1
|
Gao X, Zheng X, Wang X, Li Z, Yang L. Environmental pollutant exposure and adverse neurodevelopmental outcomes: An umbrella review and evidence grading of meta-analyses. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:137832. [PMID: 40068397 DOI: 10.1016/j.jhazmat.2025.137832] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/13/2025] [Accepted: 03/02/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Exposure to environmental pollutants during foetal and childhood development has been associated with neurodevelopmental disorders; however, existing evidence remains fragmented and lacks comprehensive credibility assessments. METHODS PubMed, Embase, PsycINFO, and the Cochrane Database of Systematic Reviews were systematically searched from database inception through January 2025, supplemented by reference list searches. Eligible studies were meta-analyses of observational research examining the associations between environmental pollutant exposure and adverse neurodevelopmental outcomes, with quality assessment performed using the AMSTAR 2 tool. Summary effect estimates were re-analysed using random-effects models, accompanied by heterogeneity I² statistics, 95 % prediction intervals, and evaluations of small-study effects and excess significance bias. Evidence was graded according to established criteria as follows: convincing, highly suggestive, suggestive, weak, or not significant. Reporting adhered to PRISMA guidelines, and the study protocol was pre-registered with PROSPERO (CRD42022339292). RESULTS A total of 45 studies, comprising 256 unique meta-analyses, were included in the umbrella review. Of these, 88 meta-analyses demonstrated statistical significance; however, 62 % of these exhibited small-study effects and/or excess significance bias. According to the quantitative grading criteria, highly suggestive evidence was identified for the association between PM2.5 exposure and attention deficit hyperactivity disorder (ADHD) (odds ratio [OR] 1.82 [1.52, 2.18]), as well as for lead (Pb) exposure (OR 1.96 [1.57, 2.46]) and ADHD. Childhood fluoride exposure was also graded as highly suggestive evidence for cognitive deficit (OR 3.80 [2.42, 5.97]). Suggestive evidence included the associations between pregnancy O3 exposure and autism spectrum disorder (ASD), childhood lead exposure with decreased intelligence quotient (IQ), decreased blood magnesium (Mg) levels in children with ADHD, and first-trimester pregnancy exposure to PM2.5 and ASD. Sensitivity analyses indicated that results derived from the Restricted Likelihood Maximum (REML) estimator demonstrated greater robustness compared to the widely used DerSimonian and Laird (DL) estimator, reflecting a 13 % increase in significant associations. CONCLUSIONS AND RELEVANCE Neurodevelopmental disorders associated with environmental pollutants from industrial and human activities pose a critical public health challenge. Future research necessitates cautious analytical strategies and enhanced methodological transparency to improve the credibility of findings.
Collapse
Affiliation(s)
- Xuping Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Health Commission Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No. 601 Huangpu Road West, Guangzhou, Guangdong 510632, China.
| | - Xiangyu Zheng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Health Commission Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xinyue Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Health Commission Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Zhiwen Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing 100191, China; Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, School of Public Health, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing 100191, China
| | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Health Commission Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
2
|
Dhiman A, Choudhary D, Mehan S, Maurya PK, Sharma AK, Kumar A, Mukherjee R, Gupta S, Khan Z, Gupta GD, Narula AS. Therapeutic potential of Baicalin against experimental obsessive compulsive disorder: Evidence from CSF, blood plasma, and brain analysis. J Neuroimmunol 2025; 403:578598. [PMID: 40168745 DOI: 10.1016/j.jneuroim.2025.578598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Obsessive-Compulsive Disorder (OCD) is a complex neuropsychiatric condition characterized by recurrent obsessions and compulsions, significantly impacting an individual's functionality and quality of life. This study aimed to explore the neuroprotective and therapeutic potential of baicalin, a flavonoid with known antioxidant, anti-inflammatory, and neurotropic properties, in an animal model of OCD induced by 8-OH-DPAT (8HPAT). The research utilized in silico docking studies and in vivo experiments to assess baicalin's interactions with key intracellular targets: SIRT-1, Nrf2, HO-1, and PPAR-gamma, and its effects on neurochemical, neurobehavioral, and histopathological parameters. In silico results indicated a strong binding affinity of baicalin for SIRT-1, Nrf2, HO-1, and PPAR-gamma, suggesting potential regulatory roles in antioxidant and anti-inflammatory pathways. In-vivo findings demonstrated that baicalin, administered at doses of 50 mg/kg and 100 mg/kg, significantly alleviated OCD-like behaviours, including excessive lever pressing, marble burying, and compulsive checking. Baicalin treatment normalized serotonin and dopamine levels and reduced glutamate levels in the brain, restoring neurotransmitter balance. Furthermore, baicalin decreased inflammatory cytokines (TNF-alpha and IL-1 beta), improved complete blood count profile, and gross morphological and histopathological alterations by restoring neuronal density and cellular integrity in affected brain regions. Combining baicalin with fluvoxamine (10 mg/kg) showed synergistic effects, further enhancing neuroprotective outcomes. These results suggest that baicalin holds promise as a potential therapeutic agent for OCD, warranting further clinical investigation to explore its efficacy and underlying mechanisms in human subjects. The findings underscore the importance of targeting intracellular pathways and neurotransmitter systems in developing effective treatments for OCD and related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abhinay Dhiman
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Divya Choudhary
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India.
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Arun Kumar Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Ritam Mukherjee
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India (Affiliated to IK Gujral Punjab Technical University), Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
3
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
4
|
Tu HF, Zierow L, Lennartsson M, Schweitzer S. A scoping review and quality assessment of machine learning techniques in identifying maternal risk factors during the peripartum phase for adverse child development. PLoS One 2025; 20:e0321268. [PMID: 40435283 PMCID: PMC12119027 DOI: 10.1371/journal.pone.0321268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/04/2025] [Indexed: 06/01/2025] Open
Abstract
Maternal exposure to environmental risk factors (e.g., heavy metal exposure) or mental health problems during the peripartum phase has been shown to lead to negative and lasting impacts on child development and life in adulthood. Given the importance of identifying early markers within highly complex and heterogeneous perinatal factors, machine learning techniques emerge as a promising tool. The main goal of the current scoping review was to summarize the evidence on the application of machine learning techniques in predicting or identifying risk factors during peripartum for child development. A critical appraisal was also conducted to evaluate various aspects, including representativeness, data leakage, validation, performance metrics, and interpretability. A systematic search was conducted in PubMed, Web of Science, Scopus, and Google Scholar to identify studies published prior to the 14th of January 2025. Review selection and data extraction were performed by three independent reviewers. After removing duplicates, the searches yielded 10,336 studies, of which 60 studies were included in the final report. Among these 60 machine learning studies, a majority were pattern-focused, using machine learning primarily as a tool to more accurately describe associations between variables, while 16 studies were prediction-focused (26.7%), exploring the predictive performance of their models. For prediction-focused machine learning studies, a diverse range of methodologies was observed. The quality assessment showed that all studies had some important criteria that were not fully met, with deviations ranging from minor to major, limiting the interpretability and generalizability of the reported findings. Future research should aim at addressing these limitations to enhance the robustness and applicability of machine learning models in this field.
Collapse
Affiliation(s)
- Hsing-Fen Tu
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
- Department of Psychology, Uppsala University, Uppsala, Sweden
- Department of Applied Educational Science, Umeå University, Umeå, Sweden
| | - Larissa Zierow
- ESB Business School, Reutlingen University, Reutlingen, Germany
- ifo Institute, CESifo, Munich, Germany
| | | | - Sascha Schweitzer
- ESB Business School, Reutlingen University, Reutlingen, Germany
- Faculty of Law and Economics, University of Bayreuth, Bayreuth, Germany
| |
Collapse
|
5
|
Bebek G, Miyagi M, Wang X, Appleby BS, Leverenz JB, Pillai JA. Protein co-aggregates of dense core amyloid plaques and CSF differ in rapidly progressive Alzheimer's disease and slower sporadic Alzheimer's disease. Alzheimers Res Ther 2025; 17:118. [PMID: 40420296 PMCID: PMC12107742 DOI: 10.1186/s13195-025-01767-x] [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: 02/11/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND The rapidly progressive phenotype of Alzheimer's disease (rpAD) remains a rare and less-studied entity. Therefore, the replication of key results from the rpAD brain and cerebrospinal fluid (CSF) is lacking. METHODS A label-free quantitative LC-MS/MS analysis of proteins co-aggregating with core-amyloid β plaques in fresh frozen tissue (FFT) from medial temporal regions of rpAD ( n = 8 ) neuropathologically characterized at the National Prion Disease Pathology Surveillance Center (NPDPSC), compared with microdissected amyloid plaques from formalin-fixed, paraffin-embedded (FFPE) tissue blocks from patients with rpAD ( n = 22 ) previously published from the NPDPSC cohort, was performed. Matched rpAD CSF from the FFT cases were compared to a previously published proteomic evaluation of CSF in the AD subtype with rapid progression. RESULTS A total of 1841 proteins were characterized in the FFT study, of which 463 were consistently identified in every rpAD patient analyzed. One thousand two hundred eighty-three proteins were shared between the FFT and the prior FFPE study. FFT offered a more comprehensive proteomic profile than the prior FFPE study and prominently included the immune system pathways. Thirty-five proteins were shared in the FFT brain tissue, matched CSF from the same subjects, in which biological processes related to immune response were again notable. These results were validated against prior published proteomic CSF AD data with a faster rate of progression to identify the top 5 potential protein biomarkers of rapid progression in AD CSF. CONCLUSIONS These results support a distinct immune-related proteomic profile in both the brain and the CSF that can be explored as potential biomarkers in the future for the clinical diagnosis of rpAD.
Collapse
Affiliation(s)
- Gurkan Bebek
- Center for Proteomics and Bioinformatics, Department of Nutrition, Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Xinglong Wang
- Department of Pathology, University of Arizona, Tucson, 85721, AZ, USA
| | - Brian S Appleby
- Department of Neurology, National Prion Disease Pathology Surveillance Center, University Hospitals Cleveland Medical Center, Cleveland, 44195, OH, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Department of Neurology, Cleveland, OH, USA
| | - Jagan A Pillai
- Lou Ruvo Center for Brain Health, Neurological Institute, Department of Neurology, Cleveland, OH, USA.
| |
Collapse
|
6
|
Moosavi Zenooz A, Eterafi M, Azarmi Giglou S, Safarzadeh E. Embracing cancer immunotherapy with manganese particles. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01070-9. [PMID: 40397376 DOI: 10.1007/s13402-025-01070-9] [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: 02/15/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
A substance integral to the sustenance and functionality of virtually all forms of life is manganese (Mn), classified as an essential trace metal. Its significance lies in its pivotal role in facilitating metabolic processes crucial for survival. Additionally, Mn exerts influence over various biological functions including bone formation and maintenance, as well as regulation within systems governing immunity, nervous signaling, and digestion. Manganese nanoparticles (Mn-NP) stand out as a beacon of promise within the realm of immunotherapy, their focus honed on intricate mechanisms such as triggering immune pathways, igniting inflammasomes, inducing immunogenic cell death (ICD), and sculpting the nuances of the tumor microenvironment. These minuscule marvels have dazzled researchers with their potential in reshaping the landscape of cancer immunotherapy - serving as potent vaccine enhancers, efficient drug couriers, and formidable allies when paired with immune checkpoint inhibitors (ICIs) or cutting-edge photodynamic/photothermal therapies. Herein, we aim to provide a comprehensive review of recent advances in the application of Mn and Mn-NP in the immunotherapy of cancer. We hope that this review will display an insightful view of Mn-NPs and provide guidance for design and application of them in immune-based cancer therapies.
