfusion Injury Through Antioxidant and Immune-Modulating Activity. Adv Healthc Mater 2025; 14:e2403846. [PMID: 39846887 PMCID: PMC11912105 DOI: 10.1002/adhm.202403846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/09/2025] [Indexed: 01/24/2025]
Abstract
Renal ischemia/reperfusion injury (IRI) is a common form of acute kidney injury. The basic mechanism underlying renal IRI is acute inflammation, where oxidative stress plays an important role. Although bilirubin exhibits potent reactive oxygen species (ROS)-scavenging properties, its clinical application is hindered by problems associated with solubility, stability, and toxicity. In this study, BX-001N, a synthetic polyethylene glycol-conjugated bilirubin 3α nanoparticle is developed and assessed its renoprotective effects in renal IRI. Intravenous administration of BX-001N led to increase uptake in the kidneys with minimal migration to the brain after IRI. Peri-IRI BX-001N administration improves renal function and attenuates renal tissue injury and tubular apoptosis to a greater extent than free bilirubin on day 1 after IRI. BX-001N suppressed renal infiltration of inflammatory cells and reduced expression of TNF-α and MCP-1. Furthermore, BX-001N increases renal tubular regeneration on day 3 and suppresses renal fibrosis on day 28. BX-001N decreases the renal expressions of dihydroethidium, malondialdehyde, and nitrotyrosine after IRI. In conclusion, BX-001N, the first Good Manufacturing Practice-grade synthetic bilirubin-based nanomedicine attenuates acute renal injury and chronic fibrosis by suppressing ROS generation and inflammation after IRI. It shows adequate safety profiles and holds promise as a new therapy for renal IRI.
Collapse
Affiliation(s)
- Ji‐Jing Yan
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hyunjin Kim
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | - Bomin Kim
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Honglin Piao
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Joon Young Jang
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Tae Kyeom Kang
- Natural Product Research CenterKorea Institute of Science & TechnologyGangneungGangwon‐do25451Republic of Korea
| | - Wook‐Bin Lee
- Natural Product Research CenterKorea Institute of Science & TechnologyGangneungGangwon‐do25451Republic of Korea
| | - Dohyeon Kim
- Department of Biological SciencesKAIST Institute for the BioCenturyCenter for Precision Bio‐NanomedicineKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Seunghyun Jo
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | | | | | - Myung L. Kim
- BILIX.Co., Ltd.YonginGyeonggi‐do16942Republic of Korea
| | - Jaeseok Yang
- Division of NephrologyDepartment of Internal MedicineYonsei University College of MedicineSeoul03722Republic of Korea
- The Research Institute for TransplantationYonsei University College of MedicineSeoul03722Republic of Korea
| | - Sangyong Jon
- Department of Biological SciencesKAIST Institute for the BioCenturyCenter for Precision Bio‐NanomedicineKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| |
Collapse
|
34
|
Van Slambrouck J, Loopmans S, Prisciandaro E, Barbarossa A, Kortleven P, Feys S, Vandervelde CM, Jin X, Cenik I, Moermans K, Fieuws S, Provoost AL, Willems A, De Leyn P, Van Veer H, Depypere L, Jansen Y, Pirenne J, Neyrinck A, Weynand B, Vanaudenaerde B, Carmeliet G, Vos R, Van Raemdonck D, Ghesquière B, Van Weyenbergh J, Ceulemans LJ. The effect of rewarming ischemia on tissue transcriptome and metabolome signatures: A clinical observational study in lung transplantation. J Heart Lung Transplant 2025; 44:437-447. [PMID: 39486771 DOI: 10.1016/j.healun.2024.10.020] [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: 05/23/2024] [Revised: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND In lung transplantation (LuTx), various ischemic phases exist, yet the rewarming ischemia time (RIT) during implantation has often been overlooked. During RIT, lungs are deflated and exposed to the body temperature in the recipient's chest cavity. Our prior clinical findings demonstrated that prolonged RIT increases the risk of primary graft dysfunction. However, the molecular mechanisms of rewarming ischemic injury in this context remain unexplored. We aimed to characterize the rewarming ischemia phase during LuTx by measuring organ temperature and comparing transcriptome and metabolome profiles in tissue obtained at the end versus the start of implantation. METHODS In a clinical observational study, 34 double-LuTx with ice preservation were analyzed. Lung core and surface temperature (n = 65 and 55 lungs) were measured during implantation. Biopsies (n = 59 lungs) were wedged from right middle lobe and left lingula at start and end of implantation. Tissue transcriptomic and metabolomic profiling were performed. RESULTS Temperature increased rapidly during implantation, reaching core/surface temperatures of 21.5°C/25.4°C within 30 minutes. Transcriptomics showed increased proinflammatory signaling and oxidative stress at the end of implantation. Upregulation of NLRP3 and NFKB1 correlated with RIT. Metabolomics indicated elevated levels of amino acids, hypoxanthine, uric acid, and cysteineglutathione disulfide alongside decreased levels of glucose and carnitines. Arginine, tyrosine, and 1-carboxyethylleucine showed a correlation with incremental RIT. CONCLUSIONS The final rewarming ischemia phase in LuTx involves rapid organ rewarming, accompanied by transcriptomic and metabolomic changes indicating proinflammatory signaling and disturbed cell metabolism. Limiting implantation time and cooling of the lung represent potential interventions to alleviate rewarming ischemic injury.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Shauni Loopmans
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Elena Prisciandaro
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Annalisa Barbarossa
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Phéline Kortleven
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, Molecular Virology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Simon Feys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Infectious and Inflammatory Disorders, KU Leuven, Leuven, Belgium; Department of Medical Intensive Care, University Hospitals Leuven, Leuven, Belgium
| | - Christelle M Vandervelde
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Xin Jin
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Ismail Cenik
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Karen Moermans
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Steffen Fieuws
- Department of Public Health, Interuniversity Center for Biostatistics and Statistical Bioinformatics, KU Leuven, Leuven, Belgium
| | - An-Lies Provoost
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Anton Willems
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Paul De Leyn
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Hans Van Veer
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Lieven Depypere
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Yanina Jansen
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Department of Microbiology, Immunology and Transplantation, Laboratory of Abdominal Transplantation, KU Leuven, Leuven, Belgium; Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne Neyrinck
- Department of Cardiovascular Sciences, Anesthesiology and Algology, KU Leuven, Leuven, Belgium; Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium; Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
| | - Bart Vanaudenaerde
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, Leuven, Belgium; Center for Cancer Biology, Metabolomics Core Facility Leuven, VIB, Leuven, Belgium
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium.
