1
|
Liu J, Guan A, Huo Z, Li X, Zhu Y, Liang H, Liu W, Zhou H, Lin Z, Yan B. Distinct neurotoxic mechanisms of thallium and lead: Calcium-mediated apoptosis and iron-induced ferroptosis in zebrafish at environmental concentrations. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138288. [PMID: 40239529 DOI: 10.1016/j.jhazmat.2025.138288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Environmental neurotoxicants, such as thallium (Tl) and lead (Pb), pose significant risks to human health, yet their toxicological mechanisms remain poorly understood. This study investigates the distinct mechanisms of Tl+- and Pb2+-induced neurotoxicity at environmentally relevant concentrations using zebrafish embryos as a model. Transcriptomic analyses revealed minimal overlap in gene expression changes between the two metals, underscoring their unique toxicological pathways. Further study demonstrated that Tl+ disrupted calcium homeostasis, activating the calcium signaling pathway and triggering apoptosis via MAPK signaling. In contrast, Pb2+ exposure triggered ferroptosis, characterized by iron overload and lipid peroxidation, with significant involvement of oxidative stress and disruption of iron metabolism. Cellular assays validated these findings, confirming the critical roles of calcium and iron dysregulation in neuronal damage. These results emphasize the complexity of metal-induced neurotoxicity and the importance of studying pollutants at realistic exposure levels. Understanding the divergent pathways of Tl+- and Pb2+-induced neurotoxicity provides critical insights for mitigating their environmental and public health impacts, highlighting the need for targeted regulatory strategies to address heavy metal pollution effectively.
Collapse
Affiliation(s)
- Jian Liu
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Solid Wastes Pollution Control and Recycling, South China University of Technology, Guangzhou, Guangdong 510006, China; Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Ai Guan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Zihui Huo
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Xin Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Yiwen Zhu
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Solid Wastes Pollution Control and Recycling, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Hongwei Liang
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Weizhen Liu
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Solid Wastes Pollution Control and Recycling, South China University of Technology, Guangzhou, Guangdong 510006, China.
| | - Hongyu Zhou
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China.
| | - Zhang Lin
- School of Metallurgy and Environment, Central South University, Changsha 410083, China; Chinese National Engineering Research Center for Control & Treatment of Heavy Metal Pollution, Changsha, Hunan 410083, China
| | - Bing Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, Guangdong 510006, China
| |
Collapse
|
2
|
Bai M, Sang Z, Cui Y, Feng H, Liu Y, Dai Z, Zhou Z, Mei X, Zhao H. Multi-omics analysis identified and confirmed TNF-α as a key initiator of the inflammatory response following spinal cord injury. Int J Biol Macromol 2025; 311:143866. [PMID: 40319972 DOI: 10.1016/j.ijbiomac.2025.143866] [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: 10/22/2024] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Spinal cord injury (SCI) is a complex central nervous system (CNS) trauma that triggers multifaceted immune responses. In recent years, zinc has garnered considerable attention for its beneficial roles in anti-apoptosis and the attenuation of oxidative stress. However, the regulatory mechanisms by which zinc influence post-SCI immune-inflammatory responses remain insufficiently understood. In this study, we employed a multi-omics approach, combining transcriptomics, metabolomics, and single-cell RNA sequencing, to elucidate the mechanisms through which zinc ions modulate immune responses in injured spinal cord tissues. Our results demonstrate that zinc ions significantly regulate the expression of TNF-α signaling molecules. By inhibiting the TNF-α signaling pathway, zinc ions effectively mitigate apoptosis and reduce immune-inflammatory responses following SCI. Furthermore, through the integration of human and murine immune cell atlases, we performed a cluster analysis of key immune cell populations and constructed an immunological landscape of spinal cord tissues post‑zinc ion treatment. Notably, we identified microglia, key CNS immune cells, as exerting a strong anti-inflammatory effect by suppressing their TNF-α signaling activity. This study not only sheds light on the pivotal immune-inflammatory regulatory mechanisms of TNF-α signaling in zinc ion-mediated SCI therapy but also provides valuable theoretical and experimental insights for advancing preclinical research on SCI.
Collapse
Affiliation(s)
- Mingyu Bai
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China
| | - Zelin Sang
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China
| | - Yang Cui
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China
| | - Huicong Feng
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Clinical Research Center for Bone Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou, Liaoning Province 121000, China
| | - Yu Liu
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China
| | - Zhen Dai
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China
| | - Zipeng Zhou
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China; Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, PLA General Hospital, Beijing, China.
| | - Xifan Mei
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China; Liaoning Vocational College of Medicine, Shenyang, Liaoning Province 121000, China; Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Clinical Research Center for Bone Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou, Liaoning Province 121000, China.
| | - Haosen Zhao
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Clinical Research Center for Bone Tissue Engineering, Jinzhou, Liaoning Province 121000, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou, Liaoning Province 121000, China.
