1
|
Amaral L, Martins M, Côrte-Real M, Outeiro TF, Chaves SR, Rego A. The neurotoxicity of pesticides: Implications for Parkinson's disease. CHEMOSPHERE 2025; 377:144348. [PMID: 40203643 DOI: 10.1016/j.chemosphere.2025.144348] [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: 09/02/2024] [Revised: 03/04/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Parkinson's disease (PD) is the fastest-growing neurodegenerative disorder worldwide, and no effective cure is currently available. Neuropathologically, PD is characterized by the selective degeneration of dopaminergic neurons in the substantia nigra and by the accumulation of alpha-synuclein (aSyn)-rich proteinaceous inclusions within surviving neurons. As a multifactorial disorder, approximately 85 % of PD cases are sporadic with unknown etiology. Among the many risk factors implicated in PD, exposure to neurotoxic pesticides stands out as a significant contributor. While the effects of many are still uncharacterized, it has already been shown that rotenone, paraquat, maneb, and dieldrin affect critical cellular pathways, including mitochondrial and proteasomal dysfunction, aSyn aggregation, autophagy dysregulation, and disruption of dopamine metabolism. With the constant rise in pesticide usage to meet the demands of a growing human population, the risk of environmental contamination and subsequent PD development is also increasing. This review explores the molecular mechanisms by which pesticide exposure influences PD development, shedding light on their role in the pathogenesis of PD and highlighting the need for preventative measures and regulatory oversight to mitigate these risks.
Collapse
Affiliation(s)
- Leslie Amaral
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal; University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Márcia Martins
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal
| | - Manuela Côrte-Real
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal
| | - Tiago F Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK; Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany; Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Susana R Chaves
- CBMA - Centre of Molecular and Environmental Biology / ARNET - Aquatic Research Network, Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal.
| | - António Rego
- Department of Biology, School of Sciences, University of Minho, 4710-057, Braga, Portugal; Solfarcos, Pharmaceutical and Cosmetic Solutions, Braga, Portugal.
| |
Collapse
|
2
|
Abstract
Both genetic and environmental factors modulate the risk of Parkinson's disease. In this article, all these pathophysiologic processes that contribute to damages at the tissue, cellular, organelle, and molecular levels, and their effects are talked about.
Collapse
Affiliation(s)
- Bin Xiao
- National Neuroscience Institute, Singapore; Duke-NUS Medical School, Singapore
| | - ZhiDong Zhou
- National Neuroscience Institute, Singapore; Duke-NUS Medical School, Singapore
| | - YinXia Chao
- National Neuroscience Institute, Singapore; Duke-NUS Medical School, Singapore
| | - Eng-King Tan
- National Neuroscience Institute, Singapore; Duke-NUS Medical School, Singapore.
| |
Collapse
|
3
|
Chen Y, Zhang X, Liu Y, Liu Z, Li Z, Li Z, Chen X, Liu J, Chen Z, Mo G, Xia Y, Mou L. A Gold Nanoparticles and MXene Nanocomposite Based Electrochemical Sensor for Point-of-Care Monitoring of Serum Biomarkers. ACS NANO 2025. [PMID: 40262058 DOI: 10.1021/acsnano.5c03194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The development of portable, cost-effective, and highly sensitive biosensors for real-time biomarker detection is crucial for advancing point-of-care testing (POCT) and wearable health monitoring. Here, we present an integrated portable electrochemical sensor (ip-ECS) that combines gold nanoparticles (AuNPs) and MXene-modified screen-printed electrodes (SPEs) with a custom-designed, low-power electronic system for point-of-care monitoring of serum biomarkers. The AuNPs and MXene nanocomposite significantly enhances the electrochemical performance of the SPE by providing a high density of active sites, improved conductivity, and catalytic activity. The detection of two model molecules (DA and UA) validated the feasibility of ip-ECS, achieving detection limits as low as 1.12 and 1.11 μM for UA and DA, respectively. Furthermore, the ip-ECS was successfully applied to detect Cys C in human serum, showing a linear response in the range of 50-5000 ng/mL and a strong correlation (ρ = 0.9556) with conventional latex immunoturbidimetry (LIA). Clinical validation using serum samples from pregnant women revealed elevated Cys C levels in gestational diabetes mellitus (GDM) patients, highlighting the sensor's potential for early GDM risk prediction. The ip-ECS represents a significant step forward in the development of next-generation biosensors for POCT, wearable diagnostics, and personalized medicine.
Collapse
Affiliation(s)
- Yongmei Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| | - Xinyu Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| | - Yan Liu
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| | - Zhengkun Liu
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Zijie Li
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Zhiqi Li
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Xiaman Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Jiayi Liu
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Zihao Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Guangquan Mo
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
| | - Yong Xia
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| | - Lei Mou
- The Key Laboratory of Advanced Interdisciplinary Studies, School of Biomedical Engineering, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Yanjiang Road, Yuexiu District, Guangzhou 510120, Guangdong, P. R. China
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou 510150, Guangdong, P. R. China
| |
Collapse
|
4
|
Xu K, Wang G, Gong J, Yang X, Cheng Y, Li D, Sheng S, Zhang F. Akkermansia muciniphila protects against dopamine neurotoxicity by modulating butyrate to inhibit microglia-mediated neuroinflammation. Int Immunopharmacol 2025; 152:114374. [PMID: 40056512 DOI: 10.1016/j.intimp.2025.114374] [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: 11/27/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
Parkinson's disease (PD) is an age-related and second most common neurodegenerative disease. To date, safe and efficient therapeutic drugs are deficient. In recent years, the relationship between gut microbiota and CNS have received more attention. Homeostatic imbalance of gut microbiota was revealed to participate in the progression of PD. This study detected that Akkermansia muciniphila (A. muciniphila) was apparently decreased in the feces of PD rats via 16S rRNA amplicon sequencing. Furtherly, we found that exogenous supplementation of A. muciniphila could improve 6-OHDA-induced motor dysfunction and dopamine (DA) neuronal damage and neuroinflammatory factors release in PD rats. Moreover, the short-chain fatty acids (SCFAs) sequencing demonstrated that A. muciniphila addition increased butyrate content both in gut and brain. The subsequent functional experiments confirmed that the exogenous supplementation of butyrate conferred neuroprotection against DA neurotoxicity. Mechanically, butyrate targeted microglia to attenuate DA neuronal injury via inhibiting microglia activation and neuroinflammatory factors production. In conclusion, A. muciniphila protected DA neuronal damage by modulating butyrate to inhibit microglia-elicited neuroinflammation. These findings provided a potential application of A. muciniphila on PD treatment.
Collapse
Affiliation(s)
- Kaifei Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Guoqing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiantao Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xinxing Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yufeng Cheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Daidi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shuo Sheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
5
|
Sun RX, Guo Y. Gene signatures and immune correlations in Parkinson's disease Braak stages. Eur J Med Res 2025; 30:278. [PMID: 40229859 PMCID: PMC11998164 DOI: 10.1186/s40001-025-02554-y] [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: 10/29/2024] [Accepted: 04/04/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD), a progressive neurodegenerative disease, still lacks disease-modifying treatment strategies. The formation of Lewy body is the typical pathological feature of PD. Pathological progression can be defined by Braak stages. However, the molecular mechanism for this ascending course of α-synuclein pathology remains unclear. METHODS In this study, weighted gene co-expression network analysis (WGCNA) was used to screen Braak stage-related gene signatures, followed by the functional enrichment analysis, including gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA). The hub genes were screened through CytoHubba and Least Absolute Shrinkage and Selection Operator (LASSO) analysis. The immune cell proportion was predicted by the ImmuCellAI. Furthermore, transcription factors (TFs) and miRNAs targeting the hub genes network were constructed. After verifying hub gene expression level through independent data sets. The validated hub gene was further analyzed to elucidate the potential molecular mechanism. RESULTS Total of 388 genes associated with Braak stages were screened out through WGCNA analysis. The KEGG analysis showed that these genes were involved in endocytosis, HIF-1 signaling pathway, synaptic vesicle cycle, dopaminergic synapse, oxytocin signaling pathway, etc. Immune infiltration analysis showed that CD4 + T cells, including nTreg, Th2, and Th17, were obviously different between different Braak stages in PD. Furthermore, eights Braak stages-related hub genes were identified, including CAMK2B, CPLX2, GAPDH, GRIN1, KCNA1, MAPK3, MAPT, and STXBP1 through the cytoHubba plugin and LASSO analysis. After verifying the expression level in three independent data sets, CPLX2 was finally identified as the most reliable Braak stages-associated hub genes in PD. CONCLUSIONS This study revealed the Braak stage-related gene signatures in PD and identified CPLX2 as a novel Braak stages-related hub gene in PD, which provided a novel target for future therapeutic interventions and disease markers. The specific molecular mechanism of CPLX2 in PD remained to be further clarified.
Collapse
Affiliation(s)
- Rui-Xue Sun
- Department of Internal Medicine, Nanjing Luhe District Zhu Town Community Health Service Center, Nanjing, Jiangsu, China.
| | - Yan Guo
- Department of Internal Medicine, Nanjing Luhe District Zhu Town Community Health Service Center, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Tuersong T, Wu QF, Chen Y, Shan Li P, Yong YX, Shataer M, Shataer S, Ma LY, Yang XL. Integrated network pharmacology, metabolomics, and microbiome studies to reveal the therapeutic effects of Anacyclus pyrethrum in PD-MCI mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156729. [PMID: 40253741 DOI: 10.1016/j.phymed.2025.156729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Anacyclus pyrethrum (l.) DC has potential value in treating Parkinson's disease (PD)-mild cognitive impairment (MCI), manifesting as impaired memory, attention, executive function, and language. However, the specific targets and modes of action of A. pyrethrum remain unclear. PURPOSE The aim of this study was to identify the active components of A. pyrethrum and examine their effectiveness in treating a mouse model of PD-MCI. METHODS We generated ethanol extracts of A. pyrethrum root (EEAP) and identified its active components and related targets using UHPLC-MS/MS and network pharmacology.The PD-MCI model was induced via intraperitoneal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP). After following continuous administration of EEAP,Altered learning or memory, as well as anxiety, were tested using the morris water maze, eight-arm radial arm maze (RAM), and open-field test,elevated plus-maze. Brain histopathology and ultrastructural changes were examined using brightfield microscopy, and electron microscopy, respectively. Furthermore, protein expression was assessed using western blotting.Stool samples were used for metabolomics analysis by UHPLC-MS/MS and for 16S rDNA sequencing to determine the compositional changes of the gut microbiota.We conducted a short-chain fatty acid targeted metabolomics experiment to study their role in the gut-brain axis in PD-MCI. RESULTS Using UPLC-MS-MS, 126 compounds were identified from A. pyrethrum samples.After searching the databases and literature reports, 31 active components and 544 drug-disease targets were screened. Biological processes and molecular functions, such as energy channels, cell signaling, and metabolism, were discovered through GO analysis. The water maze experiment showed that the average swimming distance and escape latency of mice in EEAP groups decreased. The eight-arm maze experiment showed that model had a much higher number of errors related to working memory than the control mice. In the open field experiment, compared with the control group, the mice in the EEAP group exhibited an increase in the average movement speed and total movement distance, along with a decrease in the residence time.In the elevated plus maze, the control had less anxiety than the Model. Donepezil/Levodopa(D/l) mitigated anxiety-like behavior, and EEAP (100-400 mg/kg) showed a dose-dependent increase in open-arm metrics, suggesting it may ease anxiety in mice.Hippocampal tissue of mice treated with different doses of EEAP showed intact cellular layers and the hematoxylin-eosin-stained cones were slightly better;cells were arranged neatly; their morphology was normal, and were distributed uniformly. Electron microscopy revealed that the nuclear membrane, chromatin, and nucleoli were clearly demarcated in the hippocampus of mice treated with different doses of EEAP, contrary to that in the model group. In brain extracts of the EEAP group, lighter thinner bands for amyloid precursor protein (APP) and Aβ were observed compared to those in the model group. In model mice, APP and Aβ protein expression was higher than in the blank group, as shown by stronger bands. In EEAP-treated mice, the bands were weaker, indicating reduced expression. In the model group had lower Bcl-2 and higher Bax levels. EEAP treatment increased Bcl-2 and decreased Bax expression.Compared to the control group, the model showed substantially low glutathione peroxidase (GSH-Px),superoxide dismutase(SOD),catalase (CAT)activity (p < 0.05),much higher (p < 0.05) in the EEAP-H group than that in the model. EEAP intervention significantly modulated the fecal metabolic profile of PD-MCI mice. The abundance of steroid and lipid metabolites, including linoleylethanolamine, was markedly altered in the model group compared to the control group, with EEAP treatment reversing several of these abnormalities. PLS-DA and OPLS-DA revealed significant separation between groups (Q2= 0.542, p < 0.01), confirming a dose-dependent effect. Random forest analysis identified 15 key metabolic markers, such as dose-dependent changes in d-glutamine and hydrocodone. Metabolic pathway analysis demonstrated significant enrichment in phenylalanine, tyrosine, tryptophan metabolism, and arginine biosynthesis pathways (p < 0.05). The Support Vector Machine (SVM) model achieved an AUC approaching 1, indicating substantial differences in metabolite profiles. EEAP intervention significantly influenced the composition and functional profile of the intestinal microbiota. The Venn diagram illustrates that each group shared 342 operational taxonomic units (OTUs), with the EEAP 400 group exhibiting a distinct Bacteroidetes proportion. LEfSe analysis identified g_Prevotella as the characteristic bacterium in the control group, c_Epsilonproteobacteria in the model group, and g_Adlercreutzia in the EEAP 100 group. The Faith's Phylogenetic Diversity (PD) index was highest in the EEAP 100 group, and Non-metric Multidimensional Scaling (NMDS)/Principal Coordinates Analysis (PCoA) revealed significant differences in microbial community structure. Short-chain fatty acids (SCFAs) analysis indicated that acetic acid was the predominant metabolite, while EEAP dose-dependently regulated propionic acid and isovaleric acid levels (VIP > 1, p < 0.001). These findings demonstrate that EEAP exerts its regulatory effects by reshaping the structure and metabolic functions of the gut microbiota. CONCLUSION EEAP holds great promise as a potential therapeutic agent for PD-MCI, exerting its effects through multiple mechanisms, including regulating protein expression, modulating the fecal metabolic profile, and reshaping the gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Tayier Tuersong
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Qin Fen Wu
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Yan Chen
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Pei Shan Li
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Yu Xuan Yong
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Munire Shataer
- Department of Histology and Embryology, Basic Medical College of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Samire Shataer
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Liang Ying Ma
- Department of Pharmacy, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China
| | - Xin Ling Yang
- Department of Neurology, Xinjiang Key Laboratory of Neurological Diseases, Xinjiang Clinical Research Center for Nervous System Diseases, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi 830001, Xinjiang, PR China.