| |
Collapse
|
35
|
Liao X, Tang M, Li J, Guo R, Zhong C, Chen X, Zhang X, Mo H, Que D, Yu W, Song X, Li H, Cai Y, Yang P. Acid-Triggered Cascaded Responsive Supramolecular Peptide Alleviates Myocardial Ischemia‒Reperfusion Injury by Restoring Redox Homeostasis and Protecting Mitochondrial Function. Adv Healthc Mater 2025; 14:e2404319. [PMID: 39831810 DOI: 10.1002/adhm.202404319] [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: 11/04/2024] [Revised: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Redox imbalance, including excessive production of reactive oxygen species (ROS) caused by mitochondrial dysfunction and insufficient endogenous antioxidant capacity, is the primary cause of myocardial ischemia‒reperfusion (I/R) injury. In the exploration of reducing myocardial I/R injury, it is found that protecting myocardial mitochondrial function after reperfusion not only reduces ROS bursts but also inhibits cell apoptosis triggered by the release of cytochrome c. Additionally, nuclear factor erythroid 2-related factor 2 (Nrf2) is considered a potential therapeutic target for treating myocardial I/R injury by enhancing the cellular antioxidant capacity through the induction of endogenous antioxidant enzymes. In this study, a peptide‒drug conjugate OI-FFG-ss-SS31(ISP) is developed by integrating the Nrf2 activator 4-octyl itaconate (OI) and the mitochondria-targeting protective peptide elamipretide (SS31), and its therapeutic potential for myocardial I/R injury is explored. The results showed that ISP could self-assemble into nanofibers in response to the acidic microenvironment and bind to Keap-1 with high affinity, thereby activating Nrf2 and enhancing antioxidant capacity. Simultaneously, the release of SS31 could improve mitochondrial function and reduce ROS, ultimately providing a restoration of redox homeostasis to effectively alleviate myocardial I/R injury. This study presents a promising acid-triggered peptide-drug conjugate for treating myocardial I/R injury.
Collapse
Affiliation(s)
- Xu Liao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Min Tang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Department of Cardiology of Zhuzhou Central Hospital, NO.116 Changjiang South Road Tianyuan District, Zhuzhou, 412000, P. R. China
| | - Jiejing Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Runze Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Chongbin Zhong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xiangzhou Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xuwei Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hongwei Mo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Dongdong Que
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Wenjie Yu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xudong Song
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hekai Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Yanbin Cai
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| |
Collapse
|
36
|
Ohnari Y, Ueno K, Mori K, Kawashima T, Nishida H, Higuchi A, Tokumaru O, Miyamoto S. Resorcimoline as a Novel Therapeutic Agent Attenuates Ischemia-Reperfusion Injury in Hind Extremities. Ann Vasc Surg 2025; 112:388-396. [PMID: 39732330 DOI: 10.1016/j.avsg.2024.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Acute ischemia in the hind extremities is a dangerous disease that causes irreversible damage. Revascularization procedures are important to prevent muscle damage, but these treatments may induce additional damage, also known as ischemia-reperfusion injury. The role of free radicals as pivotal mediators of ischemia-reperfusion injury remains a prominent hypothesis. We have recently revealed potent antioxidative activities of a novel free-radical scavenger named resorcimoline (RML). The present study aims to investigate RML as a new therapeutic agent to reduce muscle damage and prevent motor dysfunction of the hind extremities caused by acute limb ischemia. METHODS Ischemia was induced in rats by occluding the femoral arteries in both hind limbs for 120 min with nylon bands, followed by reperfusion for 24 h. The RML group (n = 9) received an intravenous injection of RML immediately before reperfusion, whereas the saline group (n = 9) received an equivalent volume of saline. Motor function was evaluated by counting the number of steps required to return to normal gait. Serum biomarkers, including creatine kinase (CK) and lactate dehydrogenase (LDH), were measured to evaluate muscle injury. Muscle damage was assessed histologically with hematoxylin and eosin (HE) staining. Oxidative damage to DNA in muscle was evaluated by measuring the proportions of 8-hydroxy-2'-deoxyguanosine (8-OHdG)-positive cells by immunohistochemistry. RESULTS The average number of steps required to return to normal gait in the RML group was significantly smaller compared to the saline group (P = 0.04). Serum CK and LDH levels were significantly lower in the RML group than in the saline group (P = 0.03, P = 0.005). Histologically, the RML group demonstrated a significantly lower proportion of muscle damage (P = 0.004) and positivity of 8-OHdG (P = 0.01). CONCLUSION RML attenuated muscle damage and demonstrated protective effects against motor dysfunction after limb ischemia-reperfusion injury by reducing free-radical-induced DNA damage. RML can be a novel therapeutic agent that attenuates ischemia-reperfusion injury after acute limb ischemia.
Collapse
Affiliation(s)
- Yoshito Ohnari
- Department of Cardiovascular Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Kazuhiro Ueno
- Department of Cardiovascular Surgery, Oita University Faculty of Medicine, Oita, Japan.