Collapse
Affiliation(s)
- Ali Moosavi Zenooz
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheil Azarmi Giglou
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
7
|
Khan MA. Iron responsive elements mRNA regulate Alzheimer's amyloid precursor protein translation through iron sensing. Front Aging Neurosci 2025; 17:1483913. [PMID: 40438504 PMCID: PMC12116395 DOI: 10.3389/fnagi.2025.1483913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Iron responsive element (IREs) mRNA and iron regulatory proteins (IRPs) regulate iron homeostasis. 5'-untranslated region motifs of APP IREs fold into RNA stem loops bind to IRP to control translation. Through the 5'-UTR APP IREs, iron overload accelerated the translation of the Alzheimer's amyloid precursor protein (APP). The protein synthesis activator eIF4F and the protein synthesis repressor IRP1 are the two types of proteins that IREs bind. Iron regulates the competitive binding of eIF4F and IRP1 to IRE. Iron causes the IRE and eIF4F to associate with one other, causing the dissociation of IRPs and altered translation. In order to control IRE-modulated expression of APP, messenger RNAs are becoming attractive targets for the development of small molecule therapeutics. Many mRNA interference strategies target the 2-D RNA structure, but messenger RNAs like rRNAs and tRNAs can fold into complicated, three-dimensional structures that add another level of complexity. IREs family is one of the few known 3-D mRNA regulatory elements. In this review, I present IREs structural and functional characteristics. For iron metabolism, the mRNAs encoding the proteins are controlled by this family of similar base sequences. Iron has a similar way of controlling the expression of Alzheimer's APP as ferritin IRE RNA in their 5ÚTR. Further, iron mis regulation by IRPs can be investigated and contrasted using measurements of expression levels of APP, amyloid-β and tau formation. Accordingly, IRE-modulated APP expression in Alzheimer's disease has great therapeutic potential through targeting mRNA structures.
Collapse
Affiliation(s)
- Mateen A. Khan
- Department of Life Science, College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Li J, Liu T, Xian M, Wei J. Therapeutic applications of exercise in neurodegenerative diseases: focusing on the mechanism of SIRT1. Mol Cell Biochem 2025:10.1007/s11010-025-05299-8. [PMID: 40358811 DOI: 10.1007/s11010-025-05299-8] [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: 02/06/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Neurodegenerative diseases comprise a group of central nervous system disorders marked by progressive neuronal degeneration and dysfunction. Their pathogenesis is multifactorial, involving oxidative stress, mitochondrial dysfunction, excitotoxicity, and neuroinflammation. Recent research has highlighted the potential of exercise as a non-pharmacological intervention for both the prevention and treatment of these disorders. In particular, exercise has received growing attention for its capacity to upregulate the expression and activity of SIRT1, a critical mediator of neuroprotection via downstream signaling pathways. SIRT1, a key member of the Sirtuin family, is a nicotinamide adenine dinucleotide (NAD +)-dependent class III histone deacetylase. It plays an essential role in regulating cellular metabolism, energy homeostasis, gene expression, and cellular longevity. In the context of neurodegenerative diseases, SIRT1 confers neuroprotection by modulating multiple signaling cascades through deacetylation, suppressing neuronal apoptosis, and promoting neural repair and regeneration. Exercise enhances SIRT1 expression and activity by increasing NAD + synthesis and utilization, improving intracellular redox balance, alleviating oxidative stress-induced inhibition of SIRT1, and thereby promoting its activation. Moreover, exercise may indirectly modulate SIRT1 function by influencing interacting molecular networks. This review summarizes recent advances in the therapeutic application of exercise for neurodegenerative diseases, with a focus on SIRT1 as a central mechanism. It examines how exercise mediates neuroprotection through the regulation of SIRT1 and its associated molecular mechanisms and signaling pathways. Finally, the paper discusses the potential applications and challenges of integrating exercise and SIRT1-targeted strategies in the management of neurodegenerative diseases, offering novel perspectives for the development of innovative treatments and improvements in patients' quality of life.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China.
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
9
|
Goldmann U, Wiedmer T, Garofoli A, Sedlyarov V, Bichler M, Haladik B, Wolf G, Christodoulaki E, Ingles-Prieto A, Ferrada E, Frommelt F, Teoh ST, Leippe P, Onea G, Pfeifer M, Kohlbrenner M, Chang L, Selzer P, Reinhardt J, Digles D, Ecker GF, Osthushenrich T, MacNamara A, Malarstig A, Hepworth D, Superti-Furga G. Data- and knowledge-derived functional landscape of human solute carriers. Mol Syst Biol 2025:10.1038/s44320-025-00108-2. [PMID: 40355757 DOI: 10.1038/s44320-025-00108-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
The human solute carrier (SLC) superfamily of ~460 membrane transporters remains the largest understudied protein family despite its therapeutic potential. To advance SLC research, we developed a comprehensive knowledgebase that integrates systematic multi-omics data sets with selected curated information from public sources. We annotated SLC substrates through literature curation, compiled SLC disease associations using data mining techniques, and determined the subcellular localization of SLCs by combining annotations from public databases with an immunofluorescence imaging approach. This SLC-centric knowledge is made accessible to the scientific community via a web portal featuring interactive dashboards and visualization tools. Utilizing this systematically collected and curated resource, we computationally derived an integrated functional landscape for the entire human SLC superfamily. We identified clusters with distinct properties and established functional distances between transporters. Based on all available data sets and their integration, we assigned biochemical/biological functions to each SLC, making this study one of the largest systematic annotations of human gene function and a potential blueprint for future research endeavors.
Collapse
Affiliation(s)
- Ulrich Goldmann
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andrea Garofoli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Manuel Bichler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ben Haladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Evandro Ferrada
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Fabian Frommelt
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Shao Thing Teoh
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Leippe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Gabriel Onea
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Daniela Digles
- University of Vienna, Department of Pharmaceutical Sciences, Vienna, Austria
| | - Gerhard F Ecker
- University of Vienna, Department of Pharmaceutical Sciences, Vienna, Austria
| | | | | | | | | | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
- Fondazione Ri.MED, Palermo, Italy.
| |
Collapse
|
10
|
Li J, Li X, He Y, Huang Y, Wang W, Du H, Chen C, Zhu D, Zhou X. Associations of multiple trace elements with bipolar disorder in adolescents: A case-control study. PLoS One 2025; 20:e0322958. [PMID: 40327654 PMCID: PMC12054906 DOI: 10.1371/journal.pone.0322958] [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: 10/28/2024] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Bipolar disorder (BD) is a serious mental disorder. Studies have shown an association between trace elements and mental disorders. However, this association has not been thoroughly studied in adolescents with BD. We aimed to investigated the associations between multiple trace elements and adolescent BD. METHOD This case-control study included 144 BD patients with BD and 144 matched controls. Seventeen elements in the participants' urine were measured using inductively coupled plasma mass spectrometry (ICP-MS). Least absolute shrinkage and selection operator (LASSO), multivariate logistic regression, restricted cubic spline (RCS), and Bayesian kernel machine regression (BKMR) were used to analyze the association between exposure to single and mixed elements and adolescent BD. RESULTS In the single-element models, titanium, manganese, rubidium, and iodine were negatively associated with adolescent BD. In the multi-element model selected by LASSO, titanium (OR = 0.14, 95% CI: 0.04-0.53), manganese (OR = 0.02, 95% CI: 0.01-0.08), and iodine (OR = 0.06, 95% CI: 0.02-0.22) showed a negative correlation with adolescent BD, while magnesium (OR = 11.24, 95% CI: 1.83-69.12), and nickel (OR = 6.86, 95% CI: 1.55-30.29) displayed a positive correlation. The RCS results showed a non-linear correlation between the elements titanium, manganese, iodine, magnesium, nickel, zinc, strontium and adolescent BD. In addition, the BKMR analysis showed a significant joint effect of multiple elements on adolescent BD when the concentrations of the seven elements were at or above the 55th percentile, compared with their median values. CONCLUSIONS Our findings revealed that urinary titanium, manganese, and iodine were negatively correlated with adolescent BD, whereas urinary magnesium and nickel were positively correlated with adolescent BD. These results provide evidence of an association between urinary trace elements and adolescent BD.
Collapse
Affiliation(s)
- Jie Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xuemei Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Yuqian He
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Yajie Huang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Wenjing Wang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Hang Du
- Chongqing prevention and treatment center for occupational diseases, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Dan Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyu Zhou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Martins AC, Oliveira-Paula GH, Tinkov AA, Skalny AV, Tizabi Y, Bowman AB, Aschner M. Role of manganese in brain health and disease: Focus on oxidative stress. Free Radic Biol Med 2025; 232:306-318. [PMID: 40086492 PMCID: PMC11985276 DOI: 10.1016/j.freeradbiomed.2025.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Manganese (Mn) is an essential trace element crucial for various physiological processes, but excessive exposure can lead to significant health concerns, particularly neurotoxicity. This review synthesizes current knowledge on Mn-induced oxidative stress and its role in cellular dysfunction and disease. We discuss how Mn promotes toxicity through multiple mechanisms, primarily through reactive oxygen species (ROS) generation, which leads to oxidative stress and disruption of cellular processes. The review examines key pathways affected by Mn toxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome activation, and epigenetic modifications. Recent studies have identified promising therapeutic compounds, including both synthetic and natural substances such as probucol, metformin, curcumin, resveratrol, and daidzein, which demonstrate protective effects through various mechanisms, including antioxidant enhancement, mitochondrial function preservation, and epigenetic pathway modulation. Understanding these mechanisms provides new insights into potential therapeutic strategies for Mn-induced disorders. This review also highlights future research directions, emphasizing the need for developing targeted therapies and investigating combination approaches to address multiple aspects of Mn toxicity simultaneously.
Collapse
Affiliation(s)
- Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gustavo H Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460000, Russia; IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, 20059, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
12
|
Guo Y, Li J, Liu X, Ding H, Zhang W. Potential therapeutic targets for ischemic stroke in pre-clinical studies: Epigenetic-modifying enzymes DNMT/TET and HAT/HDAC. Front Pharmacol 2025; 16:1571276. [PMID: 40356977 PMCID: PMC12066669 DOI: 10.3389/fphar.2025.1571276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and disability worldwide, driven by genetic predispositions and environmental interactions, with epigenetics playing a pivotal role in mediating these processes. Specific modifying enzymes that regulate epigenetic changes have emerged as promising targets for IS treatment. DNA methyltransferases (DNMTs), ten-eleven translocation (TET) dioxygenases, histone acetyltransferases (HATs), and histone deacetylases (HDACs) are central to epigenetic regulation. These enzymes maintain a dynamic balance between DNA methylation/demethylation and histone acetylation/deacetylation, which critically influences gene expression and neuronal survival in IS. This review is based on both in vivo and in vitro experimental studies, exploring the roles of DNMT/TET and HAT/HDAC in IS, evaluating their potential as therapeutic targets, and discussing the use of natural compounds as modulators of these enzymes to develop novel treatment strategies.
Collapse
Affiliation(s)
- Yurou Guo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaodan Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Huang Ding
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Wei Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| |
Collapse
|
13
|
Pan D, Lin C, Fu S. A systematic review and meta-analysis of the self-reported pubertal development scale's applicability to children. J Pediatr Endocrinol Metab 2025:jpem-2024-0407. [PMID: 40256953 DOI: 10.1515/jpem-2024-0407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/30/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Assessing pubertal stage can be challenging, particularly in large-scale settings, due to the sensitive nature of Tanner staging by healthcare providers (HCP) or self-reported Tanner stage through photographs or line drawings. The self-reported Pubertal Development Scale (PDS) avoids sensitive issues like genitalia or nudity, is adaptable to various settings, reduces time and cost burdens on researchers. This study aimed to explore the agreement between self-reported PDS and HCP-assessed Tanner staging. CONTENT Papers for the meta review were retrieved from Pubmed, Embase, Fang Wan, CNKI, and Cochrane Library before January 15, 2025. Quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 tool. Pooled estimates and 95 % confidence intervals (CI) were calculated using random-effects models. SUMMARY AND OUTLOOK Five studies with 6024 participants met inclusion criteria. Among stage 1-5, substantial agreement was found among girls (Wk: 0.63[0.62-0.65]) and overall participants (Wk: 0.68[0.67-0.69]), while moderate agreement was observed in boys (Wk: 0.58[0.56-0.61]). Broadening puberty criteria to stages I-III showed substantial agreement for girls (Wk: 0.66[0.64-0.68]), boys (Wk: 0.64[0.61-0.67]), and overall participants (Wk: 0.69[0.67-0.70]). For pubertal onset, using Tanner stage as the gold standard, girls showed that the area under the receiver operating characteristic curve (AUC) was 0.86(0.85-0.87), the sensitivity and positive predictive value (PPV) of self-reported PDS were 0.85 and 84.2 % respectively. Similarly, among boys, the AUC was 0.89 (95 % CI: 0.87-0.92), the sensitivity and PPV were 0.91 and 97.8 % respectively. Our findings indicate moderate to substantial agreement between the two methods, with high sensitivity and PPV for self-reported PDS in assessing puberty onset, PDS may be a reliable and cost-effective method for large-scale epidemiological studies.