| |
Collapse
|
7
|
Lang J, Xiong Z. Protective effects of harpagoside on mitochondrial functions in rotenone‑induced cell models of Parkinson's disease. Biomed Rep 2025; 22:64. [PMID: 39991000 PMCID: PMC11843190 DOI: 10.3892/br.2025.1942] [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: 05/29/2024] [Accepted: 12/16/2024] [Indexed: 02/25/2025] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. Currently, no radical treatment is available for this disease. Harpagoside is a proposed neuroprotective iridoid active ingredient that can be derived from Scrophulariae buergeriana, Scrophularia striata and Harpagophytum procumbens. The present study aimed to investigate the effects of harpagoside on mitochondrial functions in rotenone-induced cell models of Parkinson's disease (PD). Neuro-2A (N2A) cells were treated with rotenone to establish in vitro cell models of PD. Cell viability and survival were measured using a Cell Counting Kit-8 assay. Biochemical assays with spectrophotometry were used to measure complex I activity, mitochondrial swelling and caspase 3 activity. The cell survival rate was first found to be significantly decreased by rotenone (20 nmol/l) treatment. However, intervention with harpagoside (10 µmol/l) was found to increase the cell survival rate of rotenone-induced N2A cell models differentiated with 1 mmol/l of dibutyryl-cAMP. At ≥0.1 µmol/l concentration, harpagoside significantly alleviated rotenone-induced mitochondrial swelling, whereas at 1 µmol/l it significantly counteracted the inhibitory effects of rotenone on complex I activity. At 10 µmol/l harpagoside significantly inhibited rotenone-induced caspase 3 activation. These results suggest that harpagoside has the potential to protect mitochondrial functions against rotenone-induced injury in N2A cell models of PD.
Collapse
Affiliation(s)
- Juan Lang
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhongkui Xiong
- Department of Radiation Oncology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
- Department of Medical Imaging, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
8
|
Li B, Yang R, Chen X, He J, Lu Z, Li Y. Copper Mitigates Atrazine-Induced Neurotoxicity in Parkinson's Disease Models. Mol Neurobiol 2025; 62:5202-5215. [PMID: 39527184 DOI: 10.1007/s12035-024-04609-3] [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: 05/05/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Atrazine (ATR), one of the most prevalent herbicides used worldwide, has been associated with various environmental health concerns, including its potential role in promoting neurodegenerative diseases. Considering the pivotal role of metal homeostasis in the pathophysiology of neurodegenerative disorders, this study investigated the neuroprotective effects of copper supplementation in mitigating the adverse impacts of ATR in Parkinson's disease (PD). Our results reveal that ATR exposure disrupts copper and iron homeostasis within the substantia nigra pars compacta (SNpc), leading to enhanced oxidative stress and dopaminergic neuronal degeneration, which are key features of PD pathology. Remarkably, copper supplementation is able to counteract these neurotoxic effects, as demonstrated by the restoration of metal equilibrium in the SNpc, decreased lipid peroxidation, and improvements in motor function in PD rats. These results highlight the intricate relationship between environmental toxins and metal homeostasis in the genesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Bingyun Li
- School of Public Health, Shantou University, Shantou, 515063, Guangdong Province, China
| | - Ruijiao Yang
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Xiaojuan Chen
- School of Public Health, Shantou University, Shantou, 515063, Guangdong Province, China
| | - Jinyi He
- School of Public Health, Shantou University, Shantou, 515063, Guangdong Province, China
| | - Zijing Lu
- School of Public Health, Shantou University, Shantou, 515063, Guangdong Province, China
| | - Yanshu Li
- School of Public Health, Shantou University, Shantou, 515063, Guangdong Province, China.
| |
Collapse
|
9
|
Ceccarini MR, Alunni Cardinali M, Malaspina R, Libera V, Scattini G, Codini M, Chiesa I, De Maria C, Comez L, Paciaroni A, Sassi P, Valentini L. Protective effects of silk fibroin against 6-OHDA in SH-SY5Y human neuroblastoma cells and comparative study with its release from gelatin films. Int J Biol Macromol 2025; 303:140697. [PMID: 39914551 DOI: 10.1016/j.ijbiomac.2025.140697] [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: 11/28/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
In this study, we analyzed the neuroprotective action of silk fibroin (SF) regenerated with calcium chloride (CaCl2), distinguishing the effects of CaCl2 and SF, and subsequently fabricating a neuroprotective hybrid material based on SF gelatin film. Cytotoxicity induced by 6-hydroxydopamine (6-OHDA) on the human neuroblastoma SH-SY5Y cell line showed that SF had a significant shielding power against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in SH-SY5Y neuroblastoma cells, as assessed by the CCK-8 assay, cell imaging and cell cycle using flow cytometer. Specifically, the concurrent treatment with SF and 6-OHDA produced a marked neuroprotective effect. Circular dichroism analysis suggested the formation of silk III because of the interaction between the secondary structures of SF and 6-OHDA. Raman analysis was also employed to assess the impact of SF and CaCl2 on cellular metabolism, indicating the combined administration of fibroin and 6-OHDA as more effective than the use of CaCl2 alone. Subsequently, we synthesized a silk/gelatin-based film, demonstrated its ability to release SF, and confirmed its capacity to protect SH-SY5Y neuroblastoma cells.
Collapse
Affiliation(s)
- Maria Rachele Ceccarini
- Department of Pharmaceutical Science, University of Perugia, Perugia 06123, Italy; Civil and Environmental Engineering Department University of Perugia and INSTM Research Unit, Strada di Pentima, Terni 05100, Italy
| | - Martina Alunni Cardinali
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto, 8, Perugia 06123, Italy
| | - Rocco Malaspina
- Department of Physics and Geology, University of Perugia, Via Alessandro Pascoli, Perugia 06123, Italy
| | - Valeria Libera
- Department of Physics and Geology, University of Perugia, Via Alessandro Pascoli, Perugia 06123, Italy
| | - Gabriele Scattini
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo, 4, Perugia 06126, Italy
| | - Michela Codini
- Department of Pharmaceutical Science, University of Perugia, Perugia 06123, Italy
| | - Irene Chiesa
- Department of Information Engineering and Research Center E. Piaggio, University of Pisa, Largo Lucio Lazzarino, 1, 56122, Italy
| | - Carmelo De Maria
- Department of Information Engineering and Research Center E. Piaggio, University of Pisa, Largo Lucio Lazzarino, 1, 56122, Italy
| | - Lucia Comez
- Istituto Officina dei Materiali-IOM, National Research Council-CNR, Via Alessandro Pascoli, Perugia 06123, Italy
| | - Alessandro Paciaroni
- Department of Physics and Geology, University of Perugia, Via Alessandro Pascoli, Perugia 06123, Italy
| | - Paola Sassi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto, 8, Perugia 06123, Italy
| | - Luca Valentini
- Civil and Environmental Engineering Department University of Perugia and INSTM Research Unit, Strada di Pentima, Terni 05100, Italy.
| |
Collapse
|
10
|
Wang W, Liu T, Li Q, Ji E, Xu W, Qiao S, Cui Y, Li B, Xu H. Validation of a Sensitive, Simple and High-Throughput UPLC-MS/MS Method for Quantification of Catecholamines and Their Metabolites in Serum and Urine: Application in Clinical Analysis. J Chromatogr Sci 2025; 63:bmaf021. [PMID: 40237721 DOI: 10.1093/chromsci/bmaf021] [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: 01/24/2024] [Revised: 03/17/2025] [Indexed: 04/18/2025]
Abstract
A sensitive, simple and high-throughput UPLC-MS/MS method has been validated for the simultaneous quantification of catecholamines and their metabolite levels in serum and urine for clinical applications. The analytes and their isotope-labeled internal standards were extracted using a 96-well solid-phase extraction cartridge and then separated on an HSS PFP column with a 4-min gradient elution. The linear ranges were 10 ~ 5000 pg/mL for dopamine (DA), epinephrine (E), metanephrine (MN) and normetanephrine (NMN), 2 ~ 5000 pg/mL for norepinephrine (NE), and 2 ~ 2000 pg/mL for 3-methoxytyramine (3-MT). The limits of quantification were 10 pg/mL for DA and E, 5 pg/mL for MN and NMN, 2 pg/mL for 3-MT, and 20 pg/mL for NE. The accuracy was excellent with relative bias all within 10%, and the intra-day and inter-day precision values were also within the tolerance range (RSD < 15%), and the recovery was in the range of 86.0-107.7% with RSD < 15%. After correction using IS, no significant matrix effects were observed. Moreover, the discrepancies in the analyte levels between plasma and serum were investigated for the first time. The analyte levels in the two biological matrices exhibited a significant correlation (P < 0.001) and significant differences (P < 0.001).