| | - Kazuki Mori
- Department of Cardiovascular Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Takayuki Kawashima
- Department of Cardiovascular Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Haruto Nishida
- Department of Diagnostic Pathology, Oita University Faculty of Medicine, Oita, Japan
| | - Akihiro Higuchi
- Frontier Science and Social Co-creation Initiative, Kanazawa University, Ishikawa, Japan
| | - Osamu Tokumaru
- Department of Physiology, Faculty of Welfare and Health Sciences, Oita University, Oita, Japan
| | - Shinji Miyamoto
- Department of Cardiovascular Surgery, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
37
|
Peng JJ, Zhang YY, Li RF, Zhu WJ, Liu HR, Li HY, Liu B, Cao DS, Peng J, Luo XJ. Hybrid approach for drug-target interaction predictions in ischemic stroke models. Artif Intell Med 2025; 161:103067. [PMID: 39956766 DOI: 10.1016/j.artmed.2025.103067] [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: 05/20/2024] [Revised: 12/09/2024] [Accepted: 01/15/2025] [Indexed: 02/18/2025]
Abstract
Multiple cell death mechanisms are triggered during ischemic stroke and they are interconnected in a complex network with extensive crosstalk, complicating the development of targeted therapies. We therefore propose a novel framework for identifying disease-specific drug-target interaction (DTI), named strokeDTI, to extract key nodes within an interconnected graph network of activated pathways via leveraging transcriptomic sequencing data. Our findings reveal that the drugs a model can predict are highly representative of the characteristics of the database the model is trained on. However, models with comparable performance yield diametrically opposite predictions in real testing scenarios. Our analysis reveals a correlation between the reported literature on drug-target pairs and their binding scores. Leveraging this correlation, we introduced an additional module to assess the predictive validity of our model for each unique target, thereby improving the reliability of the framework's predictions. Our framework identified Cerdulatinib as a potential anti-stroke drug via targeting multiple cell death pathways, particularly necroptosis and apoptosis. Experimental validation in in vitro and in vivo models demonstrated that Cerdulatinib significantly attenuated stroke-induced brain injury via inhibiting multiple cell death pathways, improving neurological function, and reducing infarct volume. This highlights strokeDTI's potential for disease-specific drug-target identification and Cerdulatinib's potential as a potent anti-stroke drug.
Collapse
Affiliation(s)
- Jing-Jie Peng
- Department of Laboratory Medicine, the third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China
| | - Rui-Feng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China
| | - Wen-Jun Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China
| | - Hong-Rui Liu
- Department of Laboratory Medicine, the third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Hui-Yin Li
- Department of Laboratory Medicine, the third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Bin Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Dong-Sheng Cao
- Department of Pharmaceutical Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, the third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
38
|
Zhang Z, Zheng Z, Chen Y, Niu X, Ouyang T, Wang D. Mechanism of USP18-Mediated NCOA4 m6A Modification Via Maintaining FTO Stability In Regulating Ferritinophagy-Mediated Ferroptosis in Cerebral Ischemia-Reperfusion Injury. Mol Neurobiol 2025; 62:3848-3862. [PMID: 39331352 DOI: 10.1007/s12035-024-04494-w] [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: 04/13/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
This study aimed to explore whether USP18 regulates cerebral ischemia-reperfusion (I/R) injury via fat mass and obesity-associated proteins (FTO)-mediated NCOA4. Middle cerebral artery occlusion (MCAO) models were established in mice, and PC-12 cells treated with oxygen-glucose deprivation and reperfusion (OGD/R) were used as in vitro models. The USP18 lentiviral vector was transfected into cells in vitro and MCAO mice to observe its effect on ferroptosis. The relationship between USP18 and FTO was assessed using Co-IP and western blot. The effect of FTO on NCOA4 m6A modification was also elucidated. Overexpression of USP18 in MCAO models decreased cerebral infarct size and attenuated pathological conditions in mouse brain tissues. Moreover, USP18 reduced iron content, MDA, ROS, and LDH release, increased GSH levels and cell viability in both MCAO models and OGD/R cells, and promoted LC3 expression and autophagy flux. In vitro experiments on neurons showed that USP18 maintained FTO stability. The presence of FTO-m6A-YTFDH1-NCOA4 was also verified in neurons. Both in vivo and in vitro experiments showed that FTO and NCOA4 abrogated the protective effects of USP18 against ferritinophagy-mediated ferroptosis. Notably, USP18 maintains FTO stability, contributing to the removal of NCOA4 m6A modification and the suppression of NCOA4 translation, which consequently inhibits ferritinophagy-mediated ferroptosis to attenuate cerebral I/R injury.
Collapse
Affiliation(s)
- Zongyong Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Zongqing Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Yibiao Chen
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Xuegang Niu
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Taohui Ouyang
- Department of Neurosurgery, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Dengliang Wang
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China.
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
| |
Collapse
|
39
|
Öz S, Bahar MR, Şekerci G, Taşlıdere A, Tekin S. Protective Effects of Phoenixin-14 Administration Against Renal Ischemia/Reperfusion Injury in Rats. J Biochem Mol Toxicol 2025; 39:e70200. [PMID: 40025799 PMCID: PMC11873675 DOI: 10.1002/jbt.70200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Phoenixin (PNX), identified in the rat hypothalamus in 2013, has two bioactive isoforms with 14 and 20 amino acids. Initially studied for its role in reproductive regulation, research has since shown that PNX also prevents visceral pain, enhances memory, and aids heart tissue recovery. However, its role in kidney tissue remains unclear. Due to its antioxidant properties, PNX may help reduce oxidative stress and cellular damage in organs. This study was designed to determine the potential protective effects of Phoenixin-14 (PNX-14) against renal ischemia/reperfusion (I/R)-induced injury in rats. 40 male Wistar Albino rats were divided into four groups: Control, I/R, PNX-14 (50 µg/kg), and PNX-14 (100 µg/kg) (n = 10). All groups except the control group underwent 45 min of bilateral ischemia followed by 24 h of reperfusion. PNX-14 (50 and 100 μg/kg, intraperitoneally) was administered 1 h before induction of ischemia. Both doses of PNX-14 reduced the levels of acute kidney injury markers (blood urea nitrogen and creatinine) in blood tissue (p < 0.05). PNX-14 increased the activity of antioxidant enzymes (superoxide dismutase and catalase) and the levels of glutathione, while reducing malondialdehyde (p < 0.05). Histological evaluation of the I/R group revealed significant histopathological findings, and it was found that PNX-14 administration improved these histological damages (p < 0.05). These results suggest that PNX-14 provides protection against renal injury induced by I/R. After further studies, PNX-14 may be a new therapeutic strategy to prevent renal I/R injury.
