1
|
Yu J, Li X, Sun Y, Wang L, Zhang Y. Transcriptomic analysis and experiment to verify the mechanism of Xiaoyao san in the treatment of irritable bowel syndrome with depression. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119732. [PMID: 40179998 DOI: 10.1016/j.jep.2025.119732] [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: 12/10/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoyao san (XYS) is a classic traditional Chinese medicine compound first recorded in "Taiping Huimin Heji Ju Fang". Traditionally, it is used to treat irritable bowel syndrome (IBS) and depression. However, its mechanism of action in treating IBS patients with depressive symptoms is still unclear. AIM OF THE STUDY This study aimed to investigate the effects of XYS on intestinal and depressive symptoms in IBS and explore the mechanisms through transcriptomic analysis and pharmacological experiments. MATERIALS AND METHODS IBS was induced in mice through a combination of chronic unpredictable mild stress and intragastric senna leaf stimulation. We evaluated six depressive parameters, examined colon tissue with hematoxylin and eosin staining and transmission electron microscopy, analyzed related proteins using western blotting, and performed transcriptomics on brain and intestinal tissues. The possible mechanism of action was speculated by network analysis of transcriptome results and further verified using the IBS model. RESULTS The results show that XYS restored mouse weight, reduced intestinal symptoms and sensitivity, suppressed villous loss and atrophy, and enhanced the integrity of the intestinal mucosal barrier. Notably, XYS decreased the depression-like behavior. Transcriptomic analysis combined with pharmacological experiments revealed that XYS inhibited the expression of proteins related to the intestinal ACT1/TRAF6/P38MAPK/AP-1 signaling pathway while activating the brain's DRD2/TH signaling pathway and increasing dopamine release in the brain. CONCLUSIONS XYS may regulate the brain-gut axis function and improve intestinal and depressive symptoms in IBS model mice through the intestinal ACT1/TRAF6/P38MAPK/AP-1 signaling pathway and the brain DRD2/TH signaling pathway.
Collapse
Affiliation(s)
- Jiaoyan Yu
- College of Life Science, Northwest University, Xi'an, 710069, China; Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| | - Xi Li
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| | - Yantao Sun
- College of Life Science, Northwest University, Xi'an, 710069, China.
| | - Ludan Wang
- College of Life Science, Northwest University, Xi'an, 710069, China.
| | - Yajun Zhang
- College of Life Science, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
2
|
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
|
3
|
Guo X, Wang J, Fan H, Tao W, Ren Z, Li X, Liu S, Zhou P, Chen Y. Computational drug repurposing in Parkinson's disease: Omaveloxolone and cyproheptadine as promising therapeutic candidates. Front Pharmacol 2025; 16:1539032. [PMID: 40264664 PMCID: PMC12011821 DOI: 10.3389/fphar.2025.1539032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Parkinson's disease (PD), a prevalent and progressive neurodegenerative disorder, currently lacks effective and satisfactory pharmacological treatments. Computational drug repurposing represents a promising and efficient strategy for drug discovery, aiming to identify new therapeutic indications for existing pharmaceuticals. Methods: We employed a drug-target network approach to computationally repurpose FDA-approved drugs from databases such as DrugBank. A literature review was conducted to select candidates not previously reported as pharmacoprotective against PD. Subsequent in vitro evaluation utilized Cell Counting Kit-8 (CCK8) assays to assess the neuroprotective effects of the selected compounds in the SH-SY5Y cell model of Parkinson's disease induced by 1-methyl-4-phenylpyridinium (MPP+). Furthermore, an in vivo mouse model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was developed to investigate the mechanisms of action and therapeutic potential of the identified drug candidates. Results: Our approach identified 176 drug candidates, with 28 selected for their potential anti-Parkinsonian effects and lack of prior PD-related reporting. CCK8 assays showed significant neuroprotection in SH-SY5Y cells for Omaveloxolone and Cyproheptadine. In the MPTP-induced mouse model, Cyproheptadine inhibited interleukin-6 (IL-6) expression and prevented Tyrosine Hydroxylase (TH) downregulation via the MAPK/NFκB pathway, while Omaveloxolone alleviated TH downregulation, potentially through the Kelch-like ECH-associated protein 1 (KEAP1)-NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. Both drugs preserved dopaminergic neurons and improved neurological deficits in the PD model. Conclusion: This study elucidates potential drug candidates for the treatment of Parkinson's disease through the application of computational repurposing, thereby underscoring its efficacy as a drug discovery strategy.
Collapse
Affiliation(s)
- Xin Guo
- Department of Geriatric Neurology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jie Wang
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Hongyang Fan
- Department of Geriatric Neurology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Wanying Tao
- Department of Critical Care Medicine, Department of Emergency Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Zijing Ren
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Xingyue Li
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Suyu Liu
- Medical College, Nanjing University, Nanjing, China
| | - Peiyang Zhou
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yingzhu Chen
- Department of Geriatric Neurology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
4
|
Tai MDS, Ochoa L, Flydal MI, Velasco-Carneros L, Muntaner J, Santiago C, Gamiz-Arco G, Moro F, Jung-Kc K, Gil-Cantero D, Marcilla M, Kallio JP, Muga A, Valpuesta JM, Cuéllar J, Martinez A. Structural recognition and stabilization of tyrosine hydroxylase by the J-domain protein DNAJC12. Nat Commun 2025; 16:2755. [PMID: 40113792 PMCID: PMC11926245 DOI: 10.1038/s41467-025-57733-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/28/2025] [Indexed: 03/22/2025] Open
Abstract
Pathogenic variants of the J-domain protein DNAJC12 cause parkinsonism, which is associated with a defective interaction of DNAJC12 with tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine biosynthesis. In this work, we characterize the formation of the TH:DNAJC12 complex, showing that DNAJC12 binding stabilizes both TH and the variant TH-p.R202H, associated with TH deficiency. This binding delays their time-dependent aggregation in an Hsp70-independent manner, while preserving TH activity and feedback regulatory inhibition by dopamine. DNAJC12 alone barely activates Hsc70 but synergistically stimulates Hsc70 ATPase activity when complexed with TH. Cryo-electron microscopy supported by crosslinking-mass spectroscopy reveals two DNAJC12 monomers bound per TH tetramer, each embracing one of the two regulatory domain dimers, leaving the active sites available for substrate, cofactor and inhibitory dopamine interaction. Our results also reveal the key role of the C-terminal region of DNAJC12 in TH binding, explaining the pathogenic mechanism of the DNAJC12 disease variant p.W175Ter.
Collapse
Affiliation(s)
- Mary Dayne S Tai
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Lissette Ochoa
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Marte I Flydal
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Lorea Velasco-Carneros
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco (UPV/EHU), Barrio Sarriena, Leioa, Spain
| | | | - César Santiago
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Gloria Gamiz-Arco
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Fernando Moro
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco (UPV/EHU), Barrio Sarriena, Leioa, Spain
| | - Kunwar Jung-Kc
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- K.G Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
| | | | | | - Juha P Kallio
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Arturo Muga
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco (UPV/EHU), Barrio Sarriena, Leioa, Spain
| | - José María Valpuesta
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
- Unidad de Nanobiotecnología, CNB-CSIC-IMDEA Nanociencia Associated Unit, Madrid, Spain.
| | - Jorge Cuéllar
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen, Norway.
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway.
- K.G Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway.
| |
Collapse
|
5
|
Wu F, He W, Song D, Wu Z, Dai P, Zheng X, Wang H, Xie C. Ropivacaine and celecoxib-loaded injectable composite hydrogel for improved chronic pain-exacerbated myocardial ischemia-reperfusion injury. J Control Release 2025; 379:266-284. [PMID: 39800238 DOI: 10.1016/j.jconrel.2025.01.028] [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: 10/01/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Chronic pain is a prevalent condition affecting a significant portion of the global population and is known to be associated with an increased risk of cardiovascular diseases. Despite the clinical relevance, the mechanisms underlying the link between chronic pain and myocardial ischemia-reperfusion (MI/R) injury remain poorly understood. This study aimed to investigate the role of the superior cervical ganglion (SCG) in mediating the effects of chronic pain on MI/R injury and to develop a novel therapeutic strategy. We identified that chronic pain upregulated TNF-α expression and induced hyperactivity in SCG sympathetic neurons, exacerbating MI/R injury. To address this, we engineered an injectable Pluronic/alginate-based composite hydrogel loaded with celecoxib and ropivacaine (celecoxib@Laponite-dopamine-alginate-Pluronic F-127@ropivacaine, CLDAFR). This hydrogel was designed to target the SCG, providing a localized and sustained release of the therapeutic agents, thereby mitigating neuronal inflammation and inhibiting neuronal hyperactivity. The CLDAFR hydrogel demonstrated excellent biocompatibility, heat-sensitive gelation properties, and controlled drug release in vitro. In vivo studies showed that applying CLDAFR effectively reduced MI/R injury in a chronic pain model by suppressing TNF-α expression and SCG neuronal activity. In conclusion, the CLDAFR hydrogel represents a promising therapeutic material for treating chronic pain-exacerbated MI/R injury by precisely targeting the SCG and providing a sustained anti-inflammatory and analgesic effect.
Collapse
Affiliation(s)
- Fancan Wu
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Wanyou He
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Da Song
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Zhen Wu
- Department of Critical Care Medicine, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong, PR China
| | - Peng Dai
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Xueqin Zheng
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China.