| |
Collapse
|
3
|
Zhong X, Long L, Yao H, Zhu Z, Xie Z, Zhang Y, Chen F. Ammonium tetrathiomolybdate attenuates acetaminophen-induced acute liver failure by inhibiting the TRPV4/Calcium/NF-κB signaling pathway. Biochem Pharmacol 2025; 237:116917. [PMID: 40220797 DOI: 10.1016/j.bcp.2025.116917] [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: 11/16/2024] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
Acute liver failure (ALF), characterized by fulminant hepatic necrosis and excessive inflammatory-oxidative cascades, remains a critical clinical challenge with limited therapeutic options. This study investigates the therapeutic potential of ammonium tetrathiomolybdate (ATTM)-a copper-chelating agent with multimodal anti-inflammatory and antioxidant properties-in acetaminophen (APAP)-induced ALF. Utilizing APAP-challenged C57BL/6J mice, we demonstrated that ATTM administration, whether prophylactic or delayed by 2 h post-exposure, significantly attenuated hepatotoxicity, as evidenced by reduced histopathological damage and improved survival rates. These therapeutic effects were further confirmed in AML12 hepatocytes, thereby reinforcing the observed in vivo findings. RNA sequencing revealed that calcium signaling is the predominant pathway modulated by ATTM. Subsequent mechanistic validation identified Transient Receptor Potential Cation Channel Subfamily V Member 4 (TRPV4)-mediated calcium influx as the critical therapeutic target. ATTM suppressed TRPV4-dependent calcium mobilization, thereby inhibiting the sequential phosphorylation of NF-κB pathway components in both murine liver tissue and AML12 cells. Crucially, TRPV4 agonism via RN-1747 reversed the hepatoprotective effects of ATTM, thereby confirming the centrality of this axis in mediating ATTM's therapeutic actions. These findings establish ATTM as a novel modulator of the TRPV4/calcium/NF-κB signaling cascade, capable of interrupting inflammatory-oxidative loops at multiple nodes. Our work not only elucidates a previously unrecognized mechanism for copper chelators in ALF management but also positions ATTM as a promising therapeutic candidate warranting clinical translation.
Collapse
Affiliation(s)
- Xingyi Zhong
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| | - Lingzhi Long
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital, Central South University, 410013 Changsha, China.
| | - Huijun Yao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| | - Zhiqin Zhu
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| | - Ziping Xie
- Zhongshan Hospital, Fudan University (Xiamen Branch), 361015 Xiamen, China.
| | - Yangfeng Zhang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| | - Fengsheng Chen
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, 510315 Guangzhou, China.
| |
Collapse
|
4
|
Wang H, Cheng C, Ding J, Qian R, Luo T, Zheng L, Chen Y. Trifluoperazine effect on human sperm: The accumulation of reactive oxygen species and the decrease in the mitochondrial membrane potential. Reprod Toxicol 2024; 130:108730. [PMID: 39369966 DOI: 10.1016/j.reprotox.2024.108730] [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: 07/23/2024] [Revised: 09/05/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
A strong link between antipsychotic drug use and reduced human sperm quality has been reported. Trifluoperazine (TFP), a commonly used antipsychotic, is now being explored for anticancer applications. Although there are hints that TFP might affect the male reproductive system, its impact on human sperm quality remains uncertain. Using a human sperm and TFP in vitro coculture system, we examined the effect of TFP (12.5, 25, 50 and 100 μM) on human sperm function and physiological parameters. The results showed that 50 μM and 100 μM TFP induced the accumulation of reactive oxygen species (ROS) and a decrease in the mitochondrial membrane potential (MMP) of human sperm, leading to decreased sperm viability, while 25 μM TFP inhibited only the penetration ability, total sperm motility, and progressive motility. Although 12.5 μM and 25 μM TFP increased [Ca2+]i in human sperm, they did not affect capacitation or the acrosome reaction. These results may be explained by the observation that 12.5 μM and 25 μM TFP did not increase tyrosine phosphorylation in human sperm, although TFP increased [Ca2+]i in a time-course traces similar to that of progesterone. Our results indicated that TFP could cause male reproductive toxicity by inducing the accumulation of ROS and a decrease in the MMP in human sperm.
Collapse
Affiliation(s)
- Houpeng Wang
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; School of Public Health, Jiangxi Medical College, Nanchang University, China
| | - Cheng Cheng
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Jing Ding
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Ruirui Qian
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Tao Luo
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Liping Zheng
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; School of Public Health, Jiangxi Medical College, Nanchang University, China.
