1
|
Tarin M, Oryani MA, Javid H, Hashemzadeh A, Karimi-Shahri M. Advancements in chitosan-based nanocomposites with ZIF-8 nanoparticles: multifunctional platforms for wound healing applications. Carbohydr Polym 2025; 362:123656. [PMID: 40409814 DOI: 10.1016/j.carbpol.2025.123656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/25/2025]
Abstract
The integration of chitosan and zeolitic imidazolate framework-8 (ZIF-8) nanoparticles has demonstrated significant potential in enhancing wound healing through their multifunctional capabilities. This review explores recent developments in chitosan-based nanocomposites incorporating ZIF-8 nanoparticles, emphasizing their antibacterial properties, pH-responsive drug release, angiogenesis promotion, and mechanical stability. Applications span hydrogel scaffolds, electrospun nanofibers, and sprayable membranes, all tailored for addressing challenges such as bacterial resistance, delayed tissue regeneration, and chronic wound management. Key findings highlight the synergistic benefits of ZIF-8's bioactivity with chitosan's biocompatibility, yielding innovative therapeutic strategies for complex wound healing scenarios. The discussed advancements not only underline their clinical relevance but also set a foundation for future explorations in regenerative medicine.
Collapse
Affiliation(s)
- Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of medical sciences, Mashhad. Iran.
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Iran.
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
2
|
Zhang X, Wang X, Wang Y, Ma X, Geng Y, Zang S, Ban Z, Jia Y, Gao Y. Norisoboldine Alleviates Isoproterenol-Induced Myocardial Ischemic Injury via the TLR4-MyD88-Dependent NF-κB Activation Pathway and Modulation of L-Type Calcium Channels. Clin Exp Pharmacol Physiol 2025; 52:e70033. [PMID: 40011076 DOI: 10.1111/1440-1681.70033] [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: 10/13/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Norisoboldine (NIB) displays beneficial effects on cardiovascular diseases, although its protective role and underlying mechanisms in myocardial ischemia (MI) injury remain elusive. The aim of this study is to explore the potential cardioprotective mechanism of NIB on MI injury caused by isoproterenol (ISO). We administered NIB to SD rats at 20 and 40 mg/kg daily for 7 days in this study; this was followed by an ISO injection to induce MI injury. Parameters such as electrocardiogram readings, heart rate, serum concentrations of creatine kinase (CK) and creatine kinase-MB (CK-MB), levels of inflammatory markers, some histopathological assessments and oxidative stress markers were evaluated. We conducted Western blot analyses to evaluate protein expression related to apoptosis and the TLR4-MyD88-mediated NF-κB activation pathway. The L-type Ca2+ current (ICa-L) and contraction of isolated ventricular cells from rats were identified using patch-clamp methods and the IonOptix detection system. The treatment with NIB resulted in improvements in heart rate and ST-segment changes, a reduction in CK and CK-MB levels, the restoration of superoxide dismutase, catalase and glutathione levels and a decrease in malondialdehyde accumulation. Furthermore, NIB reduced the expression of inflammatory markers, lowered Ca2+ levels and reactive oxygen species production and improved myocardial tissue morphology. It also countered ISO-induced alterations in apoptosis and the TLR4-MyD88-dependent NF-κB activation pathway. Additionally, NIB considerably attenuated ICa-L and reduced the contractile function of cardiomyocytes. These results suggest that NIB effectively mitigates ISO-induced MI injury through anti-inflammatory, antioxidative, and anti-apoptotic mechanisms, potentially involving the TLR4-MyD88-dependent NF-κB activation pathway and calcium balance.