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China.
| | - Chao Xie
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
6
|
Guo M, Jiang J, Zhang A, Yu W, Huang X. Cholesterol promotes hair growth through activating sympathetic nerves and enhancing the proliferation of hair follicle stem cells. Mol Med 2025; 31:86. [PMID: 40045204 PMCID: PMC11883958 DOI: 10.1186/s10020-025-01139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
The regulatory mechanisms by which cholesterol influences hair regeneration remain incompletely understood. This study investigates the effects of cholesterol on hair follicle stem cells (HFSCs) proliferation and hair regeneration, with a focus on the underlying molecular mechanisms. Subcutaneous cholesterol injections in C57BL/6 mice significantly enhanced hair regeneration by promoting HFSCs proliferation. Hematoxylin and eosin (HE) staining revealed a greater number of hair follicles in the anagen phase in the cholesterol-treated group compared to controls. Immunofluorescence (IF) and BrdU labeling further confirmed that cholesterol significantly stimulated HFSCs proliferation. Mechanistically, cholesterol activated the PKA signaling pathway, leading to the phosphorylation of tyrosine hydroxylase (TH) at the serine 40 residue, which subsequently stimulated the sympathetic nervous system (SNS). SNS activation enhanced HFSCs proliferation and increased the proportion of hair follicles in the anagen phase. Furthermore, sympathetic nerve ablation significantly attenuated the hair regeneration-promoting effects of cholesterol, highlighting the critical regulatory role of SNS in this process. These findings provide key insights into the molecular mechanisms by which cholesterol regulates hair regeneration via the PKA-tyrosine hydroxylase-SNS pathway. Moreover, they suggest potential therapeutic applications targeting cholesterol-mediated signaling pathways to promote hair regeneration.
Collapse
Affiliation(s)
- Mengchen Guo
- Department of Dermatology, Tongji Hospital, School of Medicine, Hair Medical Center of Shanghai Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Junkun Jiang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Anke Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang Universtity, Hangzhou, China
| | - Wenjing Yu
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xin Huang
- Department of Dermatology, Tongji Hospital, School of Medicine, Hair Medical Center of Shanghai Tongji Hospital, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
7
|
Chaubey S, Singh L. Deciphering the mechanisms underlying the neuroprotective potential of kaempferol: a comprehensive investigation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2275-2292. [PMID: 39414700 DOI: 10.1007/s00210-024-03515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Neurodegenerative disorders are characterized by neuronal degradation, dysfunction, or death within the CNS. Oxidative and inflammatory stress play crucial roles in the pathogenesis of various neurodegenerative diseases. The interplay between these stressors and dysregulated cellular signaling pathways contributes to neurodegeneration. Downregulation of NRF-2 compromises antioxidant defense, exacerbating neuronal damage, while increased TLR-4/MAPK and TLR-4/NF-κB signaling promotes neuroinflammation. Excessive ROS production by NADPH oxidase leads to oxidative damage and neuronal apoptosis. The strategies targeting NRF-2, TLR-4-mediated inflammatory stress, and NADPH oxidase activity promise to mitigate neuronal damage and halt the progression of the disease. Kaempferol is a flavonoid polyphenol antioxidant found abundantly in various fruits and vegetables, including apples, grapes, tomatoes, and broccoli. It is widely found in medicinal plants including Equisetum spp., Sophora japonica, Ginkgo biloba, and Euphorbia pekinensis (Rupr.). A substantial body of in vitro and in vivo evidences have demonstrated the neuroprotective potential of kaempferol against neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Kaempferol demonstrates multifaceted potential in mitigating neuroinflammation, apoptosis, and oxidative stress in different neurodegenerative diseases through the modulation of various pathways including NRF-2, NADPH oxidase, TLR-4/MAPK, and TLR-4/NF-κB. This review article was developed through a comprehensive analysis and interpretation of research published between 2009 and 2024, sourced from multiple scientific databases, including PubMed, Scopus, ScienceDirect, and Web of Science. This review aims to provide an in-depth overview of the neuroprotective effects of kaempferol, focusing on its underlying molecular mechanisms. A total of 24 research evidence were included to elucidate the molecular pathways by which kaempferol exerts its protective effects against neurodegenerative diseases.
Collapse
Affiliation(s)
- Satyam Chaubey
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| |
Collapse
|
8
|
Gust KA, Amar SK, Gut CP, Styles RM, Karna RR, James RA, Holtzapple DM, Stricker JL, McInturf SM, Phillips EA, Honnold C, Luo X, Mumy KL, Mattie DR, Chappell MA, Mayo ML. Multi-disciplinary investigation identifies increased potency of ethyl-parathion inhaled within a soil-dust matrix to cause acetylcholinesterase-dependent molecular impacts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104615. [PMID: 39710123 DOI: 10.1016/j.etap.2024.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Neurotoxicity investigations of inhaled organophosphorus pesticide (OP), ethyl-parathion (EP), were conducted in Sprague Dawley rats comparing exposures to EP volatilized at 0, 1, 10, and 20 mg/m3 versus EP incorporated into soil dust (5 mg/m3) at 0, 0.0095, 0.09, and 0.185 mg/mg3. All exposures were sublethal, caused no respiratory effects, and no effects on balance and coordination behavior. Both volatilized and dust-incorporated EP exposures significantly decreased acetylcholinesterase (AChE) activity in plasma and hippocampus tissue. Correspondingly, plasma and hippocampal dopamine levels spiked in these exposures suggesting compensatory cholinergic / dopaminergic signal balancing. The EP exposures significantly increased expression of pro-inflammatory genes, including MAPK-14, IL6, IL1β, and TNF-α, while global RNA-seq results identified significant enrichment of inflammation, oxidative stress, and apoptosis pathways. Remarkably, dust-incorporated EP impacted similar molecular endpoints as volatilized EP but at concentrations two orders of magnitude lower highlighting potentially increased potency of EP incorporated into soil dust.
Collapse
Affiliation(s)
- Kurt A Gust
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States.
| | - Saroj K Amar
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States; Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Chet P Gut
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Renee M Styles
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Ranju R Karna
- Credere Associates LLC, Westbrook, ME located at US Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - R Arden James
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David M Holtzapple
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Joshua L Stricker
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Shawn M McInturf
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Elizabeth A Phillips
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Cary Honnold
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Xiao Luo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Karen L Mumy
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David R Mattie
- Air Force Research Laboratory/711 HPW, Wright-Patterson Air Force Base, OH, United States
| | - Mark A Chappell
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Michael L Mayo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| |
Collapse
|
9
|
Wu F, Fu C, Li Y, Wang H. TNF-α Enhanced Activity of Sympathetic Neurons in Superior Cervical Ganglion to Promote Chronic Sleep Deprivation-Related Hyperalgesia. Mol Neurobiol 2025:10.1007/s12035-025-04790-z. [PMID: 39992586 DOI: 10.1007/s12035-025-04790-z] [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: 04/23/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
The mechanisms underlying the association between sleep deprivation (SD) and hyperalgesia remain incompletely understood. In this study, the Modified Horizontal Platform Method was employed to induce chronic SD. Neuropathic pain was induced using chronic constriction injury of the sciatic nerve. Pain-like behaviors were assessed through measurements of mechanical allodynia and thermal hyperalgesia, while gait analysis was used to evaluate motor function. Immunofluorescence and western blot analyses were conducted to examine the expression of TNF-α, Iba-1, TH, neurons and c-Fos. Apoptosis was assessed using TUNEL staining. To explore anatomical connections, anterograde and retrograde tracer viruses were injected into the superior cervical ganglion (SCG) and the spinal cord, respectively. Local injection of 6-OHDA was used to ablate sympathetic neurons in the SCG, and R-7050 was administrated to block the TNF-α receptor. We found that chronic SD induced hyperalgesia in both normal and neuropathic pain model, accompanied by significant infiltration of microglia in the dorsal horn. TH expression and apoptotic cells were increased in the SCG following chronic SD. Viral tracer results demonstrated the existence of anatomical connections between the SCG and the spinal cord. Ablation of sympathetic innervation improved pain-like behaviors and reduced microglia, without affecting movement. Furthermore, chronic SD led to increased expression of TNF-α in sympathetic neurons, which was associated with heightened SCG activity. Blocking the TNF-α receptor ameliorated pain-like behaviors, decreased microglia, reduced apoptosis, lowered SCG activity. In conclusion, TNF-α enhanced the activity of sympathetic neurons in the SCG, promoting hyperalgesia related to chronic SD.
Collapse
Affiliation(s)
- Fancan Wu
- Department of Anesthesiology, The First People's Hospital of Foshan, Number 81, North of Ling Nan Road, Foshan, 528000, Guangdong, China
| | - Chao Fu
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China.
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Number 81, North of Ling Nan Road, Foshan, 528000, Guangdong, China.
| |
Collapse
|
10
|
Qiao CM, Tan LL, Ma XY, Xia YM, Li T, Li MA, Wu J, Nie X, Cui C, Zhao WJ, Shen YQ. Mechanism of S100A9-mediated astrocyte activation via TLR4/NF-κB in Parkinson's disease. Int Immunopharmacol 2025; 146:113938. [PMID: 39724736 DOI: 10.1016/j.intimp.2024.113938] [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: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Astrocyte-mediated neuroinflammation plays a key role in Parkinson's disease (PD) progression. The proinflammatory protein S100A9 is linked to various neurodegenerative diseases, but its involvement in astrocyte activation in PD remains unclear. Here, we investigate the role of S100A9 in astrocyte-mediated neuroinflammation in PD. C57BL/6J mice were intraperitoneally injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 15 mg/kg four times daily) and subsequently treated with Paquinimod, a S100A9 inhibitor (7 mg/kg, once daily for 7 days, totaling 8 doses). We observed an abnormal increase in S100A9 protein expression and a rise in S100A9-positive cells in the striatum of PD mice. Paquinimod treatment significantly improved behavioral deficits (pole test, rotarod test, traction test, and open field tests), prevented the reduction in striatal tyrosine hydroxylase (TH) protein and the loss of dopaminergic neurons (TH+) in the substantia nigra (SN) in PD mice. Interestingly, S100A9 was predominantly expressed in astrocytes (GFAP+S100A9+ cells) rather than in neurons or microglia, and its inhibition significantly reduced astrocyte activation (GFAP+ cells), reversed A1 astrocyte gene upregulation (H2-D1, C3, Serping1), and increased A2 astrocyte gene expression (Emp1, Ptx3, S100a10). Moreover, S100A9 inhibition also reduced the expression of inflammatory markers (IL-6, IL-1β, TNF-α) and suppressed the TLR4/NF-κB signaling pathway. In vitro, TLR4/NF-κB inhibitors mitigated inflammation and A1/A2 polarization of astrocytic MA cells induced by recombinant S100A9 (rS100A9). These findings suggest that S100A9 mediates astrocyte neuroinflammation and A1/A2 polarization via TLR4/NF-κB signaling, highlighting its potential as a therapeutic target for PD.
