101
|
Cunha M, Nardi A, Soares AMVM, Gil AM, Freitas R. Revealing hidden risks: in vitro analysis of PFAS hazards in Mytilus galloprovincialis gills and digestive gland. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136823. [PMID: 39694002 DOI: 10.1016/j.jhazmat.2024.136823] [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: 08/30/2024] [Revised: 11/04/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals known for their persistence and bioaccumulation, leading to widespread environmental contamination. Despite their recognised environmental risks, particularly to aquatic wildlife, including marine invertebrates, detailed impact studies are limited. PFAS can be categorised according to the length of the compound chain, with short-chain PFAS announced as a safer alternative to the more commonly used long-chain PFAS. However, recent evidence suggests that also short-chain PFAS pose significant environmental risks. The present study evaluated the adverse effects of six PFAS compounds-two short-chain (PFHxA, 6:2 FTA) and four long-chain (PFUnDA, PFDoA, PFTriDA, PFTeDA)- on the digestive gland and gills of mussels, Mytilus galloprovincialis, using in vitro assays. The results showed organ-specific responses: the digestive gland was more sensitive to PFHxA, with increased catalase activity and decreased total antioxidant capacity, and cellular damage was observed only at higher concentrations of PFTriDA. Gills were more affected by PFDoA and PFTeDA, with inhibited antioxidant enzyme activity and increased oxidative stress. PFHxA and PFTriDA also showed inhibition of acetylcholinesterase activity. 6:2 FTA had the lowest effects for both organs, while PFHxA was the most harmful. These findings underscore the need for thorough risk assessments of PFAS, considering both chain length and organ-specific effects.
Collapse
Affiliation(s)
- Marta Cunha
- Department of Biology, University of Aveiro, Aveiro 3810-193, Portugal; Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Aveiro 3810-193, Portugal
| | - Alessandro Nardi
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, Ancona, Italy; NBFC, National Biodiversity Future Center, Palermo 90131, Italy
| | - Amadeu M V M Soares
- Department of Biology, University of Aveiro, Aveiro 3810-193, Portugal; Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana M Gil
- CICECO─Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Rosa Freitas
- Department of Biology, University of Aveiro, Aveiro 3810-193, Portugal; Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Aveiro 3810-193, Portugal.
| |
Collapse
|
102
|
Khodade VS, Liu Q, Zhang C, Keceli G, Paolocci N, Toscano JP. Arylsulfonothioates: Thiol-Activated Donors of Hydropersulfides which are Excreted to Maintain Cellular Redox Homeostasis or Retained to Counter Oxidative Stress. J Am Chem Soc 2025; 147:7765-7776. [PMID: 39963866 DOI: 10.1021/jacs.4c17661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Despite their biological significance, the study of hydropersulfides (RSSH) is often limited due to their inherent instability. Here, we introduce arylsulfonothioates as thiol-activated RSSH donors and provide insight into cellular reactive sulfur species homeostasis. These precursors persulfidate physiologically relevant thiols (RSH) to form the corresponding RSSH. Real-time monitoring of hydrogen sulfide (H2S) generation via membrane inlet mass spectrometry (MIMS) was employed to follow RSSH production, revealing that electron-donating aryl substituents marginally slow RSSH release rates, whereas electron-withdrawing substituents slightly accelerate release. Furthermore, arylsulfonothioates with strong electron-withdrawing substituents offer superior protection against doxorubicin (DOX)-induced cardiotoxicity. Experiments using H9c2 cardiomyocytes affirmed the cell-permeability of arylsulfonothioates and their ability to increase intracellular RSSH levels and protein persulfidation levels. Notably, we observe the excretion of RSSH into the extracellular medium. Further investigations revealed the involvement of the cystine/glutamate antiporter SLC7A11, as cotreatment with its inhibitor, sulfasalazine, significantly reduce extracellular RSSH release. H9c2 cells exhibit tolerance to arylsulfonothioate 1g with an electron-withdrawing 4-cyano group at 1 mM; however, inhibition of the cystine antiporter results in a minor decrease in cell viability. Under oxidative stress conditions induced by DOX or hydrogen peroxide (H2O2), cotreatment with 1g diminishes the excretion of RSSH and confers cytoprotection against DOX or H2O2-mediated toxicity. Our findings show adaptive cellular responses to RSSH levels, demonstrating excretion under elevated conditions to maintain redox homeostasis and intracellular retention as a protective response during oxidative stress.
Collapse
Affiliation(s)
- Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Qi Liu
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Chengximeng Zhang
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Gizem Keceli
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Biomedical Sciences, University of Padova, Padova 35131, Italy
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
103
|
Ben Zichri- David S, Shkuri L, Ast T. Pulling back the mitochondria's iron curtain. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:6. [PMID: 40052109 PMCID: PMC11879881 DOI: 10.1038/s44324-024-00045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/09/2024] [Indexed: 03/09/2025]
Abstract
Mitochondrial functionality and cellular iron homeostasis are closely intertwined. Mitochondria are biosynthetic hubs for essential iron cofactors such as iron-sulfur (Fe-S) clusters and heme. These cofactors, in turn, enable key mitochondrial pathways, such as energy and metabolite production. Mishandling of mitochondrial iron is associated with a spectrum of human pathologies ranging from rare genetic disorders to common conditions. Here, we review mitochondrial iron utilization and its intersection with disease.
Collapse
Affiliation(s)
| | - Liraz Shkuri
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| | - Tslil Ast
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| |
Collapse
|
104
|
Mahapatra S, Shivpuje SB, Campbell HC, Wan B, Lomont J, Dong B, Ma S, Mohn KJ, Zhang C. Label-Free Quantification of Apoptosis and Necrosis Using Stimulated Raman Scattering Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.01.641010. [PMID: 40093126 PMCID: PMC11908225 DOI: 10.1101/2025.03.01.641010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Recombinant proteins are critical for modern therapeutics and diagnostics, with Chinese hamster ovary (CHO) cells serving as the primary production platform. However, environmental and chemical stressors in bioreactors often trigger cell death, particularly apoptosis, posing a significant challenge to recombinant protein manufacturing. Rapid, label-free methods to monitor cell death are essential for ensuring better production quality. Stimulated Raman scattering (SRS) microscopy offers a powerful, label-free approach to measure lipid and protein compositions in live cells. We demonstrate that SRS microscopy enables rapid and reagent-free analysis of apoptotic and necrotic transitions. Our results show that apoptotic cells exhibit higher protein concentrations, while necrotic cells show an opposite trend. To enhance analysis, we developed a quantitative single-cell analysis pipeline that extracts chemotypic and phenotypic signatures of apoptosis and necrosis, enabling the identification of subpopulations with varied responses to stressors or treatments. Furthermore, the cell death analysis was successfully generalized to other stressors and cell types. This study highlights SRS microscopy as a robust and non-invasive tool for rapid monitoring of live cell apoptotic and necrotic transitions. Our method and findings hold potential for improving quality control in CHO cell-based biopharmaceutical production and for evaluating cell death in diverse biological contexts.
Collapse
Affiliation(s)
- Shivam Mahapatra
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
| | - Shreya B Shivpuje
- Department of Statistics, Purdue University, 150 N University St, West Lafayette, IN 47907, USA
| | - Helen C Campbell
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
| | | | | | - B Dong
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, 201 S. University St., West Lafayette, IN 47907, USA
| | - Seohee Ma
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, 201 S. University St., West Lafayette, IN 47907, USA
| | - Karsten J Mohn
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
| | - Chi Zhang
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, 201 S. University St., West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, 207 S. Martin Jischke Dr., West Lafayette, IN 47907, USA
| |
Collapse
|
105
|
Zuo X, Bai HJ, Zhao QL, Zhang SH, Zhao X, Feng XZ. 17β-Trenbolone Exposure Enhances Muscle Activity and Exacerbates Parkinson's Disease Progression in Male Mice. Mol Neurobiol 2025; 62:3053-3066. [PMID: 39222261 DOI: 10.1007/s12035-024-04455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) ranks as the second most prevalent neurodegenerative disorder, and while the neuroprotective effects of estrogen are well-documented, the impact of androgens on neurological disorders remains understudied. The consequences of exposure to 17-trenbolone (17-TB), an environmental endocrine disruptor with androgen-like properties, on the mammalian nervous system have received limited attention. Therefore, in this study, we aimed to investigate the biological effects of 17-TB exposure on PD. In our investigation using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, we discovered that 17-TB exposure elevated testosterone hormone levels prevented androgen receptor (AR) reduction, upregulated the expression of muscular dystrophic factors (Atrogin1, MuRF1, Musa1, and Myostatin), improved muscle strength, and enhanced locomotor activity in the open field test. However, it is noteworthy that exposure to 17-TB also led to an upregulation of neuroinflammatory cytokines (NLRP3, IL-6, IL-1α, and IL-1β) in PD mice. Crucially, 17-TB exposure induced downregulation of nigral apoptotic proteins DJ-1 and Bcl-2 while upregulating Bax and Caspase-3 in PD mice. This exacerbated neuronal apoptosis, ultimately intensifying dopaminergic neuronal degeneration and death in the substantia nigra and striatum of PD mice. In conclusion, our findings indicate that while 17-TB mitigates muscle atrophy and enhances motor activity in PD mice, it concurrently exacerbates neuroinflammation, induces neuronal apoptosis, and worsens dopaminergic neuronal death, thereby aggravating the progression of MPTP-induced Parkinsonism. This underscores the importance of considering potential environmental risks in neurodegeneration associated with Parkinson's disease, providing a cautionary tale for our daily exposure to environmental endocrine chemical disruptors.
Collapse
Affiliation(s)
- Xiang Zuo
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Hui-Juan Bai
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qi-Li Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Shu-Hui Zhang
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China.
| | - Xi-Zeng Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
106
|
Li Y, Jiang J, Li J, Liu S, Wang C, Yu Z, Xia Y. Exosome-Derived CDC42 From Hypoxia-Pretreated Neural Stem Cells Inhibits ACSL4-Related Ferroptosis to Alleviate Vascular Injury in Parkinson's Disease Mice Models. J Neurochem 2025; 169:e70027. [PMID: 40035385 DOI: 10.1111/jnc.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that gets exacerbated by vascular injury. Neural stem cell-derived exosomes (NSC-Exos) display effective neuroprotective properties in PD models. Cell division control protein 42 (CDC42) is connected to angiogenesis, but its effects in PD remain undefined. This research aims to reveal the role of CDC42 in PD. First, we applied 1-methyl-4-phenylpyridinium (MPP+) to induce the human cerebral microvascular endothelial cells (HCMECs) model and evaluated cell viability and ferroptosis. Then, we characterized NSC-Exos. Next, to appraise the effect of hypoxia-pretreated NSC-Exos (H-NSC-Exos) on the MPP+-induced cells model, we examined angiogenesis and ferroptosis in HCMECs. Moreover, we constructed the PD mice model using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and tested the behavioral experiments and vascular injury of mice. Furthermore, we examined cellular ferroptosis and angiogenesis after knockdown of CDC42. Additionally, we investigated the interaction of CDC42 with Acyl-CoA synthetase long-chain family member 4 (ACSL4) and detected cellular ferroptosis and angiogenesis after overexpression of ACSL4. We found that H-NSC-Exos reversed the MPP+-induced decrease in HCMECs viability and migration, lowered lipid-reactive oxygen species (lipid-ROS) levels, suppressed ferroptosis, and facilitated angiogenesis. Moreover, H-NSC-Exos attenuated MPTP-induced PD development, vascular injury, and ferroptosis in mice. H-NSC-Exos with the knockdown of CDC42 reduced cell viability and angiogenesis and raised ferroptosis and lipid-ROS levels, which were reversed by ferrostatin-1 and liproxstatin-1. CDC42 interacted with ACSL4. Furthermore, overexpression of ACSL4 aggravated the above effects of H-NSC-Exos in which CDC42 was knocked down. Our study reveals that H-NSC-Exos-derived CDC42 inhibited ACSL4-related ferroptosis to alleviate vascular injury in PD mice models. CDC42 may serve as a potent therapeutic target for PD treatment.