| | - Ying Chen
- Institute of Biomedical Innovation and School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
5
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [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: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
6
|
Hu K, Xu Y, Fan J, Liu H, Di C, Xu F, Wu L, Ding K, Zhang T, Wang L, Ai H, Xie L, Wang G, Liang Y. Feasibility exploration of GSH in the treatment of acute hepatic encephalopathy from the aspects of pharmacokinetics, pharmacodynamics, and mechanism. Front Pharmacol 2024; 15:1387409. [PMID: 38887546 PMCID: PMC11181355 DOI: 10.3389/fphar.2024.1387409] [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: 02/17/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024] Open
Abstract
Our previous study highlighted the therapeutic potential of glutathione (GSH), an intracellular thiol tripeptide ubiquitous in mammalian tissues, in mitigating hepatic and cerebral damage. Building on this premise, we posited the hypothesis that GSH could be a promising candidate for treating acute hepatic encephalopathy (AHE). To verify this conjecture, we systematically investigated the feasibility of GSH as a therapeutic agent for AHE through comprehensive pharmacokinetic, pharmacodynamic, and mechanistic studies using a thioacetamide-induced AHE rat model. Our pharmacodynamic data demonstrated that oral GSH could significantly improve behavioral scores and reduce hepatic damage of AHE rats by regulating intrahepatic ALT, AST, inflammatory factors, and homeostasis of amino acids. Additionally, oral GSH demonstrated neuroprotective effects by alleviating the accumulation of intracerebral glutamine, down-regulating glutamine synthetase, and reducing taurine exposure. Pharmacokinetic studies suggested that AHE modeling led to significant decrease in hepatic and cerebral exposure of GSH and cysteine. However, oral GSH greatly enhanced the intrahepatic and intracortical GSH and CYS in AHE rats. Given the pivotal roles of CYS and GSH in maintaining redox homeostasis, we investigated the interplay between oxidative stress and pathogenesis/treatment of AHE. Our data revealed that GSH administration significantly relieved oxidative stress levels caused by AHE modeling via down-regulating the expression of NADPH oxidase 4 (NOX4) and NF-κB P65. Importantly, our findings further suggested that GSH administration significantly regulated the excessive endoplasmic reticulum (ER) stress caused by AHE modeling through the iNOS/ATF4/Ddit3 pathway. In summary, our study uncovered that exogenous GSH could stabilize intracerebral GSH and CYS levels to act on brain oxidative and ER stress, which have great significance for revealing the therapeutic effect of GSH on AHE and promoting its further development and clinical application.
Collapse
Affiliation(s)
- Kangrui Hu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yexin Xu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiye Fan
- Department of Pharmacy, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei Province, China
| | - Huafang Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chanjuan Di
- Hebei Zhitong Biopharmaceutical Co., Ltd., Gucheng, Hebei Province, China
| | - Feng Xu
- Hebei Zhitong Biopharmaceutical Co., Ltd., Gucheng, Hebei Province, China
| | - Linlin Wu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ke Ding
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Leyi Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoyu Ai
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lin Xie
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | | | - Yan Liang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
7
|
Lu B, Wei L, Shi G, Du J. Nanotherapeutics for Alleviating Anesthesia-Associated Complications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308241. [PMID: 38342603 PMCID: PMC11022745 DOI: 10.1002/advs.202308241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
Current management of anesthesia-associated complications falls short in terms of both efficacy and safety. Nanomaterials with versatile properties and unique nano-bio interactions hold substantial promise as therapeutics for addressing these complications. This review conducts a thorough examination of the existing nanotherapeutics and highlights the strategies for developing prospective nanomedicines to mitigate anesthetics-related toxicity. Initially, general, regional, and local anesthesia along with the commonly used anesthetics and related prevalent side effects are introduced. Furthermore, employing nanotechnology to prevent and alleviate the complications of anesthetics is systematically demonstrated from three aspects, that is, developing 1) safe nano-formulization for anesthetics; 2) nano-antidotes to sequester overdosed anesthetics and alter their pharmacokinetics; 3) nanomedicines with pharmacodynamic activities to treat anesthetics toxicity. Finally, the prospects and challenges facing the clinical translation of nanotherapeutics for anesthesia-related complications are discussed. This work provides a comprehensive roadmap for developing effective nanotherapeutics to prevent and mitigate anesthesia-associated toxicity, which can potentially revolutionize the management of anesthesia complications.
Collapse
Affiliation(s)
- Bin Lu
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
| | - Ling Wei
- Shanxi Bethune Hospital Center Surgery DepartmentShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan030032China
| | - Gaoxiang Shi
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
| | - Jiangfeng Du
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
- Department of Medical ImagingShanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuanShanxi Province030001China
| |
Collapse
|
8
|
Zhang Z, Zhang S, Zhang F, Zhang Q, Wei H, Xiu R, Zhao Y, Sui M. Clinical Indicators of Hepatotoxicity in Newly Diagnosed Acute Promyelocytic Leukemia Patients Undergoing Arsenic Trioxide Treatment. Biol Trace Elem Res 2024; 202:122-132. [PMID: 37097388 PMCID: PMC10764564 DOI: 10.1007/s12011-023-03676-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Arsenic trioxide (ATO)-induced hepatotoxicity is often observed in acute promyelocytic leukemia (APL) patients and decreases therapeutic effect of ATO. Thus, concerns over hepatotoxicity have been raised. The aim of this study was to explore some noninvasive clinical indicators that can be used to guide the individualized application of ATO in the future. APL patients treated with ATO were identified retrospectively via electronic health records at our hospital from August 2014 through August 2019. APL patients without hepatotoxicity were selected as controls. The association between putative risk factors and ATO-induced hepatotoxicity was estimated with ORs and 95% CIs, which were calculated using the chi-square test. The subsequent multivariate analysis was performed using logistic regression analysis. In total, 58.04% of patients experienced ATO-induced hepatotoxicity during the first week. Elevated hemoglobin (OR 8.653, 95% CI, 1.339-55.921), administration of nonprophylactic hepatoprotective agents (OR 36.455, 95% CI, 7.409-179.364), non-single-agent ATO to combat leukocytosis (OR 20.108, 95% CI, 1.357-297.893) and decreased fibrinogen (OR 3.496, 95% CI, 1.127-10.846) were found to be statistically significant risk factors for ATO-induced hepatotoxicity. The area under the ROC curve values were 0.846 for "overall ATO-induced hepatotoxicity" and 0.819 for "early ATO-induced hepatotoxicity." The results revealed that hemoglobin ≥ 80 g/L, nonprophylactic hepatoprotective agents, and non-single-agent ATO and fibrinogen < 1 g/L are risk factors for ATO-induced hepatotoxicity in newly diagnosed APL patients. These findings can enhance the clinical diagnosis of hepatotoxicity. Prospective studies should be performed in the future to validate these findings.