Collapse
Affiliation(s)
- Chen-Meng Qiao
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lu-Lu Tan
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Yu Ma
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yi-Meng Xia
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ting Li
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ming-An Li
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jian Wu
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xin Nie
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chun Cui
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei-Jiang Zhao
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yan-Qin Shen
- Laboratory of Neurodegenerative Diseases and Neuroinjury Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
11
|
Fevereiro-Martins M, Santos AC, Marques-Neves C, Guimarães H, Bicho M. Influence of Functional Variations in Genes of Neurotrophins and Neurotransmitter Systems on the Development of Retinopathy of Prematurity. Int J Mol Sci 2025; 26:898. [PMID: 39940677 PMCID: PMC11816744 DOI: 10.3390/ijms26030898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Retinal neurodevelopment, vascularization, homeostasis, and stress response are influenced by factors such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), tyrosine hydroxylase (TH), and erythropoietin (EPO). As retinopathy of prematurity (ROP) is a neurovascular retinal disease, this study analyzed the contributions of NGF (rs6330), BDNF (rs7934165), TH (rs10770141), and EPO (rs507392) genetic functional polymorphisms to the modulation of hematological and biochemical parameters of the first week of life and their association with ROP development. A multicenter cohort of 396 preterm infants (gestational age < 32 weeks or birth weight < 1500 g) was genotyped using MicroChip DNA and iPlex MassARRAY® platform. Multivariate regression followed univariate assessment of ROP risk factors. NGF (GG) genotype was associated with a higher ROP risk (OR = 1.79), which increased further (OR = 2.38) when epistatic interactions with TH (allele C) and BDNF (allele G) were present. Significant circulating biomarker differences, including bilirubin, erythrocytes, monocytes, neutrophils, lymphocytes, and platelet markers, were found between ROP and non-ROP groups, with variations depending on the polymorphism. These findings suggest that NGF (rs6330) and its interactions with related genes contribute to ROP risk, providing valuable insights into the genetic and biological mechanisms underlying the disease and identifying potential predictive biomarkers.
Collapse
Affiliation(s)
- Mariza Fevereiro-Martins
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal; (A.C.S.); (C.M.-N.); (M.B.)
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental-ISAMB, Laboratório Associado Terra, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento da Rocha Cabral 14, 1250-012 Lisboa, Portugal
- Departamento de Oftalmologia, Hospital Cuf Descobertas, Rua Mário Botas, 1998-018 Lisboa, Portugal
| | - Ana Carolina Santos
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal; (A.C.S.); (C.M.-N.); (M.B.)
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental-ISAMB, Laboratório Associado Terra, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carlos Marques-Neves
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal; (A.C.S.); (C.M.-N.); (M.B.)
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental-ISAMB, Laboratório Associado Terra, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Centro de Estudos das Ciências da Visão, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, Piso 1C, 1649-028 Lisboa, Portugal
| | - Hercília Guimarães
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
| | - Manuel Bicho
- Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal; (A.C.S.); (C.M.-N.); (M.B.)
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental-ISAMB, Laboratório Associado Terra, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Instituto de Investigação Científica Bento da Rocha Cabral, Calçada Bento da Rocha Cabral 14, 1250-012 Lisboa, Portugal
| | | |
Collapse
|
12
|
Jiao P, An Y, Wu S, Li H, Li G. Cinnamaldehyde Attenuates the Expression of IBA1 and GFAP to Inhibit Glial Cell Activation and Inflammation in the MPTP-Induced Acute Parkinson's Disease Model. PARKINSON'S DISEASE 2024; 2024:9973140. [PMID: 39741957 PMCID: PMC11688134 DOI: 10.1155/padi/9973140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/31/2024] [Accepted: 11/30/2024] [Indexed: 01/03/2025]
Abstract
Cinnamaldehyde (CA), the primary bioactive compound in cinnamon (Cinnamomum cassia Presl, Lauraceae, Cinnamomum), holds potential therapeutic benefits for Parkinson's disease (PD). To scrutinize the impact and mechanisms of CA on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD, male C57BL/6 mice were randomly allocated to CA (150, 300, and 600 mg/kg), model, Madopar, and control group (n = 12). The Open Field, Pole-jump, and Rotarod experiments assessed exercise capacity and anxiety levels. HPLC evaluated the levels of neurotransmitters. Immunohistochemistry was utilized to detect the expression of TH and GFAP. WB and RT-qPCR determine the expression levels of apoptosis-related genes and proteins in the substantia nigra and striatum. The findings revealed that CA not only enhanced motor abilities and reduced anxiety but also elevated the levels of TH, DOPAC, DA, 5-HIAA, HVA, and 5-HT in the substantia nigra and striatum. Moreover, it protected DA neurons and downregulated the expression of Bax, Casp3, and Bax/Bcl-2 mRNA and proteins, while increasing the expression of Bcl-2 mRNA compared to the model group. Furthermore, CA was observed to inhibit glial cell activation, leading to reduced levels of GFAP and IBA1 in the substantia nigra and striatum. This resulted in decreased expression of inflammatory factors such as iNOS and NF-κBp65 proteins in these regions, consequently mitigating neuroinflammation. These results suggest that CA exerts a neuroprotective effect in acute PD model mice by suppressing glial cell activation, modulating the expression of apoptotic genes, and alleviating neuroinflammation and apoptosis induced by MPTP.
Collapse
Affiliation(s)
- Panpan Jiao
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yingfeng An
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Suhui Wu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hanbing Li
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Genlin Li
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
13
|
Xu Z, Yang L, Chen H, Liu S, Li X, Li S, Ying C, Li X, Du R, Liu D. Saccharomyces cerevisiae and Kluyveromyces marxianus yeast co-cultures modulate the ruminal microbiome and metabolite availability to enhance rumen barrier function and growth performance in weaned lambs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:139-152. [PMID: 39635413 PMCID: PMC11615919 DOI: 10.1016/j.aninu.2024.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/27/2024] [Accepted: 06/03/2024] [Indexed: 12/07/2024]
Abstract
In lambs, weaning imposes stress that can contribute to impaired rumen epithelial barrier functionality and immunological dysregulation. In this study, the effects of a yeast co-culture consisting of Saccharomyces cerevisiae and Kluyveromyces marxianus (NM) on rumen health in lambs was evaluated, with a focus on parameters including growth performance, ruminal fermentation, and epithelial barrier integrity, ruminal metabolic function, and the composition of the ruminal bacteria. In total, 24 lambs were grouped into four groups of six lambs including a control (C) group fed a basal diet, and N, M, and NM groups in which lambs were fed the basal diet respectively supplemented with S. cerevisiae yeast cultures (30 g/d per head), K. marxianus yeast cultures (30 g/d per head), and co-cultures of both yeasts (30 g/d per head), the experiment lasted for 42 d. Subsequent analyses revealed that relative to the C group, the average daily gain (ADG) of lambs in the NM group was significantly greater and exhibited significant increases in a range of mRNA relative expression including monocarboxylate transporter 1 (MCT1), (Na+)/hydrogen (H+) exchanger 1 (NHE1), (Na+)/hydrogen (H+) exchanger 3 (NHE3), proton-coupled amino acid transporter 1 (PAT1), vacuolar H+-ATPase (vH+ ATPase), claudin-1, occludin in the rumen epithelium (P < 0.05). Compared with the C group, the pH of the rumen contents in the NM group was significantly decreased , and the concentrations of acetate, propionate, and butyrate were significantly increased (P < 0.05). Analysis of the rumen bacteria showed that the NM group exhibited increases in the relative abundance of Prevotella, Treponema, Moryella, Fibrobacter, CF231 and Ruminococcus (P < 0.05). Metabolomics analyses revealed an increase in the relative content of phthalic acid and cinnamaldehyde in the NM group as compared to the C group (P < 0.05), together with the greater relative content of L-tyrosine, L-dopa, rosmarinic acid, and tyrosol generated by the tyrosine metabolic pathway (P < 0.05). Spearman's correlation analyses revealed relative abundance levels of Fibrobacter and Ruminococcus were positively correlated with the mRNA relative expression levels of PAT1, NHE3, and zonula occluden-1 (ZO-1), as well as with tyrosol, phthalic acid, and cinnamaldehyde levels (P < 0.05). Ultimately, these results suggest that dietary supplementation with NM has a wide range of beneficial effects on weaned lambs and is superior to single bacterial fermentation. These effects include improvements in daily gain and rumen epithelial barrier integrity, as well as improvements in the composition of the rumen microbiome, and alterations in tyrosine metabolic pathways.