Collapse
Affiliation(s)
- Wei Wang
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Tiebing Liu
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Qingyan Li
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Enhui Ji
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Weizhe Xu
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Shi Qiao
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Yujing Cui
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Boye Li
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| | - Haishan Xu
- Civil Aviation Medical Center, Civil Aviation Administration of China, 76 Chaoyang Road, Chaoyang District, Beijing 100123, China
| |
Collapse
|
11
|
Xu L, Wang Z, Li Q. Global trends and projections of Parkinson's disease incidence: a 30-year analysis using GBD 2021 data. J Neurol 2025; 272:286. [PMID: 40131471 DOI: 10.1007/s00415-025-13030-2] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND AND OBJECTIVES Parkinson's disease (PD) is a neurodegenerative disorder marked by the progressive loss of dopaminergic neurons, leading to motor dysfunction and non-motor symptoms like cognitive decline and depression. With the aging global population, PD incidence is anticipated to rise, especially in regions with rapidly growing elderly populations. This study leverages Global Burden of Disease (GBD) 2021 data to analyze the burden of PD by region, sex, and age group, examining trends from 1992 to 2021 and projecting the future burden to 2030. METHODS Data from the GBD 2021 database for the years 1992-2021 were analyzed to assess age-standardized incidence rates (ASIR) and mortality of PD across socio-demographic index (SDI) regions, sex, and age groups. The Age-Period-Cohort (APC) model was used to explore temporal trends, while the Bayesian Age-Period-Cohort (BAPC) model projected future PD burden from 2022 to 2030. RESULTS From 1992 to 2021, global PD cases increased from 450,000 to 1.34 million, with crude incidence rates rising from 8.19 to 16.92 per 100,000 and ASIR from 11.54 to 15.63 per 100,000, indicating an annual net drift of 1.11% (95% CI 1.06%-1.17%), reflecting a growing burden driven by an aging population. All SDI regions saw a growth in PD burden, with the highest increases in middle- and high-middle-SDI regions, where male incidence was notably higher than female. Incidence rates escalated sharply in individuals aged 60 and older, peaking in those aged 85 and above. Projections suggest that by 2030, global PD cases will reach 1.93 million, with an ASIR of 27 per 100,000. DISCUSSION The findings highlight a sustained global increase in PD burden, particularly in middle- and high-income regions and among men. In low-SDI areas, PD burden may be underestimated due to limited healthcare access and diagnostic challenges. These results stress the urgent need for health policies focused on elderly populations, especially men, and call for effective prevention and intervention strategies to mitigate the future impact of PD.
Collapse
Affiliation(s)
- Libo Xu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zhenhao Wang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Qingsong Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
12
|
Kopalli SR, Behl T, Kyada A, Rekha MM, Kundlas M, Rani P, Nathiya D, Satyam Naidu K, Gulati M, Bhise M, Gupta P, Wal P, Fareed M, Ramniwas S, Koppula S, Gasmi A. Synaptic plasticity and neuroprotection: The molecular impact of flavonoids on neurodegenerative disease progression. Neuroscience 2025; 569:161-183. [PMID: 39922366 DOI: 10.1016/j.neuroscience.2025.02.007] [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: 11/08/2024] [Revised: 01/11/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Flavonoids are a broad family of polyphenolic chemicals that are present in a wide variety of fruits, vegetables, and medicinal plants. Because of their neuroprotective qualities, flavonoids have attracted a lot of interest. The potential of flavonoids to control synaptic plasticity-a crucial process underlying memory, learning, and cognitive function-is becoming more and more clear. Dysregulation of synaptic plasticity is a feature of neurodegenerative diseases such as amyotrophic lateral sclerosis (0.4 %), Parkinson's (1-2 %), Alzheimer's (5-7 %), and Huntington's ((0.2 %)). This review discusses the molecular mechanisms via which flavonoids influence synaptic plasticity as well as their therapeutic potential in neurodegenerative diseases. Flavonoids modulate key signaling pathways such as MAPK/ERK and PI3K/Akt/mTOR to support neuroprotection, synaptic plasticity, and neuronal health, while also influencing neurotrophic factors (BDNF, NGF) and their receptors (TrkB, TrkA). They regulate neurotransmitter receptors like GABA, AMPA, and NMDA to balance excitatory and inhibitory transmission, and exert antioxidant effects via the Nrf2-ARE pathway and anti-inflammatory actions by inhibiting NF-κB signaling, highlighting their potential for treating neurodegenerative diseases. These varied reactions support the preservation of synapse function and neuronal integrity in the face of neurodegenerative insults. Flavonoids can reduce the symptoms of neurodegeneration, prevent synaptic loss, and enhance cognitive function, according to experimental studies. However, there are still obstacles to using these findings in clinical settings, such as limited bioavailability and the need for consistent dose. The focus of future research should be on improving flavonoid delivery systems and combining them with conventional medications.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006 Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab 140306, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401 Punjab, India
| | - Pooja Rani
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | | | | | - Pranay Wal
- PSIT- Pranveer Singh Institute of Technology, Pharmacy Kanpur UP, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Seema Ramniwas
- University Centre for Research and Development, Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413 Punjab, India
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Amin Gasmi
- Societe Francophone de Nutritherapie et de Nutrigenetique Appliquee, Villeurbanne, France; International Institute of Nutrition and Micronutrient Sciences, Saint-Etienne, France
| |
Collapse
|
13
|
Xu L, Xie Y, Liu A, Xie L, Miao X, Hou Z, Xiang L, Jiang T, Wu A, Lin J. Innovative Applications and Perspectives of Surface-Enhanced Raman Spectroscopy Technology in Biomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409698. [PMID: 39610172 DOI: 10.1002/smll.202409698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) has become a revolutionary technique in the biomedical field, providing unparalleled sensitivity for the detection and characterization of biological samples. In this review, recent SERS innovations are comprehensively discussed, including advanced substrate materials, different SERS detection strategies, and multimodal approaches that combine SERS with other biotechnologies. Among them, the role of SERS in the accurate diagnosis of tumors is highlighted, which has promoted accurate molecular analysis and real-time monitoring of treatment effects. In addition, the growing potential of SERS in the treatment of chronic diseases such as cardiovascular disease, diabetes, and neurodegenerative diseases is discussed. Moreover, the integration with microfluidic chip systems for precise single-cell analysis is presented. To give a forward-looking view, the key challenges faced by SERS technology are also proposed, and possible solutions to overcome these obstacles are provided.
Collapse
Affiliation(s)
- Lei Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
- Department of Ultrasound Medicine, Affiliated Jinhua Hospital Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Yujiao Xie
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| | - Aochi Liu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| | - Liting Xie
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Xinyu Miao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Zhiwei Hou
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| | - Lingchao Xiang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| | - Tianan Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Ningbo, 315300, China
| |
Collapse
|
14
|
Almheiri RT, Hajjar B, Alkhaaldi SMI, Rabeh N, Aljoudi S, Abd-Elrahman KS, Hamdan H. Beyond weight loss: exploring the neurological ramifications of altered gut microbiota post-bariatric surgery. J Transl Med 2025; 23:223. [PMID: 39994634 PMCID: PMC11852891 DOI: 10.1186/s12967-025-06201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
This review discusses findings related to neurological disorders, gut microbiota, and bariatric surgery, focusing on neurotransmitters, neuroendocrine, the pathophysiology of bacteria contributing to disorders, and possible therapeutic interventions. Research on neurotransmitters suggests that their levels are heavily influenced by gut microbiota, which may link them to neurological disorders such as Alzheimer's disease, Parkinson's disease, Multiple sclerosis, Depression, and Autism spectrum disorder. The pathophysiology of bacteria that reach and influence the central nervous system has been documented. Trends in microbiota are often observed in specific neurological disorders, with a prominence of pro-inflammatory bacteria and a reduction in anti-inflammatory types. Furthermore, bariatric surgery has been shown to alter microbiota profiles similar to those observed in neurological disorders. Therapeutic interventions, including fecal microbiota transplants and probiotics, have shown potential to alleviate neurological symptoms. We suggest a framework for future studies that integrates knowledge from diverse research areas, employs rigorous methodologies, and includes long-trial clinical control groups.
Collapse
Affiliation(s)
- Rashed T Almheiri
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Baraa Hajjar
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Saif M I Alkhaaldi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Nadia Rabeh
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Sara Aljoudi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Khaled S Abd-Elrahman
- Department of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Department of Medical Sciences, College of Medicine and Health Science, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Hamdan Hamdan
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Healthcare Engineering Innovation Group (HEIG), Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
15
|
Jiménez A, Estudillo E, Guzmán-Ruiz MA, Herrera-Mundo N, Victoria-Acosta G, Cortés-Malagón EM, López-Ornelas A. Nanotechnology to Overcome Blood-Brain Barrier Permeability and Damage in Neurodegenerative Diseases. Pharmaceutics 2025; 17:281. [PMID: 40142945 PMCID: PMC11945272 DOI: 10.3390/pharmaceutics17030281] [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: 01/20/2025] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The blood-brain barrier (BBB) is a critical structure that maintains brain homeostasis by selectively regulating nutrient influx and waste efflux. Not surprisingly, it is often compromised in neurodegenerative diseases. In addition to its involvement in these pathologies, the BBB also represents a significant challenge for drug delivery into the central nervous system. Nanoparticles (NPs) have been widely explored as drug carriers capable of overcoming this barrier and effectively transporting therapies to the brain. However, their potential to directly address and ameliorate BBB dysfunction has received limited attention. In this review, we examine how NPs enhance drug delivery across the BBB to treat neurodegenerative diseases and explore emerging strategies to restore the integrity of this vital structure.
Collapse
Affiliation(s)
- Adriana Jiménez
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (A.J.); (G.V.-A.); (E.M.C.-M.)
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México 14269, Mexico;
| | - Mara A. Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Nieves Herrera-Mundo
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Georgina Victoria-Acosta
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (A.J.); (G.V.-A.); (E.M.C.-M.)
| | - Enoc Mariano Cortés-Malagón
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (A.J.); (G.V.-A.); (E.M.C.-M.)
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Ciudad de México 06800, Mexico
| | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico; (A.J.); (G.V.-A.); (E.M.C.-M.)
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Ciudad de México 06800, Mexico
| |
Collapse
|
16
|
Długosz A, Błaszak B, Czarnecki D, Szulc J. Mechanism of Action and Therapeutic Potential of Xanthohumol in Prevention of Selected Neurodegenerative Diseases. Molecules 2025; 30:694. [PMID: 39942798 PMCID: PMC11821245 DOI: 10.3390/molecules30030694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Xanthohumol (XN), a bioactive plant flavonoid, is an antioxidant, and as such, it exhibits numerous beneficial properties, including anti-inflammatory, antimicrobial, and antioxidative effects. The main dietary source of XN is beer, where it is introduced through hops. Although the concentration of XN in beer is low, the large quantities of hop-related post-production waste present an opportunity to extract XN residues for technological or pharmaceutical purposes. The presented study focuses on the role of XN in the prevention of neurodegenerative diseases, analyzing its effect at a molecular level and including its signal transduction and metabolism. The paper brings up XN's mechanism of action, potential effects, and experimental and clinical studies on Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Additionally, challenges and future research directions on XN, including its bioavailability, safety, and tolerance, have been discussed.
Collapse
Affiliation(s)
- Anna Długosz
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Błażej Błaszak
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Damian Czarnecki
- Faculty of Health Sciences, Department of Preventive Nursing, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-821 Bydgoszcz, Poland;
| | - Joanna Szulc
- Faculty of Chemical Technology and Engineering, Department of Food Industry Technology and Engineering, Bydgoszcz University of Science and Technology, 85-326 Bydgoszcz, Poland;
| |
Collapse
|
17
|
Fleige L, Capellino S. Acute stimulation of PBMCs drives switch from dopamine-induced anti- to proinflammatory phenotype of monocytes only in women. Biol Sex Differ 2025; 16:8. [PMID: 39901238 PMCID: PMC11789415 DOI: 10.1186/s13293-025-00689-5] [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: 10/09/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
Several studies report an impact of the neurotransmitter dopamine (DA) on human immune cells, with effects dependent on the immune cell type addressed and their activation status. Another contributing factor appears to be sex, as sex-specific differences in the dopaminergic pathway are described in the neurological context as well as in autoimmune diseases. However, a deeper understanding of these differences in peripheral immune cells remains limited. In this study, we investigated the effects of dopaminergic stimulation on activation and cytokine secretion of peripheral blood mononuclear cells from women and men using flow cytometry, ELISA, and multiplex assays. We found a B cell-driven downregulation in cytokine secretion of monocytes exclusively from women under physiological conditions in vitro. Moreover, B cells from men showed higher dopamine receptor (DR) expression, which was shown to be further increased by sex hormones only in men. In monocytes from women, an acute inflammatory stimulus via CpG combined with dopaminergic stimulation caused a switch to a proinflammatory phenotype, which was less pronounced in men. These novel findings in sex-specific responses to dopaminergic stimulation are crucial for understanding DA's function in the healthy and activated immune system and provide evidence to treat DA-related pathologies in a sex-specific manner.