Collapse
Affiliation(s)
- Dongxue Pan
- Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Cuilan Lin
- Bo'ai Hospital of Zhongshan, Zhongshan, Guangdong Province, P.R. China
| | - Simao Fu
- Zhongshan People's Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
14
|
Ba H, Hu P, Yuan H, Ma C, Wang Z, Shang Y, Guo Q, Wang D, Li C. RXFP2-positive mesenchymal stem cells in the antlerogenic periosteum contribute to postnatal development of deer antlers. Commun Biol 2025; 8:645. [PMID: 40263536 PMCID: PMC12015367 DOI: 10.1038/s42003-025-08085-w] [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: 11/03/2024] [Accepted: 04/12/2025] [Indexed: 04/24/2025] Open
Abstract
The postnatal development of secondary sexual characteristics is a highly complex process governed by diverse molecular signals and serves as a key marker of sexual maturity. Deer antlers exemplify such traits, distinguished not only by their unique ability to regenerate annually but also by their initiation in postnatal life. It is well established that the antlerogenic periosteum (AP) is the only tissue responsible for postnatal antler formation. Here, we identify a population of RXFP2-positive mesenchymal stem cells within the AP of both male and female deer that are crucial for antler development, primarily through the activation of canonical Wnt signaling. This process also relies on M2 macrophages recruited via IL-34 secretion. Furthermore, these cells exhibit reduced expression of HOX genes, suggesting a high degree of developmental plasticity. Our findings offer new insights into the molecular mechanisms underlying the postnatal development of secondary sexual characteristics, using deer antlers serving as a model system.
Collapse
Affiliation(s)
- Hengxing Ba
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China.
| | - Pengfei Hu
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China
| | - Hongming Yuan
- Jilin Provincial Key Laboratory of Institute of Zoonoses, Animal Genome Editing Technology Innovation Center, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Chao Ma
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China
| | - Zhen Wang
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China
| | - Yudong Shang
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China
| | - Qianqian Guo
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China
| | - Datao Wang
- Institute of Special Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, 130112, China
| | - Chunyi Li
- Jilin Provincial Key Laboratory of Deer Antler Biology, Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
15
|
Li Y, Peng JC, Fang YY, Qin LM, Aschner M, Jiang YM. Effects of Subchronic Manganese and Iron Exposure, Alone or in Combination, on Elemental Distribution in Rats. Biol Trace Elem Res 2025:10.1007/s12011-025-04626-w. [PMID: 40261544 DOI: 10.1007/s12011-025-04626-w] [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: 01/03/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Overexposure to manganese (Mn) or iron (Fe) may lead to neurological damage. The aim of this study was to investigate the effects of subchronic Mn and Fe exposure, alone or in combination, on the distribution of other elements and the relationship between Mn and Fe levels in whole blood and brain. Forty male Sprague-Dawley (SD) rats were divided into control, Mn-exposed, Fe-exposed, and combined Mn-Fe-exposed groups, with 10 rats assigned randomly to each group. The control, Mn-exposed, Fe-exposed group and the combined Mn-Fe-exposed groups were injected intraperitoneally with equal amounts of saline, 5 mg/kg MnCl2, 20 mg/kg FeSO4 or 5 mg/kg MnCl2+20 mg/kg FeSO4 once a day, 5 days a week for 8 weeks. The levels of Mn, Fe and other metallic elements [including barium (Ba), beryllium (Be), strontium (Sr), antimony (Sb), lead (Pb), vanadium (V) and copper (Cu)] in whole blood and brain tissue (including the globus pallidus, hippocampus, striatum and substantia nigra) were determined by inductively coupled plasma-mass spectrometry (ICP-MS). The results of this study show that, in whole blood, Mn levels were increased (p < 0.05) in the Mn-exposed group, Fe levels were decreased in both the Fe-exposed (p < 0.05) and Mn-exposed groups (p < 0.01), and Sb levels were increased in both the Mn-exposed and combined Mn-Fe-exposed groups (p < 0.05). In the substantia nigra, the levels of Be (p < 0.01), Sr (p < 0.05), and Cu (p < 0.001) were increased in the Fe-exposed group; the levels of Cu were also significantly increased in the Mn-exposed group (p < 0.01) and the combined Mn-Fe-exposed group (p < 0.0001); the levels of V were decreased (p < 0.05) in the combined Mn-Fe-exposed group; and the levels of V were decreased in the Fe-exposed group (p < 0.01 ), Mn-exposed group (p < 0.05) and combined Mn-Fe-exposed group (p < 0.001) had decreased Ba levels. In the pallidum, Fe levels were increased in the Mn-Fe co-exposed group (p < 0.0001); Ba (p < 0.01) and Pb (p < 0.05) levels were decreased in the Fe-exposed group; Ba (p < 0.05) levels were decreased in the Mn-exposed group; and Ba levels were increased in the Mn-Fe co-exposed group (p < 0.05). In the hippocampus, Mn (p < 0.01), Cu (p < 0.05), Sb (p < 0.01), and V (p < 0.05) levels were increased in the Fe-exposed group; Mn levels were increased in the Mn-exposed group (p < 0.01) and the combined Mn-Fe-exposed group (p < 0.0001). In the striatum, Be levels were decreased in the Mn-Fe combined exposure group (p < 0.05). Mn and Fe levels in whole blood and brain tissue can reflect the accumulation of Mn and Fe. These measurements can serve as valuable predictive biomarkers for subchronic Mn or Fe exposure and combined Mn-Fe exposure. The interactions between Mn and Fe and the distribution and abnormalities of the essential metal elements in the central nervous system and other organs need to be further investigated.
Collapse
Affiliation(s)
- Yan Li
- Guangxi Zhuang Autonomous Region Institute for the Prevention and Treatment of Occupational Disease, Nanning, 530021, China
| | - Jian-Chao Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
| | - Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China
| | - Li-Mei Qin
- Guangxi Zhuang Autonomous Region Institute for the Prevention and Treatment of Occupational Disease, Nanning, 530021, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China.
- Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Guangxi Medical University, No. 22, Shuang-yong Rd, Nanning, 530021, Guangxi, China.
| |
Collapse
|
16
|
Warden AS, Sharma N, Hutchens S, Liu C, Haggerty NR, Gurol KC, Jursa T, Smith DR, Dayne Mayfield R, Mukhopadhyay S. Elevated brain manganese induces motor disease by upregulating the kynurenine pathway of tryptophan metabolism. Proc Natl Acad Sci U S A 2025; 122:e2423628122. [PMID: 40244671 PMCID: PMC12036984 DOI: 10.1073/pnas.2423628122] [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: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
Elevated brain levels of the essential metals manganese (Mn), copper, or iron induce motor disease. However, mechanisms of metal-induced motor disease are unclear and treatments are lacking. Elucidating the mechanisms of Mn-induced motor disease is particularly important because occupational and environmental Mn overexposure is a global public health problem. To address this, here we combined unbiased transcriptomics and metabolomics with functional studies in a mouse model of human environmental Mn exposure. Transcriptomics unexpectedly revealed that Mn exposure up-regulated expression of metabolic pathways in the brain and liver. Notably, genes in the kynurenine pathway of tryptophan metabolism, which produces neuroactive metabolites that impact neurological function, were up-regulated by Mn. Subsequent unbiased metabolomics revealed that Mn treatment altered kynurenine pathway metabolites in the brain and liver. Functional experiments then demonstrated that pharmacological inhibition of the first and rate-limiting step of the kynurenine pathway fully rescued Mn-induced motor deficits. Finally, elevated Mn directly activates hypoxia-inducible factor (HIF) transcription factors, and additional mechanistic assays identified a role for HIF1, but not HIF2, in regulating expression of hepatic kynurenine pathway genes under physiological or Mn exposure conditions, suggesting that Mn-induced HIF1 activation may contribute to the dysregulation of the kynurenine pathway in Mn toxicity. These findings (1) identify the upregulation of the kynurenine pathway by elevated Mn as a fundamental mechanism of Mn-induced motor deficits; (2) provide a pharmacological approach to treat Mn-induced motor disease; and (3) should broadly advance understanding of the general principles underlying neuromotor deficits caused by metal toxicity.
Collapse
Affiliation(s)
- Anna S. Warden
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Nishant Sharma
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Steven Hutchens
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Chunyi Liu
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Noah R. Haggerty
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Kerem C. Gurol
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Roy Dayne Mayfield
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
17
|
Mateus G, Ferreira-Pêgo C, Assunção R. Mapping the Quantitative Dose-Response Relationships Between Nutrients and Health Outcomes to Inform Food Risk-Benefit Assessment. Foods 2025; 14:1420. [PMID: 40282821 PMCID: PMC12026834 DOI: 10.3390/foods14081420] [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: 03/26/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
In the context of the global food system transformation towards sustainable and healthy diets, risk-benefit assessment supported by quantitative dose-response relationships between nutrients and health outcomes is crucial for evaluating the public health impact of dietary modifications, particularly those involving novel foods. The primary aim of this review was to establish a foundational basis for RBA by compiling and synthesising quantitative dose-response relationships identified through a comprehensive literature review. This review encompassed the last 15 years up to March 2025, utilising databases such as PubMed, Scopus, and Web of Science. This review prioritised recent meta-analyses of observational studies and randomised controlled trials with a low risk of bias, as assessed using the ROBIS tool. This review identified significant dose-response relationships across 12 nutrients and their associations with various health outcomes. While dietary fibre has demonstrated broad protective effects, cereal fibre has been found to be the most beneficial for colorectal cancer prevention. Calcium has been shown to have inverse associations with several cancers, although high dairy intake may increase the risk of prostate cancer. Haem iron was linked to an increased risk of several chronic diseases, whereas non-haem iron showed less consistent associations. Zinc exhibited a potential U-shaped relationship with colorectal cancer risk. These findings underscore the complexity of nutrient-health relationships, highlighting the importance of considering dose-response gradients and nutrient sources. Although this overview primarily summarises quantitative associations without exploring in-depth mechanistic or population-specific details, it underscores the complexity of nutrient effects, including nonlinearity and source dependency. This analysis offers a valuable foundation for future risk-benefit assessments of various food scenarios, thereby informing dietary recommendations and public health strategies.
Collapse
Affiliation(s)
- Gabriel Mateus
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Caparica, 2829-511 Almada, Portugal;
| | - Cíntia Ferreira-Pêgo
- Centro Cardiovascular da Universidade de Lisboa, CCUL (CCUL@RISE), Centro Académico de Medicina de Lisboa, CAML, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
- CBIOS—Universidade Lusófona’s Research Center for Biosciences and Health Technologies, Av. Campo Grande 376, 1749-024 Lisbon, Portugal
| | - Ricardo Assunção
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Caparica, 2829-511 Almada, Portugal;
- Food and Nutrition Department, National Institute of Health Doutor Ricardo Jorge (INSA, IP), Av. Padre Cruz, 1649-016 Lisbon, Portugal
| |
Collapse
|
18
|
Asle-Rousta M, Abdollahi M, Aghajari HM, Peirovy Y. Eucalyptol Attenuates Lead-Induced Anxiety-like Behaviors by Suppressing Oxidative Stress and Neuroinflammation, Modulating SIRT1/NF-κB Signaling, and Upregulating BDNF Expression. Biol Trace Elem Res 2025:10.1007/s12011-025-04616-y. [PMID: 40205256 DOI: 10.1007/s12011-025-04616-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Lead is an environmental pollutant that possesses harmful effects on the nervous system. The current study was conducted to investigate the impacts of eucalyptol, a type of monoterpene, on anxiety behaviors, oxidative stress, neuroinflammation, and neuronal death in the hippocampus of rats exposed to lead and its possible protective mechanism. Adult male Wistar rats were divided into Control, Lead, Eucalyptol, and Lead + Eucalyptol groups. The Lead and Lead + Eucalyptol groups were given lead acetate (25 mg/kg, gavage) daily for fourteen days. The Eucalyptol and Lead + Eucalyptol groups also received eucalyptol (100 mg/kg, gavage). The results showed that eucalyptol prevented an increase in malondialdehyde levels and a decrease in glutathione levels, as well as a reduction in the activity of superoxide dismutase and glutathione peroxidase enzymes in the hippocampus of Lead + Eucalyptol animals. It also prevented an increase in the expression of pro-inflammatory cytokines tumor necrosis factor-α, interleukin (IL)- 1β, and IL- 6, and a decrease in the expression of anti-inflammatory cytokine IL- 10. In addition, this monoterpene prevented the reduction in sirtuin 1 (SIRT1) expression and the increase in nuclear factor kappa b (NF-κB) expression. It enhanced the expression of brain-derived neurotrophic factor (BDNF) at the level of mRNA and protein and reduced neuronal death in different subfields of the hippocampus. Eucalyptol also improved the performance of rats receiving lead acetate in elevated plus maze and open field tests. We concluded that eucalyptol reduces anxiety behaviors in lead-exposed rats by suppressing oxidative stress, neuroinflammation, and neuronal death in the hippocampus. The anxiolytic effect of eucalyptol in lead pollution is likely mediated by modulating SIRT1/NF-κB signaling and increasing BDNF expression.