Collapse
Affiliation(s)
- Zixuan Xu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lan Yang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hui Chen
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shixiong Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xueqiang Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Songjian Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Chun Ying
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xiao Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Rui Du
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Dacheng Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
14
|
Tu H, Yeo XY, Zhang ZW, Zhou W, Tan JY, Chi L, Chia SY, Li Z, Sim AY, Singh BK, Ma D, Zhou Z, Bonne I, Ling SC, Ng ASL, Jung S, Tan EK, Zeng L. NOTCH2NLC GGC intermediate repeat with serine induces hypermyelination and early Parkinson's disease-like phenotypes in mice. Mol Neurodegener 2024; 19:91. [PMID: 39609868 PMCID: PMC11603791 DOI: 10.1186/s13024-024-00780-2] [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: 05/07/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The expansion of GGC repeats (typically exceeding 60 repeats) in the 5' untranslated region (UTR) of the NOTCH2NLC gene (N2C) is linked to N2C-related repeat expansion disorders (NREDs), such as neuronal intranuclear inclusion disease (NIID), frontotemporal dementia (FTD), essential tremor (ET), and Parkinson's disease (PD). These disorders share common clinical manifestations, including parkinsonism, dementia, seizures, and muscle weakness. Intermediate repeat sizes ranging from 40 to 60 GGC repeats, particularly those with AGC-encoded serine insertions, have been reported to be associated with PD; however, the functional implications of these intermediate repeats with serine insertion remain unexplored. METHODS Here, we utilized cellular models harbouring different sizes of N2C variant 2 (N2C2) GGC repeat expansion and CRISPR-Cas9 engineered transgenic mouse models carrying N2C2 GGC intermediate repeats with and without serine insertion to elucidate the underlying pathophysiology associated with N2C intermediate repeat with serine insertion in NREDs. RESULTS Our findings revealed that the N2C2 GGC intermediate repeat with serine insertion (32G13S) led to mitochondrial dysfunction and cell death in vitro. The neurotoxicity was influenced by the length of the repeat and was exacerbated by the presence of the serine insertion. In 12-month-old transgenic mice, 32G13S intensified intranuclear aggregation and exhibited early PD-like characteristics, including the formation of α-synuclein fibers in the midbrain and the loss of tyrosine hydroxylase (TH)-positive neurons in both the cortex and striatum. Additionally, 32G13S induced neuronal hyperexcitability and caused locomotor behavioural impairments. Transcriptomic analysis of the mouse cortex indicated dysregulation in calcium signaling and MAPK signaling pathways, both of which are critical for mitochondrial function. Notably, genes associated with myelin sheath components, including MBP and MOG, were dysregulated in the 32G13S mouse. Further investigations using immunostaining and transmission electron microscopy revealed that the N2C intermediate repeat with serine induced mitochondrial dysfunction-related hypermyelination in the cortex. CONCLUSIONS Our in vitro and in vivo investigations provide the first evidence that the N2C-GGC intermediate repeat with serine promotes intranuclear aggregation of N2C, leading to mitochondrial dysfunction-associated hypermyelination and neuronal hyperexcitability. These changes contribute to motor deficits in early PD-like neurodegeneration in NREDs.
Collapse
Affiliation(s)
- Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Zhi-Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Wei Zhou
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore, 169856, Singapore
| | - Jayne Yi Tan
- Department of Neurology, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Li Chi
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Sook-Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Zhihong Li
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Aik Yong Sim
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117549, Singapore
| | - Brijesh Kumar Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Dongrui Ma
- Department of Neurology, Singapore General Hospital, Singapore, 169609, Singapore
| | - Zhidong Zhou
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore, 169856, Singapore
- Neuroscience & Behavioural Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Isabelle Bonne
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117549, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Immunology Translational Research Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Shuo-Chien Ling
- Neuroscience & Behavioural Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Singapore, 308433, Singapore
- Neuroscience & Behavioural Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam, 13488, Republic of Korea
| | - Eng-King Tan
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore, 169856, Singapore.
- Department of Neurology, National Neuroscience Institute, Singapore, 308433, Singapore.
- Neuroscience & Behavioural Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore.
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore.
- Neuroscience & Behavioural Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore.
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore, Novena Campus, 308232, Singapore.
| |
Collapse
|
15
|
Zhang C, Zhou J, Zhuo L, Zhang W, Lv L, Zhu L, Zhang J, Feng F, Liu W, Han L, Liao W. The TLR4/NF-κB/NLRP3 and Nrf2/HO-1 pathways mediate the neuroprotective effects of alkaloids extracted from Uncaria rhynchophylla in Parkinson's disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118391. [PMID: 38797377 DOI: 10.1016/j.jep.2024.118391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is the second most common neurodegenerative disorder with limited therapeutic options available. Neuroinflammation plays an important role in the occurrence and development of PD. Alkaloids extracted from Uncaria rhynchophylla (URA), have emerged as a potential neuroprotective agent because of its anti-inflammatory and anti-oxidant properties. Nevertheless, the underlying mechanism by which URA exerts neuroprotective effects in PD remains obscure. AIM OF THE STUDY The main aim of this study was to investigate the neuroprotective effects and underlying mechanism of URA in the treatment of PD through in vivo and in vitro models, focusing on the neuroinflammation and oxidative stress pathways. MATERIALS AND METHODS The protective effects of URA against PD were evaluated by neurobehavioral tests, immunohistochemistry, serum biochemical assays, and real-time quantitative polymerase chain reaction in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice. The role of the TLR4/NF-κB/NLRP3 pathway and the Nrf2/HO-1 pathway in URA-mediated effects was examined in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells and a microglia-neuron coculture system. RESULTS URA significantly alleviated motor deficits and dopaminergic neurotoxicity, and reversed the abnormal secretion of inflammatory and oxidative stress factors in the serum of MPTP-induced mice. URA suppressed the gene expression of Toll-like receptor 4 (TLR4), NOD-like receptor protein 3, and cyclooxygenase 2 (COX2) in the striatum of PD mice. Further studies indicated that URA inhibited activation of the TLR4/NF-κB/NLRP3 pathway and enhanced activation of the Nrf2/HO-1 pathway, reduced reactive oxygen species (ROS) production, and reversed the secretion of inflammatory mediators in LPS-stimulated BV-2 microglial cells, thereby alleviating neuroinflammatory damage to SH-SY5Y neuronal cells. CONCLUSION URA exerted neuroprotective effects against PD mainly by the inhibition of the TLR4/NF-κB/NLRP3 pathway and activation of the Nrf2/HO-1 antioxidant pathway, highlighting URA as a promising candidate for PD treatment.
Collapse
Affiliation(s)
- Chunxia Zhang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Jiayu Zhou
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Lingxin Zhuo
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Wenxin Zhang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Lingrui Lv
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Lingmeng Zhu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Jiayi Zhang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Feng Feng
- Nanjing Medical University, Nanjing, 211166, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Zhejiang Center for safety study of drug substances (Industrial Technology Innovation Platform), Hangzhou, 310018, China
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.
| | - Wenting Liao
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.
| |
Collapse
|
16
|
Douma EH, Stoop J, Lingl MVR, Smidt MP, van der Heide LP. Phosphodiesterase inhibition and Gucy2C activation enhance tyrosine hydroxylase Ser40 phosphorylation and improve 6-hydroxydopamine-induced motor deficits. Cell Biosci 2024; 14:132. [PMID: 39456033 PMCID: PMC11515495 DOI: 10.1186/s13578-024-01312-7] [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: 02/20/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Parkinson's disease is characterized by a progressive loss of dopaminergic neurons in the nigrostriatal pathway, leading to dopamine deficiency and motor impairments. Current treatments, such as L-DOPA, provide symptomatic relief but result in off-target effects and diminished efficacy over time. This study explores an alternative approach by investigating the activation of tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Specifically, we explore the effects of phosphodiesterase (PDE) inhibition and guanylate cyclase-C (GUCY2C) activation on tyrosine hydroxylase Ser40 phosphorylation and their impact on motor behavior in a 6-hydroxydopamine (6-OHDA) Parkinson's disease model. RESULTS Our findings demonstrate that increasing cyclic nucleotide levels through PDE inhibition and GUCY2C activation significantly enhances tyrosine hydroxylase Ser40 phosphorylation. In a Pitx3-deficient mouse model, which mimics the loss of dopaminergic neurons seen in Parkinson's disease, Ser40 phosphorylation remained manipulable despite reduced tyrosine hydroxylase protein levels. Moreover, we observed no evidence of tyrosine hydroxylase degradation due to Ser40 phosphorylation, challenging previous reports. Furthermore, both PDE inhibition and GUCY2C activation resulted in improved motor behavior in the 6-OHDA Parkinson's disease mouse model, highlighting the potential therapeutic benefits of these approaches. CONCLUSIONS This study underscores the therapeutic potential of enhancing tyrosine hydroxylase Ser40 phosphorylation to improve motor function in Parkinson's disease. Both PDE inhibition and GUCY2C activation represent promising non-invasive strategies to modulate endogenous dopamine biosynthesis and address motor deficits. These findings suggest that targeting cyclic nucleotide pathways could lead to novel therapeutic approaches, either as standalone treatments or in combination with existing therapies like L-DOPA, aiming to provide more durable symptom relief and potentially mitigate neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Erik H Douma
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
- Parkinnova Therapeutics B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Jesse Stoop
- Macrobian-Biotech B.V., Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Matthijs V R Lingl
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lars P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, Room C3.104, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Niu Y, Pan Y, Wang Y, Fu Y, Zhao Z, Kang L. Lead specifically declines tyrosine hydroxylase activity to induce the onset of Parkinson's disease through disrupting dopamine biosynthesis in fly models. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 356:124383. [PMID: 38897282 DOI: 10.1016/j.envpol.2024.124383] [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: 04/17/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Parkinson's disease (PD) is one of the fastest-growing neurodegenerative diseases and has been linked to the exposure to numerous environmental neurotoxins. Although lead (Pb) exposure has been related to the development of PD, the molecular target of Pb to cause the onset of PD is insufficiently investigated. Herein, we explored the effects of Pb exposure on behavior, pathophysiology, and gene expression of wild-type (WT) fly (Drosophila melanogaster) by comparison with its PD model. After exposure to Pb, the WT flies showed PD-like locomotor impairments and selective loss of dopaminergic (DAergic) neurons, displaying similar phenotypes to fly PD model (PINK1). Transcriptomic analysis showed the similarity in gene expression profiles between Pb treatment WT flies and PINK1 mutant flies. Moreover, Pb exposure resulted in endogenous dopamine deficits in WT flies. Analyses of gene expression and enzyme activity confirmed that Pb exposure reduced tyrosine hydroxylase (TH) activity and led to failure of dopamine synthesis. Furthermore, molecular dynamics simulation confirmed that Pb was adsorbed by TH and subsequently inhibited the enzymatic activity. Exogenous injection of L-dopa and melatonin could partially rescue the pathological phenotypes of Pb-exposed flies and PD fly model. Antagonist injection of microRNA-133, which negatively regulated the expression of TH gene, ultimately rescued in the manifestation of PD phenotypes in flies. Involvement of TH overexpression mutants of fly strongly promoted the resistance to Pb exposure and rescued both behavior and the number of DAergic neurons. Therefore, our study elucidates the Pb molecular target in dopamine pathway and mechanism underlying the risks of Pb exposure on the occurrence of PD at environmentally-relevant concentrations.