Collapse
Affiliation(s)
- Samet Öz
- Department of Veterinary MedicineOsmaniye Korkut Ata University, Vocational School of Health ServicesOsmaniyeTurkey
| | | | - Güldeniz Şekerci
- Department of PhysiologyInonu University, Faculty of MedicineMalatyaTurkey
| | - Aslı Taşlıdere
- Department of Histology and EmbryologyInonu University, Faculty of MedicineMalatyaTurkey
| | - Suat Tekin
- Department of PhysiologyInonu University, Faculty of MedicineMalatyaTurkey
| |
Collapse
|
40
|
Singh P, Chaudhary M, Kazmi JS, Kuschner CE, Volpe BT, Chaudhuri TD, Becker LB. Vagus nerve stimulation: A targeted approach for reducing tissue-specific ischemic reperfusion injury. Biomed Pharmacother 2025; 184:117898. [PMID: 39923406 DOI: 10.1016/j.biopha.2025.117898] [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: 09/16/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Vagus Nerve Stimulation (VNS), a neuromodulation technique of applying controlled electrical impulses to the vagus nerve, has now emerged as a potential therapeutic approach for ischemia-reperfusion insults. It provides a pivotal link in improving functional outcomes for the central nervous system and multiple target organs affected by ischemia-reperfusion injury (I/RI). Reduced blood flow during ischemia and subsequent resumption of blood supply during reperfusion to the tissue compromises cellular health because of the combination of mitochondrial dysfunction, oxidative stress, cytokine release, inflammation, apoptosis, intracellular calcium overload, and endoplasmic reticulum stress, which ultimately leads to cell death and irreversible tissue damage. Furthermore, inflammation and apoptosis also play critical roles in the acute progression of ischemic injury pathology. Emerging evidence indicates that VNS in I/RI may act in an anti-inflammatory capacity, reducing oxidative stress and apoptosis, while also improving endothelial and mitochondrial function leading to reduced infarct sizes and cytoprotection in skeletal muscle, gastrointestinal tract, liver, kidney, lung, heart, and brain tissue. In this review, we attempt to shed light on the mechanistic links between tissue-specific damage following I/RI and the therapeutic approach of VNS in attenuating damage, considering both direct and remote I/RI scenarios. Thus, we want to advance the understanding of VNS that could further warrant its clinical implementation, especially as a treatment for I/RI.
Collapse
Affiliation(s)
- Parmeshar Singh
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA
| | - Manju Chaudhary
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jacob S Kazmi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA
| | - Bruce T Volpe
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Timir D Chaudhuri
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA; Department of Emergency Medicine, Kindai University Faculty of Medicine, Osaka, Japan.
| |
Collapse
|
41
|
Zhang Y, Liu Y, Luo S, Liang H, Guo C, Du Y, Li H, Wang L, Wang X, Tang C, Zhou Y. An adoptive cell therapy with TREM2-overexpressing macrophages mitigates the transition from acute kidney injury to chronic kidney disease. Clin Transl Med 2025; 15:e70252. [PMID: 40000418 PMCID: PMC11859120 DOI: 10.1002/ctm2.70252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Macrophages have been shown to contribute to renal injury and fibrosis as well as repair. Recently, Triggering Receptor Expressed on Myeloid Cells 2 (TREM2)-positive macrophages have been shown to play important roles in regulating tissue inflammation and repair. However, it remains unclear whether they can mitigate the transition from acute kidney injury to chronic kidney disease (the AKI-CKD transition). METHODS The AKI-CKD transition was generated by unilateral ischaemia-reperfusion injury (UIRI) in wild-type (WT) and Trem2 knockout mice. F4/80 magnetic beads were used to isolate renal macrophages. Flow cytometry was used to determine the levels of TREM2 and CD11b levels. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting and histological staining were performed to determine the expression of cytokines and fibrotic markers. RNA-seq was used to investigate transcriptomic changes between WT and Trem2 knockout bone marrow-derived macrophages (BMDMs). TREM2-overexpressing macrophages were generated using lentivirus and transferred intravenously to UIRI mice. RESULTS TREM2 macrophages exhibited a strong renal protective effect on the AKI-CKD transition. Genetic deletion of Trem2 resulted in increased renal inflammation and exacerbated renal injury and fibrosis in UIRI mice. Interestingly, we found that hypoxia could increase TREM2 expression in macrophages via HIF-1α. Upregulated TREM2 expression enhanced macrophage phagocytosis and suppressed the expression of pro-inflammatory cytokines, resulting in lower levels of apoptosis and fibrosis in tubular epithelial cells. Using RNA-seq analysis, we showed that the regulatory effects of TREM2 were orchestrated by the PI3K-AKT pathway. Pharmacological regulation of the PI3K-AKT pathway could modulate the macrophage-mediated inflammation and phagocytosis. In addition, an adoptive cell therapy using TREM2-overexpressing macrophages effectively reduced the immune cell infiltration, renal injury and fibrosis in UIRI mice. CONCLUSION Our study not only provides valuable mechanistic insights into the role of Trem2 in the AKI-CKD transition but also offers a new avenue for TREM2-overexpressing macrophage-based adoptive cell therapy to treat kidney diseases. KEY POINTS TREM2 knockout worsens kidney injury and accelerates AKI-CKD transition. TREM2 is upregulated by hypoxia via HIF1α in AKI-CKD transition. An adoptive cell therapy using TREM2-overexpressing macrophages reduces kidney inflammation and fibrosis.
Collapse
Affiliation(s)
- Yating Zhang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yu Liu
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Siweier Luo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hanzhi Liang
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Chipeng Guo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yufei Du
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hongyu Li
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Le Wang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaohua Wang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Chun Tang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yiming Zhou
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
42
|
Yu HC, Chung BH, Kim Y, Lee Y, Sim H, Lee S, Hwang HP, Yu HC, Jeon S, Maeng HJ, Shin D, Kang KP, Seo SY, Bae EJ, Park BH. p21-Activated Kinase 4 and Ischemic Acute Kidney Injury in Mice and Humans. J Am Soc Nephrol 2025:00001751-990000000-00576. [PMID: 40019790 DOI: 10.1681/asn.0000000649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/14/2025] [Indexed: 04/06/2025] Open
Abstract
Key Points
p21-activated kinase 4 (PAK4) phosphorylates and inactivates GSH peroxidase 3 in the kidney after ischemia-reperfusion.Mice lacking PAK4 or treated with PAK4 proteolysis-targeting chimera were protected from kidney damage caused by ischemia-reperfusion injury.PAK4 expression in kidney tissues post-transplant inversely correlated with kidney function.