Collapse
Affiliation(s)
- Zhuo Zhang
- Central Laboratory, the First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
- Department of Hematology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Shunji Zhang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fan Zhang
- Central Laboratory, the First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China
| | - Qian Zhang
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Wei
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruolin Xiu
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanhong Zhao
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Meijuan Sui
- Central Laboratory, the First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang, China.
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
9
|
Najafi A, Asadi E, Benson JD. Comparative effects of a calcium chelator (BAPTA-AM) and melatonin on cryopreservation-induced oxidative stress and damage in ovarian tissue. Sci Rep 2023; 13:22911. [PMID: 38129642 PMCID: PMC10739950 DOI: 10.1038/s41598-023-49892-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Oncology treatments cause infertility, and ovarian tissue cryopreservation and transplantation (OTCT) is the only option for fertility preservation in prepubertal girls with cancer. However, OTCT is associated with massive follicle loss. Here, we aimed to determine the effect of supplementation of slow freezing and vitrification media with BAPTA-AM and melatonin alone and in combination on ovarian tissue viability, reactive oxygen species (ROS) levels, total antioxidant capacity (TAC), and follicular morphology and viability. Our results indicated that BAPTA-AM and melatonin can significantly improve ovarian tissue viability and the TAC/ROS ratio and reduce ROS generation in frozen-thawed ovarian tissues in slow freezing and vitrification procedures. BAPTA-AM was also found to be less effective on TAC compared to melatonin in vitrified ovarian tissue. While supplementation of slow freezing and vitrification media with BAPTA-AM and/or melatonin could increase the percentage of morphologically intact follicles in cryopreserved ovarian tissues, the differences were not significant. In conclusion, supplementation of cryopreservation media with BAPTA-AM or melatonin improved the outcome of ovarian tissue cryopreservation in both vitrification and slow freezing methods. Our data provide some insight into the importance of modulating redox balance and intracellular Ca2+ levels during ovarian tissue cryopreservation to optimize the current cryopreservation methods.
Collapse
Affiliation(s)
- Atefeh Najafi
- Department of Biology, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - Ebrahim Asadi
- Department of Biology, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - James D Benson
- Department of Biology, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada.
| |
Collapse
|
10
|
Sun X, Zhang W, Shi X, Wang Y, Zhang Y, Liu X, Xu S, Zhang J. Selenium deficiency caused hepatitis in chickens via the miR-138-5p/SelM/ROS/Ca 2+ overload pathway induced by hepatocyte necroptosis. Food Funct 2023; 14:9226-9242. [PMID: 37743830 DOI: 10.1039/d3fo00683b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Selenoprotein M (SelM), a key thioredoxin like enzyme in the endoplasmic reticulum (ER), is closely related to hepatocyte degeneration. However, the role of miR-138-5p/SelM and necroptosis in chicken SelM-deficient hepatitis and the specific biological mechanism of liver inflammation caused by SelM deficiency have not been elucidated. We established an in vivo chicken liver Se deficiency model by feeding a low-Se diet. The miR-138-5p knockdown and overexpression models and SelM knockdown models were established in LMH cells for an in vitro study. Transmission electron microscopy, H&E staining, Fluo4-AM/ER staining, and flow cytometry were used to detect the morphological changes in chicken liver tissue and the expression changes of necroptosis and inflammation in chicken liver cells. We observed that Se deficiency resulted in liver inflammation, up-regulation of miR-138-5p expression and down-regulation of SelM expression in chickens. Oxidative stress, Ca2+ overload, energy metabolism disorder and necroptosis occurred in chicken liver tissue. Importantly, ROS and the Ca2+ inhibitor could effectively alleviate the energy metabolism disorder, necroptosis and inflammatory cytokine secretion caused by miR-138-5p overexpression and SelM knockdown in LMH cells. In conclusion, selenium deficiency causes hepatitis by upregulating miR-138-5p targeting SelM. Our research findings enrich our knowledge about the biological functions of SelM and provide a theoretical basis for the lack of SelM leading to liver inflammation in chickens.