Collapse
Affiliation(s)
- Yue Niu
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yifan Pan
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yaqi Wang
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yongqi Fu
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China
| | - Zhangwu Zhao
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China
| | - Le Kang
- Institute of Life Science and Green Development/College of Life Sciences, Hebei University, Baoding 071002, China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
18
|
Douma EH, Smidt MP, van der Heide LP. Boosting endogenous dopamine production: a novel therapeutic approach for Parkinson's disease. Trends Mol Med 2024; 30:800-803. [PMID: 38926032 DOI: 10.1016/j.molmed.2024.06.002] [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: 03/31/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
Innovative therapeutic strategies are urgently needed for Parkinson's disease due to limited efficacy of current treatments and a weak therapeutic pipeline. In this forum article, we propose targeting tyrosine hydroxylase phosphorylation as a novel mechanism of action to address this critical need.
Collapse
Affiliation(s)
- Erik H Douma
- Parkinnova Therapeutics, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lars P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Roshdy M, Zaky DA, Abbas SS, Abdallah DM. Niacin, an innovative protein kinase-C-dependent endoplasmic reticulum stress reticence in murine Parkinson's disease. Life Sci 2024; 351:122865. [PMID: 38914304 DOI: 10.1016/j.lfs.2024.122865] [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: 02/18/2024] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
AIMS Niacin (NIA) supplementation showed effectiveness against Parkinson's disease (PD) in clinical trials. The depletion of NAD and endoplasmic reticulum stress response (ERSR) are implicated in the pathogenesis of PD, but the potential role for NAD precursors on ERSR is not yet established. This study was undertaken to decipher NIA molecular mechanisms against PD-accompanied ERSR, especially in relation to PKC. METHODS Alternate-day-low-dose-21 day-subcutaneous exposure to rotenone (ROT) in rats induced PD. Following the 5th ROT injection, rats received daily doses of either NIA alone or preceded by the PKC inhibitor tamoxifen (TAM). Extent of disease progression was assessed by behavioral, striatal biochemical and striatal/nigral histopathological/immunohistochemical analysis. KEY FINDINGS Via activating PKC/LKB1/AMPK stream, NIA post-treatment attenuated the ERSR reflected by the decline in ATF4, ATF6 and XBP1s to downregulate the apoptotic markers, CHOP/GADD153, p-JNK and active caspase-3. Such amendments congregated in motor activity/coordination improvements in open field and rotarod tasks, enhanced grid test latency and reduced overall PD scores, while boosting nigral/striatal tyrosine hydroxylase immunoreactivity and increasing intact neurons (Nissl stain) in both SNpc and striatum that showed less neurodegeneration (H&E stain). To different extents, TAM reverted all the NIA-related actions to prove PKC as a fulcrum in conveying the drug neurotherapeutic potential. SIGNIFICANCE PKC activation is a pioneer mechanism in the drug ERSR inhibitory anti-apoptotic modality to clarify NIA promising clinical and potent preclinical anti-PD efficacy. This kinase can be tagged as a druggable target for future add-on treatments that can assist dopaminergic neuronal aptitude against this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Merna Roshdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Samah S Abbas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
20
|
Singh A, Ansari VA, Mahmood T, Hasan SM, Wasim R, Maheshwari S, Akhtar J, Sheikh S, Vishwakarma VK. Targeting Abnormal Tau Phosphorylation for Alzheimer's Therapeutics. Horm Metab Res 2024; 56:482-488. [PMID: 38350636 DOI: 10.1055/a-2238-1384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative disorder characterized by progressive memory and cognitive decline, posing a formidable public health challenge. This review explores the intricate interplay between two pivotal players in AD pathogenesis: β-amyloid (Aβ) and tau protein. While the amyloid cascade theory has long dominated AD research, recent developments have ignited debates about its centrality. Aβ plaques and tau NFTs are hallmark pathologies in AD. Aducanumab and lecanemab, monoclonal antibodies targeting Aβ, have been approved, albeit amidst controversy, raising questions about the therapeutic efficacy of Aβ-focused interventions. On the other hand, tau, specifically its hyperphosphorylation, disrupts microtubule stability and contributes to neuronal dysfunction. Various post-translational modifications of tau drive its aggregation into NFTs. Emerging treatments targeting tau, such as GSK-3β and CDK5 inhibitors, have shown promise in preclinical and clinical studies. Restoring the equilibrium between protein kinases and phosphatases, notably protein phosphatase-2A (PP2A), is a promising avenue for AD therapy, as tau is primarily regulated by its phosphorylation state. Activation of tau-specific phosphatases offers potential for mitigating tau pathology. The evolving landscape of AD drug development emphasizes tau-centric therapies and reevaluation of the amyloid cascade hypothesis. Additionally, exploring the role of neuroinflammation and its interaction with tau pathology present promising research directions.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | | | | | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Suvaiv Sheikh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | |
Collapse
|
21
|
Tai MDS, Gamiz-Arco G, Martinez A. Dopamine synthesis and transport: current and novel therapeutics for parkinsonisms. Biochem Soc Trans 2024; 52:1275-1291. [PMID: 38813865 PMCID: PMC11346439 DOI: 10.1042/bst20231061] [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: 01/21/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
Parkinsonism is the primary type of movement disorder in adults, encompassing a set of clinical symptoms, including rigidity, tremors, dystonia, bradykinesia, and postural instability. These symptoms are primarily caused by a deficiency in dopamine (DA), an essential neurotransmitter in the brain. Currently, the DA precursor levodopa (synthetic L-DOPA) is the standard medication to treat DA deficiency, but it only addresses symptoms rather than provides a cure. In this review, we provide an overview of disorders associated with DA dysregulation and deficiency, particularly Parkinson's disease and rare inherited disorders leading predominantly to dystonia and/or parkinsonism, even in childhood. Although levodopa is relatively effective for the management of motor dysfunctions, it is less effective for severe forms of parkinsonism and is also associated with side effects and a loss of efficacy over time. We present ongoing efforts to reinforce the effect of levodopa and to develop innovative therapies that target the underlying pathogenic mechanisms affecting DA synthesis and transport, increasing neurotransmission through disease-modifying approaches, such as cell-based therapies, nucleic acid- and protein-based biologics, and small molecules.
Collapse
Affiliation(s)
| | - Gloria Gamiz-Arco
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, 5020 Bergen, Norway
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
22
|
Fitzpatrick PF, Daubner SC. Biochemical and biophysical approaches to characterization of the aromatic amino acid hydroxylases. Methods Enzymol 2024; 704:345-361. [PMID: 39300655 DOI: 10.1016/bs.mie.2024.05.009] [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: 09/22/2024]
Abstract
The aromatic amino acid hydroxylases phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase utilize a non-heme iron to catalyze the hydroxylation of the aromatic rings of their amino acid substrates, with a tetrahydropterin serving as the source of the electrons necessary for the monooxygenation reaction. These enzymes have been subjected to a variety of biochemical and biophysical approaches, resulting in a detailed understanding of their structures and mechanism. We summarize here the experimental approaches that have led to this understanding.
Collapse
Affiliation(s)
- Paul F Fitzpatrick
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, TX, United States.
| | - S Colette Daubner
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
23
|
Verma AK, Lowery S, Lin LC, Duraisami E, Lloréns JEA, Qiu Q, Hefti M, Yu CR, Albers MW, Perlman S. Persistent Neurological Deficits in Mouse PASC Reveal Antiviral Drug Limitations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.596989. [PMID: 38895239 PMCID: PMC11185538 DOI: 10.1101/2024.06.02.596989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Post-Acute Sequelae of COVID-19 (PASC) encompasses persistent neurological symptoms, including olfactory and autonomic dysfunction. Here, we report chronic neurological dysfunction in mice infected with a virulent mouse-adapted SARS-CoV-2 that does not infect the brain. Long after recovery from nasal infection, we observed loss of tyrosine hydroxylase (TH) expression in olfactory bulb glomeruli and neurotransmitter levels in the substantia nigra (SN) persisted. Vulnerability of dopaminergic neurons in these brain areas was accompanied by increased levels of proinflammatory cytokines and neurobehavioral changes. RNAseq analysis unveiled persistent microglia activation, as found in human neurodegenerative diseases. Early treatment with antivirals (nirmatrelvir and molnupiravir) reduced virus titers and lung inflammation but failed to prevent neurological abnormalities, as observed in patients. Together these results show that chronic deficiencies in neuronal function in SARS-CoV-2-infected mice are not directly linked to ongoing olfactory epithelium dysfunction. Rather, they bear similarity with neurodegenerative disease, the vulnerability of which is exacerbated by chronic inflammation.