Background
AKI after ischemia-reperfusion remains a substantial perioperative challenge lacking effective treatment. p21-activated kinase 4 (PAK4), a downstream effector of Rho GTPase, has been explored in hepatic ischemia-reperfusion injury, but its role in renal ischemia-reperfusion is unknown.
Methods
Wild-type and proximal tubule–specific Pak4 knockout mice underwent 25 minutes of ischemia followed by 24 hours of reperfusion injury. Primary tubular cells and human kidney-2 cells were exposed to hypoxia-reoxygenation injury to investigate the in vitro effect of PAK4. Selective degradation of PAK4 was employed using proteolysis-targeting chimera (PROTAC) to ameliorate AKI.
Results
Post–ischemia-reperfusion, the expression of PAK4 was upregulated through hypoxia-inducible factor 1 α in mouse kidneys. Deletion of PAK4 in proximal tubule cells, but not in myeloid cells, significantly mitigated ischemia-reperfusion–induced AKI, as evidenced by decreased levels of BUN, creatinine, tubular necrosis, apoptosis, macrophage infiltration, and lipid accumulation compared with control mice. Further investigation revealed that PAK4 phosphorylated GSH peroxidase 3 (GPx3) at T47, leading to its proteasomal degradation. In addition, pretreatment of mice with the PAK4 PROTAC preserved GPx3 and enhanced fatty acid β-oxidation, thereby protecting against AKI. In kidney tissues from people with a kidney transplant, elevated levels of PAK4 protein and phosphorylation of GPx3 at T47 were observed.
Conclusions
Renal tubular PAK4 contributes to tissue damage during ischemia-reperfusion injury, whereas PAK4 PROTAC mitigates ischemia-reperfusion injury by reducing oxidative stress and promoting fatty acid β-oxidation.
Collapse
Affiliation(s)
- Hwang Chan Yu
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| | - Byeoung Hoon Chung
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Yoejin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyunchae Sim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hong Pil Hwang
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hee Chul Yu
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seunggyu Jeon
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Eun Ju Bae
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| |
Collapse
|
43
|
Cheng L, Yin Z, Liu H, Shi S, Lv L, Wang Y, Zhou M, Li M, Guo T, Guo X, Yang G, Ma J, Yu J, Zhang Y, Duo S, Zhao L, Li R. Inhibition of LncRNA H19 Attenuates Testicular Torsion-Induced Apoptosis and Preserves Blood-Testis Barrier Integrity. Int J Mol Sci 2025; 26:2134. [PMID: 40076761 PMCID: PMC11899958 DOI: 10.3390/ijms26052134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Testicular torsion is a common emergency in adolescents, and can lead to severe ischemia reperfusion injury (IRI). LncRNA H19 has been shown to increase during ischemia, but its role in testicular IRI remains unknown. Focusing on this research gap, we utilized H19 biallelic mutant mice and Sertoli cell line (TM4) to construct in vivo and in vitro models of ischemia/reperfusion (I/R) and oxygen-glucose deprivation/reperfusion (OGD/R). Compared to WT I/R mice, H19-/- I/R mice showed milder tissue disorganization and cell loss, with a more intact blood-testis barrier (BTB). The cell viability decreased, ROS levels and apoptosis-related factors such as Bax/Bcl-2 increased in TM4 cells after OGD/R, whereas these changes were reversed when H19 was knocked down followed by OGD/R (si-H19+OGD/R). In contrast, over-expression of H19 in TM4 cells exacerbates OGD/R-induced cell apoptosis. Through in-depth analysis of KEGG-enriched pathways, the PI3K/AKT pathway was identified as a potential target of H19 modulation. Western blotting confirmed that, in OGD/R cells, elevated H19 levels were accompanied by the excessive AKT phosphorylation and the tight junction marker ZO-1 degradation; and in si-H19+OGD/R cells, the decreased AKT phosphorylation was recovered and the up-regulated ZO-1 expression was weakened simultaneously via using the AKT activator SC79. These results suggest that inhibiting H19 in OGD/R cells might preserve the integrity of the BTB by reversing the excessive phosphorylation of AKT. Moreover, H19 deficiency in si-H19+OGD/R cells alleviated the disturbances in glycolysis, fatty acid biosynthesis, and amino acid metabolism. Our study indicates that H19 might be a potential therapeutic target for clinic testicular I/R treatment.
Collapse
Affiliation(s)
- Linxin Cheng
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhibao Yin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Han Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Sijing Shi
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Limin Lv
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China;
| | - Yixi Wang
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Y.W.); (S.D.)
| | - Meng Zhou
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meishuang Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Tianxu Guo
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiyun Guo
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Guang Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Junjun Ma
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jinbo Yu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yu Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shuguang Duo
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Y.W.); (S.D.)
| | - Lihua Zhao
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China;
| |
Collapse
|
44
|
Yang L, Wen Y, Yuan Z, Zhao D, Weng P, Li Y, Chen Q, Zhang W, Hu H, Yu C. Hypoxia-mediated SUMOylation of FADD exacerbates endothelial cell injury via the RIPK1-RIPK3-MLKL signaling axis. Cell Death Dis 2025; 16:121. [PMID: 39984463 PMCID: PMC11845712 DOI: 10.1038/s41419-025-07441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 01/13/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Vascular endothelial cells are the predominant cell type in the cardiovascular system, and their dysfunction and death following hypoxic injury contribute to vascular lesions, playing an essential role in cardiovascular disease. Despite its importance, the mechanisms underlying vascular endothelial cell injury under hypoxia and potential therapeutic interventions remain poorly understood. Here, we constructed both an in vivo hypoxia model in C57BL/6 mice and an in vitro hypoxia model in HUVEC cells. Our findings demonstrated that hypoxia induces necroptosis in vascular endothelial cells and exacerbates inflammatory injury in vivo and in vitro, as evidenced by immunofluorescence and western blot. We identified FADD as a critical regulator of hypoxia-mediated necroptosis, with FADD knockdown significantly reversing hypoxia-induced necroptosis. Mechanistically, hypoxia affected protein conformation through SUMOylation of FADD and competitively inhibited its ubiquitination, leading to an increase in protein half-life and protein level of FADD. Furthermore, SUMOylation increased the interaction between FADD and RIPK1 and induced the formation of the FADD-RIPK1-RIPK3 complex, thereby promoting necroptosis in vascular endothelial cells. The SUMOylation inhibitor ginkgolic acid (GA) notably reduced hypoxia-induced vascular endothelial injury and inflammatory responses in male mice. Taken together, our research has uncovered a new process by which SUMOylation of FADD regulates hypoxia-induced necroptosis in endothelial cells, providing potential therapeutic targets for hypoxia-related cardiovascular diseases.