Collapse
Affiliation(s)
- You Li
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Junwen Jiang
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Jiameng Li
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Siliang Liu
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Chuang Wang
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Zhengtao Yu
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| |
Collapse
|
107
|
Jellinger KA. Mild cognitive impairment in amyotrophic lateral sclerosis: current view. J Neural Transm (Vienna) 2025; 132:357-368. [PMID: 39470847 DOI: 10.1007/s00702-024-02850-7] [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: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal multi-system neurodegenerative disorder with no effective treatment or cure. Although primarily characterized by motor degeneration, cognitive dysfunction is an important non-motor symptom that has a negative impact on patient and caregiver burden. Mild cognitive deficits are present in a subgroup of non-demented patients with ALS, often preceding motor symptoms. Detailed neuropsychological assessments reveal deficits in a variety of cognitive domains, including those of verbal fluency and retrieval, language, executive function, attention and verbal memory. Mild cognitive impairment (MCI), a risk factor for developing dementia, affects between 10% and over 50% of ALS patients. Neuroimaging revealed atrophy of frontal and temporal cortices, disordered white matter Integrity, volume reduction in amygdala and thalamus, hypometabolism in the frontal and superior temporal gyrus and anterior insula. Neuronal loss in non-motor brain areas, associated with TDP-43 deposition, one of the morphological hallmarks of ALS, is linked to functional disruption of frontostriatal and frontotemporo-limbic connectivities as markers for cognitive deficits in ALS, the pathogenesis of which is still poorly understood. Early diagnosis by increased cerebrospinal fluid or serum levels of neurofilament light/heavy chain or glial fibrillary acidic protein awaits confirmation for MCI in ALS. These fluid biomarkers and early detection of brain connectivity signatures before structural changes will be helpful not only in establishing early premature diagnosis but also in clarifying the pathophysiological mechanisms of MCI in ALS, which might serve as novel targets for prohibition/delay and future adequate treatment of this debilitating disorder.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, Vienna, A-1150, Austria.
| |
Collapse
|
108
|
Zhang R, Sougawa N, Mao D, Inoue H, Goda S. Signaling pathways of pro-IL-1β production induced by mechanical stress in gingival epithelial cells. J Oral Biosci 2025; 67:100626. [PMID: 39921162 DOI: 10.1016/j.job.2025.100626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES Mechanical stress on the teeth and alveolar bone caused by bruxism, orthodontics, and implants affects the periodontal tissues, causing gingival recession and alveolar bone resorption, and entire body, including the heart and vascular system. Although the same forces exerted on the alveolar bone and teeth are exerted on gingival epithelial cells, little is known about the effects of mechanical stress on these cells. This study investigated the effects of mechanical stress on gingival epithelial cells. METHODS Ca9-22 cells (human gingival epithelial cells) were used. They were seeded onto the silicone and stretched cyclically. Mechanical stress-stimulated Ca9-22 cells were evaluated for pro-inflammatory interleukin (pro-IL)-1β production using Western blotting and analyzed to assess the phosphorylation level of intracellular signaling molecules. RESULTS Mechanical stress induced pro-IL-1β upregulation in Ca9-22 cells, which was significantly inhibited by ruthenium red. Ruthenium red significantly inhibited mechanical stress-induced phosphorylation of focal adhesion kinase (FAK), P130cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2) induced by mechanical stress. Additionally, Y15 significantly inhibited the upregulation of pro-IL-1β expression and phosphorylation of FAK, P130cas, and ERK1/2 stimulated by mechanical stress. CONCLUSIONS In Ca9-22 cells, mechanical stress may increase pro-IL-1β production via mechanosensitive ion channels and FAK. These findings revealed the mechanisms of inflammation in mechanically-stressed Ca9-22 cells and may aid in the development of therapeutic approaches to prevent bone resorption.
Collapse
Affiliation(s)
- Ruixuan Zhang
- Graduate School of Dentistry, Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Nagako Sougawa
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| | - Dan Mao
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Hiroshi Inoue
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Seiji Goda
- Department of Physiology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
109
|
Ofoghi A, Kotschi S, Lemmer IL, Haas DT, Willemsen N, Bayer B, Jung AS, Möller S, Haberecht-Müller S, Krüger E, Krahmer N, Bartelt A. Activating the NFE2L1-ubiquitin-proteasome system by DDI2 protects from ferroptosis. Cell Death Differ 2025; 32:480-487. [PMID: 39384955 PMCID: PMC11893739 DOI: 10.1038/s41418-024-01398-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024] Open
Abstract
Ferroptosis is an iron-dependent, non-apoptotic form of cell death initiated by oxidative stress and lipid peroxidation. Recent evidence has linked ferroptosis to the action of the transcription factor Nuclear factor erythroid-2 derived,-like-1 (NFE2L1). NFE2L1 regulates proteasome abundance in an adaptive fashion, maintaining protein quality control to secure cellular homeostasis, but the regulation of NFE2L1 during ferroptosis and the role of the ubiquitin-proteasome system (UPS) herein are still unclear. In the present study, using an unbiased proteomic approach charting the specific ubiquitylation sites, we show that induction of ferroptosis leads to recalibration of the UPS. RSL3-induced ferroptosis inhibits proteasome activity and leads to global hyperubiquitylation, which is linked to NFE2L1 activation. As NFE2L1 resides in the endoplasmic reticulum tethered to the membrane, it undergoes complex posttranslational modification steps to become active and induce the expression of proteasome subunit genes. We show that proteolytic cleavage of NFE2L1 by the aspartyl protease DNA-damage inducible 1 homolog 2 (DDI2) is a critical step for the ferroptosis-induced feed-back loop of proteasome function. Cells lacking DDI2 cannot activate NFE2L1 in response to RSL3 and show global hyperubiquitylation. Genetic or chemical induction of ferroptosis in cells with a disrupted DDI2-NFE2L1 pathway diminishes proteasomal activity and promotes cell death. Also, treating cells with the clinical drug nelfinavir, which inhibits DDI2, sensitized cells to ferroptosis. In conclusion, our results provide new insight into the importance of the UPS in ferroptosis and highlight the role of the DDI2-NFE2L1 as a potential therapeutic target. Manipulating DDI2-NFE2L1 activity through chemical inhibition might help sensitizing cells to ferroptosis, thus enhancing existing cancer therapies.
Collapse
Affiliation(s)
- Anahita Ofoghi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Imke L Lemmer
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel T Haas
- Institute for Diabetes and Obesity (IDO), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nienke Willemsen
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Batoul Bayer
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Anna S Jung
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sophie Möller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity (IDO), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
- Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, USA.
| |
Collapse
|
110
|
Wang Y, Zhang R, Wang A, Wang X, Wang X, Zhang J, Liu G, Huang K, Liu B, Hu Y, Pan S, Ruze X, Zhai Q, Xu Y. COPB1 deficiency triggers osteoporosis with elevated iron stores by inducing osteoblast ferroptosis. J Orthop Translat 2025; 51:312-328. [PMID: 40206560 PMCID: PMC11981772 DOI: 10.1016/j.jot.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 04/11/2025] Open
Abstract
Background Osteoporosis (OP) is a systemic bone metabolic disease that results from an imbalance between bone formation and bone resorption. The accumulation of iron has been identified as an independent risk factor for osteoporosis. Ferroptosis, a novel form of programmed cell death, is driven by iron-dependent lipid peroxidation. Nevertheless, the precise role of ferroptosis in iron accumulation-induced osteoporosis remains uncertain. Methods We utilized proteomics and ELISA to screen key regulatory molecules related to iron accumulation in osteoporosis populations. HE staining was used to assess osteocyte changes in Hamp knockout (KO) iron accumulation mouse models. Western Blot, qPCR, ALP staining, and Alizarin Red staining were employed to explore the effects of siRNA-mediated gene knockdown on osteogenic differentiation in the MC3T3 cell line. ELISA, micro-CT, von Kossa staining, toluidine blue staining, TRAP staining, and calcein analysis were used to study the bone phenotype of conditional gene knockout mice. RNA-seq, endoplasmic reticulum activity probes, transmission electron microscopy (TEM), Western Blot, co-immunoprecipitation (Co-IP), flow cytometry, and ChIP-seq were employed to investigate the regulatory mechanisms of the target gene in osteogenic differentiation. OVX and Hamp KO mice were used to establish osteoporosis models, and AAV-mediated overexpression was employed to explore the intervention effects of the target gene on osteoporosis. Results The experiments demonstrate that iron accumulation can lead to changes in COPB1 expression levels in bone tissue. Cellular and animal experiments revealed that COPB1 deficiency reduces the osteogenic ability of osteoblasts. Transcriptome analysis and phenotypic experiments revealed that COPB1 deficiency induces ferroptosis and endoplasmic reticulum stress in cells. Further investigation confirmed that COPB1 plays a key role in endoplasmic reticulum stress by inhibits SLC7A11 transcription via ATF6. This reduces cystine uptake, ultimately inducing ferroptosis. Overexpression of COPB1 can restore osteogenic function in both cells and mice. Conclusion This study elucidated the essential role of COPB1 in maintaining bone homeostasis and highlights it as a potential therapeutic target for treating iron accumulation-related osteoporosis. The translational potential of this article Our data elucidate the critical role of COPB1 in maintaining bone homeostasis and demonstrate that COPB1 can directly promote bone formation, making it a potential therapeutic target for the future treatment of osteoporosis.
Collapse
Affiliation(s)
- Yike Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ruizhi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Aifei Wang
- Department of Orthopaedics, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu, China
| | - Xiao Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiongyi Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jiajun Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Gongwen Liu
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Kai Huang
- Orthopaedic Institute, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Baoshan Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yutong Hu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sheng Pan
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xieyidai Ruze
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
111
|
Chu PL, Hsiao CC, Su TC, Wang C, Lin CY. Urinary glyphosate, selenium status, and their impact on mortality: Evidence from NHANES 2013-2018. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117989. [PMID: 40023997 DOI: 10.1016/j.ecoenv.2025.117989] [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: 01/01/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE Glyphosate and glyphosate-based herbicides (GBHs), extensively used worldwide, have been associated with various health concerns, including an elevated risk of mortality. Experimental studies suggest that these herbicides may disrupt selenium homeostasis by hindering its uptake or promoting oxidative stress. However, the interplay between glyphosate exposure and selenium status remains poorly understood in epidemiological studies, particularly regarding selenium's role in modulating the mortality risk associated with glyphosate exposure in nationally representative populations. APPROACH AND RESULTS In this study, we analyzed data from the 2013-2018 National Health and Nutrition Examination Survey (NHANES), which included 6410 participants aged 3 years and older. This dataset was linked to mortality information from the National Center for Health Statistics (NCHS) for individuals aged 18 and older, with follow-up through 2019. The primary aim was to investigate the relationships between urinary glyphosate levels, whole blood selenium, selenium intake, and the influence of selenium status on glyphosate-related all-cause mortality risk. A significant negative correlation was observed between the natural logarithm (ln) of urinary glyphosate levels and the ln of whole blood selenium in the complex multiple linear regression models, with a ß coefficient of -0.010 (SE = 0.003, P = 0.003). However, no association was found between urinary glyphosate levels and selenium intake. Furthermore, the association was particularly prominent among females, non-Hispanic whites, and individuals with lower selenium intake. When examining the relationship between glyphosate exposure, whole blood selenium levels, and all-cause mortality, higher ln-urinary glyphosate levels were significantly associated with an increased risk of mortality (Hazard Ratio [HR] = 1.43; 95 % CI: 1.00-2.09). This elevated risk was especially pronounced in individuals with whole blood selenium concentrations at or above the 50th percentile. Additionally, ln-whole blood selenium was associated with a protective effect against all-cause mortality (HR = 0.01; 95 % CI: 0.00-0.18), with the strongest protective effect observed in individuals with selenium levels below the 50th percentile. CONCLUSIONS In this comprehensive analysis of NHANES data, our study identifies a potentially harmful relationship between glyphosate exposure and whole blood selenium levels. Notably, excessively high whole blood selenium levels may not only reduce the protective effects against all-cause mortality but could also increase the risk of glyphosate-related mortality, suggesting a U-shaped relationship between selenium levels and mortality risk. These findings highlight the need for further research into the health effects of glyphosate exposure and its interaction with selenium status, emphasizing the potential public health implications.