Collapse
Affiliation(s)
- Abhishek Kumar Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Shea Lowery
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Li-Chin Lin
- Iowa Neuroscience Institute, University of Iowa, IA, USA 52242
- Department of Neurology, University of Iowa, Iowa City, IA 52242
| | | | | | - Qiang Qiu
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Marco Hefti
- Department of Pathology, University of Iowa, Iowa City, IA 52242
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Mark W. Albers
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
24
|
Yu Y, Chen W, Li B, Li Z, Wang Y, Mao Y, Fan W, Bai Y, Hu H, Zhen Q, Sun L. Cutaneous Calcium/Calmodulin-Dependent Protein Kinase II-γ-Positive Sympathetic Nerves Secreting Norepinephrine Dictate Psoriasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306772. [PMID: 38544478 PMCID: PMC11187923 DOI: 10.1002/advs.202306772] [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: 09/17/2023] [Revised: 03/06/2024] [Indexed: 06/20/2024]
Abstract
Cutaneous sympathetic nerve is a crucial part of neuropsychiatric factors contributing to skin immune response, but its role in the psoriasis pathogenesis remains unclear. It is found that cutaneous calcium/calmodulin-dependent protein kinase II-γ (CAMK2γ), expressed mainly in sympathetic nerves, is activated by stress and imiquimod in mouse skin. Camk2g-deficient mice exhibits attenuated imiquimod-induced psoriasis-like manifestations and skin inflammation. CaMK2γ regulates dermal γδT-cell interleukin-17 production in imiquimod-treated mice, dependent on norepinephrine production following cutaneous sympathetic nerve activation. Adrenoceptor β1, the primary skin norepinephrine receptor, colocalises with γδT cells. CaMK2γ aggravates psoriasiform inflammation via sympathetic nerve-norepinephrine-γδT cell-adrenoceptor β1-nuclear factor-κB and -p38 axis activation. Application of alcaftadine, a small-molecule CaMK2γ inhibitor, relieves imiquimod-induced psoriasis-like manifestations in mice. This study reveals the mechanisms of sympathetic-nervous-system regulation of γδT-cell interleukin-17 secretion, and provides insight into neuropsychiatric factors dictating psoriasis pathogenesis and new potential targets for clinical psoriasis treatment.
Collapse
Affiliation(s)
- Yafen Yu
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
- The Center for Scientific Researchthe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Weiwei Chen
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Bao Li
- The Comprehensive LabCollege of BasicAnhui Medical UniversityHefei230032China
| | - Zhuo Li
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Yirui Wang
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Yiwen Mao
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Wencheng Fan
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Yuanming Bai
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| | - Hongbo Hu
- Center for Immunology and HematologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610065China
| | - Qi Zhen
- North China University of Science and Technology Affiliated HospitalTangshan063210China
- Health Science CenterNorth China University of Science and TechnologyTangshan063210China
- School of Public HealthNorth China University of Science and TechnologyTangshan063210China
| | - Liangdan Sun
- Department of Dermatologythe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- North China University of Science and Technology Affiliated HospitalTangshan063210China
- Health Science CenterNorth China University of Science and TechnologyTangshan063210China
- School of Public HealthNorth China University of Science and TechnologyTangshan063210China
- Inflammation and Immune Diseases Laboratory of North China University of Science and TechnologyTangshan063210China
- Key Laboratory of DermatologyAnhui Medical UniversityMinistry of EducationHefei230032China
| |
Collapse
|
25
|
Mahmoud AH, Elhefnawei DM, EL-Desouky MA, Kadry MO. Reciprocal crosslink among MeCP2/BDNF /CREB signaling pinpointed in autism spectrum disorder. Toxicol Rep 2024; 12:91-99. [PMID: 38229920 PMCID: PMC10789594 DOI: 10.1016/j.toxrep.2023.12.008] [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: 09/02/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024] Open
Abstract
Autism spectrum disorder, or individual disability (ID), is a condition characterized by complications in social interaction, restricted repetitive behavior, and difficulties in social communication. Neuquinon (NQ) possess a powerful therapeutic potential in various neurodegenerative disease. Nevertheless, contributing to NQ's low water solubility and bioavailability, its medicinal use has been constrained. Liposomes were supposed to be prospective drug-delivering agents for NQ, crossing the blood-brain barrier (BBB), and reaching the target organs. The current investigation aims to track the signaling pathways that govern NQ and liposomal neuquinon (LNQ) action in autistic models generated by ethyl formic acid. The neurotransmitters gamma amino-butyric acid (GABA), acetylcholine (ACh), and acetylcholinesterase (AChE) in addition to, the gene expressions of brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), and methyl-CpG-binding protein 2 (MeCP2) and the DNA damage COMET analysis at different time intervals of the study, were assessed. EFA in a dose of 500 mg/kg BW was used to induce autism in rats, and then NQ and LNQ were administered in 10 mg/kg and 2 mg/kg BW, respectively. The results revealed that NQ and LNQ significantly down-regulated BDNF, GABA, and AChE; on the other hand, they up-regulated MeCP2, CREB gene expressions, and ACh action. NQ and LNQ displayed improvement in DNA damage in almost all brain regions after EFA alterations; even better results were noticed post-LNQ therapy. Therefore, it may be concluded that neuquinon and liposomal-loaded neuquinon have a therapeutic index versus EFA-induced autism in a rat model.
Collapse
Affiliation(s)
- Ahlam H. Mahmoud
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Doaa M. Elhefnawei
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | | | - Mai O. Kadry
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| |
Collapse
|
26
|
Ji S, Guo Y, Yan W, Wei F, Ding J, Hong W, Wu X, Ku T, Yue H, Sang N. PM 2.5 exposure contributes to anxiety and depression-like behaviors via phenyl-containing compounds interfering with dopamine receptor. Proc Natl Acad Sci U S A 2024; 121:e2319595121. [PMID: 38739786 PMCID: PMC11127009 DOI: 10.1073/pnas.2319595121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Abstract
As a global problem, fine particulate matter (PM2.5) really needs local fixes. Considering the increasing epidemiological relevance to anxiety and depression but inconsistent toxicological results, the most important question is to clarify whether and how PM2.5 causally contributes to these mental disorders and which components are the most dangerous for crucial mitigation in a particular place. In the present study, we chronically subjected male mice to a real-world PM2.5 exposure system throughout the winter heating period in a coal combustion area and revealed that PM2.5 caused anxiety and depression-like behaviors in adults such as restricted activity, diminished exploratory interest, enhanced repetitive stereotypy, and elevated acquired immobility, through behavioral tests including open field, elevated plus maze, marble-burying, and forced swimming tests. Importantly, we found that dopamine signaling was perturbed using mRNA transcriptional profile and bioinformatics analysis, with Drd1 as a potential target. Subsequently, we developed the Drd1 expression-directed multifraction isolating and nontarget identifying framework and identified a total of 209 compounds in PM2.5 organic extracts capable of reducing Drd1 expression. Furthermore, by applying hierarchical characteristic fragment analysis and molecular docking and dynamics simulation, we clarified that phenyl-containing compounds competitively bound to DRD1 and interfered with dopamine signaling, thereby contributing to mental disorders. Taken together, this work provides experimental evidence for researchers and clinicians to identify hazardous factors in PM2.5 and prevent adverse health outcomes and for local governments and municipalities to control source emissions for diminishing specific disease burdens.
Collapse
Affiliation(s)
- Shaoyang Ji
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Yuqiong Guo
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Wei Yan
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu221004, People’s Republic of China
| | - Fang Wei
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Jinjian Ding
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Wenjun Hong
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Xiaoyun Wu
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Tingting Ku
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Huifeng Yue
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Nan Sang
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| |
Collapse
|
27
|
Yao JY, Li L, Xu JX, Liu YH, Shi J, Yu XQ, Kong QQ, Li K. Real-Time Monitoring of Tyrosine Hydroxylase Activity with a Ratiometric Fluorescent Probe. Anal Chem 2024; 96:7082-7090. [PMID: 38652135 DOI: 10.1021/acs.analchem.4c00382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Parkinson's disease (PD) represents the second most widespread neurodegenerative disease, and early monitoring and diagnosis are urgent at present. Tyrosine hydroxylase (TH) is a key enzyme for producing dopamine, the levels of which can serve as an indicator for assessing the severity and progression of PD. This renders the specific detection and visualization of TH a strategically vital way to meet the above demands. However, a fluorescent probe for TH monitoring is still missing. Herein, three rationally designed wash-free ratiometric fluorescent probes were proposed. Among them, TH-1 exhibited ideal photophysical properties and specific dual-channel bioimaging of TH activity in SH-SY5Y nerve cells. Moreover, the probe allowed for in vivo imaging of TH activity in zebrafish brain and living striatal slices of mice. Overall, the ratiometric fluorescent probe TH-1 could serve as a potential tool for real-time monitoring of PD in complex biosystems.
Collapse
Affiliation(s)
- Jia-Yi Yao
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Lu Li
- Orthopedic Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ji-Xuan Xu
- Orthopedic Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Jing Shi
- Orthopedic Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Asymmetric Synthesis and Chiral Technology Key Laboratory of Sichuan Province, Department of Chemistry, Xihua University, Chengdu 610039, P. R. China
| | - Qing-Quan Kong
- Orthopedic Department, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| |
Collapse
|
28
|
Thöny B, Ng J, Kurian MA, Mills P, Martinez A. Mouse models for inherited monoamine neurotransmitter disorders. J Inherit Metab Dis 2024; 47:533-550. [PMID: 38168036 DOI: 10.1002/jimd.12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Several mouse models have been developed to study human defects of primary and secondary inherited monoamine neurotransmitter disorders (iMND). As the field continues to expand, current defects in corresponding mouse models include enzymes and a molecular co-chaperone involved in monoamine synthesis and metabolism (PAH, TH, PITX3, AADC, DBH, MAOA, DNAJC6), tetrahydrobiopterin (BH4) cofactor synthesis and recycling (adGTPCH1/DRD, arGTPCH1, PTPS, SR, DHPR), and vitamin B6 cofactor deficiency (ALDH7A1), as well as defective monoamine neurotransmitter packaging (VMAT1, VMAT2) and reuptake (DAT). No mouse models are available for human DNAJC12 co-chaperone and PNPO-B6 deficiencies, disorders associated with recessive variants that result in decreased stability and function of the aromatic amino acid hydroxylases and decreased neurotransmitter synthesis, respectively. More than one mutant mouse is available for some of these defects, which is invaluable as different variant-specific (knock-in) models may provide more insights into underlying mechanisms of disorders, while complete gene inactivation (knock-out) models often have limitations in terms of recapitulating complex human diseases. While these mouse models have common phenotypic traits also observed in patients, reflecting the defective homeostasis of the monoamine neurotransmitter pathways, they also present with disease-specific manifestations with toxic accumulation or deficiency of specific metabolites related to the specific gene affected. This review provides an overview of the currently available models and may give directions toward selecting existing models or generating new ones to investigate novel pathogenic mechanisms and precision therapies.