Collapse
Affiliation(s)
- Liming Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yilin Wen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Dezhang Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Ping Weng
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yueyue Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Qingyang Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Hui Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.
| |
Collapse
|
45
|
Rodríguez-Rodríguez DR, Mendoza-Hernández OH, Cordero-Pérez P, Rivas-Galindo VM, Moreno-Peña DP, Tijerina-Márquez R, Garza-Villarreal AM, Alarcón-Galván G, Muñoz-Espinosa LE, Zapata-Chavira HA, Hernández-Guedea MA, Solis-Cruz GY, Torres-González L. Nephroprotective and Antioxidant Effects of Jatropha dioica Extract Against Ischemia-Reperfusion Injury in Wistar Rats. Int J Mol Sci 2025; 26:1838. [PMID: 40076464 PMCID: PMC11899379 DOI: 10.3390/ijms26051838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Plant extracts with antioxidant activities have shown nephroprotection against IR injury. Jatropha dioica (Jd) possesses antioxidant activity. Our aim was to evaluate the effects of a hydroalcoholic Jd extract against IR injury in Wistar rats. Rats were divided into groups (n = 6): sham (SH); no toxicity (JdTox; 300 mg/kg/day of extract for 7 days); IR (on day 7 [I: 45 min/R: 24 h]); and Jd+IR (same treatment as JdTox; same surgical procedure as IR). AST and LDH were significantly lower in the JdTox. IR exhibited significantly higher CrS, BUN, and MDA compared with SH; Jd+IR showed reductions in these markers. GSH and SOD levels were significantly lower in IR compared with SH; an increase in these markers was observed in the Jd+IR. Histologically, IR showed significant increases in medullary tubular necrosis, medullary protein casts, and medullary vascular congestion compared with SH and JdTox. In Jd+IR, a significant decrease was observed only in medullary tubular necrosis. Therefore, the evaluated hydroalcoholic Jd extract dose showed no nephrotoxicity and hepatotoxicity. Jd extract pretreatment attenuated IR-induced renal injury, as evidenced by the improved serum markers of renal damage and oxidative stress.
Collapse
Affiliation(s)
- Diana Raquel Rodríguez-Rodríguez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Oscar Humberto Mendoza-Hernández
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Paula Cordero-Pérez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Verónica Mayela Rivas-Galindo
- Department of Analytical Chemistry, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (V.M.R.-G.); (G.Y.S.-C.)
| | - Diana Patricia Moreno-Peña
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Ramiro Tijerina-Márquez
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Alondra Michelle Garza-Villarreal
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Gabriela Alarcón-Galván
- Basic Science Department, School of Medicine, Universidad de Monterrey, Monterrey 66238, Nuevo León, Mexico;
| | - Linda Elsa Muñoz-Espinosa
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| | - Homero Arturo Zapata-Chavira
- Transplant Service, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (H.A.Z.-C.); (M.A.H.-G.)
| | - Marco Antonio Hernández-Guedea
- Transplant Service, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (H.A.Z.-C.); (M.A.H.-G.)
| | - Guadalupe Yazmín Solis-Cruz
- Department of Analytical Chemistry, School of Medicine, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (V.M.R.-G.); (G.Y.S.-C.)
| | - Liliana Torres-González
- Liver Unit, Department of Internal Medicine, University Hospital “Dr. José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (D.R.R.-R.); (O.H.M.-H.); (D.P.M.-P.); (R.T.-M.); (A.M.G.-V.); (L.E.M.-E.)
| |
Collapse
|
46
|
Zhang X, Deng Z, Xu X, Zhu J, Huang Z, Ye Y, Liu J, Luo D, Liu J, Yan M, Song Y. Tetrahedral Framework Nucleic Acid-Based Delivery of DJ-1-saRNA Prevent Retinal Ischaemia-Reperfusion Injury via Inhibiting Ferroptosis. Cell Prolif 2025:e13820. [PMID: 39980149 DOI: 10.1111/cpr.13820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Retinal ischaemia/reperfusion injury (RI/RI) is the primary pathophysiological mechanism underlying retinal ischaemic diseases, potentially resulting in significant and irreversible visual impairment. Currently, there are no effective treatments available for RI/RI, and oxidative stress is a critical factor that contributes to the associated damage. DJ-1, an important endogenous antioxidant, has been proposed as a promising therapeutic agent for RI/RI owing to its potential for overexpression. In this study, tetrahedral frame nucleic acids (tFNAs) were utilised as an effective delivery vehicle for DJ-1 small activating RNA (saRNA), resulting in the synthesis of a novel nanocomposite (tFNAs-DJ-1-saRNA). In vitro experiments demonstrated that tFNAs effectively delivered DJ-1-saRNA to R28 cells, thus exerting a repair effect on oxidative stress injury. In vivo investigations revealed that the intravitreal injection of tFNAs-DJ-1-saRNA facilitated retinal DJ-1 gene expression and mitigated retinal atrophy induced by RI/RI. Mechanistically, tFNAs-DJ-1-saRNA activated the xCT/GPX4 pathway, thereby inhibiting ferroptosis, reducing ganglion cell damage and protecting the retinal tissue. In conclusion, this study demonstrated that the tFNAs-DJ-1-saRNA complex can ameliorate RI/RI by inhibiting ferroptosis, suggesting its potential as a novel agent for the treatment of retinal ischaemic diseases.