Collapse
Affiliation(s)
- Leonie Fleige
- Department of Immunology, Research Group of Neuroimmunology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, Ardeystraße 67, 44139, Dortmund, Germany
| | - Silvia Capellino
- Department of Immunology, Research Group of Neuroimmunology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, Ardeystraße 67, 44139, Dortmund, Germany.
| |
Collapse
|
18
|
Adam H, Gopinath SCB, Krishnan H, Adam T, Fakhri MA, Salim ET, Shamsher A, Subramaniam S, Chen Y. Selective detection of alpha synuclein amyloid fibrils by faradaic and non-faradaic electrochemical impedance spectroscopic approaches. Bioelectrochemistry 2025; 161:108800. [PMID: 39241513 DOI: 10.1016/j.bioelechem.2024.108800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
This study utilized faradaic and non-faradaic electrochemical impedance spectroscopy to detect alpha synuclein amyloid fibrils on gold interdigitated tetraelectrodes (AuIDTE), providing valuable insights into electrochemical reactions for clinical use. AuIDE was purchased, modified with zinc oxide for increased hydrophobicity. Functionalization was conducted with hexacyanidoferrate and carbonyldiimidazole. Faradaic electrochemical impedance spectroscopy has been extensively explored in clinical diagnostics and biomedical research, providing information on the performance and stability of electrochemical biosensors. This understanding can help develop more sensitive, selective, and reliable biosensing platforms for the detection of clinically relevant analytes like biomarkers, proteins, and nucleic acids. Non-faradaic electrochemical impedance spectroscopy measures the interfacial capacitance at the electrode-electrolyte interface, eliminating the need for redox-active species and simplifying experimental setups. It has practical implications in clinical settings, like real-time detection and monitoring of biomolecules and biomarkers by tracking changes in interfacial capacitance. The limit of detection (LOD) for normal alpha synuclein in faradaic mode is 2.39-fM, The LOD for aggregated alpha synuclein detection is 1.82-fM. The LOD for non-faradaic detection of normal alpha synuclein is 2.22-fM, and the LOD for nonfaradaic detection of aggregated alpha synuclein is 2.40-fM. The proposed EIS-based AuIDTEs sensor detects alpha synuclein amyloid fibrils and it is highly sensitive.
Collapse
Affiliation(s)
- Hussaini Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia
| | - Subash C B Gopinath
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia; Center for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602 105, Tamil Nadu, India; Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600 Arau, Perlis, Malaysia; Department of Technical Sciences, Western Caspian University, Baku, AZ 1075, Azerbaijan; Centre for Chemical Biology, Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia; Department of Computer Science and Engineering, Faculty of Science and Information Technology, Daffodil International University, Daffodil Smart City, Birulia, Savar, Dhaka 1216, Bangladesh.
| | - Hemavathi Krishnan
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia
| | - Tijjani Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia; Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis, 02600 Arau, Perlis, Malaysia
| | - Makram A Fakhri
- Laser and Optoelectronics Eng. Department, University of Technology-Iraq, Baghdad 10066, Iraq
| | - Evan T Salim
- Applied Science Department, University of Technology-Iraq, Baghdad 10066, Iraq
| | - A Shamsher
- Electrical Engineering Department, Seberang Perai Polytechnic, 13500 Permatang Pauh, Penang, Malaysia
| | - Sreeramanan Subramaniam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia; Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600 Arau, Perlis, Malaysia; Centre for Chemical Biology, Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia; School of Biological Sciences, Universiti Sains Malaysia, Georgetown, 11800 Penang, Malaysia
| | - Yeng Chen
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
19
|
Tenchov R, Sasso JM, Zhou QA. Evolving Landscape of Parkinson's Disease Research: Challenges and Perspectives. ACS OMEGA 2025; 10:1864-1892. [PMID: 39866628 PMCID: PMC11755173 DOI: 10.1021/acsomega.4c09114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects movement. It occurs due to a gradual deficit of dopamine-producing brain cells, particularly in the substantia nigra. The precise etiology of PD is not fully understood, but it likely involves a combination of genetic and environmental factors. The therapies available at present alleviate symptoms but do not stop the disease's advancement. Research endeavors are currently directed at inventing disease-controlling therapies that aim at the inherent mechanisms of PD. PD biomarker breakthroughs hold enormous potential: earlier diagnosis, better monitoring, and targeted treatment based on individual response could significantly improve patient outcomes and ease the burden of this disease. PD research is an active and evolving field, focusing on understanding disease mechanisms, identifying biomarkers, developing new treatments, and improving care. In this report, we explore data from the CAS Content Collection to outline the research progress in PD. We analyze the publication landscape to offer perspective into the latest expertise advancements. Key emerging concepts are reviewed and strategies to fight disease evaluated. Pharmacological targets, genetic risk factors, as well as comorbid diseases are explored, and clinical usage of products against PD with their production pipelines and trials for drug repurposing are examined. This review aims to offer a comprehensive overview of the advancing landscape of the current understanding about PD, to define challenges, and to assess growth prospects to stimulate efforts in battling the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
20
|
Xie S, Yuan Y, Wang J, Bai Y, Wang T, Qiu B, Yang Y, Lin SC. Optimal dose and type of exercise improve walking velocity in adults with Parkinson's disease: a systematic review and Bayesian network meta-analysis. Sci Rep 2025; 15:2239. [PMID: 39824889 PMCID: PMC11742409 DOI: 10.1038/s41598-025-85456-7] [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: 09/07/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
To examine the dose-response relationship between specific types of exercise for improving walking velocity in Parkinson's disease (PD). This systematic review and network meta-analysis included searches of PubMed, Medline, Embase, PsycINFO, Cochrane Library, and Web of Science were searched from inception until February 18th, 2024. Data analysis was performed using R software with the MBNMA and RJAGS packages. Outcome indicators were measured as mean standard deviation (SMD) along with 95% confidence intervals (95% CrI). The network's risk of bias was independently evaluated by two reviewers employing the ROB2 tool. Our review encompassed 54 studies with 2,828 PD patients, examining the dose-response effects of different exercises on walking velocity. Aerobic Exercise (AE) demonstrated the greatest improvement at an optimal dose of 1,400 METs-min/week (SMD:1.215, 95% Crl: 0.113 to 2.306). Both Multicomponent Exercise (Mul) (SMD: 1.202, 95% Crl: 0.193 to 2.231) and Sensory Exercise (SE) (SMD: 0.649, 95% Crl: 0.139 to 1.183) showed optimal outcomes at a dose of 1,000 METs-min/week. Resistance Training (RT) was most effective at 750 METs-min/week (SMD:0.778, 95% Crl: 0.062 to 1.549), while Mind-Body Exercise (MBE) yielded significant improvements at a lower optimal dose of 500 METs-min/week (SMD: 0.580, 95% Crl: 0.218 to 1.137), offering valuable insights for exercise prescription in PD management. Various types of exercise showed specific optimal benefits at corresponding doses, among which AE was the most effective in improving the walking speed of PD patients at 1,400 Mets*min/week.Trial registration: PROSPERO (CRD42024506919).
Collapse
Affiliation(s)
- Shaoqing Xie
- School of Physical Education and Sport, West Anhui University, Shanghai, 237012, China
| | - Yuan Yuan
- Department of Physical Education, Kunsan National University, Kunsan, 541150, South Korea
| | - Junyu Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
- Officers College of PAP, Chengdu, 610200, China
| | - Ying Bai
- Department of Physical Education, Kunsan National University, Kunsan, 541150, South Korea
| | - Tao Wang
- School of Physical Education and Institute of Sports Biology, Shaanxi Normal University, Xian, 710119, Shaanxi, China
| | - Bopeng Qiu
- School of Strength and Conditioning Training, Beijing Sport University, Beijing, 100084, China
| | - Yong Yang
- Laboratory of Kinesiology and Rehabilitation, School of Physical Education and Sport, Chaohu University, No. 1 Xuefu Road, Chaohu Economic Development Zone, Hefei, 238000, Anhui, China.
| | - Shu-Cheng Lin
- School of Gerontology and Long-Term Care, Taipei Medical University, Taipei City 110, Taiwan (R.O.C.), Taipei, Taiwan.
| |
Collapse
|
21
|
Wang Z, Zhu H, Xiong W. Metabolism and metabolomics in senescence, aging, and age-related diseases: a multiscale perspective. Front Med 2025:10.1007/s11684-024-1116-0. [PMID: 39821730 DOI: 10.1007/s11684-024-1116-0] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
The pursuit of healthy aging has long rendered aging and senescence captivating. Age-related ailments, such as cardiovascular diseases, diabetes, and neurodegenerative disorders, pose significant threats to individuals. Recent studies have shed light on the intricate mechanisms encompassing genetics, epigenetics, transcriptomics, and metabolomics in the processes of senescence and aging, as well as the establishment of age-related pathologies. Amidst these underlying mechanisms governing aging and related pathology metabolism assumes a pivotal role that holds promise for intervention and therapeutics. The advancements in metabolomics techniques and analysis methods have significantly propelled the study of senescence and aging, particularly with the aid of multiscale metabolomics which has facilitated the discovery of metabolic markers and therapeutic potentials. This review provides an overview of senescence and aging, emphasizing the crucial role metabolism plays in the aging process as well as age-related diseases.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongying Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Wei Xiong
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| |
Collapse
|
22
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
23
|
Omejc N, Stankovski T, Peskar M, Kalc M, Manganotti P, Gramann K, Dzeroski S, Marusic U. Cortico-Muscular Phase Connectivity During an Isometric Knee Extension Task in People with Early Parkinson's Disease. IEEE Trans Neural Syst Rehabil Eng 2025; PP:488-501. [PMID: 40030955 DOI: 10.1109/tnsre.2025.3527578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Parkinson's disease (PD) is characterized by enhanced beta-band activity (13-30 Hz) in the motor control regions. Simultaneously, cortico-muscular (CM) connectivity in the beta-band during iso-metric contractions tends to decline with age, in various diseases, and under dual-task conditions. OBJECTIVE This study aimed to characterize electroencephalograph (EEG) and electromyograph (EMG) power spectra during a motor task, assess CM phase connectivity, and explore how these measures are modulated by an additional cognitive task. Specifically, we focused on the beta-band to explore the relationship between heightened beta amplitude and reduced beta CM connectivity. METHODOLOGY Early-stage people with PD and age-matched controls performed an isometric knee extension task, a cognitive task, and a combined dual task, while EEG (128ch) and EMG (2x32ch) were recorded. CM phase connectivity was assessed through phase coherence and a phase dynamics model. RESULTS The EEG power spectrum revealed no cohort differences in the beta-band. EMG also showed no differences up to 80 Hz. However, the combined EEG-EMG analysis uncovered reduced beta phase coherence in people with early PD during the motor task. CM phase coherence exhibited distinct scalp topography and frequency ranges compared to the EEG power spectrum, suggesting different mechanisms for pathological beta increase and CM connectivity. Additionally, phase dynamics modelling indicated stronger directional coupling from the cortex to the active muscle and less prominent phase coupling across people with PD. Despite high inter-individual variability, these metrics may prove useful for personalized assessments, particularly in people with heightened CM connectivity.