Collapse
Affiliation(s)
| | - Mojdeh Abdollahi
- Nanobiotechnology Research Center, Za.C., Islamic Azad University, Zanjan, Iran
| | | | - Yasaman Peirovy
- Nanobiotechnology Research Center, Za.C., Islamic Azad University, Zanjan, Iran
| |
Collapse
|
19
|
Zhao Z, Peng Y, Yang Y, Li S, Ling J, Zhu Z, He C. ATP13A2 as a prognostic biomarker and its correlation with immune infiltration in cervical cancer: A retrospective study. J Cell Mol Med 2025; 29:e70097. [PMID: 40197818 PMCID: PMC11976316 DOI: 10.1111/jcmm.70097] [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: 02/20/2024] [Revised: 06/11/2024] [Accepted: 09/03/2024] [Indexed: 04/10/2025] Open
Abstract
While the oncogene ATP13A2 is reportedly involved in colorectal cancer, its role in cervical cancer (CC) has yet to be fully characterized. In this study, we investigated ATP13A2 as a potential prognostic biomarker of CC. To this end, we compared CC tissues with normal tissues to identify differentially expressed genes, identifying ATP13A2 as a potential marker of CC. Elevated ATP13A2 expression levels were identified in CC samples compared to noncancerous samples across various data sets, with further immunohistochemical validation. Functional enrichment analysis revealed that ATP13A2 plays an essential role in the CXCL12-activated CXCR4 signalling pathway and chemotaxis regulation, which may alter immune infiltration. Notably, increased ATP13A2 levels were associated with poor overall survival. Furthermore, multiple clinical characteristics were significantly associated with ATP13A2 expression. Additionally, tumour bacterial infiltration was assessed using weighted co-expression network analysis, revealing a relationship between ATP13A2 expression and bacteria in the CC tumour microenvironment. Our results suggest that ATP13A2 is a promising diagnostic and prognostic marker for CC. However, further large-scale studies are needed to fully elucidate the mechanisms underlying the involvement of ATP13A2 in CC.
Collapse
Affiliation(s)
- Zhi Zhao
- Zhengzhou Yihe Hospital, Postdoctoral Innovation Practice BaseHenan UniversityZhengzhouHenanChina
- Department of Breast SurgeryGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Yijie Peng
- Department of Hepatobiliary and Pancreatic SurgeryThe Central Hospital of ShaoyangShaoyangHunanChina
| | - Yuanyuan Yang
- Clinical Research Center for Women's Reproductive Health in Hunan ProvinceChangshaHunanChina
- Reproductive Medicine CenterXiangya Hospital of Central South UniversityChangshaChina
| | - Shuaiyu Li
- School of Information ScienceKyushu UniversityFukuokaJapan
| | - Jiang Ling
- Department of Forensic Science, School of Basic Medical SciencesCentral South UniversityChangshaHunanChina
| | - Zhenyu Zhu
- Department of Breast SurgeryGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Chenfeng He
- Department of Integrative BioanalyticsInstitute of Development, Aging and Cancer (IDAC), Tohoku UniversitySendaiJapan
| |
Collapse
|
20
|
Wang J, Wang DZ, Li BH, Yang S, Guo FQ, Zheng B, Wang JH. Elevated circulating cathepsin S levels are associated with cognitive decline and neurodegeneration in a cohort of patients reporting memory complaints. J Alzheimers Dis 2025; 104:1147-1153. [PMID: 40095668 DOI: 10.1177/13872877251322809] [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] [Indexed: 03/19/2025]
Abstract
BackgroundAs a member of the cysteine protease family, cathepsin S has been implicated in the pathogenesis of various diseases, including Alzheimer's disease (AD), primarily by promoting inflammation.ObjectiveCurrent evidence regarding the role of cathepsin S primarily comes from animal studies. This study aims to explore the clinical relevance of cathepsin S in AD.MethodsIn a cohort of older adults aged 60 or above with memory complaints, we examined baseline plasma levels of cathepsin S and assessed their association with cognitive decline and biomarkers of neurodegeneration during a 36-month follow-up.ResultsPlasma levels of cathepsin S were significantly higher in individuals experiencing longitudinal cognitive decline compared to those without cognitive decline. Furthermore, plasma levels of cathepsin S were associated with declines in Mini-Mental State Examination (MMSE) scores and increases in neurofilament light and pTau181 levels. Higher plasma cathepsin S levels were linked to an increased risk of longitudinal cognitive decline (decrease in MMSE scores of 3 or more), adjusting for age, sex, education, APOE genotype, alcohol consumption, smoking, and comorbidities.ConclusionsThis study provides additional evidence supporting the potential role of cathepsin S in the pathogenesis of AD from a clinical perspective.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Ya'an People's Hospital, Ya'an, Sichuan Province, China
| | - Duo-Zi Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Bing-Hu Li
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Shu Yang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Fu-Qiang Guo
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Bo Zheng
- Department of Neurology, Ya'an People's Hospital, Ya'an, Sichuan Province, China
| | - Jian-Hong Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| |
Collapse
|
21
|
Ouisselsat M, El Maouaki A, Maidoumi S, François Y, Pineau A, Sedki A. Assessment of Essential and Toxic Element Levels in the Toenails of Children with Autism Spectrum Disorder. Biol Trace Elem Res 2025; 203:2394-2408. [PMID: 39042314 DOI: 10.1007/s12011-024-04319-w] [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: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Autism spectrum disorder (ASD) has become a global public health concern, impacting the quality of life. The question of gene-environment interaction in the emergence of ASD remains a subject of ongoing debate, and exploring its pathophysiology is thoroughly related to metals as a risk factor. Therefore, this study aims to assess the levels of toxic (Al, Cd, Hg, and Pb) and essential (Cr, Mn, Fe, Ni, Cu, Zn, and Se) elements in toenail samples collected in children with ASD and neurotypical children, by ICP-MS. Parallelly, we will discuss the use of toenails as an exposure indicator. The study involved 208 children aged 3 to 14 from Marrakech, Morocco. One hundred two were diagnosed with ASD and 106 were neurotypical children. Significant statistical differences in the concentration of Cr, Mn, and Fe were documented between the two groups. Higher levels of Pb in toenails compared to reference values have been reported. No association was established between concentrations of elements and age. Spearman correlation coefficients revealed a significantly different pattern of mutual dependence for toxic and essential elements between the two groups. The strongest positive correlations were found in the neurotypical group (Fe-Mn (ρ = 0.750), and Se-Zn (ρ = 0.800)). These results provide additional, although inconclusive, evidence on the probable role of element disturbance in the pathogenesis of ASD. Further studies should be performed to explore other nutritional, cultural, sociodemographic, environmental, and methodological factors that may impact the levels of these elements in the nails and their possible correlation with the incidence of ASD.
Collapse
Affiliation(s)
- Mariam Ouisselsat
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco.
| | - Amal El Maouaki
- Laboratory of Clinical, Experimental and Environmental Neurosciences, Faculty of Medicine and Pharmacy, Cadi Ayyad University, 274, 40000, Marrakech, Morocco
| | - Sana Maidoumi
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco
- Higher Institute of Nursing Professions and Health Techniques, Rue Abdelouahab Derraq, 40000, Marrakech, Morocco
| | - Yannick François
- Mineral Element Dosing Centre, UFR of Pharmaceutical and Biological Sciences, University of Nantes, 9, Rue Bias, 44035 - 44000, Nantes Cedex 1, France
| | - Alain Pineau
- Mineral Element Dosing Centre, UFR of Pharmaceutical and Biological Sciences, University of Nantes, 9, Rue Bias, 44035 - 44000, Nantes Cedex 1, France
| | - Azeddine Sedki
- Laboratory of Water, Biodiversity and Climate Change, Department of Biology, Faculty of Sciences - Semlalia, Cadi Ayyad University, Bd Moulay Abdellah BP, 2390-40001, Marrakech, Morocco
| |
Collapse
|
22
|
Das S, Murumulla L, Ghosh P, Challa S. Heavy metal-induced disruption of the autophagy-lysosomal pathway: implications for aging and neurodegenerative disorders. Biometals 2025; 38:371-417. [PMID: 39960543 DOI: 10.1007/s10534-025-00665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/19/2025] [Indexed: 04/03/2025]
Abstract
Heavy metals such as lead, mercury, cadmium, magnesium, manganese, arsenic, copper pose considerable threats to neuronal health and are increasingly recognized as factors contributing to aging-related neurodegeneration. Exposure to these environmental toxins disrupts cellular homeostasis, resulting in oxidative stress and compromising critical cellular processes, particularly the autophagy-lysosomal pathway. This pathway is vital for preserving cellular integrity by breaking down damaged proteins and organelles; however, toxicity from heavy metals can hinder this function, leading to the buildup of harmful substances, inflammation, and increased neuronal injury. As individuals age, the consequences of neurodegeneration become more significant, raising the likelihood of developing disorders like Alzheimer's and Parkinson's disease. This review explores the intricate relationship between heavy metal exposure, dysfunction of the autophagy-lysosomal pathway, and aging-related neurodegeneration, emphasizing the urgent need for a comprehensive understanding of these mechanisms. The insights gained from this analysis are crucial for creating targeted therapeutic approaches aimed at alleviating the harmful effects of heavy metals on neuronal health and improving cellular resilience in aging populations.
Collapse
Affiliation(s)
- Shrabani Das
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Pritha Ghosh
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
23
|
Zhou X, Xia X, Li L, Ye Y, Chen Q, Ke M, Cui Q, He Y, Chen Y, Lin S, Liu W, Wang J. Evaluation of Heavy Metals and Essential Minerals in the Hair of Children with Autism Spectrum Disorder and Their Association with Symptom Severity. Biol Trace Elem Res 2025:10.1007/s12011-025-04588-z. [PMID: 40153150 DOI: 10.1007/s12011-025-04588-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/17/2025] [Indexed: 03/30/2025]
Abstract
The exact cause of Autism spectrum disorder (ASD) remains unclear. The accumulation of heavy metals and the imbalance of trace elements are believed to play a key role in the pathogenesis of ASD. This study aimed to compare the levels of trace elements and heavy metals in the hair of 1-16-year-old children with varying ASD severity. We included a control group of 57 children, as well as 124 children with autism, consisting of 53 with mild to moderate autism and 71 with severe autism. Questionnaires and hair samples were collected, and 21 chemical elements were analyzed using inductively coupled plasma mass spectrometry (ICP-MS). Children with severe autism showed significantly higher levels of the trace elements copper (Cu) and heavy metals vanadium (V), cobalt (Co), nickel (Ni), arsenic (As), cadmium (Cd), and lead (Pb) in their bodies compared to the control group. Boys with severe autism showed significantly higher levels of Cu, As, Cd, and Pb compared to the control group, while girls with severe autism exhibited significantly lower levels of potassium (K) and increased levels of Pb. Severely autistic children under 7 years old had significantly increased levels of Mn, Cu, V, Co, Ni, As, Cd, and Pb. Children with severe autism aged 7-16 years typically showed significantly higher levels of Cu and As. These findings underscore the importance of heavy metals and essential minerals as environmental factors in the severity of ASD disease.