Collapse
Affiliation(s)
- Pei-Lun Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei 242, Taiwan
| | - Ching-Chung Hsiao
- Department of Nephrology, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei 100, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan; Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei 100, Taiwan; School of Medicine, College of Medicine, National Cheng-Kung University, Tainan 700, Taiwan
| | - Chikang Wang
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Chien-Yu Lin
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan; Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan; Department of Internal Medicine, En Chu Kong Hospital, New Taipei City 237, Taiwan.
| |
Collapse
|
112
|
Yamashita N, Yokoyama Y, Kumagai A, Fukushima R, Yamagata R, Hwang G. SRXN1 is a novel protective factor against methylmercury-induced apoptosis in C17.2 mouse neural stem cells. Toxicol Res 2025; 41:167-173. [PMID: 40013080 PMCID: PMC11850658 DOI: 10.1007/s43188-024-00277-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/18/2024] [Indexed: 02/28/2025] Open
Abstract
Methylmercury is an environmental pollutant that exhibits severe cerebral neurotoxicity and remains a worldwide problem. Motor and mental development disorders have been observed in children born to women who consumed relatively large amounts of methylmercury via contaminated fish during pregnancy. We previously found by RNA-sequencing analysis that treatment of C17.2 mouse neural stem cells with methylmercury induced the expression of SRXN1 (sulfiredoxin-1), a redox regulator. In this study, we examined the effect of methylmercury on SRXN1 expression and the role of SRXN1 in methylmercury-induced cell death. After C17.2 cells were treated with methylmercury, both mRNA and protein expression of SRXN1 increased in a time- and concentration-dependent manner. Because the induction of SRXN1 expression by methylmercury was suppressed by pretreatment with a transcription inhibitor, we searched the upstream region of the SRXN1 gene and found a binding sequence for transcription factor 3 (TCF3). Interestingly, the induction of SRXN1 expression by methylmercury was attenuated in cells in which TCF3 expression was suppressed by siRNA compared with control cells. This suggests that TCF3 is involved in the induction of SRXN1 expression by methylmercury. We previously reported that TCF3 overexpression suppressed methylmercury-induced apoptosis; in the present study, we found that SRXN1 overexpression also suppressed methylmercury-induced apoptosis, as assessed by cleaved caspase-3 expression. In summary, our results indicate that SRXN1 induced by TCF3-mediated expression is a novel protective factor against methylmercury-induced apoptosis. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00277-6.
Collapse
Affiliation(s)
- Naoya Yamashita
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Yukie Yokoyama
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Ayano Kumagai
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Ryoko Fukushima
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Ryota Yamagata
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| | - Gi‑Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-Ku, Sendai, Miyagi 981-8558 Japan
| |
Collapse
|
113
|
Yang L, Chen J, Yao Z, Cai J, Zhang H, Wang Z, Guo H, Zha Y. Associations of multiple plasma metals with the risk of type 2 diabetes in Chinese adults: A cross-sectional study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117941. [PMID: 40009940 DOI: 10.1016/j.ecoenv.2025.117941] [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: 08/26/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Evidence regarding the associations between co-exposure to multiple metals and diabetes risk was scarce. This study aimed to evaluate the associations of multiple metals with diabetes risk using multiple statistical methods. This cross-sectional study included 192 diabetic patients and 189 healthy subjects. We employed inductively coupled plasma mass spectrometry (ICP-MS) to determine the plasma concentrations of 18 metals. Least absolute shrinkage and selection operator (LASSO) regression, logistic regression, and Bayesian kernel machine regression (BKMR) were applied to evaluate associations of multiple metals with diabetes risk comprehensively. These models consistently suggested that aluminium and selenium were positively associated with diabetes risk, while manganese, rubidium, and lead were negatively associated with diabetes risk. Age-specific differences of selenium and sex-specific differences of manganese in diabetes risk were also observed based on stratified analyses. According to RCS analyses, we obtained dose-response relationships between metals and diabetes risk:(1) there were inverted U-shaped associations of plasma aluminium and selenium with diabetes risk, with the threshold close to 20.5µg/L and 75.9µg/L, respectively (both P for overall < 0.05; both P for non-linearity < 0.05). (2) There were L-shaped associations of rubidium and lead with diabetes risk, with the turning point close to 144.5µg/L and 2.5µg/L, respectively (both P for overall < 0.05; both P for non-linearity < 0.05). (3) Manganese was linearly and negatively correlated with diabetes risk when concentrations of manganese were less than approximately 4.2 μg/L (P for overall < 0.05; P for non-linearity = 0.268). The BKMR model also revealed a negative combined effect of metal mixtures on diabetes risk and potential interactions between six pairs of metals (aluminium-manganese, aluminium-selenium, aluminium-rubidium, aluminium-lead, manganese-selenium, and manganese-rubidium). In summary, we need to pay attention to the role of low plasma levels of aluminium, selenium, manganese, rubidium, and lead in diabetes, especially regarding their safety windows.
Collapse
Affiliation(s)
- Liting Yang
- Biomedical Research Institute of Hubei University of Medicine, Shiyan 442000, China
| | - Jin Chen
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Zijun Yao
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China; Public Health Service Center of Wuhan East Lake Scenic District, Wuhan 430077, China
| | - Junwei Cai
- Department of Endocrinology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Han Zhang
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Zhen Wang
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Huailan Guo
- School of Public Health, Hubei University of Medicine, Shiyan 442000, China.
| | - Yongjiu Zha
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
114
|
Huang F, Li S, Zhang A, Zhao J, Zhang S, Liu D, Chen W. Identification of ferroptosis-related genes in heart tissues of patients with hypertrophic cardiomyopathy. Medicine (Baltimore) 2025; 104:e41525. [PMID: 40020138 PMCID: PMC11875581 DOI: 10.1097/md.0000000000041525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND This study aims to investigate the role of ferroptosis in hypertrophic cardiomyopathy (HCM), a genetic disorder characterized by abnormal thickening of the heart muscle. The objective is to identify differentially expressed genes associated with ferroptosis in HCM and understand the potential molecular mechanisms underlying the disease. METHODS Comprehensive genomic analysis was conducted to identify differentially expressed genes associated with ferroptosis in HCM. The analysis focused on TFRC, SCD, SLC2A1, EGR1, GDF15, SNCA, PLIN2, and NQO1 as hub genes regulating ferroptosis. Functional enrichment analysis was performed to uncover their involvement in pathways such as ferroptosis, ubiquinone biosynthesis, and HIF-1 signaling. In addition, immune cell infiltration patterns in HCM were explored, and associations between the hub genes and immune infiltration were identified. RESULTS The analysis revealed TFRC, SCD, SLC2A1, EGR1, GDF15, SNCA, PLIN2, and NQO1 as hub genes involved in the regulation of ferroptosis in HCM. Functional enrichment analysis indicated their contribution to key pathways related to ferroptosis, ubiquinone biosynthesis, and HIF-1 signaling. Furthermore, associations between the hub genes and immune infiltration in HCM were observed. CONCLUSION This study provides valuable insights into the molecular basis of HCM by identifying differentially expressed genes associated with ferroptosis. The findings suggest potential molecular mechanisms underlying the development of HCM. These findings contribute to a better understanding of HCM and may pave the way for the development of targeted therapies and improved diagnostic approaches for this debilitating cardiac disorder.
Collapse
Affiliation(s)
- Fang Huang
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Shujuan Li
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Ailei Zhang
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Jihuai Zhao
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Shaoqiang Zhang
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Dongwei Liu
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| | - Wei Chen
- Department of Cardiology, Qingdao West Coast New Area People’s Hospital, Cardiovascular Internal Medicine, Qingdao, Shandong, China
| |
Collapse
|
115
|
Lee JG, Jang JY, Baik SM. Selenium as an Antioxidant: Roles and Clinical Applications in Critically Ill and Trauma Patients: A Narrative Review. Antioxidants (Basel) 2025; 14:294. [PMID: 40227249 PMCID: PMC11939285 DOI: 10.3390/antiox14030294] [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: 02/03/2025] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
Selenium plays an indispensable role in antioxidant defense through its incorporation into selenoproteins, including glutathione peroxidase (GPx) and thioredoxin reductase. In the context of trauma and critical illness, systemic inflammation and oxidative stress frequently deplete selenium reserves, compromising the body's antioxidant defenses. This deficiency exacerbates immune dysfunction, elevates the risk of multi-organ dysfunction syndrome, and increases susceptibility to infections and mortality. Observational studies have consistently shown that lower selenium levels correlate with poorer clinical outcomes, such as extended stays in intensive care units and higher mortality rates. Supplementation of selenium has demonstrated promise in restoring GPx activity, reducing oxidative stress markers, and supporting recovery, particularly in patients with pre-existing selenium deficiency. While the impact on mortality remains variable across clinical trials, early and targeted supplementation appears to be beneficial, especially when combined with other micronutrients like vitamins C and E or zinc. These combinations enhance the antioxidant response and tackle the complex oxidative pathways in critically ill and trauma patients. Importantly, the clinical benefits of selenium supplementation appear to be influenced by baseline selenium status, with patients exhibiting severe deficiency deriving the most pronounced improvements in oxidative stress markers, immune function, and recovery. This review highlights the critical importance of addressing selenium deficiency, advocating for personalized therapeutic strategies. However, further large-scale studies are essential to optimize dosing regimens, refine combination therapies, and validate selenium's therapeutic potential in trauma and critical care settings.
Collapse
Affiliation(s)
- Jae-Gil Lee
- Department of Surgery, Ewha Womans University Mokdong Hospital, Seoul 07985, Republic of Korea;
| | - Ji-Young Jang
- Department of Surgery, National Health Insurance Service Ilsan Hospital, Goyang 10444, Republic of Korea;
| | - Seung-Min Baik
- Department of Surgery, Ewha Womans University Mokdong Hospital, Seoul 07985, Republic of Korea;
| |
Collapse
|
116
|
Scarpellini C, Klejborowska G, Lanthier C, Toye A, Musiałek K, Van San E, Walravens M, Berg M, Hassannia B, Van der Veken P, De Winter H, Vanden Berghe T, Augustyns K. Oxazole-Based Ferroptosis Inhibitors with Promising Properties to Treat Central Nervous System Diseases. J Med Chem 2025; 68:4908-4928. [PMID: 39913870 PMCID: PMC11874020 DOI: 10.1021/acs.jmedchem.4c03149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Ferroptosis plays an important role in the occurrence and development of many diseases, including neurodegenerative diseases. Thus, ferroptosis inhibitors able to cross the blood-brain barrier may have therapeutic potential. The best ferroptosis inhibitors so far are lipophilic radical trapping antioxidants (RTAs) that block lipid peroxidation in membranes. Several generations of ferrostatins have been synthesized, among which UAMC-3203 showed high potency in animal models with improved properties compared to ferrostatin-1. To further improve its pharmacokinetics properties, drug-likeness, and permeability, we modified UAMC-3203 by decreasing the size of the molecule and reducing its polarity by replacing the sulfonamide first by amide groups and subsequently by isosteric oxazoles. Herein, we present the design, synthesis, and biological evaluation of a novel series of oxazole RTAs with high potency, excellent oral bioavailability, and high concentrations in brain tissue.
Collapse
Affiliation(s)
- Camilla Scarpellini
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Greta Klejborowska
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Caroline Lanthier
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Ariane Toye
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Karolina Musiałek
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
- Department
of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznań 61-614, Poland
| | - Emily Van San
- Cell Death
Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Magali Walravens
- Cell Death
Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Maya Berg
- Infla-Med
Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Behrouz Hassannia
- Cell Death
Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Molecular
Signalling and Cell Death Unit, VIB Center
for Inflammation Research, Ghent 9052, Belgium
- Department
of Biomedical Molecular Biology, Ghent University, Ghent 9000, Belgium
| | - Pieter Van der Veken
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Hans De Winter
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Tom Vanden Berghe
- Cell Death
Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
- Infla-Med
Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
- Molecular
Signalling and Cell Death Unit, VIB Center
for Inflammation Research, Ghent 9052, Belgium
- Department
of Biomedical Molecular Biology, Ghent University, Ghent 9000, Belgium
| | - Koen Augustyns
- Laboratory
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Faculty
of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp 2610, Belgium
- Infla-Med
Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
117
|
Li Q, Liu L, Liu Y, Zheng T, Chen N, Du P, Ye H. Exploration of key genes associated with oxidative stress in polycystic ovary syndrome and experimental validation. Front Med (Lausanne) 2025; 12:1493771. [PMID: 40083347 PMCID: PMC11904916 DOI: 10.3389/fmed.2025.1493771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction The current study demonstrated that oxidative stress (OS) is closely related to the pathogenesis of polycystic ovary syndrome (PCOS). However, there are numerous factors that lead to OS, therefore, identifying the key genes associated with PCOS that contribute to OS is crucial for elucidating the pathogenesis of PCOS and selecting appropriate treatment strategies. Methods Four datasets (GSE95728, GSE106724, GSE138572, and GSE145296) were downloaded from the gene expression omnibus (GEO) database. GSE95728 and GSE106724 were combined to identify differentially expressed genes (DEGs) in PCOS. weighted gene correlation network analysis (WGCNA) was used to screen key module genes associated with PCOS. Differentially expressed OS related genes (DE-OSRGs) associated with PCOS were obtained by overlapping DEGs, key module genes, and OSRGs. Subsequently, the optimal machine model was obtained to identify key genes by comparing the performance of the random forest model (RF), support vector machine model (SVM), and generalized linear model (GLM). The molecular networks were constructed to reveal the non-coding regulatory mechanisms of key genes based on GSE138572 and GSE145296. The Drug-Gene Interaction Database (DGIdb) was used to predict the potential therapeutic agents of key genes for PCOS. Finally, the expression of key OSRGs was validated by RT-PCR. Results In this study, 8 DE-OSRGs were identified. Based on the residuals and root mean square error of the three models, the best performance of RF was derived and 7 key genes (TNFSF10, CBL, IFNG, CP, CASP8, APOA1, and DDIT3) were identified. The GSEA enrichment analysis revealed that TNFSF10, CP, DDIT3, and INFG are all enriched in the NOD-like receptor signaling pathway and natural killer cell-mediated cytotoxicity pathways. The molecular regulatory network uncovered that both TNFSF10 and CBL are regulated by non-coding RNAs. Additionally, 70 potential therapeutic drugs for PCOS were predicted, with ibuprofen associated with DDIT3 and IFNG. RT-qPCR validation confirmed the expression trends of key genes IFNG, DDIT3, and APOA1 were consistent with the dataset, and the observed differences were statistically significant (P < 0.05). Conclusion The identification of seven key genes and molecular regulatory networks through bioinformatics analysis is of great significance for exploring the pathogenesis and therapeutic strategies of PCOS.