Collapse
Affiliation(s)
- Xin Zhang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xinliu Wang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuanyuan Wang
- College of Basic Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, China
| | | | - Yunyun Geng
- College of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuxian Zang
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Ziyun Ban
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yugai Jia
- College of Basic Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, China
| | - Yonggang Gao
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
3
|
Ye M, He Y, Xia Y, Zhong Z, Kong X, Zhou Y, Xia W, Wang W, Fan H, Chen L, Wu X, Li Q. Association Between Serum Zinc and Non-Alcoholic Fatty Liver Disease and Advanced Liver Fibrosis: NHANES 2011-2016. Biol Trace Elem Res 2025; 203:1305-1316. [PMID: 38861177 DOI: 10.1007/s12011-024-04261-x] [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: 03/21/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Limited and inconclusive evidence exists regarding the correlation between serum zinc levels and non-alcoholic fatty liver disease (NAFLD) and advanced fibrosis. The objective of this cross-sectional study was to investigate the association between serum zinc concentration and both NAFLD and advanced liver fibrosis among the United States (US) adults. 3398 subjects from National Health and Nutrition Examination Survey (NHANES) 2011-2016 were included. Serum zinc concentration was measured by inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS). NAFLD was diagnosed with Hepatic Steatosis Index (HSI), and advanced fibrosis risk was assessed by NAFLD Fibrosis Score (NFS). Weighted logistic regression and restricted cubic splines (RCS) were used to examine the association between serum zinc concentration and NAFLD and advanced fibrosis. Linear trend tests were conducted by incorporating the median of serum zinc quartiles as a continuous variable in the models. We employed sensitivity analysis and subgroup analysis to enhance the robustness of our results. The results from the RCS regression revealed no evident nonlinear relationship between serum zinc concentration and the presence of NAFLD and advanced fibrosis (p-nonlinear > 0.05). Compared with those in the lowest quartile (Q1) of serum zinc concentrations, the odds ratios (95% confidence intervals) of NAFLD were 1.49 (0.89,2.49) in Q2, 0.99 (0.68,1.45) in Q3, and 2.00 (1.40,2.86) in Q4 (p-trend = 0.002). Similarly, the odds ratios (95% confidence intervals) for advanced fibrosis in Q2-4 compared to Q1 were 0.86 (0.50,1.47), 0.60 (0.26,1.39), and 0.41 (0.21,0.77), respectively (p-trend = 0.006). Subgroup analyses and sensitivity analyses reinforce the same conclusion. The investigation revealed a positive linear relationship between serum zinc concentrations and the probability of developing NAFLD. Conversely, an inverse correlation was observed between serum zinc concentrations and the incidence of advanced liver fibrosis among individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Miaomin Ye
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yin Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Ziyi Zhong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yunting Zhou
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wenqing Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Weiping Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Huan Fan
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Lu Chen
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaohui Wu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
4
|
Li K, Zhou Z, Cao Y. Effects of orally exposed SiO 2 nanoparticles on lipid profiles in gut-liver axis of mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117580. [PMID: 39708451 DOI: 10.1016/j.ecoenv.2024.117580] [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: 03/06/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Recently we proposed the possibility of orally exposed nanoparticles (NPs) to alter metabolite homeostasis by changing metabolism pathways, in addition to intestinal damages, but relatively few studies investigated the changes of metabolite profiles in multi-organs. This study investigated the influences of orally exposed SiO2 NPs on lipid profiles in gut-liver axis. To this end, we treated mice with 16, 160 or 1600 mg/kg bodyweight SiO2 NPs via intragastric route. After 5 days exposure (once a day), we observed that SiO2 NPs induced minimal pathological changes but increased most of the trace elements. Furthermore, lipid staining was gradually decreased in intestines and livers with the increase of NP levels. Consistently, lipidomics results showed that most of the lipid classes in mouse intestines and livers were decreased following SiO2 NP administration. We further identified the lipid classes significantly decreased in both intestines and livers, such as phosphatidylserine (PS), phosphatidylglycerol (PG), and phosphatidylethanolamine (PE). Only a few lipid classes, such as anandamide, showed opposite trends in these organs. For metabolism pathway, SiO2 NPs suppressed autophagy, showing as a significant decrease of microtubule-associated protein 1 A/1B light chain 3 (LC3) and adipose triglyceride lipase (Atgl), accompanying with an accumulation of P62, in both intestines and livers. However, lysosomal-associated membrane protein 2 (Lamp2) showed different trend, that it was significantly increased in intestines but decreased in livers. Combined, our results indicated that intragastric administration of SiO2 NPs altered trace element balance and lipid profiles, accompanying with a change of autophagic lipolysis proteins, in mouse gut-liver axis.
Collapse
Affiliation(s)
- Kuanhang Li
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zhengzheng Zhou
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
5
|
Li T, Yu C. Metal-Dependent Cell Death in Renal Fibrosis: Now and in the Future. Int J Mol Sci 2024; 25:13279. [PMID: 39769044 PMCID: PMC11678559 DOI: 10.3390/ijms252413279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Renal fibrosis is a common final pathway underlying nearly almost all progressive kidney diseases. Metal ions are essential trace elements in organisms and are involved in important physiological activities. However, aberrations in intracellular metal ion metabolism may disrupt homeostasis, causing cell death and increasing susceptibility to various diseases. Accumulating evidence suggests a complex association between metal-dependent cell death and renal fibrosis. In this article, we provide a comprehensive overview of the specific molecular mechanisms of metal-dependent cell death and their crosstalk, up-to-date evidence supporting their role in renal fibrosis, therapeutic targeting strategies, and research needs, aiming to offer a rationale for future clinical treatment of renal fibrosis.