Collapse
Affiliation(s)
- Xianggui Zhang
- The First School of Clinical Medical, Southern Medical University, Guangzhou, China
| | - Zhende Deng
- The First School of Clinical Medical, Southern Medical University, Guangzhou, China
| | - Xiaoxiao Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Zhu
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Zhen Huang
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Ya Ye
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Jingying Liu
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Delun Luo
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinnan Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming Yan
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Yanping Song
- The First School of Clinical Medical, Southern Medical University, Guangzhou, China
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| |
Collapse
|
47
|
Yang L, Tang H, Wang J, Xu D, Xuan R, Xie S, Xu P, Li X. O-GlcNAcylation attenuates ischemia-reperfusion-induced pulmonary epithelial cell ferroptosis via the Nrf2/G6PDH pathway. BMC Biol 2025; 23:32. [PMID: 39901237 PMCID: PMC11792224 DOI: 10.1186/s12915-025-02126-w] [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: 07/24/2024] [Accepted: 01/09/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Lung ischemia-reperfusion (I/R) injury is a common clinical pathology associated with high mortality. The pathophysiology of lung I/R injury involves ferroptosis and elevated protein O-GlcNAcylation levels, while the effect of O-GlcNAcylation on lung I/R injury remains unclear. This research aimed to explore the effect of O-GlcNAcylation on reducing ferroptosis in pulmonary epithelial cells caused by I/R. RESULTS First, we identified O-GlcNAc transferase 1 (Ogt1) as a differentially expressed gene in lung epithelial cells of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) patients, using single-cell sequencing, and Gene Ontology analysis (GO analysis) revealed the enrichment of the ferroptosis process. We found a time-dependent dynamic alteration in lung O-GlcNAcylation during I/R injury. Proteomics analysis identified the differentially expressed proteins enriched in ferroptosis and multiple redox-related pathways based on KEGG annotation. Thus, we generated Ogt1-conditional knockout mice and found that Ogt1 deficiency aggravated ferroptosis, as evidenced by lipid reactive oxygen species (lipid ROS), malondialdehyde (MDA), Fe2+, as well as alterations in critical protein expression glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11). Consistently, we found that elevated O-GlcNAcylation inhibited ferroptosis sensitivity in hypoxia/reoxygenation (H/R) injury-induced TC-1 cells via O-GlcNAcylated NF-E2-related factor-2 (Nrf2). Furthermore, both the chromatin immunoprecipitation (ChIP) assay and the dual-luciferase reporter assay indicated that Nrf2 could bind with translation start site (TSS) of glucose-6-phosphate dehydrogenase (G6PDH) and promote its transcriptional activity. As an important rate-limiting enzyme in the pentose phosphate pathway (PPP), elevated G6PDH provided a mass of nicotinamide adenine dinucleotide phosphate (NADPH) to improve the redox state of glutathione (GSH) and eventually led to ferroptosis resistance. Rescue experiments proved that Nrf2 knockdown or Nrf2-T334A (O-GlcNAcylation site) mutation abolished the protective effect of ferroptosis resistance. CONCLUSIONS In summary, we revealed that O-GlcNAcylation could protect against I/R lung injury by reducing ferroptosis sensitivity via the Nrf2/G6PDH pathway. Our work will provide a new basis for clinical therapeutic strategies for pulmonary ischemia-reperfusion-induced acute lung injury.
Collapse
Affiliation(s)
- Liuqing Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Dawei Xu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Rui Xuan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Pengfei Xu
- Department of Hepatobiliary and Pancreatic Surgery, School of Pharmaceutical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
48
|
Yuan Y, Chen T, Yang Y, Han H, Xu L. E2F1/CDK5/DRP1 axis mediates microglial mitochondrial division and autophagy in the pathogenesis of cerebral ischemia-reperfusion injury. Clin Transl Med 2025; 15:e70197. [PMID: 39968698 PMCID: PMC11836619 DOI: 10.1002/ctm2.70197] [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: 06/17/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The integrity of brain function is at stake due to cerebral ischemia-reperfusion injury (CIRI), which encompasses mitochondrial dysfunction, autophagy, and neuroinflammation. The role of E2F1 in mediating these processes in microglia during CIRI remains unclear. METHODS A CIRI mouse model was utilized for single-cell RNA transcriptome sequencing of brain tissues. The research comprised diverse gene expression, gene ontology (GO), and the enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Experimental techniques included oxygen-glucose deprivation (OGD/R) cell models, RT-qPCR, Western Blot, ChIP assays, and microglia-neuron co-cultures. RESULTS A significant aspect highlighted in the study was the involvement of CDK5 in the induction of mitochondrial abnormalities associated with CIRI. Upregulation of E2F1 and CDK5 in post-CIRI microglia was observed. E2F1 facilitated CDK5 transcription, leading to DRP1 phosphorylation, exacerbating neurotoxic effects. Silencing E2F1 improved neurobehavioral outcomes in CIRI mice. CONCLUSIONS Activation of E2F1-mediated CDK5 drives mitochondrial division while inhibiting mitophagy in microglia, triggering inflammation, neuronal apoptosis, and exacerbating CIRI damage. Targeting this pathway could offer novel therapeutic strategies for mitigating CIRI-induced brain injury. KEY POINTS Identification of the E2F1/CDK5/DRP1 Axis in CIRI This study reveals that the E2F1 transcription factor upregulates CDK5 expression, which in turn phosphorylates DRP1, promoting excessive mitochondrial fission and inhibiting mitophagy in microglia. This mechanism plays a critical role in cerebral ischemia-reperfusion injury (CIRI). Mitochondrial Dysfunction and Neuroinflammation The activation of DRP1 leads to mitochondrial fragmentation and excessive ROS accumulation, triggering microglial activation and inflammatory responses, exacerbating neuronal apoptosis and brain injury in CIRI. Therapeutic Potential of E2F1 Silencing Knockdown of E2F1 in microglia effectively reduces mitochondrial damage, restores mitophagy, suppresses inflammation, and improves neurological outcomes in a CIRI mouse model, highlighting a promising therapeutic target for ischemic stroke intervention.