Collapse
Affiliation(s)
- Qinhua Li
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Obstetrics and Gynecology, Yichang Central People’s Hospital, Yichang, China
- Institute of Obstetrics and Gynecology, China Three Gorges University, Yichang, China
| | - Lei Liu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Yuhan Liu
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- China Three Gorges University, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University and Yichang Central People’s Hospital, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Tingting Zheng
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Obstetrics and Gynecology, Yichang Central People’s Hospital, Yichang, China
- Institute of Obstetrics and Gynecology, China Three Gorges University, Yichang, China
| | - Ningjing Chen
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Obstetrics and Gynecology, Yichang Central People’s Hospital, Yichang, China
- Institute of Obstetrics and Gynecology, China Three Gorges University, Yichang, China
| | - Peiyao Du
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Obstetrics and Gynecology, Yichang Central People’s Hospital, Yichang, China
- Institute of Obstetrics and Gynecology, China Three Gorges University, Yichang, China
| | - Hong Ye
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Department of Obstetrics and Gynecology, Yichang Central People’s Hospital, Yichang, China
- Institute of Obstetrics and Gynecology, China Three Gorges University, Yichang, China
| |
Collapse
|
118
|
Chen SY, Shyu IL, Chi JT. NINJ1 in Cell Death and Ferroptosis: Implications for Tumor Invasion and Metastasis. Cancers (Basel) 2025; 17:800. [PMID: 40075648 PMCID: PMC11898531 DOI: 10.3390/cancers17050800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
NINJ1 was initially recognized for its role in nerve regeneration and cellular adhesion. Subsequent studies have uncovered its participation in cancer progression, where NINJ1 regulates critical steps in tumor metastasis, such as cell migration and invasion. More recently, NINJ1 has emerged as a multifunctional protein mediating plasma membrane rupture (PMR) in several lytic cell death processes, including apoptosis, necroptosis, and pyroptosis. However, its role in ferroptosis-an iron-dependent form of lytic cell death characterized by lipid peroxidation-remained unclear until 2024. Ferroptosis is a tumor suppression mechanism that may be particularly relevant to detached and metastatic cancer cells. This review explores the role of NINJ1 in tumor invasion and metastasis, focusing on its regulation of ferroptosis via a non-canonical mechanism distinct from other cell deaths. We discuss the process of ferroptosis and its implications for cancer invasion and metastasis. Furthermore, we review recent studies highlighting the diverse roles of NINJ1 in ferroptosis regulation, including its canonical function in PMR and its non-canonical function of modulating intracellular levels of glutathione (GSH) and coenzyme A (CoA) via interaction with xCT anti-porter. Given that ferroptosis has been associated with tumor suppression, metastasis, the elimination of treatment-resistant cancer cells, and tumor dormancy, NINJ1's modulation of ferroptosis presents a promising therapeutic target for inhibiting metastasis. Understanding the dual role of NINJ1 in promoting or restraining ferroptosis depending on cellular context could open avenues for novel anti-cancer strategies to enhance ferroptotic vulnerability in metastatic tumors.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA;
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ing-Luen Shyu
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan 710, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
119
|
You Y, Li Y, Ye L, Xu F, Fan J. Interleukins-27 Aggravates Liver Injury by Impairing the Antimicrobial Response of Macrophages via the Promotion of Mitochondrial Dysfunction in the Context of Sepsis. Mediators Inflamm 2025; 2025:6608718. [PMID: 40046257 PMCID: PMC11882325 DOI: 10.1155/mi/6608718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 05/13/2025] Open
Abstract
Background and Aims: Plasma interleukin (IL)-27 is an important mediator of acute hepatic injury (AHI) associated with sepsis. Mitochondria contribute to the proper regulation of macrophage phagocytosis. In this study, we investigated the effect of IL-27 on mitochondrial function and the antimicrobial response of macrophages in sepsis-associated AHI. Methods: Wild-type (WT) and IL-27 receptor WSX-1 deficient (IL-27R-/-) mice underwent cecal ligation and puncture (CLP). The severity of hepatic injury, inflammatory cytokine levels, hepatic pyroptosis, and bacterial load in the liver and blood were assessed 24 h after CLP. In vitro, RAW264.7 cells and peritoneal macrophages were treated with lipopolysaccharide (LPS) and/or IL-27. The phagocytosis and killing functions of macrophages were detected. Mitochondrial function and mitophagy were detected using western blot, glutathione (GSH)/malondialdehyde (MDA) content measurement, fluorescence staining, and JC-1 staining in vivo and in vitro. After treatment with nicotinamide mononucleotide (NMN, NAD + precursor), a pharmacologic agent that improves mitochondrial function, the inflammatory response, hepatic injury, and hepatic pyroptosis were assessed. Results: IL-27R-/- mice exhibited a marked reduction in hepatic injury, pyroptosis (based on cleaved GSDMD and cleaved Caspases 1 protein levels), and systemic inflammation (based on serum IL-6, IL-10, and TNF-α levels) compared to WT mice following CLP. After CLP, mice lacking IL-27R displayed significantly higher bacterial clearance and greater local infection control. Subsequent studies demonstrated that IL-27 directly impaired the LPS-induced bacterial phagocytosis, killing capacity, and mitochondrial function of macrophages. Finally, enhanced mitochondrial function using NMN in vivo significantly alleviated pathological liver injury and inflammation. Conclusions: These findings indicated that IL-27 impairs the bacterial phagocytosis capacity of macrophages by aggravating mitochondrial dysfunction to aggravate AHI during sepsis.
Collapse
Affiliation(s)
- Yuehua You
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuyan Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Critical Care Medicine, The First People's Hospital of Chongqing High-tech Zone, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Ye
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Fan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
120
|
Kuhn BN, Cannella N, Chitre AS, Nguyen KMH, Cohen K, Chen D, Peng B, Ziegler KS, Lin B, Johnson BB, Missfeldt Sanches T, Crow AD, Lunerti V, Gupta A, Dereschewitz E, Soverchia L, Hopkins JL, Roberts AT, Ubaldi M, Abdulmalek S, Kinen A, Hardiman G, Chung D, Polesskaya O, Solberg Woods LC, Ciccocioppo R, Kalivas PW, Palmer AA. Genome-wide association study reveals multiple loci for nociception and opioid consumption behaviors associated with heroin vulnerability in outbred rats. Mol Psychiatry 2025:10.1038/s41380-025-02922-4. [PMID: 40000848 DOI: 10.1038/s41380-025-02922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/20/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
The increased prevalence of opioid use disorder (OUD) makes it imperative to disentangle the biological mechanisms contributing to individual differences in OUD vulnerability. OUD shows strong heritability, however genetic variants contributing to vulnerability remain poorly defined. We performed a genome-wide association study using over 850 male and female heterogeneous stock (HS) rats to identify genes underlying behaviors associated with OUD such as nociception, as well as heroin-taking, extinction and seeking behaviors. By using an animal model of OUD, we were able to identify genetic variants associated with distinct OUD behaviors while maintaining a uniform environment, an experimental design not easily achieved in humans. Furthermore, we used a novel non-linear network-based clustering approach to characterize rats based on OUD vulnerability to assess genetic variants associated with OUD susceptibility. Our findings confirm the heritability of several OUD-like behaviors, including OUD susceptibility. Additionally, several genetic variants associated with nociceptive threshold prior to heroin experience, heroin consumption, escalation of intake, and motivation to obtain heroin were identified. Tom1, a microglial component, was implicated for nociception. Several genes involved in dopaminergic signaling, neuroplasticity and substance use disorders, including Brwd1, Pcp4, Phb1l2 and Mmp15 were implicated for the heroin traits. Additionally, an OUD vulnerable phenotype was associated with genetic variants for consumption and break point, suggesting a specific genetic contribution for OUD-like traits contributing to vulnerability. Together, these findings identify novel genetic markers related to the susceptibility to OUD-relevant behaviors in HS rats.
Collapse
Affiliation(s)
- Brittany N Kuhn
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Nazzareno Cannella
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Khai-Minh H Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Katarina Cohen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Denghui Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Beverly Peng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Kendra S Ziegler
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Bonnie Lin
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Benjamin B Johnson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Ayteria D Crow
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Veronica Lunerti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Arkobrato Gupta
- The Interdisciplinary Ph.D. Program in Biostatistics, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Eric Dereschewitz
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Laura Soverchia
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Jordan L Hopkins
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Analyse T Roberts
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Massimo Ubaldi
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Sarah Abdulmalek
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Analia Kinen
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Gary Hardiman
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Departments of Medicine and Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Dongjun Chung
- The Interdisciplinary Ph.D. Program in Biostatistics, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
121
|
Vinceti M, Mazzoli R, Wise LA, Veneri F, Filippini T. Calling for a comprehensive risk assessment of selenium in drinking water. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 966:178700. [PMID: 39923476 DOI: 10.1016/j.scitotenv.2025.178700] [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: 01/04/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
In the last two decades, research has elucidated that selenium, a trace element, has both nutritional and toxicological effects on human health, depending on its dose and chemical form. Recent animal, laboratory, and human studies have shown harmful effects of certain selenium species at specific exposure levels, prompting the need to reassess overall exposure to this element, including that occurring through drinking water, a primary source of inorganic selenium. Drinking water selenium standards worldwide are scarce and existing standards are inconsistent, likely because they have been informed by an incomplete and outdated assessment of the scientific evidence. Incorporating all the available human and laboratory evidence into a precautionary regulatory framework indicates that a drinking water limit of around 5 μg/L of selenium is needed to protect human health, i.e. with an uncertainty factor of 2 versus the lowest adverse effect level observed in human studies, and that higher values may pose unacceptable risks to humans. Despite the rarity of such high levels of selenium in underground and potable waters, coal mining and other sources of environmental pollution as well as geological factors may raise drinking water selenium content above a safe threshold, triggering the need to protect consumers, and to face challenging technological issues for selenium removal, currently under active investigation.