Collapse
Affiliation(s)
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
6
|
Zhang P, Zhong D, Yu Y, Wang L, Li Y, Liang Y, Shi Y, Duan M, Li B, Niu H, Xu Y. Integration of STING activation and COX-2 inhibition via steric-hindrance effect tuned nanoreactors for cancer chemoimmunotherapy. Biomaterials 2024; 311:122695. [PMID: 38954960 DOI: 10.1016/j.biomaterials.2024.122695] [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: 04/17/2024] [Revised: 06/01/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Integrating immunotherapy with nanomaterials-based chemotherapy presents a promising avenue for amplifying antitumor outcomes. Nevertheless, the suppressive tumor immune microenvironment (TIME) and the upregulation of cyclooxygenase-2 (COX-2) induced by chemotherapy can hinder the efficacy of the chemoimmunotherapy. This study presents a TIME-reshaping strategy by developing a steric-hindrance effect tuned zinc-based metal-organic framework (MOF), designated as CZFNPs. This nanoreactor is engineered by in situ loading of the COX-2 inhibitor, C-phycocyanin (CPC), into the framework building blocks, while simultaneously weakening the stability of the MOF. Consequently, CZFNPs achieve rapid pH-responsive release of zinc ions (Zn2+) and CPC upon specific transport to tumor cells overexpressing folate receptors. Accordingly, Zn2+ can induce reactive oxygen species (ROS)-mediated cytotoxicity therapy while synchronize with mitochondrial DNA (mtDNA) release, which stimulates mtDNA/cGAS-STING pathway-mediated innate immunity. The CPC suppresses the chemotherapy-induced overexpression of COX-2, thus cooperatively reprogramming the suppressive TIME and boosting the antitumor immune response. In xenograft tumor models, the CZFNPs system effectively modulates STING and COX-2 expression, converting "cold" tumors into "hot" tumors, thereby resulting in ≈ 4-fold tumor regression relative to ZIF-8 treatment alone. This approach offers a potent strategy for enhancing the efficacy of combined nanomaterial-based chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Di Zhong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Yongbo Yu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lupeng Wang
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Yifan Li
- Department of Breast Center of the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong Province, China
| | - Ye Liang
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yanfeng Shi
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Meilin Duan
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, Shandong Province, China.
| | - Haitao Niu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Yuanhong Xu
- Department of Urology, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
7
|
Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: focus on mitochondrial dysfunction. Angiogenesis 2024; 27:623-639. [PMID: 39060773 PMCID: PMC11564294 DOI: 10.1007/s10456-024-09938-4] [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: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
As a vital component of blood vessels, endothelial cells play a key role in maintaining overall physiological function by residing between circulating blood and semi-solid tissue. Various stress stimuli can induce endothelial injury, leading to the onset of corresponding diseases in the body. In recent years, the importance of mitochondria in vascular endothelial injury has become increasingly apparent. Mitochondria, as the primary site of cellular aerobic respiration and the organelle for "energy information transfer," can detect endothelial cell damage by integrating and receiving various external stress signals. The generation of reactive oxygen species (ROS) and mitochondrial dysfunction often determine the evolution of endothelial cell injury towards necrosis or apoptosis. Therefore, mitochondria are closely associated with endothelial cell function, helping to determine the progression of clinical diseases. This article comprehensively reviews the interconnection and pathogenesis of mitochondrial-induced vascular endothelial cell injury in cardiovascular diseases, renal diseases, pulmonary-related diseases, cerebrovascular diseases, and microvascular diseases associated with diabetes. Corresponding therapeutic approaches are also provided. Additionally, strategies for using clinical drugs to treat vascular endothelial injury-based diseases are discussed, aiming to offer new insights and treatment options for the clinical diagnosis of related vascular injuries.