Collapse
Affiliation(s)
- Xinyue Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Wenyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Yuqi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Yilei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Xiaojing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Jiuli Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
- Heilongjiang Polytechnic, Harbin 150030, P. R. China.
| |
Collapse
|
11
|
Li Z, Fan X, Fan J, Zhang W, Liu J, Liu B, Zhang H. Delivering drugs to tubular cells and organelles: the application of nanodrugs in acute kidney injury. Nanomedicine (Lond) 2023; 18:1477-1493. [PMID: 37721160 DOI: 10.2217/nnm-2023-0200] [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] [Indexed: 09/19/2023] Open
Abstract
Acute kidney injury (AKI) is a common clinical syndrome with limited treatment options and high mortality rates. Proximal tubular epithelial cells (PTECs) play a key role in AKI progression. Subcellular dysfunctions, including mitochondrial, nuclear, endoplasmic reticulum and lysosomal dysfunctions, are extensively studied in PTECs. These studies have led to the development of potential therapeutic drugs. However, clinical development of those drugs faces challenges such as low solubility, short circulation time and severe systemic side effects. Nanotechnology provides a promising solution by improving drug properties through nanocrystallization and enabling targeted delivery to specific sites. This review summarizes advancements and limitations of nanoparticle-based drug-delivery systems in targeting PTECs and subcellular organelles, particularly mitochondria, for AKI treatment.
Collapse
Affiliation(s)
- Zhi Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China
| | - Xiao Fan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China
| | - Jun Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
- Department of Physiology & Pathophysiology, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, 410013, China
| |
Collapse
|
12
|
Oh BC. Phosphoinositides and intracellular calcium signaling: novel insights into phosphoinositides and calcium coupling as negative regulators of cellular signaling. Exp Mol Med 2023; 55:1702-1712. [PMID: 37524877 PMCID: PMC10474053 DOI: 10.1038/s12276-023-01067-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 08/02/2023] Open
Abstract
Intracellular calcium (Ca2+) and phosphoinositides (PIPs) are crucial for regulating cellular activities such as metabolism and cell survival. Cells maintain precise intracellular Ca2+ and PIP levels via the actions of a complex system of Ca2+ channels, transporters, Ca2+ ATPases, and signaling effectors, including specific lipid kinases, phosphatases, and phospholipases. Recent research has shed light on the complex interplay between Ca2+ and PIP signaling, suggesting that elevated intracellular Ca2+ levels negatively regulate PIP signaling by inhibiting the membrane localization of PIP-binding proteins carrying specific domains, such as the pleckstrin homology (PH) and Ca2+-independent C2 domains. This dysregulation is often associated with cancer and metabolic diseases. PIPs recruit various proteins with PH domains to the plasma membrane in response to growth hormones, which activate signaling pathways regulating metabolism, cell survival, and growth. However, abnormal PIP signaling in cancer cells triggers consistent membrane localization and activation of PIP-binding proteins. In the context of obesity, an excessive intracellular Ca2+ level prevents the membrane localization of the PIP-binding proteins AKT, IRS1, and PLCδ via Ca2+-PIPs, contributing to insulin resistance and other metabolic diseases. Furthermore, an excessive intracellular Ca2+ level can cause functional defects in subcellular organelles such as the endoplasmic reticulum (ER), lysosomes, and mitochondria, causing metabolic diseases. This review explores how intracellular Ca2+ overload negatively regulates the membrane localization of PIP-binding proteins.
Collapse
Affiliation(s)
- Byung-Chul Oh
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon College of Medicine, Incheon, 21999, Republic of Korea.
| |
Collapse
|
13
|
Yan J, Zhang J, Wang Y, Liu H, Sun X, Li A, Cui P, Yu L, Yan X, He Z. Rapidly Inhibiting the Inflammatory Cytokine Storms and Restoring Cellular Homeostasis to Alleviate Sepsis by Blocking Pyroptosis and Mitochondrial Apoptosis Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207448. [PMID: 36932048 PMCID: PMC10190643 DOI: 10.1002/advs.202207448] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/10/2023] [Indexed: 05/18/2023]
Abstract
Pyroptosis, systemic inflammation, and mitochondrial apoptosis are the three primary contributors to sepsis's multiple organ failure, the ultimate cause of high clinical mortality. Currently, the drugs under development only target a single pathogenesis, which is obviously insufficient. In this study, an acid-responsive hollow mesoporous polydopamine (HMPDA) nanocarrier that is highly capable of carrying both the hydrophilic drug NAD+ and the hydrophobic drug BAPTA-AM, with its outer layer being sealed by the inflammatory targeting peptide PEG-LSA, is developed. Once targeted to the region of inflammation, HMPDA begins depolymerization, releasing the drugs NAD+ and BAPTA-AM. Depletion of polydopamine on excessive reactive oxygen species production, promotion of ATP production and anti-inflammation by NAD+ replenishment, and chelation of BAPTA (generated by BA-AM hydrolysis) on overloaded Ca2+ can comprehensively block the three stages of sepsis, i.e., precisely inhibit the activation of pyroptosis pathway (NF-κB-NLRP3-ASC-Casp-1), inflammation pathway (IL-1β, IL-6, and TNF-α), and mitochondrial apoptosis pathway (Bcl-2/Bax-Cyt-C-Casp-9-Casp-3), thereby restoring intracellular homeostasis, saving the cells in a state of "critical survival," further reducing LPS-induced systemic inflammation, finally restoring the organ functions. In conclusion, the synthesis of this agent provides a simple and effective synergistic drug delivery nanosystem, which demonstrates significant therapeutic potential in a model of LPS-induced sepsis.