Collapse
Affiliation(s)
- Marco Vinceti
- CREAGEN - Environmental, Genetic and Nutritional Epidemiology Research Center, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia Medical School, Modena, Italy; Department of Epidemiology, School of Public Health, Boston University, Boston, MA, United States of America.
| | - Riccardo Mazzoli
- CREAGEN - Environmental, Genetic and Nutritional Epidemiology Research Center, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia Medical School, Modena, Italy
| | - Lauren A Wise
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, United States of America
| | - Federica Veneri
- CREAGEN - Environmental, Genetic and Nutritional Epidemiology Research Center, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia Medical School, Modena, Italy; Unit of Dentistry and Oral-Maxillo-Facial Surgery, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Filippini
- CREAGEN - Environmental, Genetic and Nutritional Epidemiology Research Center, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia Medical School, Modena, Italy; Department of Epidemiology, School of Public Health, University of California Berkeley, Berkeley, CA, United States of America
| |
Collapse
|
122
|
Schwab A, Brabletz T. Grease, fuel and target - polyunsaturated lipids in metastasis. Cell Res 2025:10.1038/s41422-025-01089-5. [PMID: 40000773 DOI: 10.1038/s41422-025-01089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Affiliation(s)
- Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| |
Collapse
|
123
|
Kacemi R, Campos MG. Bee Pollen Potential to Modulate Ferroptosis: Phytochemical Insights for Age-Related Diseases. Antioxidants (Basel) 2025; 14:265. [PMID: 40227202 PMCID: PMC11939620 DOI: 10.3390/antiox14030265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 04/15/2025] Open
Abstract
Bee pollen (BP) is one of the richest known natural resources of micronutrients and bioactive phytochemicals. Some captivating bioactivities of BP compounds, although being largely investigated for the latter as individual molecules, remain very scarcely investigated or completely uninvestigated in bee pollen as a whole product. Among the most intriguing of these bioactivities, we identified ferroptosis as a major one. Ferroptosis, a recently discovered form of cell death (connecting oxidative stress and inflammation), is a complex pathophysiological process and one of the most crucial and perplexing events in current challenging human diseases such as cancer, neurodegeneration, and general aging diseases. Many BP compounds were found to intricately modulate ferroptosis depending on the cellular context by inducing this cell death mechanism in malignant cells and preventing it in non-malignant cells. Since research in both fields, i.e., BP and ferroptosis, is still recent, we deemed it necessary to undertake this review to figure out the extent of BP potential in modulating ferroptosis mechanisms. Our research proved that a wide range of BP compounds (polyphenols, phenolamides, carotenoids, vitamins, minerals, and others) substantially modulate diverse ferroptosis mechanisms. Accordingly, these phytochemicals and nutrients showed interesting potential in preclinical studies to lead to ferroptosis-mediated outcomes in important pathophysiological processes, including many aging-related disorders. One of the most paramount challenges that remain to be resolved is to determine how different BP compounds act on ferroptosis in different biological and pathophysiological contexts, either through synergistic or antagonistic behaviors. We hope that our current work constitutes a valuable incentive for future investigations in this promising and very relevant research avenue.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3000-548 Coimbra, Portugal
| |
Collapse
|
124
|
Yuan R, Zhang Y, Han J. The association of selenium exposure with the odds of metabolic syndrome: a dose-response meta-analysis. BMC Endocr Disord 2025; 25:49. [PMID: 39994710 PMCID: PMC11853193 DOI: 10.1186/s12902-025-01858-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Selenium is a key regulator of metabolic homeostasis. It has been proposed that exposure to selenium might be associated with metabolic syndrome (MetS). However, the results are contradictory. This meta-analysis was carried out to analyze the relationships between selenium levels in biological samples and odds of Mets. METHODS We searched Scopus and PubMed databases up until September 2024 to identify relevant studies. Odds ratio (OR) and 95% confidence interval was used to pool the data using a random effects model. RESULTS The meta-analysis encompassed 18 observational studies involving 21,481 participants. It found that higher selenium exposure was related to an elevated likelihood of MetS (OR = 1.30, 95% CI = 1.12-1.51), even after controlling for covariates, such as smoking, age, alcohol consumption, and physical activity. Heterogeneity was significant among the studies (I² = 88.9%, P = 0.001). While elevated serum selenium levels linked to a higher odds of MetS, no such relationship was observed for selenium in urine or toenails. Subgroup analyses indicated that this association was evident only in females (OR = 2.0, 95% CI = 1.17-1.43) and particularly pronounced in individuals aged ≥ 50 years. A dose-response relationship was identified, showing a 6% increase in MetS odds for each additional 10 µg/L of serum selenium, with the odds rising non-linearly when serum levels surpassed 80 µg/L. CONCLUSIONS This study suggests that selenium may associated with the odds of MetS, following a dose-response relationship.
Collapse
Affiliation(s)
- Rongrong Yuan
- Department of Endocrinology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Yu Zhang
- Department of Nephrology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Jiakai Han
- Department of Endocrinology, Huaihe Hospital of Henan University, Kaifeng, 475000, China.
| |
Collapse
|
125
|
Gollowitzer A, Pein H, Rao Z, Waltl L, Bereuter L, Loeser K, Meyer T, Jafari V, Witt F, Winkler R, Su F, Große S, Thürmer M, Grander J, Hotze M, Harder S, Espada L, Magnutzki A, Gstir R, Weinigel C, Rummler S, Bonn G, Pachmayr J, Ermolaeva M, Harayama T, Schlüter H, Kosan C, Heller R, Thedieck K, Schmitt M, Shimizu T, Popp J, Shindou H, Kwiatkowski M, Koeberle A. Attenuated growth factor signaling during cell death initiation sensitizes membranes towards peroxidation. Nat Commun 2025; 16:1774. [PMID: 40000627 PMCID: PMC11861335 DOI: 10.1038/s41467-025-56711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Cell death programs such as apoptosis and ferroptosis are associated with aberrant redox homeostasis linked to lipid metabolism and membrane function. Evidence for cross-talk between these programs is emerging. Here, we show that cytotoxic stress channels polyunsaturated fatty acids via lysophospholipid acyltransferase 12 into phospholipids that become susceptible to peroxidation under additional redox stress. This reprogramming is associated with altered acyl-CoA synthetase isoenzyme expression and caused by a decrease in growth factor receptor tyrosine kinase (RTK)-phosphatidylinositol-3-kinase signaling, resulting in suppressed fatty acid biosynthesis, for specific stressors via impaired Akt-SREBP1 activation. The reduced availability of de novo synthesized fatty acids favors the channeling of polyunsaturated fatty acids into phospholipids. Growth factor withdrawal by serum starvation mimics this phenotype, whereas RTK ligands counteract it. We conclude that attenuated RTK signaling during cell death initiation increases cells' susceptibility to oxidative membrane damage at the interface of apoptosis and alternative cell death programs.
Collapse
Affiliation(s)
- André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Konstantin Loeser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Tobias Meyer
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Vajiheh Jafari
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916, Badalona, Spain
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Silke Große
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Maria Thürmer
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ronald Gstir
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Günther Bonn
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Johanna Pachmayr
- Institute of Pharmacy, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur - CNRS UMR7275 - Inserm U1323, 06560, Valbonne, France
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
- Department Metabolism, Senescence and Autophagy, Research Center One Health Ruhr, University Alliance Ruhr & University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
- German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147, Essen, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo 141-0021, Japan
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
126
|
Li X, Hu X, You H, Zheng K, Tang R, Kong F. Regulation of pattern recognition receptor signaling by palmitoylation. iScience 2025; 28:111667. [PMID: 39877903 PMCID: PMC11772949 DOI: 10.1016/j.isci.2024.111667] [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] [Indexed: 01/31/2025] Open
Abstract
Pattern recognition receptors (PRRs), consisting of Toll-like receptors, RIG-I-like receptors, cytosolic DNA sensors, and NOD-like receptors, sense exogenous pathogenic molecules and endogenous damage signals to maintain physiological homeostasis. Upon activation, PRRs stimulate the sensitization of nuclear factor κB, mitogen-activated protein kinase, TANK-binding kinase 1-interferon (IFN) regulatory factor, and inflammasome signaling pathways to produce inflammatory factors and IFNs to activate Janus kinase/signal transducer and activator of transcription signaling pathways, resulting in anti-infection, antitumor, and other specific immune responses. Palmitoylation is a crucial type of post-translational modification that reversibly alters the localization, stability, and biological activity of target molecules. Here, we discuss the available knowledge on the biological roles and underlying mechanisms linked to protein palmitoylation in modulating PRRs and their downstream signaling pathways under physiological and pathological conditions. Moreover, recent advances in the use of palmitoylation as an attractive therapeutic target for disorders caused by the dysregulation of PRRs were summarized.
Collapse
Affiliation(s)
- Xiaocui Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaofang Hu
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
127
|
do Carmo Santos ML, Silva Santos A, Pereira Silva de Novais D, dos Santos Lopes N, Pirovani CP, Micheli F. The family of glutathione peroxidase proteins and their role against biotic stress in plants: a systematic review. FRONTIERS IN PLANT SCIENCE 2025; 16:1425880. [PMID: 40051871 PMCID: PMC11882536 DOI: 10.3389/fpls.2025.1425880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/09/2025] [Indexed: 03/09/2025]
Abstract
Introduction Glutathione peroxidases (GPXs) are extensively studied for their indispensable roles in eliminating reactive oxygen species by catalyzing the reduction of hydrogen peroxide or lipid peroxides to prevent cell damage. However, knowledge of GPXs in plants still has many gaps to be filled. Thus, we present the first systematic review (SR) aimed at examining the function of GPXs and their protective role against cell death in plants subjected to biotic stress. Methods To guide the SR and avoid bias, a protocol was developed that contained inclusion and exclusion criteria based on PRISMA guidelines. Three databases (PubMed, Science Direct, and Springer) were used to identify relevant studies for this research were selected. Results A total of 28 articles related to the proposed objective. The results highlight the importance of GPXs in plant defense against biotic stress, including their role in protecting against cell death, similar to the anti-apoptotic GPXs in animals. Data from gene expression and protein accumulation studies in plants under various biotic stresses reveal that GPXs can both increase resistance and susceptibility to pathogens. In addition to their antioxidant functions, GPXs act as sensors and transmitters of H2O2 signals, integrating with the ABA signaling pathway during stress. Discussion These findings show that GPXs delay senescence or reinforce physical barriers, thereby modulating resistance or susceptibility to pathogens. Additionally, their functions are linked to their cellular localization, which demonstrates an evolutionary relationship between the studied isoforms and their role in plant defense. This information broadens the understanding of molecular strategies involving GPX isoforms and provides a foundation for discussions and actions aimed at controlling necrotrophic and/or hemibiotrophic pathogens.
Collapse
Affiliation(s)
- Maria Luíza do Carmo Santos
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
| | - Ariana Silva Santos
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
| | - Diogo Pereira Silva de Novais
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
- Instituto Federal de Educação Ciência e Tecnologia da Bahia (IFBA), Bahia, Brazil
| | - Natasha dos Santos Lopes
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
| | - Carlos Priminho Pirovani
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
| | - Fabienne Micheli
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Ilhéus, Brazil
- CIRAD, UMR AGAP, Montpellier, France
| |
Collapse
|
128
|
Zhu Z, Nie G, Peng X, Zhan X, Ding D. KAT8 catalyzes the acetylation of SEPP1 at lysine 247/249 and modulates the activity of CD8 + T cells via LRP8 to promote anti-tumor immunity in pancreatic cancer. Cell Biosci 2025; 15:24. [PMID: 39972392 PMCID: PMC11841300 DOI: 10.1186/s13578-025-01356-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) remains one of the most lethal malignancies with unfavorable prognosis globally. Bioinformatics analysis predicted that SEPP1 was low expressed in PC and related to tumor immune microenvironment, but its biological function was still unclear. METHODS PC xenograft and liver metastasis mouse models, as well as PC cell-MDSCs co-culture system, were established for in vivo and in vitro studies, respectively. The expression and localization of key molecules were detected by qRT-PCR, western blot, immunohistochemistry and immunofluorescence. Flow cytometry was employed to assess the abundance of immune cells and cell apoptosis. The interactions among KAT8, SEPP1 and LRP8 were detected by co-IP. Cell viability, migration and invasion were monitored by CCK-8 and transwell assays. RESULTS SEPP1 was downregulated in pancreatic tumors, and it was positively correlated with the abundance of CD8+ T cells. In vivo overexpression of SEPP1 impaired PC tumor growth and liver metastasis via modulating the abundance of CD8+ T cell and MDSCs. KAT8 upregulated SEPP1 transcription and protein level via catalyzing the acetylation at K247/249 on SEPP1, and SEPP1 impaired MDSCs survival via its receptor LRP8, thus regulating CD8+ T cell-mediated immune responses in PC. In vivo studies further revealed that SEPP1 recombinant protein enhanced the efficacy of anti-PD-1 therapy in PC xenograft mouse model. CONCLUSION KAT8 catalyzed the acetylation of SEPP1 at K247/249 and modulated the activity of CD8+ T cells via LRP8 to promote anti-tumor immunity in PC.