Collapse
|
24
|
Yi LX, Tan EK, Zhou ZD. The α-Synuclein Seeding Amplification Assay for Parkinson's Disease. Int J Mol Sci 2025; 26:389. [PMID: 39796243 PMCID: PMC11720040 DOI: 10.3390/ijms26010389] [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: 12/04/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. Currently, PD is incurable, and the diagnosis of PD mainly relies on clinical manifestations. The central pathological event in PD is the abnormal aggregation and deposition of misfolded α-synuclein (α-Syn) protein aggregates in the Lewy body (LB) in affected brain areas. Behaving as a prion-like seeding, the misfolded α-syn protein can induce and facilitate the aggregation of native unfolded α-Syn protein to aggravate α-Syn protein aggregation, leading to PD progression. Recently, in a blood-based α-Syn seeding amplification assay (SAA), Kluge et al. identified pathological α-Syn seeding activity in PD patients with Parkin (PRKN) gene variants. Additionally, pathological α-syn seeding activity was also identified in sporadic PD and PD patients with Leucine-rich repeat kinase 2 (LRRK2) or glucocerebrosidase (GBA) gene variants. Principally, the α-Syn SAA can be used to detect pathological α-Syn seeding activity, which will significantly enhance PD diagnosis, progression monitoring, prognosis prediction, and anti-PD therapy. The significance and future strategies of α-Syn SAA protocol are highlighted and proposed, whereas challenges and limitations of the assay are discussed.
Collapse
Affiliation(s)
- Ling-Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore;
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore;
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| | - Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore;
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
25
|
Teng WB, Deng HW, Lv BH, Zhou SD, Li BR, Hu RT. Exploring and validating key genetic biomarkers for diagnosis of Parkinson's disease. Brain Res Bull 2025; 220:111165. [PMID: 39667505 DOI: 10.1016/j.brainresbull.2024.111165] [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: 10/21/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurological condition characterized by complex genetic basic, and the reliable diagnosis of PD remained limited. OBJECTIVE To identify genes crucial to PD and assess their potential as diagnostic markers. METHODS Differentially expressed genes (DEGs) were screened from the PD tissue dataset and blood dataset. Two machine learning methods were used to identify key PD-related genes. The genes were validated in an independent dataset. Further validation using 120 peripheral blood mononuclear cells (PBMCs) from PD patients. The clinical significance and the diagnostic value of the genes was determined. The function of genes was analyzed and verified by cells experiments. RESULTS Thirteen common upregulated genes were identified between PD tissue dataset and blood dataset. Two machine learning methods identify three key PD-related genes (GPX2, CR1, ZNF556). An independent dataset and PBMCs samples results showed increased expression in PD patients. Clinical analysis showed that GPX2 and CR1 expression correlated with early-stage PD. The validated dataset of blood samples revealed each three gene showed moderate diagnostic potential for PD, with combined analysis outperforming individual gene analysis (AUC:0.701). The PBMCs samples showed similar diagnostic value of each gene, and the combination of the three genes presented better diagnostic value (AUC:0.801). Functional studies highlighted the involvement of these genes in key pathways in PD pathology. The results of SH-SY5Y cells showed that these three genes increased from PD cell model. CONCLUSIONS GPX2, CR1, ZNF556 were critical to the development of PD and might serve as diagnostic markers for PD.
Collapse
Affiliation(s)
- Wen-Bin Teng
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Hao-Wei Deng
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Bing-Hua Lv
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Shao-Dan Zhou
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Bin-Ru Li
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| | - Rui-Ting Hu
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| |
Collapse
|
26
|
Singh L. Daidzein's potential in halting neurodegeneration: unveiling mechanistic insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:243-259. [PMID: 39158734 DOI: 10.1007/s00210-024-03356-5] [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: 05/21/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Neurological conditions encompassing a wide range of disorders pose significant challenges globally. The complex interactions among signaling pathways and molecular elements play pivotal roles in the initiation and progression of neurodegenerative diseases. Isoflavones have emerged as a promising candidate to fight against neurodegenerative diseases. Daidzein, a 7-hydroxy-3-(4-hydroxyphenyl)-chromen-4-one, belongs to the isoflavone class and exhibits a diverse pharmacological profile. It is found primarily in soybeans and soy products, as well as in some other legumes and herbs. Investigations into daidzein have revealed that it confers neuroprotection by inhibiting oxidative stress, inflammation, and apoptosis, which are key contributors to neuronal damage and degeneration. Activating pathways like PI3K/Akt/mTOR and promoting neurotrophic factors like BDNF by daidzein underscore its potential in supporting neuronal function and combating neurodegeneration. Daidzein's effects on dopamine provide further avenues for intervention in conditions like Parkinson's disease. Additionally, the modulation of inflammatory and NRF-2-antioxidant signaling by daidzein reinforces its neuroprotective role. Moreover, daidzein's interaction with receptors and cellular processes like ER-β, GPR30, MAO, VEGF, and GnRH highlights its multifaceted effects across multiple pathways involved in neuroprotection and neuronal function. This review article delves into the mechanistic interplay of various mediators in mediating the neuroprotective effects of daidzein. The review article consolidates and analyzes research published over nearly two decades (2005-2024) from various databases, including PubMed, Scopus, ScienceDirect, and Web of Science, to provide a comprehensive understanding of daidzein's effects and mechanisms in neuroprotection.
Collapse
Affiliation(s)
- Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
27
|
Mushta I, Koks S, Popov A, Lysenko O. Exploring the Potential Imaging Biomarkers for Parkinson's Disease Using Machine Learning Approach. Bioengineering (Basel) 2024; 12:11. [PMID: 39851285 PMCID: PMC11762086 DOI: 10.3390/bioengineering12010011] [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: 11/04/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor and neuropsychiatric symptoms resulting from the loss of dopamine-producing neurons in the substantia nigra pars compacta (SNc). Dopamine transporter scan (DATSCAN), based on single-photon emission computed tomography (SPECT), is commonly used to evaluate the loss of dopaminergic neurons in the striatum. This study aims to identify a biomarker from DATSCAN images and develop a machine learning (ML) algorithm for PD diagnosis. Using 13 DATSCAN-derived parameters and patient handedness from 1309 individuals in the Parkinson's Progression Markers Initiative (PPMI) database, we trained an AdaBoost classifier, achieving an accuracy of 98.88% and an area under the receiver operating characteristic (ROC) curve of 99.81%. To ensure interpretability, we applied the local interpretable model-agnostic explainer (LIME), identifying contralateral putamen SBR as the most predictive feature for distinguishing PD from healthy controls. By focusing on a single biomarker, our approach simplifies PD diagnosis, integrates seamlessly into clinical workflows, and provides interpretable, actionable insights. Although DATSCAN has limitations in detecting early-stage PD, our study demonstrates the potential of ML to enhance diagnostic precision, contributing to improved clinical decision-making and patient outcomes.
Collapse
Affiliation(s)
- Illia Mushta
- Department of Electronic Computational Equipment Design, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 03056 Kyiv, Ukraine;
| | - Sulev Koks
- Perron Institute for Neurological and Translational Science, Murdoch University, Nedlands, WA 6009, Australia;
| | - Anton Popov
- Department of Electronic Engineering, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 03056 Kyiv, Ukraine;
- Faculty of Applied Sciences, Ukrainian Catholic University, 79026 Lviv, Ukraine
| | - Oleksandr Lysenko
- Department of Electronic Computational Equipment Design, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 03056 Kyiv, Ukraine;
| |
Collapse
|
28
|
Jiao Y, Yang L, Wang R, Song G, Fu J, Wang J, Gao N, Wang H. Drug Delivery Across the Blood-Brain Barrier: A New Strategy for the Treatment of Neurological Diseases. Pharmaceutics 2024; 16:1611. [PMID: 39771589 PMCID: PMC11677317 DOI: 10.3390/pharmaceutics16121611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The blood-brain barrier (BBB) serves as a highly selective barrier between the blood and the central nervous system (CNS), and its main function is to protect the brain from foreign substances. This physiological property plays a crucial role in maintaining CNS homeostasis, but at the same time greatly limits the delivery of drug molecules to the CNS, thus posing a major challenge for the treatment of neurological diseases. Given that the high incidence and low cure rate of neurological diseases have become a global public health problem, the development of effective BBB penetration technologies is important for enhancing the efficiency of CNS drug delivery, reducing systemic toxicity, and improving the therapeutic outcomes of neurological diseases. This review describes the physiological and pathological properties of the BBB, as well as the current challenges of trans-BBB drug delivery, detailing the structural basis of the BBB and its role in CNS protection. Secondly, this paper reviews the drug delivery strategies for the BBB in recent years, including physical, biological and chemical approaches, as well as nanoparticle-based delivery technologies, and provides a comprehensive assessment of the effectiveness, advantages and limitations of these delivery strategies. It is hoped that the review in this paper will provide valuable references and inspiration for future researchers in therapeutic studies of neurological diseases.
Collapse
Affiliation(s)
- Yimai Jiao
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Luosen Yang
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Rujuan Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Guoqiang Song
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Jingxuan Fu
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300401, China;
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| | - Na Gao
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Hui Wang
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300401, China; (Y.J.); (R.W.); (G.S.); (J.F.); (J.W.)
| |
Collapse
|
29
|
Ruggieri E, Di Domenico E, Locatelli AG, Isopo F, Damanti S, De Lorenzo R, Milan E, Musco G, Rovere-Querini P, Cenci S, Vénéreau E. HMGB1, an evolving pleiotropic protein critical for cellular and tissue homeostasis: Role in aging and age-related diseases. Ageing Res Rev 2024; 102:102550. [PMID: 39427887 DOI: 10.1016/j.arr.2024.102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Aging is a universal biological process characterized by a progressive, cumulative decline in homeostatic capabilities and physiological functions, which inevitably increases vulnerability to diseases. A number of molecular pathomechanisms and hallmarks of aging have been recognized, yet we miss a thorough understanding of their complex interconnectedness. This review explores the molecular and cellular mechanisms underlying human aging, with a focus on the multiple roles of high mobility group Box 1 protein (HMGB1), the archetypal damage-associated molecular pattern (DAMP) molecule. In the nucleus, this non-histone chromatin-associated protein functions as a DNA chaperone and regulator of gene transcription, influencing DNA structure and gene expression. Moreover, this versatile protein can translocate to the cytoplasm to orchestrate other processes, such as autophagy, or be unconventionally secreted into the extracellular environment, where it acts as a DAMP, combining inflammatory and regenerative properties. Notably, lower expression of HMGB1 within the cell and its heightened extracellular release have been associated with diverse age-associated traits, making it a suitable candidate as a universal biomarker of aging. In this review, we outline the evidence implicating HMGB1 in aging, also in light of an evolutionary perspective on its functional pleiotropy, and propose critical issues that need to be addressed to gauge the value of HMGB1 as a potential biomarker across age-related diseases and therapeutic target to promote healthy longevity.