Collapse
Affiliation(s)
- Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Jingwen Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Hong Liu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Xueping Sun
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Aixin Li
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Pengfei Cui
- College of Marine Life SciencesOcean University of ChinaQingdao266003China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Xuefeng Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systemand Key Laboratory of Marine Chemistry Theory and TechnologyMinistry of EducationOcean University of ChinaQingdao266100China
- College of Chemistry and Chemical EngineeringOcean University of ChinaQingdao266100China
| |
Collapse
|
14
|
Yan J, Wang Y, Zhang J, Liu X, Yu L, He Z. Rapidly Blocking the Calcium Overload/ROS Production Feedback Loop to Alleviate Acute Kidney Injury via Microenvironment-Responsive BAPTA-AM/BAC Co-Delivery Nanosystem. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206936. [PMID: 36719986 DOI: 10.1002/smll.202206936] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/14/2023] [Indexed: 06/18/2023]
Abstract
Calcium overload and ROS overproduction, two major triggers of acute kidney injury (AKI), are self-amplifying and mutually reinforcing, forming a complicated cascading feedback loop that induces kidney cell "suicide" and ultimately renal failure. There are currently no clinically effective drugs for the treatment of AKI, excluding adjuvant therapy. In this study, a porous silicon-based nanocarrier rich in disulfide bond skeleton (<50 nm) is developed that enables efficient co-loading of the hydrophilic drug borane amino complex and the hydrophobic drug BAPTA-AM, with its outer layer sealed by the renal tubule-targeting peptide PEG-LTH. Once targeted to the kidney injured site, the nanocarrier structure collapses in the high glutathione environment of the early stage of AKI, releasing the drugs. Under the action of the slightly acidic inflammatory environment and intracellular esterase, the released drugs produce hydrogen and BAPTA, which can rapidly eliminate the excess ROS and overloaded Ca2+ , blocking endoplasmic reticulum/mitochondrial apoptosis pathway (ATF4-CHOP-Bax axis, Casp-12-Casp-3 axis, Cyt-C-Casp-3 axis) and inflammatory pathway (TNF-α-NF-κB axis) from the source, thus rescuing the renal cells in the "critical survival" state and further restoring the kidney function. Overall, this nanoparticle shows substantial clinical promise as a potential therapeutic strategy for I/R injury-related diseases.
Collapse
Affiliation(s)
- Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| | - Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| | - Jingwen Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| | - Xiaohu Liu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, and Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao, 266100, P. R. China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, P. R. China
| |
Collapse
|
15
|
Wang Y, Pu M, Yan J, Zhang J, Wei H, Yu L, Yan X, He Z. 1,2-Bis(2-aminophenoxy)ethane- N, N, N', N'-tetraacetic Acid Acetoxymethyl Ester Loaded Reactive Oxygen Species Responsive Hyaluronic Acid-Bilirubin Nanoparticles for Acute Kidney Injury Therapy via Alleviating Calcium Overload Mediated Endoplasmic Reticulum Stress. ACS NANO 2023; 17:472-491. [PMID: 36574627 DOI: 10.1021/acsnano.2c08982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Calcium overload is one of the early determinants of the core cellular events that contribute to the pathogenesis of acute kidney injury (AKI), which include oxidative stress, ATP depletion, calcium overload, and inflammatory response with self-amplifying and interactive feedback loops that ultimately lead to cellular injury and renal failure. Excluding adjuvant therapy, there are currently no approved pharmacotherapies for the treatment of AKI. Using an adipic dihydride linker, we modified the hyaluronic acid polymer chain with a potent antioxidant, bilirubin, to produce an amphiphilic conjugate. Subsequently, we developed a kidney-targeted and reactive oxygen species (ROS)-responsive drug delivery system based on the flash nanocomplexation method to deliver a well-known intracellular calcium chelator, 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM, BA), with the goal of rescuing renal cell damage via rapidly scavenging of intracellularly overloaded Ca2+. In the ischemia-reperfusion (I/R) induced AKI rat model, a single dose of as-prepared formulation (BA 100 μg·kg-1) 6 h post-reperfusion significantly reduced renal function indicators by more than 60% within 12 h, significantly alleviated tissular pathological changes, ameliorated tissular oxidative damage, significantly inhibited apoptosis of renal tubular cells and the expression of renal tubular marker kidney injury molecule 1, etc., thus greatly reducing the risk of kidney failure. Mechanistically, the treatment with BA-loaded NPs significantly inhibited the activation of the ER stress cascade response (IRE1-TRAF2-JNK, ATF4-CHOP, and ATF6 axis) and regulated the downstream apoptosis-related pathway while also reducing the inflammatory response. The BA-loaded NPs hold great promise as a potential therapy for I/R injury-related diseases.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Minju Pu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Jingwen Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Huichao Wei
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Xuefeng Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| |
Collapse
|
16
|