Collapse
Affiliation(s)
- Xulan Zhou
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Xiaochun Xia
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Liming Li
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Yaohui Ye
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Qihui Chen
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Mingyue Ke
- Department of Laboratory Medicine, Siming District Center for Disease Control and Prevention, Xiamen, China
| | - Qian Cui
- Department of Laboratory Medicine, Siming District Center for Disease Control and Prevention, Xiamen, China
| | - Yuling He
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Yiting Chen
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China
| | - Shaoqing Lin
- Department of Medical Record, Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Wenlong Liu
- Department of Child Development and Behavior, Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China.
| | - Juan Wang
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, China.
| |
Collapse
|
24
|
Cai Y, Zeng X, Wu M, Chen H, Sun M, Yang H. TET1 mitigates prenatal fluoride-induced cognition impairment by modulating Bcl2 DNA hydroxymethylation level. Mol Med 2025; 31:117. [PMID: 40133886 PMCID: PMC11938627 DOI: 10.1186/s10020-025-01174-w] [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: 06/02/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Fluoride exposure during pregnancy commonly compromises fetal neurodevelopment and largely results in a broad spectrum of cognitive deficiencies in the adult offspring. However, the precise mechanisms underlying these effects remain to be fully elucidated. Herein, we investigate the impacts of fluoride on neural excitability and apoptosis, synaptic plasticity, and cognitive function, as well as possible underlying mechanisms. Our results indicated that exposure to a high sodium fluoride (100 mg/L) during pregnancy in the mouse can cause the cognitive deficits of their offspring, accompanied by a decrease in the expression of Tet-eleven translocation protein 1 (TET1), an enzyme responsible for DNA hydroxymethylation. Additionally, there is a reduction in the dendritic spine density and the expression of postsynaptic density protein-95 (PSD95) in the hippocampal regions of male offspring. Furthermore, in vitro fluoride treatment significantly exacerbates neuronal apoptosis and reduces the frequency of spikes in spontaneous action potential. More significantly, we also found that TET1 could directly bind to the promotor region of Bcl2, altering its DNA hydroxymethylation and Bcl2 expression. Intriguingly, Tet1 knock-out mice exhibited cognitive deficits similar to those observed in male animals exposed to high levels of fluoride. Furthermore, the down-regulation of TET1 protein, along with the consequent alteration in Bcl2 hydroxymethylation and increased neuronal apoptosis, are likely mechanisms underlying the impact of prenatal fluoride exposure on the neurodevelopment of male offspring. These findings provide novel insights into the molecular mechanisms by which fluoride exposure induces neurodevelopmental impairment of the male offspring.
Collapse
Affiliation(s)
- Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
25
|
Xie J, Luo D, Xing P, Ding W. The Dual Roles of STAT3 in Ferroptosis: Mechanism, Regulation and Therapeutic Potential. J Inflamm Res 2025; 18:4251-4266. [PMID: 40144540 PMCID: PMC11938932 DOI: 10.2147/jir.s506964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Ferroptosis, an iron-dependent programmed mechanism of cell death that is driven by lipid peroxidation, is an important pathogenic factor in oncological and non-oncological disorders. Dysregulation of iron and lipid metabolism profoundly influences disease progression through ferroptosis modulation. Signal transducer and activator of transcription 3 (STAT3), a transcriptional regulator, regulates ferroptosis by binding to promoters of key molecules such as solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), and ferritin heavy chain 1 (FTH1). In this review, we described the role of STAT3 in supporting tumors survival by suppressing ferroptosis in malignancies, and bidirectionally regulating ferroptosis in non-tumors to regulate the development of the disease. We also reported emerging therapeutic strategies that target STAT3-mediated ferroptosis, including natural phytochemicals, inhibitors, and nanotechnology-enabled drug delivery systems. These advancements deepen the mechanistic understanding of ferroptosis regulation, and provide new theoretical bases and strategies to treat ferroptosis-related diseases.
Collapse
Affiliation(s)
- Jinghui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Dan Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Pengfei Xing
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
26
|
Song J, Li H, Fang X. The inflection point: α-Klotho levels and the risk of all-cause mortality. Front Endocrinol (Lausanne) 2025; 16:1405003. [PMID: 40134808 PMCID: PMC11932894 DOI: 10.3389/fendo.2025.1405003] [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: 03/22/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose The controversial nature of the association between α-Klotho and mortality risk in the general population warrants further investigation. This study aims to examine the correlation between circulating α-Klotho levels and the risk of all-cause mortality. Methods A sample size of 13,748 individuals from the NAHNES 2005-2016 cycles was included in this study. The effect of different α-Klotho levels (divided into quartiles) on survival was assessed using Kaplan-Meier (KM) curves. Cox proportional hazards models were used to analyze the linear relationship between log α-Klotho and the risk of all-cause mortality. Restricted cubic spline Cox proportional hazards regression model was used to analyze the non-linear relationship between log α-Klotho and risk of all-cause mortality. Threshold effect analysis was performed to determine the most favorable inflection point for log α-Klotho. Stratification and sensitivity analyses were performed to assess the robustness of the results. Results A total of 1,569 deaths were reported during the median follow-up period of 5.33 years (2.83-7.83 years). Among the log α-Klotho quartile groups, quartile 1 had the highest mortality rate compared to quartiles 2, 3, and 4. Multifactorial Cox regression analysis revealed a weak association between log α-Klotho and a 44% reduction in the risk of all-cause mortality (p=0.0473). We also found a U-shaped non-linear association between log α-Klotho and risk of all-cause mortality, with an optimal inflection point identified at 2.89 pg/mL. The stability of the U-shaped association between log α-Klotho and mortality risk was observed in various stratification and sensitivity analyses. Conclusion This study identified a U-shaped association between circulating α-Klotho levels and risk of all-cause mortality, with a notable inflection point at 2.89 pg/mL. Further investigation is warranted to fully elucidate the potential mechanisms underlying the association between α-Klotho and risk of all-cause mortality in the broader population.
Collapse
Affiliation(s)
- Jianling Song
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hong Li
- Department of Medical Records, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
27
|
Espitia-Pérez P, Espitia-Pérez L, Peñata-Taborda A, Brango H, Pastor-Sierra K, Galeano-Páez C, Arteaga-Arroyo G, Humanez-Alvarez A, Rodríguez Díaz R, Salas Osorio J, Valderrama LA, Saint’Pierre TD. Genetic Damage and Multi-Elemental Exposure in Populations in Proximity to Artisanal and Small-Scale Gold (ASGM) Mining Areas in North Colombia. TOXICS 2025; 13:202. [PMID: 40137529 PMCID: PMC11946375 DOI: 10.3390/toxics13030202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 03/29/2025]
Abstract
This study evaluates DNA damage and multi-element exposure in populations from La Mojana, a region of North Colombia heavily impacted by artisanal and small-scale gold mining (ASGM). DNA damage markers from the cytokinesis-block micronucleus cytome (CBMN-Cyt) assay, including micronucleated binucleated cells (MNBN), nuclear buds (NBUDs) and nucleoplasmic bridges (NPB), were assessed in 71 exposed individuals and 37 unexposed participants. Exposed individuals had significantly higher MNBN frequencies (PR = 1.26, 95% CI: 1.02-1.57, p = 0.039). Principal Component Analysis (PCA) identified the "Soil-Derived Mining-Associated Elements" (PC1), including V, Fe, Al, Co, Ba, Se and Mn, as being strongly associated with high MNBN frequencies in the exposed population (PR = 10.45, 95% CI: 9.75-12.18, p < 0.001). GAMLSS modeling revealed non-linear effects of PC1, with greater increases in MNBN at higher concentrations, especially in exposed individuals. These results highlight the dual role of essential and toxic elements, with low concentrations being potentially protective but higher concentrations increasing genotoxicity. Women consistently exhibited higher MNBN frequencies than men, suggesting sex-specific susceptibilities. This study highlights the compounded risks of chronic metal exposure in mining-impacted regions and underscores the urgent need for targeted interventions to mitigate genotoxic risks in vulnerable populations.
Collapse
Affiliation(s)
- Pedro Espitia-Pérez
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Lyda Espitia-Pérez
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Ana Peñata-Taborda
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Hugo Brango
- Facultad de Educación y Ciencias, Departamento de Matemáticas, Universidad de Sucre, Sincelejo 700003, Colombia;
| | - Karina Pastor-Sierra
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Claudia Galeano-Páez
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Gean Arteaga-Arroyo
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Alicia Humanez-Alvarez
- Grupo de Investigación Biomédica y Biología Molecular, Facultad de Ciencias de la Salud, Universidad del Sinú, Montería 230001, Colombia; (L.E.-P.); (A.P.-T.); (K.P.-S.); (C.G.-P.); (G.A.-A.); (A.H.-A.)
| | - Ruber Rodríguez Díaz
- Hospital Alma Máter, Unidad de Cuidados Intensivos (UCI), Medellín 050001, Colombia;
| | - Javier Salas Osorio
- Hospital Alma Máter, Servicios Ambulatorios, Coordinación Médica, Medellín 050001, Colombia;
| | | | | |
Collapse
|
28
|
Kouroumalis E, Tsomidis I, Voumvouraki A. HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease. Biomedicines 2025; 13:683. [PMID: 40149659 PMCID: PMC11940282 DOI: 10.3390/biomedicines13030683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete Medical School, 71500 Heraklion, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
29
|
Neha V, Parithathvi A, Dsouza HS. Ameliorative role of bioactive compounds against lead-induced neurotoxicity. Neuroscience 2025; 568:46-56. [PMID: 39805419 DOI: 10.1016/j.neuroscience.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/04/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Lead (Pb) is an environmental toxin ubiquitously present in the human environment due to anthropogenic activities and industrialization. Lead can enter the human body through various sources and pathways, such as inhalation, ingestion and dermal contact, leading to detrimental health effects. The majority of lead that enters the body is removed by urine or feces; however, under chronic exposure conditions, lead is not efficient, as lead is absorbed and transferred to numerous organs, such as the brain, liver, kidney, muscles, and heart, and it is ultimately stored in mineralizing tissues such as bones and teeth. The central nervous system is the most affected among all the organs and systems affected, as lead is a known neurotoxin. Lead absorption is elevated in the fasting state than in the fed state. Chelation therapy, which is used to treat lead poisoning, has various adverse effects, making this treatment detrimental because it disrupts the levels of other essential elements and redistributes lead to various tissues. One of the main mechanisms by which lead induces toxicity is through the generation of reactive oxygen species. Hence, bioactive compounds that are the source of antioxidants if consumed along with ongoing lead exposure can ameliorate the toxic effects of lead.
Collapse
Affiliation(s)
- Venkatesan Neha
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Aluru Parithathvi
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences,Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Herman Sunil Dsouza
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
30
|
Quan Q, Ma X, Feng J, Li W, Li X. Ginsenoside Rg1 improves autophagy dysfunction to ameliorate Alzheimer's disease via targeting FGR proto-oncogene. Neuropeptides 2025; 111:102514. [PMID: 40073763 DOI: 10.1016/j.npep.2025.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
Alzheimer's disease (AD) is a neurodegeneration driven by beta-amyloid (Aβ) deposits in the brain involving autophagy dysfunction. Ginsenoside Rg1, a pharmacologically active compound found in ginseng, has possible therapeutic effects for AD. This study discovered that FGR proto-oncogene (FGR) was a therapeutic target of Rg1 in AD and it was possibly involved in autophagy. C57BL/6 J mice were injected with 5 μL (1 μg/mL) Aβ1-42 in the right lateral ventricle to establish an AD model. AD mouse hippocampus had high FGR expression. Intragastrically administered Rg1 (40 mg/kg) decreased FGR protein levels in AD mice's hippocampus and improved memory function in AD mice. Both sides of the mice hippocampal fissure were administered with 2 μL lentiviral particles (1 × 107 TU) containing FGR overexpression plasmids. FGR overexpression rendered Rg1 ineffectual in restoring memory function and reducing hippocampal neuron damage. We injected 2 μL lentiviral particles (1 × 107 TU) containing short hairpin RNA plasmids targeting FGR to the mice hippocampal fissures. FGR knockdown improved spatial memory function of AD mice, reduced hippocampal neuron apoptosis, and prevented Aβ accumulation. HT22 cells were transfected with small interfering RNA targeting FGR. FGR knockdown increased the viability of Aβ1-42 treated HT22 cells. BACE1 and LC3II/I protein levels were decreased and p62 and SIRT1 were increased in AD mice and cells with FGR knockdown. LC3 was down-regulated after inhibiting FGR expression in Aβ1-42 treated hippocampal neurons. In conclusion, Rg1 exerts anti-AD functions by targeting FGR and downregulating its expression.