Collapse
Affiliation(s)
- Boxian Pang
- Beijing University of Chinese Medicine, Beijing, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | | | - Tieliang Pang
- Beijing Anding hospital, Capital Medical University, Beijing, China
| | - Xinyao Sun
- Beijing University of Chinese Medicine, Beijing, China
| | - Xin Liu
- Bioscience Department, University of Nottingham, Nottingham, UK
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiagge, Xicheng District, Beijing, China.
| |
Collapse
|
8
|
Zhang M, Zhou H, Liu L, Song W. Biological effect of U(VI) exposure on lung epithelial BEAS-2B cells. CHEMOSPHERE 2024; 366:143451. [PMID: 39362378 DOI: 10.1016/j.chemosphere.2024.143451] [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: 08/02/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024]
Abstract
In this study, the biological effects of U(VI) exposure on lung epithelial cells were investigated by MTT assay, immunofluorescence, flow cytometry, and Western blotting. U(VI)-induced stress triggers oxidative stress in cells, activates MAPK signaling pathways, and promotes inflammation. Additionally, U(VI) causes damage to the cell membrane structure and severe DNA injury, impacting the accuracy of transcription and translation. The results demonstrate that U(VI) exposure significantly inhibits cell proliferation and migration. This is attributed to the disruption of the PI3K/AKT/GSK-3β/β-catenin signaling pathway and the reduction in CyclinD1 expression, leading to a delayed cell cycle, decreased growth rate, mitochondrial damage, and reduced energy metabolism. This study provides a comprehensive understanding of the molecular mechanisms underlying uranium-induced cellular toxicity in lung epithelial cells.
Collapse
Affiliation(s)
- Mingxia Zhang
- College of Life Sciences, Anhui Normal University, Wuhu 241000, China; Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Han Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lei Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Wencheng Song
- College of Life Sciences, Anhui Normal University, Wuhu 241000, China; Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China; Collaborative Innovation Center of Radiation Medicine, Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
9
|
Dai Y, Duan S, Wang R, He P, Zhang Z, Li M, Shen Z, Chen Y, Zhao Y, Yang H, Li X, Zhang R, Sun J. Associations between multiple urinary metals and metabolic syndrome: Exploring the mediating role of liver function in Chinese community-dwelling elderly. J Trace Elem Med Biol 2024; 85:127472. [PMID: 38823271 DOI: 10.1016/j.jtemb.2024.127472] [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: 03/10/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Multiple metals exposure has been revealed to be related to metabolic syndrome (MetS). However, the associations and interactions between multiple metals exposure and MetS are remains controversial, and the potential mechanism of the above-mentioned is still unclear. METHODS The associations between urinary metals and the MetS were analyzed by multivariable logistic regression model and restricted cubic spline (RCS). Bayesian kernel machine regression (BKMR) model and quantile-based g-computation (qgcomp) were applied to explore the mixed exposure and interaction effect of metals. Mediation analysis was used to explore the role of liver function. RESULTS In the single metal model, multiple metals were significantly associated with MetS. RCS analysis further verified the associations between 8 metals and MetS. BKMR model and qgcomp showed that zinc (Zn), iron (Fe), and tellurium (Te) were the main factors affecting the overall effect. In addition, mediation analysis indicated that serum alanine aminotransferase (ALT) mediated 21.54% and 13.29% in the associations of vanadium (V) and Zn with the risk of MetS, respectively. CONCLUSIONS Elevated urinary concentration of Zn, V, Te, copper (Cu), molybdenum (Mo), and thallium (Tl) were related to the increased risk of MetS. Conversely, Fe and selenium (Se) may be protective factors for MetS in mixed exposure. Liver function may play a key role in the association of V and Zn exposure with MetS.
Collapse
Affiliation(s)
- Yuqing Dai
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Siyu Duan
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Rui Wang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Pei He
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Zhongyuan Zhang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Meiyan Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Zhuoheng Shen
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Yue Chen
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Yi Zhao
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Huifang Yang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China
| | - Xiaoyu Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China.
| | - Rui Zhang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China.
| | - Jian Sun
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia 750004, PR China.