Collapse
Affiliation(s)
- Zhongfei Zhu
- Department of Hepatobiliary, Pancreatic and Spleen Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Gang Nie
- Department of Hepatobiliary, Pancreatic and Spleen Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, The First Affiliated Hospital of Naval Medical University, No. 168, Changhai Road, Yangpu District, Shanghai, 200433, China.
| | - Xianbao Zhan
- Department of Oncology, The First Affiliated Hospital of Naval Medical University, No. 168, Changhai Road, Yangpu District, Shanghai, 200433, China.
| | - Dan Ding
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, No. 168, Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
129
|
Lemay SE, Mougin M, Sauvaget M, El Kabbout R, Valasarajan C, Yamamoto K, Martineau S, Pelletier A, Bilodeau C, Grobs Y, Bourgeois A, Romanet C, Breuils-Bonnet S, Montesinos MS, Lu M, Chen H, Gilbert M, Théberge C, Potus F, Pullamsetti S, Provencher S, Bonnet S, Boucherat O. Unraveling AURKB as a potential therapeutic target in pulmonary hypertension using integrated transcriptomic analysis and pre-clinical studies. Cell Rep Med 2025; 6:101964. [PMID: 39933527 PMCID: PMC11866512 DOI: 10.1016/j.xcrm.2025.101964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/29/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Despite advances in treatment, the prognosis for patients with pulmonary arterial hypertension (PAH) remains dismal, highlighting the need for further therapeutic advances. By using RNA sequencing on pulmonary artery smooth muscle cells (PASMCs), functional enrichment, and connectivity map analyses, we identify Aurora kinase B (AURKB) as a candidate therapeutic target. We show that AURKB inhibition blocks cell cycle progression and reverses the gene signature of PAH-PASMCs. We also report that PAH-PASMCs that escape apoptosis acquire a senescence-associated secretory phenotype. In vivo, AURKB inhibition using barasertib improves hemodynamics in two preclinical models of established PAH by attenuating pulmonary vascular remodeling. A therapeutic effect is also observed in human precision-cut lung slices. Finally, we demonstrate that the combination of barasertib with a p21 attenuator is more effective in reducing vascular remodeling than either drug alone. These findings provide insight into strategies for therapeutic manipulation.
Collapse
MESH Headings
- Humans
- Animals
- Aurora Kinase B/antagonists & inhibitors
- Aurora Kinase B/metabolism
- Aurora Kinase B/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Vascular Remodeling/drug effects
- Gene Expression Profiling
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- Transcriptome/genetics
- Mice
- Male
- Apoptosis/drug effects
- Disease Models, Animal
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
Collapse
Affiliation(s)
- Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Manon Mougin
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Mélanie Sauvaget
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Reem El Kabbout
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Chanil Valasarajan
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Keiko Yamamoto
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Andréanne Pelletier
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Coralie Bilodeau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Yann Grobs
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Charlotte Romanet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | | | - Min Lu
- Morphic Therapeutic, Inc, Waltham, MA, USA
| | | | - Mégan Gilbert
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Charlie Théberge
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - François Potus
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, German Center for Lung Research (DZL), Bad Nauheim, Germany; Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Justus Liebig University, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada.
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada; Department of Medicine, Laval University, Québec City, QC, Canada.
| |
Collapse
|
130
|
Liu S, Pi J, Zhang Q. Origins of Ultrasensitivity and Complex Signaling Dynamics of Cellular Hydrogen Peroxide and Peroxiredoxin. Antioxidants (Basel) 2025; 14:235. [PMID: 40002419 PMCID: PMC11852172 DOI: 10.3390/antiox14020235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Hydrogen peroxide (H2O2) plays a crucial role in cell signaling in response to physiological and environmental perturbations. H2O2 can oxidize typical 2-Cys peroxiredoxin (PRX) first into a sulfenic acid, which resolves into a disulfide that can be reduced by thioredoxin (TRX)/TRX reductase (TR). At high levels, H2O2 can also hyperoxidize sulfenylated PRX into a sulfinic acid that can be reduced by sulfiredoxin (SRX). Therefore, PRX, TRX, TR, and SRX (abbreviated as PTRS system here) constitute the coupled sulfenylation and sulfinylation cycle (CSSC), where certain oxidized PRX and TRX forms also function as redox signaling intermediates. Earlier studies have revealed that the PTRS system is capable of rich signaling dynamics, including linearity, ultrasensitivity/switch-like response, nonmonotonicity, circadian oscillation, and possibly, bistability. However, the origins of ultrasensitivity, which is fundamentally required for redox signal amplification, have not been adequately characterized, and their roles in enabling complex nonlinear dynamics of the PTRS system remain to be determined. Through in-depth mathematical modeling analyses, here we revealed multiple sources of ultrasensitivity that are intrinsic to the CSSC, including zero-order kinetic cycles, multistep H2O2 signaling, and a mechanism arising from diminished H2O2 removal at high PRX hyperoxidation state. The CSSC, structurally a positive feedback loop, is capable of bistability under certain parameter conditions, which requires embedding multiple sources of ultrasensitivity identified. Forming a negative feedback loop with cytosolic SRX as previously observed in energetically active cells, the mitochondrial PTRS system (where PRX3 is expressed) can produce sustained circadian oscillations through supercritical Hopf bifurcations. In conclusion, our study provided novel quantitative insights into the dynamical complexity of the PTRS system and improved appreciation of intracellular redox signaling.
Collapse
Affiliation(s)
- Shengnan Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
131
|
Nomoto H, Furusawa S, Yokoyama H, Suzuki Y, Izumihara R, Oe Y, Takahashi K, Miya A, Kameda H, Cho KY, Takeuchi J, Kurihara Y, Nakamura A, Atsumi T. Improvement of β-Cell Function After Switching From DPP-4 Inhibitors to Oral Semaglutide: SWITCH-SEMA2 Post Hoc Analysis. J Clin Endocrinol Metab 2025; 110:e583-e591. [PMID: 38695547 DOI: 10.1210/clinem/dgae213] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Indexed: 02/19/2025]
Abstract
CONTEXT Whether continuation of dipeptidyl peptidase-4 inhibitors (DPP-4is) or switching to oral semaglutide is more beneficial for β-cell function is unclear. OBJECTIVE To assess the efficacy of switching from DPP-4is to oral semaglutide for β-cell function compared with DPP-4i continuation. METHODS Post hoc analysis of SWITCH-SEMA 2, a multicenter prospective randomized controlled trial on the switch to oral semaglutide vs DPP-4i continuation without dose adjustment for 24 weeks in subjects with type 2 diabetes treated with DPP-4is, was conducted. Changes in markers for glucose metabolism, including homeostatic model assessment (HOMA2) scores and disposition index (DI), were compared between the groups. RESULTS A total of 146 subjects (semaglutide group, 69; DPP-4i group, 77) were analyzed. In the semaglutide group, glycemic control, liver enzyme deviations, and lipid profiles improved after 24 weeks. Regarding indices for β-cell function, changes in HOMA2-β as well as DI, reflecting the ability of β-cells to compensate for insulin resistance, were significantly higher in the semaglutide group compared with the DPP-4i group (mean change, +10.4 vs +0.6 in HOMA2-β [P = .001] and +0.09 vs 0.0 in DI [P < .001]). Improvement in DI in the semaglutide group was correlated significantly to changes in body mass index (BMI), HbA1c, and fatty liver index reflecting liver steatosis. Multiple linear regression analysis revealed that dose of semaglutide (≥ 7 mg/day), reduction in fatty liver index, and metformin nonuse were independently associated with improvement of DI. CONCLUSION Switching to oral semaglutide ameliorated β-cell function compared with DPP-4is, presumably via tissue-to-tissue crosstalk between liver and β-cells.
Collapse
Affiliation(s)
- Hiroshi Nomoto
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Sho Furusawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | | | - Yuka Suzuki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Rimi Izumihara
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yuki Oe
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kiyohiko Takahashi
- Division of Diabetes and Endocrinology, Department of Medicine, Hakodate Central General Hospital, Hakodate 040-8585, Japan
| | - Aika Miya
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Hiraku Kameda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kyu Yong Cho
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Jun Takeuchi
- Sapporo Diabetes and Thyroid Clinic, Sapporo 060-0807, Japan
| | | | - Akinobu Nakamura
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
132
|
Usuda J, Yagyu K, Tanaka H, Hori M, Ishikawa K, Takahashi Y. Nanoscale visualization of the anti-tumor effect of a plasma-activated Ringer's lactate solution. Faraday Discuss 2025; 257:212-223. [PMID: 39470167 DOI: 10.1039/d4fd00116h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Plasma-activated Ringer's lactate solutions (PALs), which are Ringer's lactate solutions treated with non-thermal atmospheric-pressure plasma, have an anti-tumor effect and can be used for chemotherapy. As the anti-tumor effect of the PAL is influenced by the cell-treatment time, it is necessary to monitor the structural changes of the cell surface with non-invasive, nanoscale, and time-lapse imaging to understand the anti-tumor effect. In this study, to characterize the anti-tumor effect of the PAL, we used scanning ion conductance microscopy (SICM), using glass nanopipettes as probes, to visualize the structural changes of the cell surface. SICM time-lapse topographic imaging visualized a decrease in the movement of lamellipodia in normal cells and cancer cells after the PAL treatment. Furthermore, in normal cells, protrusive structures were observed on the cell surface. Time-lapse imaging using SICM allowed us to characterize the differences in the morphological changes between the normal and cancer cells upon exposure to the PAL.
Collapse
Affiliation(s)
- Junichi Usuda
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Kenshin Yagyu
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Hiromasa Tanaka
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Masaru Hori
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kenji Ishikawa
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yasufumi Takahashi
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| |
Collapse
|
133
|
Urano Y, Iwagaki A, Takeishi A, Uchiyama N, Noguchi N. Downregulation of the SREBP pathways and disruption of redox status by 25-hydroxycholesterol predispose cells to ferroptosis. Free Radic Biol Med 2025; 228:319-328. [PMID: 39778605 DOI: 10.1016/j.freeradbiomed.2025.01.010] [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: 11/22/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Enzymatically formed side-chain oxysterols function as signaling molecules regulating cholesterol homeostasis and act as intermediates in the biosynthesis of bile acids. In addition to these physiological functions, an imbalance in oxysterol homeostasis has been implicated in pathophysiology. Cholesterol 25-hydroxylase (CH25H) and its product 25-hydroxycholesterol (25-OHC), also formed by autoxidation, are associated with amyotrophic lateral sclerosis. However, the effects of 25-OHC on cell viability in glial cells remain unclear. This study demonstrates that 25-OHC induces ferroptosis, an iron-dependent programmed cell death, in mouse Schwann IMS32 cells. Mechanistically, 25-OHC suppressed the expression of selenoprotein glutathione peroxidase 4 (GPX4) at both the transcriptional and translational levels by inhibiting the processing of sterol regulatory element-binding proteins (SREBPs). In addition, 25-OHC upregulated the expression of NADH-cytochrome b5 reductase 1 (CYB5R1) and NADPH-cytochrome P450 reductase (POR), enzymes that promote lipid peroxidation. We further found that 25-OHC increases the expression of glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) and decreases glutathione levels. Importantly, non-cytotoxic concentrations of 25-OHC enhanced cellular sensitivity to ferroptosis inducers by downregulating GPX4 expression. These findings reveal a multifaceted approach whereby 25-OHC induces ferroptosis through SREBP pathway suppression and redox imbalance in mouse Schwann IMS32 cells.
Collapse
Affiliation(s)
- Yasuomi Urano
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Anan Iwagaki
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Arisa Takeishi
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Nazuna Uchiyama
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Noriko Noguchi
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan.
| |
Collapse
|
134
|
Xu H, Yang L, Wu Y, Lei H. Double-edged sword effect of GPX4 in skin homeostasis and diseases. Arch Dermatol Res 2025; 317:404. [PMID: 39951160 DOI: 10.1007/s00403-025-03903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 05/09/2025]
Abstract
Glutathione peroxidase 4 (GPX4) is a crucial antioxidant enzyme that plays a vital role in protecting cells from oxidative damage and lipid peroxidation. In the context of skin diseases, GPX4 serves as a key regulator of oxidative stress and inflammation, both of which are significant features of various skin conditions. By preventing lipid peroxidation and maintaining membrane integrity, GPX4 acts as a safeguard against cell death pathways, particularly ferroptosis, in skin diseases. Dysregulation of GPX4 in conditions such as dermatitis, psoriasis, and skin cancer is linked to heightened oxidative stress, inflammation, and tissue damage. Understanding the role of GPX4 and its intricate interactions in skin disease pathogenesis can aid in more effectively targeting oxidative stress and inflammation, leading to promising therapeutic interventions. This review summarizes the role of GPX4 in maintaining skin homeostasis and its involvement in disease, proposing strategies to target GPX4, including its post-translational modifications. Investigate the precise mechanism through which GPX4 influences the onset of skin diseases, and utilize GPX4 agonists or inhibitors as potential treatments.