Collapse
Affiliation(s)
- Qiankun Quan
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinxin Ma
- Department of Psychology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - JianJun Feng
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanni Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
31
|
Zhang SY, Yang N, Hao PH, Wen R, Zhang TN. Targeting sirtuins in neurological disorders: A comprehensive review. Int J Biol Macromol 2025; 292:139258. [PMID: 39736297 DOI: 10.1016/j.ijbiomac.2024.139258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025]
Abstract
The sirtuin (SIRT) family is a group of seven conserved nicotinamide adenine dinucleotide-dependent histone deacetylases (SIRT1-SIRT7), which play crucial roles in various fundamental biological processes, including metabolism, aging, stress responses, inflammation, and cell survival. The role of SIRTs in neuro-pathophysiology has recently attracted significant attention. Notably, SIRT1-SIRT3 have been identified as key players in neuroprotection as they reduce neuroinflammation and regulate mitochondrial function. This review summarizes the latest research advancements in the role of the SIRT family in neurological diseases, mainly including neurodegenerative diseases, ischemia-related diseases, bleeding-related diseases, nervous system injury and other nervous system diseases, emphasizing their critical functions and associated signaling pathways, (e.g., AMPK/SIRT1/PGC-1α, AMPK/SIRT1/IL-1β/NF-κB, STAT2-SIRT4-mTOR, SIRT3/FOXO3α, and other signaling pathways in disease progression, particularly their protective roles in neurodegenerative diseases, ischemic injuries, and neural damage. Additionally, this review discusses progress in clinical studies targeting SIRT-specific small-molecule agonists and inhibitors. Further research on SIRTs may provide new insights into potential therapeutic strategies for the prevention and treatment of neurological disorders.
Collapse
Affiliation(s)
- Sen-Yu Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Peng-Hui Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
32
|
Yi X, Wei R, Huang S, Wei P, Li H, Li Z, Aschner M, Jiang Y, Ou S, Li S. The effect of resveratrol on lead-induced oxidative damage and apoptosis in HT-22 cells. Food Chem Toxicol 2025; 197:115274. [PMID: 39864579 DOI: 10.1016/j.fct.2025.115274] [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: 11/12/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVE The purpose of this work was to investigate whether resveratrol affects lead-induced oxidative damage in HT-22 cells, characterizing mechanisms and strategies for preventing and treating lead-induced neurotoxicity. METHODS Various lead and resveratrol concentrations were applied to HT-22 cells over different time periods. First, we established the lead treatment (12.5, 50 and 200 μmol/L) and resveratrol (40 μmol/L) intervention model for the study. MTT was used to analyze HT-22 cell survival rate. The rates of cell death, mitochondrial membrane potential, lipid peroxidation, and reactive oxygen species (ROS) generation were all measured by flow cytometry. Cellular oxidant (MDA) and antioxidant (SOD, GSH-Px) levels were measured with test kits. Western blotting was used to assess the expression of proteins related to autophagy and apoptosis. RESULTS Lead reduced HT-22 cell viability in a concentration/time-dependent manner. In addition, lead (200 μmol/L) decreased the protein expression of BCL2, while increasing PARP and BAX expression and apoptotic rate. Moreover, the lead-exposed group had significantly higher levels of ROS, lipid-ROS, and MDA than the control group. This was accompanied by increased MDA levels and decreased SOD, GSH-Px, and MMP levels in the lead-exposed cells. Furthermore, lead lowered SIRT1 protein expression, while increasing the levels of autophagy-related proteins, including P62, ATG5, Beclin-1 and LC3 Ⅱ/Ⅰ. Resveratrol (40 μmol/L), an agonist of SIRT1, restored the effects of lead (200 μmol/L) to levelsindistinguishable from controls. CONCLUSION Resveratrol inhibited mitochondrial damage and restored the lead-induced block of autophagic flux and oxidative stress by activating SIRT1, thereby alleviating lead-induced damage in HT-22 cells.
Collapse
Affiliation(s)
- Xiang Yi
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; School of Public Health, Xiangnan University, Chenzhou, 423000, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Ruokun Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaoni Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Peiqi Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - HuiShuai Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Zhenning Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shiyan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
33
|
Jiang W, Zhang B, Xu J, Xue L, Wang L. Current status and perspectives of esophageal cancer: a comprehensive review. Cancer Commun (Lond) 2025; 45:281-331. [PMID: 39723635 PMCID: PMC11947622 DOI: 10.1002/cac2.12645] [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: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Esophageal cancer (EC) continues to be a significant global health concern, with two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Prevention and changes in etiology, improvements in early detection, and refinements in the treatment have led to remarkable progress in the outcomes of EC patients in the past two decades. This seminar provides an in-depth analysis of advances in the epidemiology, disease biology, screening, diagnosis, and treatment landscape of esophageal cancer, focusing on the ongoing debate surrounding multimodality therapy. Despite significant advancements, EC remains a deadly disease, underscoring the need for continued research into early detection methods, understanding the molecular mechanisms, and developing effective treatments.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| | - Bo Zhang
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jiaqi Xu
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Liyan Xue
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Luhua Wang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| |
Collapse
|
34
|
Yu H, Li X, Ning B, Feng L, Ren Y, Li S, Kang Y, Ma J, Zhao M. SIRT1: a potential therapeutic target for coronary heart disease combined with anxiety or depression. J Drug Target 2025; 33:328-340. [PMID: 39470049 DOI: 10.1080/1061186x.2024.2422882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Coronary heart disease (CHD) combined with anxiety or depression is increasingly receiving attention in the clinical field of cardiology, and exploring the comorbidity pathological mechanisms of cardiovascular disease combined with psychological disorders is a hot research topic for scholars in this field. Current research suggests that Silent Information Regulatory Factor 1 (SIRT1) may serve as a potential biomarker for the comorbidity mechanism and treatment of CHD with anxiety or depression. SIRT1 is considered a promising therapeutic target for CHD combined with anxiety or depression, with the ability to regulate inflammatory cytokine levels, alleviate oxidative stress damage, activate multiple signalling pathways, reduce platelet hyperresponsiveness, and exert neuroprotective and cardioprotective effects. In this comprehensive review, we deeply studied the structure, function, and mechanism of SIRT1, and discussed its protective effects in the cardiovascular and nervous system. The latest progress in the mechanism of SIRT1's role in CHD combined with anxiety or depression was emphasised, including its specific mechanisms in regulating inflammatory response, alleviating oxidative stress, and mediating various signalling pathways. In addition, this article also summarises the therapeutic potential of SIRT1 as a potential biomarker in patients with CHD combined with anxiety or depression.
Collapse
Affiliation(s)
- Hubin Yu
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinping Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lanshuan Feng
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yaolong Ren
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shilin Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yalong Kang
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Mingjun Zhao
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
35
|
Liu Z, Lai K, Li P, Gong Y, Fu H, Dong H, Yang Z, Qin R, Guo L. Enhanced Anticancer Selectivity of Cyclometalated Imidazole/Pyrazole-Imine Iridium III Complexes Through the Switch from Cationic to Zwitterionic Forms. Inorg Chem 2025; 64:2837-2856. [PMID: 39895267 DOI: 10.1021/acs.inorgchem.4c04937] [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: 02/04/2025]
Abstract
Cyclometalated iridiumIII complexes have shown promising anticancer properties, with variations in charge and ligand substitution significantly influencing their biological activity. However, zwitterionic iridiumIII complexes remain scarcely explored. Herein, we report a series of zwitterionic cyclometalated imidazole/pyrazole-imine iridiumIII complexes and compare their biological activity to analogous cationic complexes with sulfonate counteranions. X-ray crystallography confirmed the structural differences between the cationic and zwitterionic forms. These complexes exhibited cytotoxicity against A549, HeLa, and HepG2 cancer cells, with IC50 values ranging from 14.35 to 69.12 μM. While cationic complexes showed higher cytotoxicity, zwitterionic complexes demonstrated enhanced selectivity for A549 cancer cells over BEAS-2B normal cells (selectivity index: 3.72-5.90 for zwitterionic forms vs 1.16-1.44 for cationic forms). This selectivity is attributed to distinct cellular uptake mechanisms: zwitterionic complexes use an energy-dependent pathway in cancer cells and an energy-independent pathway in normal cells, leading to differences in cellular accumulation and redox activity. Mechanistic studies revealed that both complex types induce ROS generation and mitochondrial membrane depolarization (MMP), with apoptosis as the primary cell death pathway.
Collapse
Affiliation(s)
- Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Ruixin Qin
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
36
|
Aleksandrova Y, Neganova M. Antioxidant Senotherapy by Natural Compounds: A Beneficial Partner in Cancer Treatment. Antioxidants (Basel) 2025; 14:199. [PMID: 40002385 PMCID: PMC11851806 DOI: 10.3390/antiox14020199] [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: 01/30/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Aging is a general biological process inherent in all living organisms. It is characterized by progressive cellular dysfunction. For many years, aging has been widely recognized as a highly effective mechanism for suppressing the progression of malignant neoplasms. However, in recent years, increasing evidence suggests a "double-edged" role of aging in cancer development. According to these data, aging is not only a tumor suppressor that leads to cell cycle arrest in neoplastic cells, but also a cancer promoter that ensures a chronic proinflammatory and immunosuppressive microenvironment. In this regard, in our review, we discuss recent data on the destructive role of senescent cells in the pathogenesis of cancer. We also identify for the first time correlations between the modulation of the senescence-associated secretory phenotype and the antitumor effects of naturally occurring molecules.
Collapse
Affiliation(s)
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia;
| |
Collapse
|
37
|
Shi M, Li H, Liang R, Lin H, Tang Q. The transcription factor STAT3 and aging: an intermediate medium. Biogerontology 2025; 26:55. [PMID: 39920354 DOI: 10.1007/s10522-025-10193-3] [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: 12/24/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Aging is a physiological/pathological process accompanied by progressive impairment of cellular function, leading to a variety of aging-related diseases. STAT3 is one of the core regulatory factors of aging. It is involved in body metabolism, development and senescence, cell apoptosis and so on. During the aging process, the changes of growth factors and cytokines will cause the activation of STAT3 to varying degrees, regulate the inflammatory pathways related to aging, regulate body inflammation, mitochondrial function, cell aging and autophagy to regulate and influence the aging process. Drugs targeting STAT3 can treat senescence related diseases. This review summarizes the role of STAT3 signaling factors in the pathogenesis of aging, including mitochondrial function, cellular senescence, autophagy, and chronic inflammation mediated by inflammatory pathways. Finally, the key regulatory role of STAT3 in senescence related diseases is emphasized. In summary, we reveal that drug development and clinical application targeting STAT3 is one of the key points in delaying aging and treating aging-related diseases in the future.
Collapse
Affiliation(s)
- Min Shi
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Honyu Li
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Haiyan Lin
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Qiang Tang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China.
| |
Collapse
|
38
|
Xu Y, Reheman A, Feng W. Recent research progress on metal ions and metal-based nanomaterials in tumor therapy. Front Bioeng Biotechnol 2025; 13:1550089. [PMID: 39991139 PMCID: PMC11842396 DOI: 10.3389/fbioe.2025.1550089] [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: 12/22/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Tumors, as a disease that seriously threatens human health, have always been a major challenge in the field of medicine. Currently, the main methods of tumor treatment include surgery, radiotherapy, chemotherapy, etc., but these traditional treatment methods often have certain limitations. In addition, tumor recurrence and metastasis are also difficult problems faced in clinical treatment. In this context, the importance of metal-based nanomaterials in tumor therapy is increasingly highlighted. Metal-based nanomaterials possess unique physical, chemical, and biological properties, providing new ideas and methods for tumor treatment. Metal-based nanomaterials can achieve targeted therapy for tumors through various mechanisms, reducing damage to normal tissues; they can also serve as drug carriers, improving the stability and bioavailability of drugs; at the same time, some metal-based nanomaterials also have photothermal, photodynamic, and other characteristics, which can be used for phototherapy of tumors. This review examines the latest advances in the application of metal-based nanomaterials in tumor therapy within past 5 years, and presents prospective insights into the future applications.