| |
Collapse
|
10
|
Zhu H, Yu Y, Li Y, Chang S, Liu Y. Puerarin ameliorates high glucose-induced MIN6 cell injury by activating PINK1/Parkin-mediated mitochondrial autophagy. Heliyon 2024; 10:e36176. [PMID: 39224278 PMCID: PMC11367457 DOI: 10.1016/j.heliyon.2024.e36176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The dysfunction of pancreatic β-cells plays a pivotal role in the pathogenesis of type 2 diabetes mellitus (T2DM). Despite numerous studies demonstrating the anti-inflammatory and antioxidant properties of puerarin, the protective effects of puerarin on β-cells remain poorly understood. Hence, this study aimed to explore the effects of puerarin on β-cell dysfunction in a hyperglycemic environment via the PINK/Parkin-mediated mitochondrial autophagy pathway. The alterations in cell viability of MIN6 cells exposed to glucose concentrations of 5 mM, 10 mM, 20 mM, and 30 mM for 24 h, 48 h, and 72 h, respectively, were assessed using the CCK-8 assay to optimize the modeling conditions. Subsequently, cellular insulin secretion was measured using enzyme-linked immunosorbent assay (ELISA), apoptosis rate by flow cytometry, mitochondrial membrane potential alteration by JC-1, cellular ROS production by the DCFH-DA fluorescent probe, and fusion of cellular autophagosomes and lysosomes through adenoviral infection analysis. Furthermore, gene and protein expression levels of the PINK/Parkin-mediated mitochondrial autophagy pathway and mitochondrial apoptosis pathway were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot, respectively. Results indicated a significant decrease in MIN6 cell viability following 48 h of exposure to 30 mM glucose concentration. Puerarin intervention markedly attenuated ROS production, restored mitochondrial membrane potential, induced PINK/Parkin-mediated mitochondrial autophagy, suppressed activation of the mitochondrial apoptotic pathway, mitigated apoptosis, and enhanced insulin secretion in a high glucose (HG) environment. The findings of this investigation contribute to a deeper understanding of the precise mechanism underlying the protective effects of puerarin on β-cells and offer a theoretical foundation for advancing puerarin-based therapeutics aimed at ameliorating T2DM.
Collapse
Affiliation(s)
- Hongyang Zhu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - You Yu
- The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Yuting Li
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shiyao Chang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yuhui Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
11
|
Lai Y, Gao FF, Ge RT, Liu R, Ma S, Liu X. Metal ions overloading and cell death. Cell Biol Toxicol 2024; 40:72. [PMID: 39162885 PMCID: PMC11335907 DOI: 10.1007/s10565-024-09910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Cell death maintains cell morphology and homeostasis during development by removing damaged or obsolete cells. The concentration of metal ions whithin cells is regulated by various intracellular transporters and repositories to maintain dynamic balance. External or internal stimuli might increase the concentration of metal ions, which results in ions overloading. Abnormal accumulation of large amounts of metal ions can lead to disruption of various signaling in the cell, which in turn can produce toxic effects and lead to the occurrence of different types of cell deaths. In order to further study the occurrence and development of metal ions overloading induced cell death, this paper reviewed the regulation of Ca2+, Fe3+, Cu2+ and Zn2+ metal ions, and the internal mechanism of cell death induced by overloading. Furthermore, we found that different metal ions possess a synergistic and competitive relationship in the regulation of cell death. And the enhanced level of oxidative stress was present in all the processes of cell death due to metal ions overloading, which possibly due to the combination of factors. Therefore, this review offers a theoretical foundation for the investigation of the toxic effects of metal ions, and presents innovative insights for targeted regulation and therapeutic intervention. HIGHLIGHTS: • Metal ions overloading disrupts homeostasis, which in turn affects the regulation of cell death. • Metal ions overloading can cause cell death via reactive oxygen species (ROS). • Different metal ions have synergistic and competitive relationships for regulating cell death.
Collapse
Affiliation(s)
- Yun Lai
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Fen Fen Gao
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Ruo Ting Ge
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Rui Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
| | - Xiaodong Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
12
|
Luan M, Feng Z, Zhu W, Xing Y, Ma X, Zhu J, Wang Y, Jia Y. Mechanism of metal ion-induced cell death in gastrointestinal cancer. Biomed Pharmacother 2024; 174:116574. [PMID: 38593706 DOI: 10.1016/j.biopha.2024.116574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024] Open
Abstract
Gastrointestinal (GI) cancer is one of the most severe types of cancer, with a significant impact on human health worldwide. Due to the urgent demand for more effective therapeutic strategies against GI cancers, novel research on metal ions for treating GI cancers has attracted increasing attention. Currently, with accumulating research on the relationship between metal ions and cancer therapy, several metal ions have been discovered to induce cell death. In particular, the three novel modes of cell death, including ferroptosis, cuproptosis, and calcicoptosis, have become focal points of research in the field of cancer. Meanwhile, other metal ions have also been found to trigger cell death through various mechanisms. Accordingly, this review focuses on the mechanisms of metal ion-induced cell death in GI cancers, hoping to provide theoretical support for further GI cancer therapies.
Collapse
Affiliation(s)
- Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Zhaotian Feng
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Jingyu Zhu
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|