Collapse
Affiliation(s)
- Beat Thöny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Joanne Ng
- Genetic Therapy Accelerator Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - Manju A Kurian
- Zayed Centre for Research into Rare Disease in Children, GOS Institute of Child Health, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Philippa Mills
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Aurora Martinez
- Department of Biomedicine and Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
29
|
Jung-Kc K, Tristán-Noguero A, Altankhuyag A, Piñol Belenguer D, Prestegård KS, Fernandez-Carasa I, Colini Baldeschi A, Sigatulina Bondarenko M, García-Cazorla A, Consiglio A, Martinez A. Tetrahydrobiopterin (BH 4) treatment stabilizes tyrosine hydroxylase: Rescue of tyrosine hydroxylase deficiency phenotypes in human neurons and in a knock-in mouse model. J Inherit Metab Dis 2024; 47:494-508. [PMID: 38196161 DOI: 10.1002/jimd.12702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Proteostatic regulation of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine biosynthesis, is crucial for maintaining proper brain neurotransmitter homeostasis. Variants of the TH gene are associated with tyrosine hydroxylase deficiency (THD), a rare disorder with a wide phenotypic spectrum and variable response to treatment, which affects protein stability and may lead to accelerated degradation, loss of TH function and catecholamine deficiency. In this study, we investigated the effects of the TH cofactor tetrahydrobiopterin (BH4) on the stability of TH in isolated protein and in DAn- differentiated from iPSCs from a human healthy subject, as well as from THD patients with the R233H variant in homozygosity (THDA) and R328W and T399M variants in heterozygosity (THDB). We report an increase in TH and dopamine levels, and an increase in the number of TH+ cells in control and THDA cells. To translate this in vitro effect, we treated with BH4 a knock-in THD mouse model with Th variant corresponding to R233H in patients. Importantly, treatment with BH4 significantly improved motor function in these mice, as demonstrated by increased latency on the rotarod test and improved horizontal activity (catalepsy). In conclusion, our study demonstrates the stabilizing effects of BH4 on TH protein levels and function in THD neurons and mice, rescuing disease phenotypes and improving motor outcomes. These findings highlight the therapeutic potential of BH4 as a treatment option for THDA patients with specific variants and provide insights into the modulation of TH stability and its implications for THD management.
Collapse
Affiliation(s)
- Kunwar Jung-Kc
- Department of Biomedicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
| | - Alba Tristán-Noguero
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology Department, Institut Pediàtric de Recerca and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Molecular Physiology of the Synapse, Institut de Recerca Sant Pau (IR Sant Pau), Universitat Autònoma Barcelona, Barcelona, Spain
| | | | - David Piñol Belenguer
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | | | - Irene Fernandez-Carasa
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Arianna Colini Baldeschi
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Maria Sigatulina Bondarenko
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology Department, Institut Pediàtric de Recerca and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Angeles García-Cazorla
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology Department, Institut Pediàtric de Recerca and MetabERN, Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica En Red Enfermedades Raras (CIBERER), Madrid, Spain
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
30
|
Jung-Klawitter S, Richter P, Yuan Y, Welzel K, Kube M, Bähr S, Leibner A, Flory E, Opladen T. Tyrosine hydroxylase variants influence protein expression, cellular localization, stability, enzymatic activity and the physical interaction between tyrosine hydroxylase and GTP cyclohydrolase 1. J Inherit Metab Dis 2024; 47:517-532. [PMID: 38084654 DOI: 10.1002/jimd.12690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/17/2023] [Accepted: 11/03/2023] [Indexed: 05/16/2024]
Abstract
Tyrosine hydroxylase (TH) is the rate-limiting enzyme in dopamine biosynthesis catalyzing the tetrahydrobiopterin (BH4)-dependent hydroxylation of tyrosine to L-DOPA. Here, we analyzed 25 TH variants associated with various degrees of dopa-responsive dystonia and evaluate the effect of each variant on protein stability, activity and cellular localization. Furthermore, we investigated the physical interaction between TH and human wildtype (wt) GTP cyclohydrolase 1 (GTPCH) and the effect of variants on this interaction. Our in vitro results classify variants according to their resistance to proteinase K digestion into three groups (stable, intermediate, unstable). Based on their cellular localization, two groups of variants can be identified, variant group one with cytoplasmic distribution and variant group two forming aggregates. These aggregates do not correlate with loss of enzymatic activity but nevertheless might be a good target for molecular chaperones. Unfortunately, no obvious correlation between the half-life of a variant and its enzymatic activity or between solubility, stability and enzymatic activity of a given variant could be found. Excitingly, some variants disrupt the physical interaction between TH and human wildtype GTPCH, thereby interfering with enzymatic activity and offering new druggable targets for therapy. Taken together, our results highlight the importance of an in-depth molecular analysis of each variant in order to be able to classify groups of disease variants and to find specific therapies for each subgroup. Stand-alone in silico analyses predict less precise the effect of specific variants and should be combined with other in vitro analyses in cellular model systems.
Collapse
Affiliation(s)
- Sabine Jung-Klawitter
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Petra Richter
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Yuheng Yuan
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Karin Welzel
- Division of Hematology, Cell and Gene Therapy, Paul-Ehrlich-Institute, Langen, Germany
| | - Marie Kube
- Institute of Molecular Biology, Mainz, Germany
| | - Stella Bähr
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Alexander Leibner
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| | - Egbert Flory
- Division of Hematology, Cell and Gene Therapy, Paul-Ehrlich-Institute, Langen, Germany
| | - Thomas Opladen
- Medical Faculty, Center for Child and Adolescent Medicine, Pediatric Clinic I, Section of Neuropediatrics and Metabolic Medicine, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
31
|
Molnár Z, Koplányi G, Farkas R, Péli N, Kenéz B, Decsi B, Katona G, Balogh GT, Vértessy BG, Balogh-Weiser D. Immobilization of human tyrosine hydroxylase onto magnetic nanoparticles - A novel formulation of a therapeutic enzyme. Int J Biol Macromol 2024; 268:131939. [PMID: 38692555 DOI: 10.1016/j.ijbiomac.2024.131939] [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: 09/27/2023] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
Human tyrosine hydroxylase (hTH) has key role in the production of catecholamine neurotransmitters. The structure, function and regulation of hTH has been extensively researched area and the possibility of enzyme replacement therapy (ERT) involving hTH through nanocarriers has been raised as well. However, our understanding on how hTH may interact with nanocarriers is still lacking. In this work, we attempted to investigate the immobilization of hTH on magnetic nanoparticles (MNPs) with various surface linkers in quantitative and mechanistic detail. Our results showed that the activity of hTH was retained after immobilization via secondary and covalent interactions as well. The colloidal stability of hTH could be also enhanced proved by Dynamic light scattering and Zeta potential analysis and a homogenous enzyme layer could be achieved, which was investigated by Raman mapping. The covalent attachment of hTH on MNPs via aldehyde or epoxy linkers provide irreversible immobilization and 38.1 % and 16.5 % recovery (ER). The hTH-MNPs catalyst had 25 % ER in average in simulated nasal electrolyte solution (SNES). This outcome highlights the relevance of immobilization applying MNPs as a potential formulation tool of sensitive therapeutic enzymes offering new opportunities for ERT related to neurodegenerative disorders.
Collapse
Affiliation(s)
- Zsófia Molnár
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary; Institue of Enzymology, Research Center of Natural Science, Eötvös Loránd Research Network, Magyar tudósok körútja 2., H-1117 Budapest, Hungary
| | - Gábor Koplányi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Réka Farkas
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Noémi Péli
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Balázs Kenéz
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Balázs Decsi
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - György T Balogh
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary; Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes E. Street 7-9, H-1092 Budapest, Hungary
| | - Beáta G Vértessy
- Institue of Enzymology, Research Center of Natural Science, Eötvös Loránd Research Network, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; Department Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Diána Balogh-Weiser
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary; Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary.
| |
Collapse
|
32
|
Liu G, Li M, Zeng Z, Fan Q, Ren X, Wang Z, Sun Y, He Y, Sun L, Deng Y, Liu S, Zhong C, Gao J. Tyrosine hydroxylase inhibits HCC progression by downregulating TGFβ/Smad signaling. Eur J Med Res 2024; 29:228. [PMID: 38610044 PMCID: PMC11015545 DOI: 10.1186/s40001-024-01703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/31/2024] [Indexed: 04/14/2024] Open
Abstract
The alteration of metabolic processes has been found to have significant impacts on the development of hepatocellular carcinoma (HCC). Nevertheless, the effects of dysfunction of tyrosine metabolism on the development of HCC remains to be discovered. This research demonstrated that tyrosine hydroxylase (TH), which responsible for the initial and limiting step in the bio-generation of the neuro-transmitters dopamine and adrenaline, et al. was shown to be reduced in HCC. Increased expression of TH was found facilitates the survival of HCC patients. In addition, decreased TH indicated larger tumor size, much more numbers of tumor, higher level of AFP, and the presence of cirrhosis. TH effectively impairs the growth and metastasis of HCC cells, a process dependent on the phosphorylation of serine residues (S19/S40). TH directly binds to Smad2 and hinders the cascade activation of TGFβ/Smad signaling with the treatment of TGFβ1. In summary, our study uncovered the non-metabolic functions of TH in the development of HCC and proposes that TH might be a promising biomarker for diagnosis as well as an innovative target for metastatic HCC.