Collapse
Affiliation(s)
- Yongcheng Xu
- The Second School of Clinical Medicine, Shenyang Medical College, Shenyang, China
| | - Aikebaier Reheman
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Wenhua Feng
- Department of Human Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Liaoning Province Key Laboratory for Phenomics of Human Ethnic Specificity and Critical Illness, Shenyang, China
- Shenyang Key Laboratory for Phenomics, Shenyang Medical College, Shenyang, China
| |
Collapse
|
39
|
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
|
40
|
Qu H, Yu Q, Ye L, Zheng J. SLC39A14 promotes the development of esophageal squamous cell carcinoma through PI3K/Akt/mTOR signaling pathway. Int Immunopharmacol 2025; 146:113831. [PMID: 39700956 DOI: 10.1016/j.intimp.2024.113831] [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: 10/12/2024] [Revised: 11/25/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES This study aims to investigate the expression of solute carrier family 39 member 14 (SLC39A14) in esophageal squamous cell carcinoma (ESCC) tissues and its prognosis, as well as the impact of SLC39A14 expression on the biological behavior of ESCC cells and associated mechanisms. METHODS Bioinformatics analysis was utilized to compare the differential expression of SLC39A14 mRNA between esophageal cancer tissues and adjacent non-cancerous tissues. Immunohistochemistry was employed to evaluate SLC39A14 protein expression in human ESCC tissues and normal esophageal tissues, followed by an analysis of its association with clinicopathological parameters in esophageal cancer patients. Through cell proliferation, migration, invasion, and Western blot assays, we deeply evaluated the specific effects of SLC39A14 gene knockdown (or overexpression) on ESCC cells and explored its potential biological functions in ESCC. Subsequently, we validated the role of SLC39A14 in ESCC in a xenograft model. Furthermore, LY294002 drug intervention was used to verify the regulatory effect of SLC39A14 on PI3K/Akt/mTOR signaling pathway. RESULTS Both mRNA and protein levels of SLC39A14 were significantly elevated in tumor tissues from ESCC patients compared to adjacent normal tissues. Notably, higher levels of SLC39A14 expression positively correlated with ESCC tumor size (p = 0.010) and clinical T stage (p = 0.025), while exhibiting a negative correlation with overall patient survival rates (p = 0.023). In vitro experiments demonstrated that knocking down SLC39A14 significantly inhibited cell proliferation, migration and invasion. In vivo study showed that SLC39A14 facilitated progression within murine models bearing ESCC tumors. Mechanistic analyses suggested that pro-carcinogenic effects exerted by SLC39A14 are mediated through activation of the PI3K/Akt/mTOR signaling pathway. CONCLUSIONS Our findings suggest that SLC39A14 may serve as a potential biomarker for ESCC due to its pro-oncogenic role during ESCC progression.
Collapse
Affiliation(s)
- Hangshuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, China
| | - Luxia Ye
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Jingmin Zheng
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China.
| |
Collapse
|
41
|
Assari S, Najand B, Zare H. Puberty Onset and Positive Urgency Explain Diminished Returns of Family Income on Tobacco and Marijuana Use. OPEN JOURNAL OF PSYCHOLOGY 2025; 5:1141. [PMID: 39886641 PMCID: PMC11780720 DOI: 10.31586/ojp.2025.1141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Background Puberty is a crucial developmental milestone that involves significant physiological, emotional, and behavioral changes. Early puberty onset, influenced by both biological and social factors, is associated with an increased risk of engaging in substance use, such as tobacco and marijuana. While high family income is generally linked to delayed puberty onset and lower behavioral risks, these benefits may not be equally protective for Black youth due to the phenomenon of Minorities' Diminished Returns (MDRs). MDRs suggest that higher family income does not offer the same protective effects for Black youth as it does for White youth, potentially leading to earlier puberty and increased substance use among high-income Black adolescents. Objective This study aimed to investigate whether early puberty onset and associated positive urgency (impulsivity) mediate the relationship between family income and the initiation of tobacco and marijuana use over a six-year follow-up period among adolescents. Additionally, the study examined whether the effects of family income on early puberty onset differ by race, testing the hypothesis that high-income Black youth would experience earlier puberty onset compared to their high-income White peers. Methods Data were sourced from the Adolescent Brain Cognitive Development (ABCD) Study. Participants were 9-10-year-old adolescents at baseline, followed over a period of six years. Structural equation modeling (SEM) was used to assess whether early puberty onset mediated the effects of family income on substance use behaviors. Interaction terms between race and family income were included to test whether the impact of family income varies by race. Results Early puberty onset and associated positive urgency partially explained the relationship between family income and the initiation of tobacco and marijuana use. High-income Black youth showed earlier puberty onset compared to their White counterparts. Earlier puberty onset then predicted higher positive urgency. These factors, in turn, were linked to higher rates of tobacco and marijuana initiation. Conclusions This study provides additional evidence that the benefits of high family income do not extend equally to Black adolescents, particularly regarding delaying puberty onset and its consequences for substance use.
Collapse
Affiliation(s)
- Shervin Assari
- Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
- Marginalization-Related Diminished Returns (MDRs) Center, Los Angeles, CA, United States
| | - Babak Najand
- Marginalization-Related Diminished Returns (MDRs) Center, Los Angeles, CA, United States
| | - Hossein Zare
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- University of Maryland Global Campus (UMGC), Adelphi, MD, United States
| |
Collapse
|
42
|
Zhu L, Yang M, Fan L, Yan Q, Zhang L, Mu P, Lu F. Interaction between resveratrol and SIRT1: role in neurodegenerative diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:89-101. [PMID: 39105797 DOI: 10.1007/s00210-024-03319-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024]
Abstract
Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease, pose significant health challenges and economic burdens worldwide. Recent studies have emphasized the potential therapeutic value of activating silent information regulator-1 (SIRT1) in treating these conditions. Resveratrol, a compound known for its ability to potently activate SIRT1, has demonstrated promising neuroprotective effects by targeting the underlying mechanisms of neurodegeneration. In this review, we delve into the crucial role of resveratrol-mediated SIRT1 upregulation in improving neurodegenerative diseases. The role of the activation of SIRT1 by resveratrol was reviewed. Moreover, network pharmacology was used to elucidate the possible mechanisms of resveratrol in these diseases. Activation of SIRT1 by resveratrol had positive effects on neuronal function and survival and alleviated the hallmark features of these diseases, such as protein aggregation, oxidative stress, neuroinflammation, and mitochondrial dysfunction. In terms of network pharmacology, the signaling pathways by which resveratrol protects against different neurodegenerative diseases were slightly different. Although the precise mechanisms underlying the neuroprotective effects of resveratrol and SIRT1 activation remain under investigation, these findings offer valuable insights into potential therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang, 113004, People's Republic of China
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang, 113004, People's Republic of China
| | - Miaomiao Yang
- Department of Biochemistry and Molecular Biology, Shenyang Medical College, Shenyang, 113004, People's Republic of China
- Basic Medical College, Shenyang Medical College, Shenyang, 113004, People's Republic of China
| | - Lehao Fan
- Basic Medical College, Shenyang Medical College, Shenyang, 113004, People's Republic of China
| | - Qiuying Yan
- Basic Medical College, Shenyang Medical College, Shenyang, 113004, People's Republic of China
| | - Lifeng Zhang
- Department of Public Health, Shenyang Medical College, Shenyang, 113004, People's Republic of China.
| | - Ping Mu
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang, 113004, People's Republic of China.
- Department of Physiology, Shenyang Medical College, Shenyang, 113004, People's Republic of China.
| | - Fangjin Lu
- Department of Pharmaceutical Analysis, Shenyang Medical College, Shenyang, 113004, People's Republic of China.
| |
Collapse
|
43
|
Li X, Liu Q, Wu M, Wang H, Yang J, Mu X, Zhang XD. Artificially Engineered Nanoprobes for Ultrasensitive Magnetic Resonance Imaging. Adv Healthc Mater 2025; 14:e2403099. [PMID: 39562174 DOI: 10.1002/adhm.202403099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/11/2024] [Indexed: 11/21/2024]
Abstract
Magnetic resonance imaging (MRI) is a noninvasive and radiation-free technique used for soft tissue. However, there are some limitations of the MRI modality, such as low sensitivity and poor image resolution. Artificially engineered magnetic nanoprobes have been extensively explored as a versatile platform for ultrasensitive MRI contrast agents due to their unique physiochemical characteristics and tunable magnetic properties. In this review, the emphasis is on recent progress in MRI nanoprobes with different structures and elements, including gadolinium-, iron-, manganese-based and metal-free nanoprobes. The key influencing factors and advanced engineering strategies for modulating the relaxation ratio of MRI nanoprobes are systematically condensed. Furthermore, the widespread and noninvasive visualization applications of MRI nanoprobes for real time monitoring of major organs and accurate disease diagnosing, such as cerebrovascular, ischemia, Alzheimer's disease, liver fibrosis, whole-body tumors, inflammation, as well as multi-mode imaging applications are summarized. Finally, the challenges and prospects for the future development of MRI nanoprobes are discussed, and promising strategies are specifically emphasized for improving biocompatibility, precisely engineering of optimal size, AI-driven prediction and design, and multifunctional self-assembly to enhance diagnostics. This review will provide new inspiration for artificial engineering and nanotechnology-based molecular probes for medical diagnosis and therapy with ultrasensitive MRI.
Collapse
Affiliation(s)
- Xuyan Li
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qingshan Liu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Menglin Wu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jiang Yang
- School of Medicine, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Science, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Science, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
44
|
Wang R, Deng L, Wang Y, Liu N, Yang M, Qiu J, Chen C. Synergistic effects of combined lead and iprodione exposure on P53 signaling-mediated hepatotoxicity, enterotoxicity and transgenerational toxicity in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178127. [PMID: 39708747 DOI: 10.1016/j.scitotenv.2024.178127] [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: 06/13/2024] [Revised: 11/04/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Environmental heavy metal contamination, combined with inappropriate use of fungicides, has led to the co-existence of lead (Pb) and iprodione (IPR), presenting signification risks to ecosystems and human health. The toxic effects resulting from concurrent exposure to Pb and IPR, however, remain poorly understood. In the study, we conducted a comprehensive 60-day subchronic study to investigate the toxic effects on the liver and gut in parental male zebrafish through employing multi-omics analyses. We also explored the potential transgenerational toxicity to unexposed offspring embryos. The results demonstrated that exposure to both Pb and IPR exacerbated intestinal pathological damage, decreased the expression of intestinal tight junction molecules, and activated the expression of intestinal inflammatory molecules in the gut. Metabolic and microbial analyses, utilizing 16S rRNA sequencing and non-targeted metabolic profiling, revealed alterations in the intestinal flora structure and disruptions in metabolite synthesis. Notably, we observed a significant negative correlation between the abundance of the Lactobacillus genus and uracil synthesis. Furthermore, liver RNA-seq analysis identified a marked enrichment of the P53 signaling pathway, confirmed by the activation of P53-mediated apoptotic markers, which was consistent with the observed increase in inflammatory infiltration and pathological damage within the liver. Importantly, P53-mediated apoptosis and inflammatory responses were activated in offspring embryos, suggesting that long-term parental exposure to Pb and IPR may induce transgenerational toxicity, potentially impacting offspring health. Despite the identification of these molecular changes, the phenotypic effects remain to be elucidated. Future studies are necessary to evaluate the potential phenotypic changes in offspring to fully understand the long-term effects of Pb and IPR exposure. Overall, these findings enhance the understanding of the molecular mechanisms underlying the toxic effects of Pb and IPR and emphasize the importance of a comprehensive risk assessment of environmental pollutants.