Collapse
Affiliation(s)
- Ya‐Jing Yuan
- Department of AnesthesiaTianjin Medical University Cancer Institute &HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and Therapy, TianjinTianjinChina
| | - Tingting Chen
- Department of Radiation OncologyTianjin Medical University Cancer Institute &HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and Therapy, TianjinTianjinChina
| | - Yan‐Ling Yang
- Department of Radiation OncologyTianjin Medical University Cancer Institute &HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and Therapy, TianjinTianjinChina
| | - Hao‐Nan Han
- Hubei Key Laboratory of Tumor Microenvironment and ImmunotherapyCollege of Basic Medical SciencesChina Three Gorges UniversityYichangChina
| | - Li‐Ming Xu
- Department of Radiation OncologyTianjin Medical University Cancer Institute &HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and Therapy, TianjinTianjinChina
| |
Collapse
|
49
|
Demirdas E, Arslan G, Kartal H, Erol G, Ozdem T, Yavuz BB, Gunay C, Oz BS. Melatonin as a shield against skeletal muscle damage: A study on ischemia-reperfusion injury. ULUS TRAVMA ACIL CER 2025; 31:103-111. [PMID: 39963912 PMCID: PMC11843425 DOI: 10.14744/tjtes.2025.44890] [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: 11/20/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025]
Abstract
We evaluated the protective effects of melatonin against skeletal muscle ischemia-reperfusion injury, a significant cause of skeletal muscle damage. Ischemia-reperfusion (I/R) injury occurs due to a temporary restriction of blood flow (ischemia) followed by its restoration (reperfusion), triggering oxidative stress, inflammation, and cell death. Although current treatments are limited, melatonin's antioxidant and anti-inflammatory properties suggest potential benefits. METHODS We studied 30 male mice divided into five groups: control, melatonin control, I/R, melatonin + I/R, and dimethyl sulfoxide control. After the designated treatments, we assessed muscle tissue for antioxidant capacity (total antioxidant status [TAS]), oxidative stress markers (total oxidative status [TOS] and malondialdehyde [MDA]), inflammation (myeloperoxidase [MPO]), and cell death (terminal deoxynucleotidyl transferase dUTP nick-end labeling [TUNEL] assay and histological analysis). RESULTS Melatonin significantly increased antioxidant capacity (TAS) compared to all other groups. Conversely, oxidative stress (TOS) was significantly lower in the melatonin + I/R group compared to the I/R group alone. Histological analysis revealed greater necrosis, edema, inflammation, and cell death in the I/R group compared to others. Interestingly, the melatonin + I/R group exhibited significantly less damage than the I/R group, highlighting melatonin's protective effect. CONCLUSION This study demonstrates that exogenous melatonin effectively reduces oxidative stress, inflammation, and cell death in skeletal muscle tissue subjected to I/R injury. These findings suggest that melatonin may be a promising therapeutic agent for mitigating I/R-induced complications in skeletal muscle injury.
Collapse
Affiliation(s)
- Ertan Demirdas
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Gokhan Arslan
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Hakan Kartal
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Gokhan Erol
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Tayfun Ozdem
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Basak Buyuk Yavuz
- Department of Histology and Embryology, Izmir Democracy University Faculty of Medicine, Izmir-Türkiye
| | - Celalettin Gunay
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| | - Bilgehan Savas Oz
- Department of Cardiovascular Surgery, University of Health Sciences Gulhane Faculty of Medicine, Ankara-Türkiye
| |
Collapse
|
50
|
Ishikawa K, Murao A, Aziz M, Wang P. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 (MOP3) attenuates inflammation and improves survival in hepatic ischemia/reperfusion injury. Surgery 2025; 178:108872. [PMID: 39455391 PMCID: PMC11717596 DOI: 10.1016/j.surg.2024.09.029] [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: 06/21/2024] [Revised: 08/20/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Hepatic ischemia/reperfusion injury is a severe clinical condition leading to high mortality as the result of excessive inflammation, partially triggered by released damage-associated molecular patterns. Extracellular cold-inducible RNA-binding protein is a new damage-associated molecular pattern. Current clinical management of hepatic ischemia/reperfusion injury is limited to supportive therapy, necessitating the development of novel and effective treatment strategies. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 is a newly invented oligopeptide originating from milk fat globule-epidermal growth factor-VIII. This peptide acts as an opsonic compound that specifically binds to extracellular cold-inducible RNA-binding protein to facilitate its clearance by phagocytes, thereby attenuating inflammation. In this study, we hypothesized that milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 attenuated hepatic ischemia/reperfusion injury by inhibiting extracellular cold-inducible RNA-binding protein-induced inflammation in Kupffer cells. METHODS We treated Kupffer cells isolated from male C57BL/6 mice with extracellular cold-inducible RNA-binding protein and various doses of milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 for 4 hours, then measured cytokines in the culture supernatants. In addition, mice underwent 70% hepatic ischemia for 60 minutes immediately followed by the intravenous administration of either vehicle or milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3. Blood and ischemic liver tissues were collected 24 hours later, and inflammatory markers including cytokines, liver enzymes, chemokines, myeloperoxidase activity, and Z-DNA-binding protein 1 were measured. Hepatic tissue damage and cell death were evaluated histologically. Survival rates were monitored for 10 days posthepatic ischemia/reperfusion. RESULTS The release of interleukin-6 and tumor necrosis factor-α from extracellular cold-inducible RNA-binding protein-challenged Kupffer cells was significantly reduced by milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 in a dose-dependent manner. In hepatic ischemia/reperfusion mice, milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment significantly decreased serum levels of extracellular cold-inducible RNA-binding protein, interleukin-6, tumor necrosis factor-α, aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment also significantly reduced mRNA levels of interleukin-6, tumor necrosis factor-α, interleukin-1β, Z-DNA-binding protein 1, and chemokine macrophage inflammatory protein-2, as well as myeloperoxidase activity in hepatic tissues. Histologic evaluation demonstrated that treatment with milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 significantly attenuated tissue damage and cell death in the liver of hepatic ischemia/reperfusion mice. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment significantly improved the survival rate of hepatic ischemia/reperfusion mice. CONCLUSION Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 significantly attenuated inflammation and liver tissue damage and improved survival after hepatic ischemia/reperfusion. Thus, milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 holds promise as a potential future therapeutic strategy for hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Kouhei Ishikawa
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| |
Collapse
|