Shi M, Zhou Z, Zhou Z, Shen L, Shen J, Zhou G, Zhu R. Identification of key genes and infiltrating immune cells among acetaminophen-induced acute liver failure and HBV-associated acute liver failure. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:775. [PMID: 35965803 PMCID: PMC9372688 DOI: 10.21037/atm-22-2742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Acute liver failure (ALF) is a life-threatening complication that is relatively uncommon. ALF causes severe hepatocyte damage and necrosis, which can lead to liver dysfunction and even multi-organ failure. A growing body of evidence suggests that immune cell infiltration and some abnormally expressed genes are associated with ALF development. However, in ALF, they have yet to be thoroughly investigated. METHODS The Gene Expression Omnibus (GEO) database was used to obtain microarray datasets such as GSE74000, GSE120652, GSE38941, and GSE14668, which were then examined via GEO2R to determine differentially expressed genes (DEGs) associated with ALF. Metascape was employed to annotate the underlined genes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The mechanism of IGF1 in 2 different kinds of ALF including acetaminophen-induced ALF and hepatitis B virus (HBV)-induced ALF was studied using gene set enrichment analysis (GSEA). Next, immune cell infiltration was investigated and differentiated in ALF using CIBERSORT. RESULTS Six genes (HAO2, IGF1, PLA2G7, SC5D, GNE, SLC1A1) were found to be abnormally expressed in the 2 distinct types of ALF i.e., acetaminophen-induced ALF and HBV-induced ALF. IGF1 was identified as a hub gene in ALF and was found to be associated with several developmental cascades including immune responses, inflammatory responses, and intracellular calcium homeostasis. Additionally, the number of CD4 naive T cells, CD8 T cells, and follicular helper T cells was increased in acetaminophen-induced ALF, whereas the number of activated NK cells, resting NK cells, and plasma cells was increased in HBV-induced ALF. CONCLUSIONS The present study determined a potential molecular target, namely IGF1, in acetaminophen-induced ALF and HBV-induced ALF, which may provide novel insights into the pathophysiology and management of ALF. Concurrently, the putative immunological pathways have been found.
Collapse
Affiliation(s)
- Min Shi
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Zhuyi Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Zhongxia Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong, China
| | - Lijuan Shen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Jianbo Shen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Renfei Zhu
- Department of Hepatobiliary Surgery, Affiliated Nantong Third Hospital of Nantong University, Nantong, China
| |
Collapse
|
17
|
Yin Q, Zang G, Li N, Sun C, Du R. Agonist-induced Piezo1 activation promote mitochondrial-dependent apoptosis in vascular smooth muscle cells. BMC Cardiovasc Disord 2022; 22:287. [PMID: 35751027 PMCID: PMC9233385 DOI: 10.1186/s12872-022-02726-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/15/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Mechanical damage plays an essential role in the progression of atherosclerosis. Piezo1 is a new mechanically sensitive ion channel. The present study investigated the vascular smooth muscle cells (VSMCs) apoptosis induced by Piezo1 activation and explored its underlying mechanism. METHODS We evaluated cell viability and apoptosis rate with cell counting kit-8 (CCK-8) and Annexin V-FITC/PI flow cytometry assay, respectively. And then Western blot was performed to measure the relative protein. Reactive oxygen species (ROS) and intracellular Ca2+ were assessed via fluorescence microscope, and the mitochondrial transmembrane potential was monitored by JC-10 staining. RESULTS Our in vitro study revealed that mice in the ApoE-/- group compared with control mice showed higher Piezo1 expression(P < 0.05). Besides, Yoda1, a Piezo1 agonist, triggered Ca2+ overload, mitochondrial damage, accumulation of ROS, and VSMCs apoptosis in a dose-depend manner. Furthermore, BAPT-AM (an intracellular Ca2+ chelator) and NAC (an antioxidant) suppressed the mitochondrial damage and attenuated the VSMCs apoptosis. CONCLUSION Our study suggested that Piezo1 induced VSMCs apoptosis because of Ca2+ overload, excessive ROS generation, and mitochondrial dysfunction, which indicated that Piezo1 has potential value in treating vascular diseases.
Collapse
Affiliation(s)
- Qing Yin
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China.,School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Guangyao Zang
- School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Nannan Li
- School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Chenchen Sun
- School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Rongzeng Du
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China.
| |
Collapse
|
18
|
CHEN W, LI K, ZHU S, LUO X, WANG Y, LIU Z, FANG Y, XIA Z. Glutathione ameliorates Hypoxia/Reoxygenation (H/R) induced hepatocyte oxidative damage via regulating HO-1 signaling. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.61221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Wuye CHEN
- First People Hospital of Huizhou, China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Park S, Yun BH, Bae H, Lim W, Song G. Reproductive toxicity of folpet through deregulation of calcium homeostasis in porcine trophectoderm and luminal epithelial cells during early pregnancy. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 179:104974. [PMID: 34802524 DOI: 10.1016/j.pestbp.2021.104974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/22/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
Folpet, a fungicide, is utilized even in cosmetics and pharmaceuticals. The LD50 of folpet in mammals, birds, and fish is relatively high. Recently, several negative effects of folpet on the respiratory system and cornea have been reported. However, there is no study on the negative effects of folpet on maternal-fetus interactions. In the present study, we used porcine trophectoderm (pTr) cells and porcine luminal epithelial (pLE) cells to investigate the toxic effects of folpet during implantation. Folpet treatment decreased cell proliferation and promoted apoptosis with cell cycle arrest. In addition, the ERK, JNK, and AKT signal pathways were activated by folpet treatment. Folpet treatment induced calcium overload in pTr and pLE cells mediating antimigratory and antiadhesive effects in both cell lines. Co-treatment with calcium chelates decreased the anti-implantation effect of folpet. Overall, our results demonstrated potential reproductive toxicity of folpet in pig.