Collapse
Affiliation(s)
- Elena Ruggieri
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Erika Di Domenico
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Flavio Isopo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Enrico Milan
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| | - Emilie Vénéreau
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
30
|
Zhang F, Ye Z, Xie Y, Liu M, Zhang L, Zhang J, Xu Z. Levodopa-induced dyskinesia: brain iron deposition as a new hypothesis. Biometals 2024; 37:1307-1323. [PMID: 39212870 DOI: 10.1007/s10534-024-00628-8] [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: 07/08/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in the older adults. The main pathological change in PD is the degenerative death of dopamine (DA) neurons in the midbrain substantia nigra, which causes a significant decrease in the DA content of the striatum. However, the exact etiology of this pathological change remains unclear. Genetic factors, environmental factors, aging, and oxidative stress may be involved in the degenerative death of dopaminergic neurons in PD. Pharmacological treatment using levodopa (L-DOPA) remains the main treatment for PD. Most patients with PD consuming L-DOPA for a long time usually develop levodopa-induced dyskinesia (LID) after 6.5 years of use, and LID seriously affects the quality of life and increases the risk of disability. Recently, studies have revealed that cerebral iron deposition may be involved in LID development and that iron deposition has neurotoxic effects and accelerates disease onset. However, the relationship between cerebral iron deposition and LID remains unclear. Herein, we reviewed the mechanisms by which iron deposition may be associated with LID development, which are mainly related to oxidative stress, neuroinflammation, and mitochondrial and lysosomal dysfunction. Using iron as an important target, the search and development of safe and effective brain iron scavengers, and thus the alleviation and treatment of LID, has a very important scientific and clinical value, as well as a good application prospect.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Zhuofan Ye
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
- Department of Neurology, Guizhou Pronvincial People's Hospital, Guiyang, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
31
|
Sánchez-Camacho JV, Gómez-Chavarín M, Galindo-Solano N, Padilla-Cortés P, Maldonado-García JL, Pérez-Sánchez G, Pavón L, Ramírez-Santos J, Roldán Roldán G, Gómez-López M, Gutierrez-Ospina G. Non-Categorical Analyses Identify Rotenone-Induced 'Parkinsonian' Rats Benefiting from Nano-Emulsified Punicic Acid (Nano-PSO) in a Phenotypically Diverse Population: Implications for Translational Neurodegenerative Therapies. Int J Mol Sci 2024; 25:12635. [PMID: 39684350 PMCID: PMC11640963 DOI: 10.3390/ijms252312635] [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: 07/28/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 12/18/2024] Open
Abstract
The pursuit of nutraceuticals to improve the quality of life for patients with neurodegenerative conditions is a dynamic field within neuropharmacology. Unfortunately, many nutraceuticals that show promise in preclinical studies fail to demonstrate significant clinical benefits in human trials, leading to their exclusion as therapeutic options. This discrepancy may stem from the categorical interpretation of preclinical and clinical results. Basic researchers often assume that non-human experimental animals exhibit less phenotypic variability than humans. This belief overlooks interindividual phenotype variation, thereby leading to categorical conclusions being drawn from experiments. Consequently, when human clinical trials are conducted, the researchers expect similarly conclusive results. If these results are not achieved, the nutraceutical is deemed ineffective for clinical use, even if numerous individuals might benefit. In our study, we evaluated whether analyzing phenotype variability and similarity through non-categorical methods could help identify rotenone (ROT)-treated rats that might benefit from consuming nano-emulsified punicic acid (Nano-PSO), even if the prevention of "parkinsonism" or the restoration of neurometabolic function is inconsistent across individuals. Our findings supported this hypothesis. The benefits of Nano-PSO were not categorical; however, analyzing phenotype variance allowed us to identify ROT rats with varying degrees of benefit from Nano-PSO consumption. Hence, the translational potential of results from basic science studies testing nutraceuticals as pharmaceutical products against neurodegeneration may improve if researchers also interpret their results using non-categorical methods of data analysis for population screening, even if the overall therapeutic outcomes for the entire population show internal inconsistencies.
Collapse
Affiliation(s)
| | - Margarita Gómez-Chavarín
- Laboratorio de Medicina Regenerativa y Canales Iónicos, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Nuria Galindo-Solano
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (N.G.-S.); (J.R.-S.)
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Patricia Padilla-Cortés
- Unidad de Cromatografía, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñíz”, Ciudad de México 14370, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñíz”, Ciudad de México 14370, Mexico
| | - Jesús Ramírez-Santos
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (N.G.-S.); (J.R.-S.)
| | - Gabriel Roldán Roldán
- Laboratorio de Neurología Conductual, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Modesto Gómez-López
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
| | - Gabriel Gutierrez-Ospina
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (N.G.-S.); (J.R.-S.)
- Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
32
|
Wang Y, Jiang Z, Chu C, Zhang Z, Wang J, Li D, He N, Fietkiewicz C, Zhou C, Kaiser M, Bai X, Zhang C, Liu C. Push-pull effects of basal ganglia network in Parkinson's disease inferred by functional MRI. NPJ Parkinsons Dis 2024; 10:224. [PMID: 39567536 PMCID: PMC11579490 DOI: 10.1038/s41531-024-00835-7] [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: 04/15/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
Deep brain stimulation (DBS) can ameliorate motor symptoms in Parkinson's disease (PD), but its mechanism remains unclear. This work constructs a multi-scale brain model using the fMRI data from 27 PD patients with subthalamic DBS and 30 healthy controls. The model fits microscopic coupling parameters in the cortico-basal ganglia-thalamic neural loop to match individual connectivity, finding the "push-pull" effect of basal ganglia network. Specifically, increased GABAergic projection into the thalamus from basal ganglia worsens rigidity, while reduced GABAergic projection within the cortex exacerbates bradykinesia, suggesting that the dopamine deficiency induces the chain coupling variations to "push" the network to an abnormal state. Conversely, DBS can alleviate rigidity by enhancing GABAergic projections within the basal ganglia, and improve bradykinesia by reducing cortical projections to basal ganglia, exhibiting that DBS "pulls" the network to a healthy state. This work combines the microscopic and macroscopic neural information for understanding PD and its treatment.
Collapse
Affiliation(s)
- Yuxin Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Zhiqi Jiang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Chunguang Chu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Zhen Zhang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Jiang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Dianyou Li
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Naying He
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chris Fietkiewicz
- Department of Mathematics & Computer Science, Hobart and William Smith Colleges, Geneva, NY, USA
| | - Changsong Zhou
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Marcus Kaiser
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Xuze Bai
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Clinical Neuroscience Center, Ruijin Hospital LuWan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Technology, Shanghai, China.
| | - Chen Liu
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China.
| |
Collapse
|
33
|
Grimaldi L, Bovi E, Formisano R, Sancesario G. ApoE: The Non-Protagonist Actor in Neurological Diseases. Genes (Basel) 2024; 15:1397. [PMID: 39596597 PMCID: PMC11593850 DOI: 10.3390/genes15111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Apolipoprotein E (APOE = gene, ApoE = protein) is a glycoprotein involved in the biological process of lipid transportation and metabolism, contributing to lipid homeostasis. APOE has been extensively studied for its correlation with neurodegenerative diseases, in particular Alzheimer's disease (AD), where the possession of the epsilon 4 (E4) allele is established as a risk factor for developing AD in non-familiar sporadic forms. Recently, evidence suggests a broad involvement of E4 also in other neurological conditions, where it has been shown to be a predictive marker for worse clinical outcomes in Parkinson's disease (PD), brain trauma, and disturbances of consciousness. The mechanisms underlying these associations are complex and involve amyloid-β (Aβ) peptide accumulation and neuroinflammation, although many others have yet to be identified. OBJECTIVES The aim of this review is to overview the current knowledge on ApoE as a non-protagonist actor in processes underlying neurodegenerative diseases and its clinical significance in AD, PD, acquired brain trauma, and Disorders of Consciousness (DoC). Ethical implications of genetic testing for APOE variants and information disclosure will also be briefly discussed.
Collapse
Affiliation(s)
- Lorenzo Grimaldi
- Clinical Neurochemistry Unit and Biobank, IRCCS Santa Lucia Foundation, Via Ardeatina, 306/354, 00179 Rome, Italy
- European Center for Brain Research, Via del Fosso del Fiorano, 00143 Rome, Italy
| | - Eleonora Bovi
- Clinical Neurochemistry Unit and Biobank, IRCCS Santa Lucia Foundation, Via Ardeatina, 306/354, 00179 Rome, Italy
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy
| | - Rita Formisano
- Post-Coma Unit and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina, 306/354, 00179 Rome, Italy
| | - Giulia Sancesario
- Clinical Neurochemistry Unit and Biobank, IRCCS Santa Lucia Foundation, Via Ardeatina, 306/354, 00179 Rome, Italy
- European Center for Brain Research, Via del Fosso del Fiorano, 00143 Rome, Italy
| |
Collapse
|
34
|
Zhao K, Wang D, Wang D, Chen P, Wei Y, Tu L, Chen Y, Tang Y, Yao H, Zhou B, Lu J, Wang P, Liao Z, Chen Y, Han Y, Zhang X, Liu Y. Macroscale connectome topographical structure reveals the biomechanisms of brain dysfunction in Alzheimer's disease. SCIENCE ADVANCES 2024; 10:eado8837. [PMID: 39392880 PMCID: PMC11809497 DOI: 10.1126/sciadv.ado8837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/11/2024] [Indexed: 10/13/2024]
Abstract
The intricate spatial configurations of brain networks offer essential insights into understanding the specific patterns of brain abnormalities and the underlying biological mechanisms associated with Alzheimer's disease (AD), normal aging, and other neurodegenerative disorders. This study investigated alterations in the topographical structure of the brain related to aging and neurodegenerative diseases by analyzing brain gradients derived from structural MRI data across multiple cohorts (n = 7323). The analysis identified distinct gradient patterns in AD, aging, and other neurodegenerative conditions. Gene enrichment analysis indicated that inorganic ion transmembrane transport was the most significant term in normal aging, while chemical synaptic transmission is a common enrichment term across various neurodegenerative diseases. Moreover, the findings show that each disorder exhibits unique dysfunctional neurophysiological characteristics. These insights are pivotal for elucidating the distinct biological mechanisms underlying AD, thereby enhancing our understanding of its unique clinical phenotypes in contrast to normal aging and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Research Institute of Shandong University: Magnetic Field-free Medicine & Functional Imaging, Jinan, China
- Shandong Key Laboratory: Magnetic Field-free Medicine & Functional Imaging (MF), Jinan, China
| | - Dong Wang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Pindong Chen
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| | - Yongbin Wei
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Liyun Tu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yuqi Chen
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yi Tang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongxiang Yao
- Department of Radiology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhengluan Liao
- Department of Psychiatry, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yan Chen
- Department of Psychiatry, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xi Zhang
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
35
|
Xie L, Liu Y, Gao Y, Zhou J. Functional Near-Infrared Spectroscopy in neurodegenerative disease: a review. Front Neurosci 2024; 18:1469903. [PMID: 39416953 PMCID: PMC11479976 DOI: 10.3389/fnins.2024.1469903] [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: 07/24/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
In recent years, with the aggravation of aging, the incidence of neurodegenerative diseases is increasing year by year, and the prognosis of patients is poor. Functional Near-Infrared Spectroscopy (fNIRS) is a new and non-invasive neuroimaging technology, which has been gradually deepened in the application research of neurodegenerative diseases by virtue of its unique neurooxygen signal brain functional imaging characteristics in monitoring the disease condition, making treatment plans and evaluating the treatment effect. In this paper, the mechanism of action and technical characteristics of fNIRS are briefly introduced, and the application research of fNIRS in different neurodegenerative diseases is summarized in order to provide new ideas for future related research and clinical application.
Collapse
Affiliation(s)
| | - Yong Liu
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | | | | |
Collapse
|
36
|
Bizup B, Tzounopoulos T. On the genesis and unique functions of zinc neuromodulation. J Neurophysiol 2024; 132:1241-1254. [PMID: 39196675 PMCID: PMC11495185 DOI: 10.1152/jn.00285.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/30/2024] Open
Abstract
In addition to the essential structural and catalytic functions of zinc, evolution has adopted synaptic zinc as a neuromodulator. In the brain, synaptic zinc is released primarily from glutamatergic neurons, notably in the neocortex, hippocampus, amygdala, and auditory brainstem. In these brain areas, synaptic zinc is essential for neuronal and sensory processing fine-tuning. But what niche does zinc fill in neural signaling that other neuromodulators do not? Here, we discuss the evolutionary history of zinc as a signaling agent and its eventual adoption as an essential neuromodulator in the mammalian brain. We then attempt to describe the unique roles that zinc has carved out of the vast and diverse landscape of neuromodulators.