Collapse
Affiliation(s)
- Ruike Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Ligang Deng
- Institute of Agricultural Quality Standards and Testing Technology Research, Shandong Academy of Agricultural Sciences, Jinan, China; Shandong Provincial Key Laboratory of Test Technology on Food Quality and Safety, Jinan, China
| | - Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Na Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Menglian Yang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Chen Chen
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
45
|
Li H, Cui Y, Hu Y, Zhao M, Li K, Pang X, Sun F, Zhou B. Mammalian SLC39A13 promotes ER/Golgi iron transport and iron homeostasis in multiple compartments. Nat Commun 2024; 15:10838. [PMID: 39738060 PMCID: PMC11685566 DOI: 10.1038/s41467-024-55149-2] [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: 05/17/2023] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Iron is a potent biochemical, and accurate homeostatic control is orchestrated by a network of interacting players at multiple levels. Although our understanding of organismal iron homeostasis has advanced, intracellular iron homeostasis is poorly understood, including coordination between organelles and iron export into the ER/Golgi. Here, we show that SLC39A13 (ZIP13), previously identified as a zinc transporter, promotes intracellular iron transport and reduces intracellular iron levels. ZIP13 loss causes an iron deficiency in the ER/Golgi and other intracellular compartments, such as lysosomes and mitochondria, as well as elevating iron in the cytosol. ZIP13 overexpression has the opposite effect, increasing iron in organellar compartments. We suggest that ZIP13 gatekeeps an iron trafficking route that shunts iron from the cytosol to the ER/Golgi hub. Zip13-knockout male mice have iron deposition in several tissues. These data demonstrate that mammalian ZIP13 is crucial for iron homeostasis and suggest a potential iron transport function.
Collapse
Affiliation(s)
- Huihui Li
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanmei Cui
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yule Hu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengran Zhao
- Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Xiaoyun Pang
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fei Sun
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Zhou
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen, 518055, China.
| |
Collapse
|
46
|
Verdecchia A, de la Torre IC, Diaz IM, Sanz VG, Mesa YG, Cobo T, Gallardo VP. Analysis of the Relationship Between Body Mass Index (BMI) and Dento-Skeletal Maturation: A Cross-Sectional Case-Control Study. Dent J (Basel) 2024; 13:8. [PMID: 39851584 PMCID: PMC11764128 DOI: 10.3390/dj13010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: The aim of this cross-sectional study was to establish if there is a relationship between body mass index (BMI) and skeletodental development in young obese patients in comparison with normal-weight patients. Methods: The sample consisted of 178 individuals (115 normal weight, 37 overweight and 26 obese), aged 6 to 16 years, with a mean biological age of 11.96 ± 2.44 years. Dental maturation (dental age) was determined by using Demerjian's method; craniofacial growth pattern, skeletal and dental class were determined by using cephalometric measurements; skeletal maturation was determined by using Baccetti's method. Data were statistically analyzed. Results: According to Demirjian's method, the mean dental age of the overweight and obese subjects was significantly higher than that of the normal-weight subjects (p = 0.001 and p = 0.02, respectively). A higher rate of dental class III was detected in the overweight group (p = 0.002). Concerning cephalometric records, statistically significant differences (p = 0.018) were observed in the distribution of SNA and SNB values, higher values being found in obese subjects. No difference was detected in the distribution of the ANB angle between the groups. As regards craniofacial growth pattern, no significant differences were found between the groups. Children with obesity presented more advanced skeletal maturation, reaching statistically significant differences (p = 0.02), in comparison with the normal-weight subjects. Conclusions: In conclusion, obese children showed increased tooth and skeletal development in comparison to the normal-weight subjects. These findings may be of interest for orthodontists, who should consider weight status when performing orthodontic treatment in children and adolescents.
Collapse
Affiliation(s)
- Alessio Verdecchia
- Orthodontics Division, Universidad de Oviedo, Instituto Asturiano de Odontologia, 33006 Oviedo, Spain; (I.M.D.); (T.C.)
| | - Inmaculada Coronado de la Torre
- Orthodontics Teaching Unit, Department of Stomatology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (I.C.d.l.T.); (V.P.G.)
| | - Ivan Menéndez Diaz
- Orthodontics Division, Universidad de Oviedo, Instituto Asturiano de Odontologia, 33006 Oviedo, Spain; (I.M.D.); (T.C.)
| | - Veronica García Sanz
- Orthodontics Teaching Unit, Department of Stomatology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (I.C.d.l.T.); (V.P.G.)
| | - Yolanda García Mesa
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain;
| | - Teresa Cobo
- Orthodontics Division, Universidad de Oviedo, Instituto Asturiano de Odontologia, 33006 Oviedo, Spain; (I.M.D.); (T.C.)
| | - Vanessa Paredes Gallardo
- Orthodontics Teaching Unit, Department of Stomatology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain; (I.C.d.l.T.); (V.P.G.)
| |
Collapse
|
47
|
Khan MA. Targeting Iron Responsive Elements (IREs) of APP mRNA into Novel Therapeutics to Control the Translation of Amyloid-β Precursor Protein in Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:1669. [PMID: 39770511 PMCID: PMC11677800 DOI: 10.3390/ph17121669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The hallmark of Alzheimer's disease (AD) is the buildup of amyloid-β (Aβ), which is produced when the amyloid precursor protein (APP) misfolds and deposits as neurotoxic plaques in the brain. A functional iron responsive element (IRE) RNA stem loop is encoded by the APP 5'-UTR and may be a target for regulating the production of Alzheimer's amyloid precursor protein. Since modifying Aβ protein expression can give anti-amyloid efficacy and protective brain iron balance, targeted regulation of amyloid protein synthesis through modulation of 5'-UTR sequence function is a novel method for the prospective therapy of Alzheimer's disease. Numerous mRNA interference strategies target the 2D RNA structure, even though messenger RNAs like tRNAs and rRNAs can fold into complex, three-dimensional structures, adding even another level of complexity. The IRE family is among the few known 3D mRNA regulatory elements. This review seeks to describe the structural and functional aspects of IREs in transcripts, including that of the amyloid precursor protein, that are relevant to neurodegenerative diseases, including AD. The mRNAs encoding the proteins involved in iron metabolism are controlled by this family of similar base sequences. Like ferritin IRE RNA in their 5'-UTR, iron controls the production of APP in their 5'-UTR. Iron misregulation by iron regulatory proteins (IRPs) can also be investigated and contrasted using measurements of the expression levels of tau production, Aβ, and APP. The development of AD is aided by iron binding to Aβ, which promotes Aβ aggregation. The development of small chemical therapeutics to control IRE-modulated expression of APP is increasingly thought to target messenger RNAs. Thus, IRE-modulated APP expression in AD has important therapeutic implications by targeting mRNA structures.
Collapse
Affiliation(s)
- Mateen A Khan
- Department of Life Science, College of Science and General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
48
|
Hu R, Li G, Hu P, Niu H, Li W, Jiang S, Guan G, Xu Q, Liu M, Chen L. bmp10 maintains cardiac function by regulating iron homeostasis. J Genet Genomics 2024; 51:1459-1473. [PMID: 39414074 DOI: 10.1016/j.jgg.2024.10.003] [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: 06/23/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024]
Abstract
Heart disease remains the leading cause of death worldwide. Iron imbalance, whether deficiency or overload, contributes to heart failure. However, the molecular mechanisms governing iron homeostasis in the heart are poorly understood. Here, we demonstrate that mutation of bmp10, a heart-born morphogen crucial for embryonic heart development, results in severe anemia and cardiac hypertrophy in zebrafish. Initially, bmp10 deficiency causes cardiac iron deficiency, which later progresses to iron overload due to the dysregulated hepcidin/ferroportin axis in cardiac cells, leading to ferroptosis and heart failure. Early iron supplementation in bmp10-/- mutants rescues erythropoiesis, while iron chelation in juvenile fishes significantly alleviates cardiac hypertrophy. We further demonstrate that the interplay between HIF1α-driven hypoxic signaling and the IL6/p-STAT3 inflammatory pathways is critical for regulating cardiac iron metabolism. Our findings reveal BMP10 as a key regulator of iron homeostasis in the vertebrate heart and highlight the potential of targeting the BMP10-hepcidin-iron axis as a therapeutic strategy for iron-related cardiomyopathy.
Collapse
Affiliation(s)
- Ruiqin Hu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Genfang Li
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Peng Hu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Hongbo Niu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Wenhao Li
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Shouwen Jiang
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Guijun Guan
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Qianghua Xu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, College of Marine Science, Shanghai Ocean University, Shanghai 201306, China
| | - Mingli Liu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Liangbiao Chen
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
49
|
Fathima A, Bagang N, Kumar N, Dastidar SG, Shenoy S. Role of SIRT1 in Potentially Toxic Trace Elements (Lead, Fluoride, Aluminum and Cadmium) Associated Neurodevelopmental Toxicity. Biol Trace Elem Res 2024; 202:5395-5412. [PMID: 38416341 PMCID: PMC11502598 DOI: 10.1007/s12011-024-04116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/17/2024] [Indexed: 02/29/2024]
Abstract
The formation of the central nervous system is a meticulously planned and intricate process. Any modification to this process has the potential to disrupt the structure and operation of the brain, which could result in deficiencies in neurological growth. When neurotoxic substances are present during the early stages of development, they can be exceptionally dangerous. Prenatally, the immature brain is extremely vulnerable and is therefore at high risk in pregnant women associated with occupational exposures. Lead, fluoride, aluminum, and cadmium are examples of possibly toxic trace elements that have been identified as an environmental concern in the aetiology of a number of neurological and neurodegenerative illnesses. SIRT1, a member of the sirtuin family has received most attention for its potential neuroprotective properties. SIRT1 is an intriguing therapeutic target since it demonstrates important functions to increase neurogenesis and cellular lifespan by modulating multiple pathways. It promotes axonal extension, neurite growth, and dendritic branching during the development of neurons. Additionally, it contributes to neurogenesis, synaptic plasticity, memory development, and neuroprotection. This review summarizes the possible role of SIRT1 signalling pathway in potentially toxic trace elements -induced neurodevelopmental toxicity, highlighting some molecular pathways such as mitochondrial biogenesis, CREB/BDNF and PGC-1α/NRF1/TFAM.
Collapse
Affiliation(s)
- Aqsa Fathima
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Newly Bagang
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial area Hajipur, Vaishali, Bihar, 844102, India
| | - Somasish Ghosh Dastidar
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
50
|
Gupta K, Soni N, Nema RK, Sahu N, Srivastava RK, Ratre P, Mishra PK. Microcystin-LR in drinking water: An emerging role of mitochondrial-induced epigenetic modifications and possible mitigation strategies. Toxicol Rep 2024; 13:101745. [PMID: 39411183 PMCID: PMC11474209 DOI: 10.1016/j.toxrep.2024.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Algal blooms are a serious menace to freshwater bodies all over the world. These blooms typically comprise cyanobacterial outgrowths that produce a heptapeptide toxin, Microcystin-LR (MC-LR). Chronic MC-LR exposure impairs mitochondrial-nuclear crosstalk, ROS generation, activation of DNA damage repair pathways, apoptosis, and calcium homeostasis by interfering with PC/MAPK/RTK/PI3K signaling. The discovery of the toxin's biosynthesis pathways paved the way for the development of molecular techniques for the early detection of microcystin. Phosphatase inhibition-based bioassays, high-performance liquid chromatography, and enzyme-linked immunosorbent tests have recently been employed to identify MC-LR in aquatic ecosystems. Biosensors are an exciting alternative for effective on-site analysis and field-based characterization. Here, we present a synthesis of evidence supporting MC-LR as a mitotoxicant, examine various detection methods, and propose a novel theory for the relevance of MC-LR-induced breakdown of mitochondrial machinery and its myriad biological ramifications in human health and disease.
Collapse
Affiliation(s)
- Kashish Gupta
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Nikita Soni
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Ram Kumar Nema
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Neelam Sahu
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pooja Ratre
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
| | - Pradyumna Kumar Mishra
- Division of Environmental Biotechnology, Genetics & Molecular Biology (EBGMB), ICMR-National Institute for Research in Environmental Health (NIREH), Bhopal, India
- Faculty of Medical Research, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|