Collapse
Affiliation(s)
- Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathophysiology, Research Unit of Stress and Cancer, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
135
|
Batyrova G, Taskozhina G, Umarova G, Umarov Y, Morenko M, Iriskulov B, Kudabayeva K, Bazargaliyev Y. Unveiling the Role of Selenium in Child Development: Impacts on Growth, Neurodevelopment and Immunity. J Clin Med 2025; 14:1274. [PMID: 40004804 PMCID: PMC11856779 DOI: 10.3390/jcm14041274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Selenium (Se) is a vital trace element for children, playing a crucial role in numerous physiological processes, including antioxidant defense, immune regulation, thyroid function, and bone metabolism. Emerging evidence highlights its potential impact on child development and growth while also underscoring the complexity of its mechanisms and the global variations in Se intake. The aim of this review is to comprehensively elucidate the significance of Se in various biological processes within the human body, with a focus on its role in child development and growth; its biochemical effects on the nervous system, thyroid function, immune system, and bone tissue; and the implications of Se deficiency and toxicity. This review integrates findings from experimental models, epidemiological studies, and clinical trials to explore Se's role in neurodevelopment, growth regulation, and immune competence in children. Selenoproteins, which regulate oxidative stress and thyroid hormone and bone metabolism, are essential for normal growth and cognitive development in children. Se deficiency and toxicity has been linked to impaired immune function, growth retardation, and decreased immune function. The findings underscore Se's influence on various biological pathways that are critical for healthy child development and its broader importance for child health. Public health strategies aimed at optimizing selenium intake may play a pivotal role in improving pediatric health outcomes worldwide.
Collapse
Affiliation(s)
- Gulnara Batyrova
- Department of Clinical Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Gulaim Taskozhina
- Department of Clinical Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Gulmira Umarova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan
| | - Yeskendir Umarov
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Marina Morenko
- Department of Children’s Diseases, Astana Medical University, Astana 010000, Kazakhstan;
| | - Bakhtiyar Iriskulov
- Department of Normal and Pathological Physiology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan;
| | - Khatimya Kudabayeva
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (K.K.); (Y.B.)
| | - Yerlan Bazargaliyev
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (K.K.); (Y.B.)
| |
Collapse
|
136
|
Dixon S, Leak L, Wang Z, Lee WC, Johnson B, Millner A, Ko PJ, Decosto C, Magtanong L, Ritho J, Skouta R, Atilla-Gokcumen E, Myers C, Moffat J, Boone C, Bensinger S, Moding E, Joseph A, Chan A, Chitkara S, Salinas J, Nathanson D. A clinical drug candidate that triggers non-apoptotic cancer cell death. RESEARCH SQUARE 2025:rs.3.rs-4138879. [PMID: 39989975 PMCID: PMC11844650 DOI: 10.21203/rs.3.rs-4138879/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Small molecules that induce non-apoptotic cell death are of fundamental mechanistic interest and may be useful to treat certain cancers. Here, we report that tegavivint, a drug candidate undergoing human clinical trials, can activate a unique mechanism of non-apoptotic cell death in sarcomas and other cancer cells. This lethal mechanism is distinct from ferroptosis, necroptosis and pyroptosis and requires the lipid metabolic enzyme trans-2,3-enoyl-CoA reductase (TECR). TECR is canonically involved in the synthesis of very long chain fatty acids but appears to promote non-apoptotic cell death in response to CIL56 and tegavivint via the synthesis of the saturated long-chain fatty acid palmitate. These findings outline a lipid-dependent non-apoptotic cell death mechanism that can be induced by a drug candidate currently being tested in humans.
Collapse
|
137
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [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: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
138
|
Fan S, Hu Y, Shi J. Role of ferroptosis in atrial fibrillation: a review. Front Pharmacol 2025; 16:1362060. [PMID: 39981174 PMCID: PMC11839810 DOI: 10.3389/fphar.2025.1362060] [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: 12/27/2023] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Cardiovascular disease remains the leading cause of mortality, with atrial fibrillation emerging as one of the most common conditions encountered in clinical practice. However, its underlying mechanisms remain poorly understood, prompting ongoing research. Ferroptosis, a recently discovered form of regulated cell death characterized by lipid peroxidation and disrupted cellular redox balance leading to cell death due to iron overload, has attracted significant attention. Since its identification, ferroptosis has been extensively studied in various contexts, including cancer, stroke, myocardial ischemia/reperfusion injury, and heart failure. Growing evidence suggests that ferroptosis may also play a critical role in the onset and progression of atrial fibrillation, though research in this area is still limited. This article provides a concise overview of the potential mechanisms by which ferroptosis may contribute to the pathogenesis of atrial fibrillation.
Collapse
Affiliation(s)
- Shaowei Fan
- Lugouqiao Second Community Health Service Center, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Jingjing Shi
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
139
|
Kim SG, Keum M, Choe YM, Suh GH, Lee BC, Kim HS, Lee JH, Hwang J, Yi D, Kim JW. Selenium and Episodic Memory: The Moderating Role of Apolipoprotein E ε4. Nutrients 2025; 17:595. [PMID: 39940451 PMCID: PMC11819958 DOI: 10.3390/nu17030595] [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: 01/21/2025] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Selenium (Se), a vital trace element, plays a neuroprotective role by mitigating oxidative stress through selenoproteins and regulating metal balance. The apolipoprotein E ε4 allele (APOE4), a significant genetic risk factor for Alzheimer's disease (AD), has been linked to reduced Se levels and weakened antioxidant capacity. This research explores the association between serum Se concentrations and cognitive performance, with an emphasis on how APOE4 status influences this relationship. Methods: This study included 196 older adults from community and memory clinic settings, who underwent assessments for episodic memory, global cognition, and non-memory functions using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological battery, with serum selenium levels analyzed via inductively coupled plasma-mass spectrometry (ICP-MS) and APOE genotyping conducted to determine allele status. Results: Higher serum Se levels were associated with better episodic memory score (EMS) (B = 0.065, 95% CI = 0.020-0.110, p = 0.005) and CERAD total score (TS) (B = 0.119, 95% CI = 0.046-0.193, p = 0.002). However, the interaction between Se and APOE4 status significantly affected EMS (B = -0.074, 95% CI = -0.109 to -0.039, p < 0.001), with significant benefits observed in APOE4-negative participants. Conclusions: This study highlights the genotype-specific impact of Se on cognitive health, emphasizing the need for personalized nutritional interventions targeting Se levels, particularly for APOE4-negative individuals. Future research should further elucidate the mechanisms of Se's effects and assess its therapeutic potential in aging populations.
Collapse
Affiliation(s)
- Shin Gyeom Kim
- Department of Neuropsychiatry, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Musung Keum
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
| | - Young Min Choe
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| | - Guk-Hee Suh
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| | - Boung Chul Lee
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul 07247, Republic of Korea
| | - Hyun Soo Kim
- Department of Laboratory Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea;
| | - Jun Hyung Lee
- Department of Laboratory Medicine, Green Cross Laboratories (GC Labs), Yongin 16924, Gyeonggi, Republic of Korea;
| | - Jaeuk Hwang
- Department of Psychiatry, Soonchunhyang University Hospital Seoul, Seoul 04401, Republic of Korea;
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul 03080, Republic of Korea;
| | - Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| |
Collapse
|
140
|
Li M, Hu Y, Wu X, Tong J, Tao J, Tang A, Ji Y, Yao Y, Tao F, Liang C. Placental Ferroptosis May Be Involved in Prenatal Arsenic Exposure Induced Cognitive Impairment in Offspring. Biol Trace Elem Res 2025:10.1007/s12011-025-04525-0. [PMID: 39912999 DOI: 10.1007/s12011-025-04525-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025]
Abstract
The association between prenatal arsenic (As) exposure and offspring's cognition is still unclear, and the underlying etiology has also not been elucidated. Based on the Ma'anshan Birth Cohort (MABC) study in China, 1814 mother-child pairs were included in this study, and the association of As levels in cord serum with preschoolers' intelligence scores was explored. To validate the results from population study, in vivo models were adopted to observe the association between prenatal As exposure and spatial learning and memory abilities of mice offsprings. The As-exposure induced ferroptosis in the placenta of human beings as well as C57BL/6 J mice and HTR-8/SVneo cells was explored in order to clarify the potential cause of impairment of offspring's cognition related to As exposure, respectively. In the population study, we observed a significant inverse association between natural logarithm transformed (ln) As levels and preschoolers' intelligence scores, especially for the fluid reasoning index (FRI) [(β (95%CI): - 1.07 (- 1.98, - 0.16)] and working memory index (WMI) [β (95%CI): - 1.51 (- 2.76, - 0.25)]. Meanwhile, the data from in vivo models revealed that the learning and memory abilities of offspring mice decreased after prenatal As exposure. The occurrence of ferroptosis-like characteristics in the placenta and HTR-8/SVneo cells after As exposure was observed, accompanying with evident oxidative stress, iron accumulation, mitochondrial damage, and decreased protein levels of GPX4, xCT, and FTH1 (or FPN1). Notably, the ferroptosis-like alterations induced by NaAsO2 can be effectively alleviated by N-acetylcysteine (NAC) and ferrostatin-1 (Fer-1) treatment in HTR-8/SVneo cells, respectively. In conclusion, prenatal As exposure associates with impairment of offspring's cognition, and placental ferroptosis may be involved in the association. Further studies are needed to confirm the findings.
Collapse
Affiliation(s)
- Mengzhu Li
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yuan Hu
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Jiulongpo District Center for Disease Control and Prevention, Chongqing, 400039, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jiajing Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Anni Tang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yanli Ji
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Yuyou Yao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Chunmei Liang
- School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
141
|
Chen Y, Li H, Peng Y, Li T, Li X, Wang C, Xiao R, Dong J, Du X. Nanoization of Technical Pesticides: Facile and Smart Pesticide Nanocapsules Directly Encapsulated through "On Site" Metal-Polyphenol Coordination Assembly for Improved Efficacy and Biosafety. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2865-2879. [PMID: 39869849 DOI: 10.1021/acs.jafc.4c10948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Facile pesticide nanocapsules were successfully prepared by directly encapsulating the antisolvent precipitation of pesticides through instantaneous "on site" coordination assembly of tannic acid and Fe3+, avoiding tedious preparation, time consumption, and large amounts of organic solvents. The pesticide nanocapsules showed excellent resistance to ultraviolet photolysis and rainwater washing owing to the nanocapsule walls. The smart pesticide nanocapsules exhibited the controlled release of pesticides under multidimensional stimuli, such as acidic/alkaline pH, glutathione, H2O2, phytic acid, laccase, tannase, and sunlight, which were related to the physiological and natural environments of crops, pests, and pathogens. The tebuconazole nanocapsules not only enhanced the fungicidal activity against Fusarium graminearum and effective control efficacy in wheat powdery mildew through foliar spray and seed coating, but also improved the biosafety of target plant growth and nontarget organisms. The facile, smart, efficient, safe, and green pesticide nanocapsules using the universal strategy have broad application prospects in ecoagriculture.
Collapse
Affiliation(s)
- Yuxia Chen
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Hang Li
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuhui Peng
- Henan Engineering Research Center of Green Pesticide Creation & Intelligent Pesticide Residue Sensor Detection, College of Resources and Environment, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Tongtong Li
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiaona Li
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chen Wang
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ruixi Xiao
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jiangtao Dong
- Henan Engineering Research Center of Green Pesticide Creation & Intelligent Pesticide Residue Sensor Detection, College of Resources and Environment, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xuezhong Du
- State Key Laboratory of Coordination Chemistry, Key Laboratory of Mesoscopic Chemistry (Ministry of Education), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
142
|
Zhao G, Li X, Zhang Y, Wang X, Deng L, Xu J, Jin S, Zuo Z, Xun L, Luo M, Yang F, Qi J, Fu P. Intricating connections: the role of ferroptosis in systemic lupus erythematosus. Front Immunol 2025; 16:1534926. [PMID: 39967676 PMCID: PMC11832682 DOI: 10.3389/fimmu.2025.1534926] [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: 11/26/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory and autoimmune disease with multiple tissue damage. However, the pathology remains elusive, and effective treatments are lacking. Multiple types of programmed cell death (PCD) implicated in SLE progression have recently been identified. Although ferroptosis, an iron-dependent form of cell death, has numerous pathophysiological features similar to those of SLE, such as intracellular iron accumulation, mitochondrial dysfunction, lipid metabolism disorders and concentration of damage associated-molecular patterns (DAMPs), only a few reports have demonstrated that ferroptosis is involved in SLE progression and that the role of ferroptosis in SLE pathogenesis continues to be neglected. Therefore, this review elucidates the potential intricate relationship between SLE and ferroptosis to provide a reliable theoretical basis for further research on ferroptosis in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Guowang Zhao
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xinghai Li
- Department of Minimal Invasive Intervention Radiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi, China
| | - Ying Zhang
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xingzi Wang
- Department of Nephrology, Yueyang Central Hospital, Yueyang, Hunan, China
| | - Li Deng
- Department of Internal Medicine, Community Health Service Station of Dian Mian Avenue, Kunming, Yunnan, China
| | - Juan Xu
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Shumei Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kunming, Yunnan, China
| | - Zan Zuo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Linting Xun
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mei Luo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Fan Yang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ping Fu
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
143
|
Lu Y, Li T, Shu Y, Lu C, Luo Z, Wang J, Xiong H, Li W. Lipid peroxidation and sarcopenia: molecular mechanisms and potential therapeutic approaches. Front Med (Lausanne) 2025; 12:1525205. [PMID: 39963429 PMCID: PMC11831367 DOI: 10.3389/fmed.2025.1525205] [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: 11/09/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Sarcopenia is an age-related condition characterized by the progressive loss of skeletal muscle mass and strength. With the global aging population, its incidence is rapidly increasing. Lipid peroxidation is a critical biochemical process that generates reactive oxygen species (ROS), leading to the destruction of muscle cell structure and function. It plays a pivotal role in the onset and progression of sarcopenia. This review summarizes the mechanisms by which lipid peroxidation contributes to sarcopenia, with a focus on its regulatory effects on cell membrane damage, mitochondrial dysfunction, and cell death. In addition, we discuss the protective role of antioxidant factors such as GPX4 (glutathione peroxidase 4) and antioxidant peptides like SS peptides in mitigating lipid peroxidation and delaying the progression of sarcopenia. Finally, the potential of various strategies, including natural compounds, supplements, natural extracts, and lifestyle interventions, in inhibiting lipid peroxidation and promoting muscle health is explored.