Collapse
Affiliation(s)
- Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
37
|
Weishaupt AK, Ruecker L, Meiners T, Schwerdtle T, Silva Avila D, Aschner M, Bornhorst J. Copper-mediated neurotoxicity and genetic vulnerability in the background of neurodegenerative diseases in C. elegans. Toxicol Sci 2024; 201:254-262. [PMID: 39067045 PMCID: PMC11424883 DOI: 10.1093/toxsci/kfae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
The mechanisms associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), have yet to be fully characterized, and genetic as well as environmental factors in their disease etiology are underappreciated. Although mutations in genes such as PARKIN and LRRK2 have been linked to PD, the idiopathic component of the disease suggests a contribution of environmental risk factors, including metals, such as copper (Cu). Cu overexposure has been reported to cause oxidative stress and neurotoxicity, but its role in neurodegenerative diseases is rarely studied. Using Caenorhabditis elegans (C. elegans) as a model organism for neurotoxicity, we assessed the effects of Cu oversupply in AD and PD models. Our findings reveal that although copper treatment did not induce neurodegeneration in wild-type worms or the AD model, it significantly exacerbated neurodegeneration in the PD-associated mutants PARKIN and LRRK2. These results suggest that genetic predisposition for PD enhances the sensitivity to copper toxicity, highlighting the multifactorial nature of neurodegenerative diseases. Furthermore, our study provides insight into the mechanisms underlying Cu-induced neurotoxicity in PD models, including disruptions in dopamine levels, altered dopamine-dependent behavior and degraded dopaminergic neurons. Overall, our novel findings contribute to a better understanding of the complex interactions between genetic susceptibility, environmental factors, and neurodegenerative disease pathogenesis, emphasizing the importance of a tightly regulated Cu homeostasis in the etiology of PD. Copper oversupply exacerbated neurodegeneration in Caenorhabditis elegans models of Parkinson's disease, highlighting the genetic susceptibility and emphasizing the crucial role of tightly regulated copper homeostasis in Parkinson's disease pathogenesis.
Collapse
Affiliation(s)
- Ann-Kathrin Weishaupt
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| | - Lysann Ruecker
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Torben Meiners
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Tanja Schwerdtle
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Daiana Silva Avila
- Laboratory of Toxicology and Biochemistry in Caenorhabditis elegans, Universidade Federal do Pampa, 97501-970 Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Julia Bornhorst
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| |
Collapse
|
38
|
Razali K, Kumar J, Mohamed WMY. Characterizing the adult zebrafish model of Parkinson's disease: a systematic review of dynamic changes in behavior and physiology post-MPTP administration. Front Neurosci 2024; 18:1432102. [PMID: 39319314 PMCID: PMC11420122 DOI: 10.3389/fnins.2024.1432102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/13/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Adult zebrafish are increasingly used in Parkinson's disease (PD) research due to their well-characterized dopaminergic system. Among the toxin-based models, the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is widely utilized to induce parkinsonism in adult zebrafish. Therefore, this review presents an overview of the procedures and the dynamic changes in behavior and physiology observed in the adult zebrafish PD model following a single intraperitoneal injection of MPTP. Methods A systematic literature search in the PubMed and Google Scholar databases was conducted to identify relevant articles. Of the 165 articles identified, 9 were included in this review. These chosen articles are original works published before March 2024, all of which utilized adult zebrafish induced with MPTP as the model for PD. Other articles were excluded based on factors such as limited relevance, utilization of zebrafish embryos or larvae instead of adults, and variations in MPTP deliveries. Results Studies indicated that the ideal model entails the utilization of mixed gender zebrafish aged between 4 and 6 months from the wild-type strain. The acceptable MPTP doses ranges between 20 μg/g (lowest) and 225 μg/g (highest) and doses above 292 μg/g are lethal. Furthermore, noticeable parkinsonian symptoms appear 1 day after administration and persist for more than 1 week. Discussion Mitochondrial dysfunction precedes dopaminergic neurodegeneration within this experimental regime. A single administration of MPTP effectively induces PD in adult zebrafish. This study aids in crafting the adult zebrafish PD model, outlining the progressive behavioral and physiological changes ensuing from MPTP administration.
Collapse
Affiliation(s)
- Khairiah Razali
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wael M. Y. Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
39
|
Zhang C, Shao W, Yuan H, Xiao R, Zhang Y, Wei C, Ni X, He N, Chen G, Gui S, Cheng Z, Wang Q. Engineered Extracellular Vesicle-Based Nanoformulations That Coordinate Neuroinflammation and Immune Homeostasis, Enhancing Parkinson's Disease Therapy. ACS NANO 2024; 18:23014-23031. [PMID: 39145985 DOI: 10.1021/acsnano.4c04674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Although conventional intervention to microglia can mitigate neuroinflammation in the short term, immune disorders by peripheral inflammatory cells can infiltrate continuously, resulting in an overactivated immune microenvironment of Parkinson's disease (PD). Here, we design engineered extracellular vesicle-based nanoformulations (EVNs) to address multiple factors for the management of PD. Specifically, EVN is developed by coating CCR2-enriched mesenchymal stem cell-derived extracellular vesicles (MSCCCR2 EVs) onto a dihydrotanshinone I-loaded nanocarrier (MSeN-DT). The MSCCCR2 EVs (the shell of EVN) can actively show homing to specific chemokines CCL2 in the substantia nigra, which enables them to block the infiltration of peripheral inflammatory cells. Interestingly, MSeN-DT (the core of EVN) can promote the Nrf2-GPX4 pathway for the suppression of the source of inflammation by inhibiting ferroptosis in microglia. In the PD model mice, a satisfactory therapeutic effect is achieved, with inhibition of peripheral inflammatory cell infiltration, precise regulation of inflammatory microglia in the substantia nigra, as well as promotion of behavioral improvement and repairing damaged neurons. In this way, the combinatorial code of alleviation of inflammation and modulation of immune homeostasis can reshape the immune microenvironment in PD, which bridges internal anti-inflammatory and external immunity. This finding reveals a comprehensive therapeutic paradigm for PD that breaks the vicious cycle of immune overactivation.
Collapse
Affiliation(s)
- Chuan Zhang
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Wei Shao
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Hao Yuan
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Ru Xiao
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Yaru Zhang
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Chaoqi Wei
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Xinyi Ni
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Ning He
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Guangliang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Shuangying Gui
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Zhifei Cheng
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Qi Wang
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| |
Collapse
|
40
|
Salama RM, Darwish SF, Yehia R, Eissa N, Elmongy NF, Abd-Elgalil MM, Schaalan MF, El Wakeel SA. Apilarnil exerts neuroprotective effects and alleviates motor dysfunction by rebalancing M1/M2 microglia polarization, regulating miR-155 and miR-124 expression in a rotenone-induced Parkinson's disease rat model. Int Immunopharmacol 2024; 137:112536. [PMID: 38909495 DOI: 10.1016/j.intimp.2024.112536] [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: 02/06/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Microglial activation contributes to the neuropathology of Parkinson's disease (PD). Inhibiting M1 while simultaneously boosting M2 microglia activation may therefore be a potential treatment for PD. Apilarnil (API) is a bee product produced from drone larvae. Recent research has demonstrated the protective effects of API on multiple body systems. Nevertheless, its impact on PD or the microglial M1/M2 pathway has not yet been investigated. Thus, we intended to evaluate the dose-dependent effects of API in rotenone (ROT)-induced PD rat model and explore the role of M1/M2 in mediating its effect. Seventy-two Wistar rats were equally grouped as; control, API, ROT, and groups in which API (200, 400, and 800 mg/kg, p.o.) was given simultaneously with ROT (2 mg/kg, s.c.) for 28 days. The high dose of API (800 mg/kg) showed enhanced motor function, higher expression of tyrosine hydroxylase and dopamine levels, less dopamine turnover and α-synuclein expression, and a better histopathological picture when compared to the ROT group and the lower two doses. API's high dose exerted its neuroprotective effects through abridging the M1 microglial activity, illustrated in the reduced expression of miR-155, Iba-1, CD36, CXCL10, and other pro-inflammatory markers' levels. Inversely, API high dose enhanced M2 microglial activity, witnessed in the elevated expression of miR-124, CD206, Ym1, Fizz1, arginase-1, and other anti-inflammatory indices, in comparison to the diseased group. To conclude, our study revealed a novel neuroprotective impact for API against experimentally induced PD, where the high dose showed the highest protection via rebalancing M1/M2 polarization.
Collapse
Affiliation(s)
- Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt.
| | - Rana Yehia
- Pharmacology and Toxicology Department, Faculty of Pharmacy, British University in Egypt (BUE), Cairo, Egypt.
| | - Nermin Eissa
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates.
| | - Noura F Elmongy
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt.
| | - Mona M Abd-Elgalil
- Histology and Cell Biology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt.
| | - Mona F Schaalan
- Clinical Pharmacy Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Sara A El Wakeel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| |
Collapse
|
41
|
Suzuki T, Bono H. A systematic exploration of unexploited genes for oxidative stress in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:160. [PMID: 39154038 PMCID: PMC11330442 DOI: 10.1038/s41531-024-00776-1] [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: 03/12/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Human disease-associated gene data are accessible through databases, including the Open Targets Platform, DisGeNET, miRTex, RNADisease, and PubChem. However, missing data entries in such databases are anticipated because of curational errors, biases, and text-mining failures. Additionally, the extensive research on human diseases has led to challenges in registering comprehensive data. The lack of essential data in databases hinders knowledge sharing and should be addressed. Therefore, we propose an analysis pipeline to explore missing entries of unexploited genes in the human disease-associated gene databases. Using this pipeline for genes in Parkinson's disease with oxidative stress revealed two unexploited genes: nuclear protein 1 (NUPR1) and ubiquitin-like with PHD and ring finger domains 2 (UHRF2). This methodology enhances the identification of underrepresented disease-associated genes, facilitating easier access to potential human disease-related functional genes. This study aims to identify unexploited genes for further research and does not include independent experimental validation.
Collapse
Affiliation(s)
- Takayuki Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan
| | - Hidemasa Bono
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
- Database Center for Life Science (DBCLS), Joint Support-Center for Data Science Research, Research Organization of Information and Systems (ROIS), 178-4-4 Wakashiba, Kashiwa, Chiba, 277-0871, Japan.
| |
Collapse
|
42
|
Ploper D, Pernicone AO, Tomas-Grau RH, Manzano VE, Socías SB, Teran MDM, Budeguer Isa V, Sosa-Padilla B, González-Lizárraga F, Avila CL, Guayán ML, Chaves S, Cruz H, Vera Pingitore E, Varela O, Chehín R. Design, Synthesis, and Evaluation of a Novel Conjugate Molecule with Dopaminergic and Neuroprotective Activities for Parkinson's Disease. ACS Chem Neurosci 2024; 15:2795-2810. [PMID: 38991155 DOI: 10.1021/acschemneuro.4c00169] [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: 07/13/2024] Open
Abstract
The escalating prevalence of Parkinson's disease (PD) underscores the need for innovative therapeutic interventions since current palliative measures, including the standard l-Dopa formulations, face challenges of tolerance and side effects while failing to address the underlying neurodegenerative processes. Here, we introduce DAD9, a novel conjugate molecule that aims to combine symptomatic relief with disease-modifying strategies for PD. Crafted through knowledge-guided chemistry, the molecule combines a nonantibiotic doxycycline derivative with dopamine, preserving neuroprotective attributes while maintaining dopaminergic agonism. This compound exhibited no off-target effects on PD-relevant cell functions and sustained antioxidant and anti-inflammatory properties of the tetracycline precursor. Furthermore, it effectively interfered with the formation and seeding of toxic α-synuclein aggregates without producing detrimental oxidative species. In addition, DAD9 was able to activate dopamine receptors, and docking simulations shed light onto the molecular details of this interaction. These findings position DAD9 as a potential neuroprotective dopaminergic agonist, promising advancements in PD therapeutics.
Collapse
Affiliation(s)
- Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Agustín O Pernicone
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rodrigo H Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Verónica E Manzano
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Del Milagro Teran
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Valentina Budeguer Isa
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Bernardo Sosa-Padilla
- Instituto de Química del Noroeste Argentino (INQUINOA) (CONICET), Universidad Nacional de Tucumán (UNT), Ayacucho 471, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - César L Avila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Laura Guayán
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Silvina Chaves
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Hernán Cruz
- Instituto de Química Física, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Lorenzo 456, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Esteban Vera Pingitore
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Oscar Varela
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| |
Collapse
|
43
|
Peck EG, Holleran KM, Curry AM, Holter KM, Estave PM, Sens JP, Locke JL, Ortelli OA, George BE, Dawes MH, West AM, Alexander NJ, Kiraly DD, Farris SP, Gould RW, McCool BA, Jones SR. Synaptogyrin-3 Prevents Cocaine Addiction and Dopamine Deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.27.605436. [PMID: 39211138 PMCID: PMC11361146 DOI: 10.1101/2024.07.27.605436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Synaptogyrin-3, a functionally obscure synaptic vesicle protein, interacts with vesicular monoamine and dopamine transporters, bringing together dopamine release and reuptake sites. Synaptogyrin-3 was reduced by chronic cocaine exposure in both humans and rats, and synaptogyrin-3 levels inversely correlated with motivation to take cocaine in rats. Synaptogyrin-3 overexpression in dopamine neurons reduced cocaine self-administration, decreased anxiety-like behavior, and enhanced cognitive flexibility. Overexpression also enhanced nucleus accumbens dopamine signaling and prevented cocaine-induced deficits, suggesting a putative therapeutic role for synaptogyrin-3 in cocaine use disorder.