Collapse
Affiliation(s)
- Sunwoo Park
- Department of Plant and Biomaterials Science, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Bo Hyun Yun
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hyocheol Bae
- Department of Oriental Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul 02707, Republic of Korea..
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
20
|
Csomos A, Kontra B, Jancsó A, Galbács G, Deme R, Kele Z, Rózsa BJ, Kovács E, Mucsi Z. A Comprehensive Study of the Ca
2+
Ion Binding of Fluorescently Labelled BAPTA Analogues. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Attila Csomos
- Department of Chemistry Femtonics Ltd. 1094 Budapest Hungary
| | - Bence Kontra
- Department of Chemistry Femtonics Ltd. 1094 Budapest Hungary
| | - Attila Jancsó
- Department of Inorganic and Analytical Chemistry University of Szeged Szeged 6720 Hungary
| | - Gábor Galbács
- Department of Inorganic and Analytical Chemistry University of Szeged Szeged 6720 Hungary
| | - Ruth Deme
- Department of Organic Chemistry Semmelweis University Budapest H-1092 Hungary
| | - Zoltán Kele
- Institute of Medical Chemistry, University of Szeged Szeged H-6720 Hungary
| | - Balázs József Rózsa
- Two-Photon Measurement Technology Research Group The Faculty of Information Technology Pázmány Péter Catholic University Budapest 1083 Hungary
- Laboratory of 3D Functional Imaging of Neuronal Networks and Dendritic Integration Institute of Experimental Medicine Budapest 1083 Hungary
| | - Ervin Kovács
- Department of Chemistry Femtonics Ltd. 1094 Budapest Hungary
- Polymer Chemistry Research Group Research Centre for Natural Sciences Budapest 1117 Hungary
| | - Zoltán Mucsi
- Department of Chemistry Femtonics Ltd. 1094 Budapest Hungary
- Faculty of Materials Science and Engineering University of Miskolc Miskolc 3515 Hungary
| |
Collapse
|
21
|
Ye L, Zeng Q, Ling M, Ma R, Chen H, Lin F, Li Z, Pan L. Inhibition of IP3R/Ca2+ Dysregulation Protects Mice From Ventilator-Induced Lung Injury via Endoplasmic Reticulum and Mitochondrial Pathways. Front Immunol 2021; 12:729094. [PMID: 34603302 PMCID: PMC8479188 DOI: 10.3389/fimmu.2021.729094] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Rationale Disruption of intracellular calcium (Ca2+) homeostasis is implicated in inflammatory responses. Here we investigated endoplasmic reticulum (ER) Ca2+ efflux through the Inositol 1,4,5-trisphosphate receptor (IP3R) as a potential mechanism of inflammatory pathophysiology in a ventilator-induced lung injury (VILI) mouse model. Methods C57BL/6 mice were exposed to mechanical ventilation using high tidal volume (HTV). Mice were pretreated with the IP3R agonist carbachol, IP3R inhibitor 2-aminoethoxydiphenyl borate (2-APB) or the Ca2+ chelator BAPTA-AM. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected to measure Ca2+ concentrations, inflammatory responses and mRNA/protein expression associated with ER stress, NLRP3 inflammasome activation and inflammation. Analyses were conducted in concert with cultured murine lung cell lines. Results Lungs from mice subjected to HTV displayed upregulated IP3R expression in ER and mitochondrial-associated-membranes (MAMs), with enhanced formation of MAMs. Moreover, HTV disrupted Ca2+ homeostasis, with increased flux from the ER to the cytoplasm and mitochondria. Administration of carbachol aggravated HTV-induced lung injury and inflammation while pretreatment with 2-APB or BAPTA-AM largely prevented these effects. HTV activated the IRE1α and PERK arms of the ER stress signaling response and induced mitochondrial dysfunction-NLRP3 inflammasome activation in an IP3R-dependent manner. Similarly, disruption of IP3R/Ca2+ in MLE12 and RAW264.7 cells using carbachol lead to inflammatory responses, and stimulated ER stress and mitochondrial dysfunction. Conclusion Increase in IP3R-mediated Ca2+ release is involved in the inflammatory pathophysiology of VILI via ER stress and mitochondrial dysfunction. Antagonizing IP3R/Ca2+ and/or maintaining Ca2+ homeostasis in lung tissue represents a prospective treatment approach for VILI.
Collapse
Affiliation(s)
- Liu Ye
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qi Zeng
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Maoyao Ling
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Riliang Ma
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haishao Chen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhao Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|