Collapse
Affiliation(s)
- Yifan Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Tiao Li
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Yang Shu
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Zhiqiang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Jingrui Wang
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wangyang Li
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
144
|
Plangger I, Mühlsteiger A, Berger J, Feilner J, Wurst K, Koeberle A, Koeberle SC, Magauer T. Development of a Synthetic Platform for Ent-Pimaranes Reveals their Potential as Novel Non-Redox Active Ferroptosis Inhibitors. Chemistry 2025; 31:e202403811. [PMID: 39665294 PMCID: PMC7617283 DOI: 10.1002/chem.202403811] [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: 10/14/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/13/2024]
Abstract
We present a comprehensive account on the evolution of a synthetic platform for a subfamily of ent-pimaranes. For the most complex member, norflickinflimiod C, five distinct strategies relying on either cationic or radical polyene cyclizations to construct the requisite tricyclic carbon scaffold were explored. Insights from early and late stage oxidative and reductive dearomatization studies ultimately led to a mild, rhodium-catalyzed arene hydrogenation for the final synthetic route. A Sharpless asymmetric dihydroxylation was found to be suitable to render the platform enantioselective and diversification of a late-stage key intermediate culminated in the total synthesis of eight ent-pimaranes in 11-16 steps. These compounds were found to inhibit the formation of pro-inflammatory leukotrienes and other 5-lipoxygenase products. Notably, three ent-pimaranes exhibited low micromolar, non-redox active ferroptosis inhibition with remarkable structural specificity.
Collapse
Affiliation(s)
- Immanuel Plangger
- Department of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80–82, 6020Innsbruck (Austria)
| | - Alex Mühlsteiger
- Department of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80–82, 6020Innsbruck (Austria)
| | - Julia Berger
- Michael Popp Institute, University of Innsbruck, Mitterweg 24, 6020Innsbruck (Austria)
| | - Julian Feilner
- Department of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80–82, 6020Innsbruck (Austria)
| | - Klaus Wurst
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80–82, 6020Innsbruck (Austria)
| | - Andreas Koeberle
- Michael Popp Institute, University of Innsbruck, Mitterweg 24, 6020Innsbruck (Austria)
- Pharmacognosy / Institute of Pharmaceutical Sciences, University of Graz, Beethovenstraße 8, 8010Graz (Austria)
| | - Solveigh C. Koeberle
- Michael Popp Institute, University of Innsbruck, Mitterweg 24, 6020Innsbruck (Austria)
- Pharmacognosy / Institute of Pharmaceutical Sciences, University of Graz, Beethovenstraße 8, 8010Graz (Austria)
| | - Thomas Magauer
- Department of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80–82, 6020Innsbruck (Austria)
| |
Collapse
|
145
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
146
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Selenoproteins: Zoom-In to Their Metal-Binding Properties in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:1305. [PMID: 39941073 PMCID: PMC11818150 DOI: 10.3390/ijms26031305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
Selenoproteins contain selenium (Se), which is included in the 21st proteinogenic amino acid selenocysteine (Sec). Selenium (Se) is an essential trace element that exerts its biological actions mainly through selenoproteins. Selenoproteins have crucial roles in maintaining healthy brain activity. At the same time, brain-function-associated selenoproteins may also be involved in neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The selenoproteins GPx4 (glutathione peroxidase 4), GPx1 (glutathione peroxidase 1), SELENOP (selenoprotein P), SELENOK (selenoprotein K), SELENOS (selenoprotein S), SELENOW (selenoprotein W), and SELENOT (selenoprotein T) are highly expressed, specifically in AD-related brain regions being closely correlated to brain function. Only a few selenoproteins, mentioned above (especially SELENOP), can bind transition and heavy metals. Metal ion homeostasis accomplishes the vital physiological function of the brain. Dyshomeostasis of these metals induces and entertains neurodegenerative diseases. In this review, we described some of the proposed and established mechanisms underlying the actions and properties of the above-mentioned selenoproteins having the characteristic feature of binding transition or heavy metals.
Collapse
Affiliation(s)
| | | | - Carmen Beatrice Dogaru
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.M.); (I.S.)
| | | |
Collapse
|
147
|
Dansereau SJ, Sheng J. Heavy Chalcogen Properties of Sulfur and Selenium Enhance Nucleic Acid-Based Therapeutics. Biomolecules 2025; 15:218. [PMID: 40001521 PMCID: PMC11853670 DOI: 10.3390/biom15020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
The Group 16 elements of the periodic table have a characteristic valence shell configuration instrumental to their chemical properties and reactivities. The electrostatic potentials of these so-called chalcogens have been exploited in the design of materials that require the efficient passage of electrons including supermagnets, photocatalytic dyes, and solar panels. Likewise, the incorporation of the heavy chalcogen selenium into organic frameworks has been shown to increase the reactivities of double bonds and heterocyclic rings, while its interactions with aromatic side chains in the hydrophobic core of proteins via selenomethionine impart a stabilizing effect. Typically present in the active site, the hypervalence of selenocysteine enables it to further stabilize the folded protein and mediate electron transfer. Selenium's native occurrence in bacterial tRNA maintains base pair fidelity, most notably during oxidative stress, through its electronic and steric effects. Such native modifications at the positions 2 and 5 of uridine render these sites relevant in the design of RNA-based therapeutics. Innocuous selenium substitution for oxygen in the former and the standard methods of selenium-derivatized oligonucleotide synthesis and detection have led to the establishment of a novel class of therapeutics. In this review, we summarize some progress in this area.
Collapse
Affiliation(s)
| | - Jia Sheng
- Department of Chemistry and the RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
148
|
Jones LR. Intra-host variability of SARS-CoV-2: Patterns, causes and impact on COVID-19. Virology 2025; 603:110366. [PMID: 39724740 DOI: 10.1016/j.virol.2024.110366] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Intra-host viral variability is related to pathogenicity, persistence, drug resistance, and the emergence of new clades. This work reviews the large amount of data on SARS-CoV-2 intra-host variability accumulated to date, addressing known and potential implications in COVID-19 and the emergence of VOCs and lineage-defining mutations. Topics covered include the distribution of intra-host polymorphisms across the genome, the corresponding mutational signatures, their patterns of emergence and extinction throughout infection, and the processes governing their abundance, frequency, and type (synonymous, nonsynonymous, indels, nonsense). Besides, evidence is reviewed that the virus can replicate and mutate in isolation at different anatomical compartments, which may imply that what we have learned from respiratory samples could be part of a broader picture.
Collapse
Affiliation(s)
- Leandro R Jones
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida Rivadavia 1917, C1083ACA Ciudad Autónoma de Buenos Aires, Argentina; Laboratorio de Virología y Genética Molecular (LVGM), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Belgrano 160, Trelew, CP, 9100, Argentina.
| |
Collapse
|
149
|
Varlamova EG, Gudkov SV, Turovsky EA. Differential effect of cerium nanoparticles on the viability, redox-status and Ca 2+-signaling system of cancer cells of various origins. Arch Biochem Biophys 2025; 764:110261. [PMID: 39645139 DOI: 10.1016/j.abb.2024.110261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The present study aims to understand the molecular mechanism underlying the therapeutic effect of cerium nanoparticles (CeNPs) in oncology. Cancer cells were treated with different concentrations of pure nanocerium of different sizes synthesized by laser ablation. Due to the not insignificant influence of surface defects and oxygen species on the ROS-modulating properties of cerium nanoparticles, the nanoparticles were not coated with surfactants or organic molecules during synthesis, which could potentially inhibit a number of pro-oxidative effects. Reactive oxygen species (ROS) production, expression of genes encoding redox-status proteins, selenoproteins and proteins regulating cell death and endoplasmic reticulum stress (ER-stress) were investigated as indicators of the molecular mechanism of cancer cell death. Studies were conducted on the effects of cerium nanoparticles on the Ca2+ signaling system of cancer cells of different origins. Mouse fibroblasts (L-929 cell line) were used as non-cancerous ("normal") cells for which a whole series of experiments were performed, and a comparative analysis of the effects of nanoceria. It was found that 75 nm-sized cerium nanoparticles did not affect the redox-status and ROS production of cancer cells. In fibroblast cells, however, this nanoparticle diameter led to a deterioration of the cellular redox status and ROS production in a wide range of nanoparticle concentrations. Larger nanoparticles (100 nm-sized and 160 nm-sized), on the other hand, showed a different effect on cancer cells of different origins. In mouse fibroblast L-929 cells, however, 100 nm-sized or 160 nm-sized CeNPs acted in a high concentration range to disrupt mitochondrial membrane potential and activate early apoptosis. High concentrations of CeNPs were required to increase ROS production, reduce redox-status and induce apoptosis in human A-172 glioblastoma cells compared to the hepatocellular carcinoma cell line HepG2 and the breast cancer cell line MCF-7. In the A-172 glioblastoma cells, ER-stress was also not activated and their Ca2+ signaling system was activated by a significantly higher concentration of CeNPs, which could also contribute to the formation of tolerance of this cancer cell line to nanoceria. The Ca2+ signaling system of mouse fibroblasts was found to be highly sensitive to activation by nanoceria and the cells produced Ca2+ signals with higher amplitude compared to A-172 and MCF-7 cells.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia
| | - Egor A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| |
Collapse
|
150
|
Liu F, Wang K, Nie J, Deng MG. Unraveling the Link: Mendelian Randomization Reveals Causal Relationship Between Selenium and Metabolic Syndrome. Biol Trace Elem Res 2025; 203:790-798. [PMID: 38776021 DOI: 10.1007/s12011-024-04237-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/15/2024] [Indexed: 11/28/2024]
Abstract
Observational studies have linked selenium and metabolic syndrome (MetS), but the causality remains unclear. Therefore, this study intends to determine the causal relationship between selenium and the risk of MetS and its component features [body mass index (BMI), waist circumference adjusted for BMI (WCadjBMI), triglycerides (TC), HDL-cholesterol (HDL-C), fasting blood glucose (FBG), fasting blood insulin (FBI), systolic blood pressure (SBP), and diastolic blood pressure (DBP)]. This study was designed as the two-sample Mendelian randomization (MR), and genetic variants were obtained from the genome-wide association studies. The inverse variance weighted (IVW) was applied as the primary method, and the MR-Egger, weighted median, and MR-PRESSO were supplemented to assess its robustness. The Bonferroni method was used to correct p-values for multiple tests. Genetically incremented selenium level was related to higher odds ratios of developing the MetS (OR = 1.054, 95% CI = 1.016-1.094, p = 0.0049). As for components, significant causal links were identified between selenium and BMI (β = 0.015, p = 1.321 × 10-5), WCadjBMI (β = 0.033, p = 2.352 × 10-4), HDL-C (β = -0.036, p = 1.352 × 10-8), FBG (β = 0.028, p = 0.001), and FBI (β = 0.028, p = 0.002). No significant association was discovered for SBP (β = -0.076, p = 0.218) and DBP (β = 0.054, p = 0.227). These results were generally supported by the weighted median and MR-PRESSO methods. Our study provided evidence of the causal effect of selenium on MetS risk from the genetic perspective in the European population, and further investigation across diverse populations was warranted.
Collapse
Affiliation(s)
- Fang Liu
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Kai Wang
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, 430000, China
| | - Jiaqi Nie
- Xiaogan Municipal Center for Disease Control and Prevention, Xiaogan, 432000, Hubei, China
| | - Ming-Gang Deng
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei, China.
| |
Collapse
|