Collapse
|
44
|
Chen H, Wang X, Chang Z, Zhang J, Xie D. Evidence for genetic causality between iron homeostasis and Parkinson's disease: A two-sample Mendelian randomization study. J Trace Elem Med Biol 2024; 84:127430. [PMID: 38484633 DOI: 10.1016/j.jtemb.2024.127430] [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: 02/01/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a degenerative disease of the central nervous system, and its specific etiology is still unclear. At present, it is believed that the main pathological basis is the reduction of dopamine concentration in the brain striatum. Although many previous studies have believed that iron as an important nutrient element participates in the occurrence and development of PD, whether there is a causal correlation between total iron binding capacity(TIBC), transferring saturation(TSAT), ferritin and serum iron in iron homeostasis indicators and PD, there has been a lack of effective genetic evidence. METHODS We used Mendelian randomization (MR) as an analytical method to effectively evaluate the genetic association between exposure and outcome, based on the largest genome-wide association study (GWAS) data to date. By using randomly assigned genetic instrumental variables (SNPs, Single Nucleotide Polymorphisms) that are not affected by any causal relationship, we effectively evaluated the causal relationship between iron homeostasis indicators and PD while controlling for confounding factors. RESULTS By coordinated analysis of 86 SNPs associated with iron homeostasis markers and 12,858,066 SNPs associated with PD, a total of 56 SNPs were finally screened for genome-wide significance of iron homeostasis associated with PD. The results of inverse variance weighting(IVW) analysis suggested that iron( β = - 0.524; 95%cl=-0.046 to -0.002; P=0.032) was considered to have a genetic causal relationship with PD. Cochran's Q, Egger intercept and MR-PRESSO global tests did not detect the existence of heterogeneity and pleiotropy (P>0.05). Mr Steiger directionality test further confirmed our estimation of the potential causal direction of iron and PD (P=0.001). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.387; 95%Cl=-1.179-0.405; P=0.338) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.101; 95%Cl=--0.987 to -0.405; P=0.823) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). TIBC (P=0.008), TSAT (P=0.000) and ferritin (P=0.013) were all consistent with the estimation of MR Steiger directivity test. CONCLUSION Our study found that among the four iron homeostasis markers, there was a genetic causal association between serum iron and PD, and the serum iron level was negatively correlated with the risk of PD. In addition, TIBC, TSAT, ferritin had no genetic causal relationship with PD.
Collapse
Affiliation(s)
- Hong Chen
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ze Chang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100089, China
| | - Juan Zhang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China.
| | - Daojun Xie
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
| |
Collapse
|
45
|
Selvaraj DB, Panneerselvam A, Vergil Andrews JF, Kandasamy M. Cysteamine HCl Administration Impedes Motor and Olfactory Functions, Accompanied by a Reduced Number of Dopaminergic Neurons, in Experimental Mice: A Preclinical Mimetic Relevant to Parkinson's Disease. Brain Sci 2024; 14:632. [PMID: 39061373 PMCID: PMC11275195 DOI: 10.3390/brainsci14070632] [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: 06/05/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Cysteamine hydrochloride (Cys-HCl) has been established as a potent ulcerogenic agent of the gastrointestinal (GI) system. GI dysfunction and olfactory deficits are the most common clinical symptoms of many movement disorders, including Parkinson's disease (PD). Cys-HCl has been shown to interfere with dopamine, a neurotransmitter crucial for motor, olfactory, and cognitive functions. However, the reports on the effect of Cys-HCl treatment on the behavioral aspects and functions of the dopamine system appear to be inconsistent. Therefore, we revisited the impact of Cys-HCl on the motor function in experimental mice using a battery of behavioral tests, such as the pole test (PT), beam-walking test (BWT), and rotarod test (RDT), while the olfactory ability and cognitive functions were examined through the buried-food test (BFT) and Y-maze test. Furthermore, we investigated the effect of Cys-HCl on the number of dopaminergic tyrosine hydroxylase (TH)-positive cells in the substantia nigra (SN) and olfactory bulb (OB) of the experimental mice using immunohistochemistry. The results revealed that Cys-HCl administration in the mice induced significant impairments in their motor balance and coordination, as their movement-related performances were markedly reduced in terms of the behavioral tasks. Mice exposed to Cys-HCl showed pronounced reductions in their odor discrimination abilities as well as cognitive impairments. Strikingly, the number of TH-positive neurons was found to be reduced in the SN and OB of the Cys-HCl-treated group, which is a bonafide neuropathogenic hallmark of PD. This study highlights the potential neurotoxic effects of Cys-HCl in experimental brains and suggests further investigation into its role in the pathogenesis of Parkinsonism.
Collapse
Affiliation(s)
- Divya Bharathi Selvaraj
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
| | - Anusiya Panneerselvam
- Department of Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India;
| | - Jemi Feiona Vergil Andrews
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (D.B.S.); (J.F.V.A.)
- University Grants Commission-Faculty Recharge Programme (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
46
|
Meca AD, Boboc IKS, Mititelu-Tartau L, Bogdan M. Unlocking the Potential: Semaglutide's Impact on Alzheimer's and Parkinson's Disease in Animal Models. Curr Issues Mol Biol 2024; 46:5929-5949. [PMID: 38921025 PMCID: PMC11202139 DOI: 10.3390/cimb46060354] [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: 05/17/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Semaglutide (SEM), a glucagon-like peptide-1 receptor agonist, has garnered increasing interest for its potential therapeutic effects in neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). This review provides a comprehensive description of SEM's mechanism of action and its effects in preclinical studies of these debilitating conditions. In animal models of AD, SEM has proved beneficial effects on multiple pathological hallmarks of the disease. SEM administration has been associated with reductions in amyloid-beta plaque deposition and mitigation of neuroinflammation. Moreover, SEM treatment has been shown to ameliorate behavioral deficits related to anxiety and social interaction. SEM-treated animals exhibit improvements in spatial learning and memory retention tasks, as evidenced by enhanced performance in maze navigation tests and novel object recognition assays. Similarly, in animal models of PD, SEM has demonstrated promising neuroprotective effects through various mechanisms. These include modulation of neuroinflammation, enhancement of mitochondrial function, and promotion of neurogenesis. Additionally, SEM has been shown to improve motor function and ameliorate dopaminergic neuronal loss, offering the potential for disease-modifying treatment strategies. Overall, the accumulating evidence from preclinical studies suggests that SEM holds promise as a novel therapeutic approach for AD and PD. Further research is warranted to elucidate the underlying mechanisms of SEM's neuroprotective effects and to translate these findings into clinical applications for the treatment of these devastating neurodegenerative disorders.
Collapse
Affiliation(s)
- Andreea Daniela Meca
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania; (A.D.M.); (I.K.S.B.)
| | - Ianis Kevyn Stefan Boboc
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania; (A.D.M.); (I.K.S.B.)
| | - Liliana Mititelu-Tartau
- Department of Pharmacology, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Maria Bogdan
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania; (A.D.M.); (I.K.S.B.)
| |
Collapse
|
47
|
Yi LX, Tan EK, Zhou ZD. Tyrosine Hydroxylase Inhibitors and Dopamine Receptor Agonists Combination Therapy for Parkinson's Disease. Int J Mol Sci 2024; 25:4643. [PMID: 38731862 PMCID: PMC11083272 DOI: 10.3390/ijms25094643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
There are currently no disease-modifying therapies for Parkinson's disease (PD), a progressive neurodegenerative disorder associated with dopaminergic neuronal loss. There is increasing evidence that endogenous dopamine (DA) can be a pathological factor in neurodegeneration in PD. Tyrosine hydroxylase (TH) is the key rate-limiting enzyme for DA generation. Drugs that inhibit TH, such as alpha-methyltyrosine (α-MT), have recently been shown to protect against neurodegeneration in various PD models. DA receptor agonists can activate post-synaptic DA receptors to alleviate DA-deficiency-induced PD symptoms. However, DA receptor agonists have no therapeutic effects against neurodegeneration. Thus, a combination therapy with DA receptor agonists plus TH inhibitors may be an attractive therapeutic approach. TH inhibitors can protect and promote the survival of remaining dopaminergic neurons in PD patients' brains, whereas DA receptor agonists activate post-synaptic DA receptors to alleviate PD symptoms. Additionally, other PD drugs, such as N-acetylcysteine (NAC) and anticholinergic drugs, may be used as adjunctive medications to improve therapeutic effects. This multi-drug cocktail may represent a novel strategy to protect against progressive dopaminergic neurodegeneration and alleviate PD disease progression.
Collapse
Affiliation(s)
- Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
- Department of Neurology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| | - Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore;
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
48
|
Kalinderi K, Papaliagkas V, Fidani L. GLP-1 Receptor Agonists: A New Treatment in Parkinson's Disease. Int J Mol Sci 2024; 25:3812. [PMID: 38612620 PMCID: PMC11011817 DOI: 10.3390/ijms25073812] [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: 02/13/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Recent data highlight similarities between neurodegenerative diseases, including PD and type 2 diabetes mellitus (T2DM), suggesting a crucial interplay between the gut-brain axis. Glucagon-like peptide-1 receptor (GLP-1R) agonists, known for their use in T2DM treatment, are currently extensively studied as novel PD modifying agents. For this narrative review article, we searched PubMed and Scopus databases for peer-reviewed research, review articles and clinical trials regarding GLP-1R agonists and PD published in the English language with no time restrictions. We also screened the references of the selected articles for possible additional articles in order to include most of the key recent evidence. Many data on animal models and preclinical studies show that GLP1-R agonists can restore dopamine levels, inhibit dopaminergic loss, attenuate neuronal degeneration and alleviate motor and non-motor features of PD. Evidence from clinical studies is also very promising, enhancing the possibility of adding GLP1-R agonists to the current armamentarium of drugs available for PD treatment.
Collapse
Affiliation(s)
- Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece;
| | - Liana Fidani
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
49
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
50
|
Thangavel R, Kaur H, Dubova I, Selvakumar GP, Ahmed ME, Raikwar SP, Govindarajan R, Kempuraj D. Parkinson's Disease Dementia Patients: Expression of Glia Maturation Factor in the Brain. Int J Mol Sci 2024; 25:1182. [PMID: 38256254 PMCID: PMC11154259 DOI: 10.3390/ijms25021182] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disease characterized by the presence of dopaminergic neuronal loss and motor disorders. PD dementia (PDD) is a cognitive disorder that affects many PD patients. We have previously demonstrated the proinflammatory role of the glia maturation factor (GMF) in neuroinflammation and neurodegeneration in AD, PD, traumatic brain injury (TBI), and experimental autoimmune encephalomyelitis (EAE) in human brains and animal models. The purpose of this study was to investigate the expression of the GMF in the human PDD brain. We analyzed the expression pattern of the GMF protein in conjunction with amyloid plaques (APs) and neurofibrillary tangles (NFTs) in the substantia nigra (SN) and striatum of PDD brains using immunostaining. We detected a large number of GMF-positive glial fibrillary acidic protein (GFAP) reactive astrocytes, especially abundant in areas with degenerating dopaminergic neurons within the SN and striatum in PDD. Additionally, we observed excess levels of GMF in glial cells in the vicinity of APs, and NFTs in the SN and striatum of PDD and non-PDD patients. We found that the majority of GMF-positive immunoreactive glial cells were co-localized with GFAP-reactive astrocytes. Our findings suggest that the GMF may be involved in the pathogenesis of PDD.
Collapse
|