Collapse
Affiliation(s)
- Guoqian Liu
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, Hunan, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Mengwei Li
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, Hunan, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zimei Zeng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Qi Fan
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xinxin Ren
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Zhexin Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yaoqi Sun
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yulin He
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lunquan Sun
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, Hunan, China
| | - Yuezhen Deng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Shupeng Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Institute of Gynecological Minimally Invasive Medicine, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chenxi Zhong
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Jie Gao
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, Hunan, China.
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Xiangya Cancer Center, Xiangya Hospital, Central South University, 87th of Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
33
|
Duda J, Podlewska S. Prediction of probability distributions of molecular properties: towards more efficient virtual screening and better understanding of compound representations. Mol Divers 2024; 28:437-448. [PMID: 36586082 DOI: 10.1007/s11030-022-10589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/18/2022] [Indexed: 01/01/2023]
Abstract
Various in silico approaches to predict activity and properties of chemical compounds constitute nowadays the basis of computer-aided drug design. While there is a general focus on the predictions of values, mathematically more appropriate is the prognosis of probability distributions, which offers additional possibilities, such as the evaluation of uncertainty, higher moments, and quantiles. In this study, we applied the Hierarchical Correlation Reconstruction approach to assess several ADMET properties of chemical compounds. It uses multiple linear regression to independently assess multiple moments, which are then finally combined into predicted probability distribution. The method enables inexpensive selection of compounds with properties nearly certain to fall into the particular range during virtual screening and automatic rejection of predictions characterized by high rate of uncertainty; however, unlike to the currently used virtual screening methods, it focuses on the prediction of the property distribution, not its actual value. Moreover, the presented protocol enables detection of structural features, which should be carefully considered when optimizing compounds towards particular property, as well as it provides deeper understanding of the examined compound representations.
Collapse
Affiliation(s)
- Jarosław Duda
- Faculty of Mathematics and Computer Science, Jagiellonian University, Łojasiewicza 6, 30-348, Kraków, Poland
| | - Sabina Podlewska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| |
Collapse
|
34
|
Liao XL, Chen ZF, Ou SP, Liu QY, Lin SH, Zhou JM, Wang Y, Cai Z. Neurological impairment is crucial for tire rubber-derived contaminant 6PPDQ-induced acute toxicity to rainbow trout. Sci Bull (Beijing) 2024; 69:621-635. [PMID: 38185590 DOI: 10.1016/j.scib.2023.12.045] [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: 09/23/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6PPDQ) has attracted significant attention due to its highly acute lethality to sensitive salmonids. However, studies investigating the mechanisms underlying its acute toxicity have been lacking. In this work, we demonstrated the sensitivity of rainbow trout to 6PPDQ-induced mortality. Moribund trout exhibited significantly higher brain concentrations of 6PPDQ compared to surviving trout. In an in vitro model using human brain microvascular endothelial cells, 6PPDQ can penetrate the blood-brain barrier and enhance blood-brain barrier permeability without compromising cell viability. The time spent in the top of the tank increased with rising 6PPDQ concentrations, as indicated by locomotion behavior tests. Furthermore, 6PPDQ influenced neurotransmitter levels and mRNA expression of neurotransmission-related genes in the brain and exhibited strong binding affinity to target neurotransmission-related proteins using computational simulations. The integrated biomarker response value associated with neurotoxicity showed a positive linear correlation with trout mortality. These findings significantly contribute to filling the knowledge gap between neurological impairments and apical outcomes, including behavioral effects and mortality, induced by 6PPDQ.
Collapse
Affiliation(s)
- Xiao-Liang Liao
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhi-Feng Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Shi-Ping Ou
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Qian-Yi Liu
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Shan-Hong Lin
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Jia-Ming Zhou
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Yujie Wang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zongwei Cai
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China; State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China.
| |
Collapse
|
35
|
Maheshwari S, Singh A, Ansari VA, Mahmood T, Wasim R, Akhtar J, Verma A. Navigating the dementia landscape: Biomarkers and emerging therapies. Ageing Res Rev 2024; 94:102193. [PMID: 38215913 DOI: 10.1016/j.arr.2024.102193] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
The field of dementia research has witnessed significant developments in our understanding of neurodegenerative disorders, with a particular focus on Alzheimer's disease (AD) and Frontotemporal Dementia (FTD). Dementia, a collection of symptoms arising from the degeneration of brain cells, presents a significant healthcare challenge, especially as its prevalence escalates with age. This abstract delves into the complexities of these disorders, the role of biomarkers in their diagnosis and monitoring, as well as emerging neurophysiological insights. In the context of AD, anti-amyloid therapy has gained prominence, aiming to reduce the accumulation of amyloid-beta (Aβ) plaques in the brain, a hallmark of the disease. Notably, Leqembi recently received full FDA approval, marking a significant breakthrough in AD treatment. Additionally, ongoing phase 3 clinical trials are investigating novel therapies, including Masitinib and NE3107, focusing on cognitive and functional improvements in AD patients. In the realm of FTD, research has unveiled distinct neuropathological features, including the involvement of proteins like TDP-43 and progranulin, providing valuable insights into the diagnosis and management of this heterogeneous condition. Biomarkers, including neurofilaments and various tau fragments, have shown promise in enhancing diagnostic accuracy. Neurophysiological techniques, such as transcranial magnetic stimulation (TMS), have contributed to our understanding of AD and FTD. TMS has uncovered unique neurophysiological signatures, highlighting impaired plasticity, hyperexcitability, and altered connectivity in AD, while FTD displays differences in neurotransmitter systems, particularly GABAergic and glutamatergic circuits. Lastly, ongoing clinical trials in anti-amyloid therapy for AD, such as Simufilam, Solanezumab, Gantenerumab, and Remternetug, offer hope for individuals affected by this devastating disease, with the potential to alter the course of cognitive decline. These advancements collectively illuminate the evolving landscape of dementia research and the pursuit of effective treatments for these challenging conditions.
Collapse
Affiliation(s)
- Shubhrat Maheshwari
- Faculty of Pharmaceutical Sciences Rama University Mandhana, Bithoor Road, Kanpur, Uttar Pradesh 209217, India; Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| | - Aditya Singh
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Vaseem Ahamad Ansari
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Tarique Mahmood
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Rufaida Wasim
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Juber Akhtar
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Amita Verma
- Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| |
Collapse
|
36
|
Kawahata I, Fukunaga K. Pathogenic Impact of Fatty Acid-Binding Proteins in Parkinson's Disease-Potential Biomarkers and Therapeutic Targets. Int J Mol Sci 2023; 24:17037. [PMID: 38069360 PMCID: PMC10707307 DOI: 10.3390/ijms242317037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease is a neurodegenerative condition characterized by motor dysfunction resulting from the degeneration of dopamine-producing neurons in the midbrain. This dopamine deficiency gives rise to a spectrum of movement-related symptoms, including tremors, rigidity, and bradykinesia. While the precise etiology of Parkinson's disease remains elusive, genetic mutations, protein aggregation, inflammatory processes, and oxidative stress are believed to contribute to its development. In this context, fatty acid-binding proteins (FABPs) in the central nervous system, FABP3, FABP5, and FABP7, impact α-synuclein aggregation, neurotoxicity, and neuroinflammation. These FABPs accumulate in mitochondria during neurodegeneration, disrupting their membrane potential and homeostasis. In particular, FABP3, abundant in nigrostriatal dopaminergic neurons, is responsible for α-synuclein propagation into neurons and intracellular accumulation, affecting the loss of mesencephalic tyrosine hydroxylase protein, a rate-limiting enzyme of dopamine biosynthesis. This review summarizes the characteristics of FABP family proteins and delves into the pathogenic significance of FABPs in the pathogenesis of Parkinson's disease. Furthermore, it examines potential novel therapeutic targets and early diagnostic biomarkers for Parkinson's disease and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
- BRI Pharma Inc., Sendai 982-0804, Japan
| |
Collapse
|
37
|
Li SJ, Lo YC, Tseng HY, Lin SH, Kuo CH, Chen TC, Chang CW, Liang YW, Lin YC, Wang CY, Cho TY, Wang MH, Chen CT, Chen YY. Nucleus accumbens deep brain stimulation improves depressive-like behaviors through BDNF-mediated alterations in brain functional connectivity of dopaminergic pathway. Neurobiol Stress 2023; 26:100566. [PMID: 37664874 PMCID: PMC10474237 DOI: 10.1016/j.ynstr.2023.100566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023] Open
Abstract
Major depressive disorder (MDD), a common psychiatric condition, adversely affects patients' moods and quality of life. Despite the development of various treatments, many patients with MDD remain vulnerable and inadequately controlled. Since anhedonia is a feature of depression and there is evidence of leading to metabolic disorder, deep brain stimulation (DBS) to the nucleus accumbens (NAc) might be promising in modulating the dopaminergic pathway. To determine whether NAc-DBS alters glucose metabolism via mitochondrial alteration and neurogenesis and whether these changes increase neural plasticity that improves behavioral functions in a chronic social defeat stress (CSDS) mouse model. The Lab-designed MR-compatible neural probes were implanted in the bilateral NAc of C57BL/6 mice with and without CSDS, followed by DBS or sham stimulation. All animals underwent open-field and sucrose preference testing, and brain resting-state functional MRI analysis. Meanwhile, we checked the placement of neural probes in each mouse by T2 images. By confirming the placement location, mice with incorrect probe placement (the negative control group) showed no significant therapeutic effects in behavioral performance and functional connectivity (FC) after receiving electrical stimulation and were excluded from further analysis. Western blotting, seahorse metabolic analysis, and electron microscopy were further applied for the investigation of NAc-DBS. We found NAc-DBS restored emotional deficits in CSDS-subjected mice. Concurrent with behavioral amelioration, the CSDS DBS-on group exhibited enhanced FC in the dopaminergic pathway with increased expression of BDNF- and NeuN-positive cells increased dopamine D1 receptor, dopamine D2 receptors, and TH in the medial prefrontal cortex, NAc, ventral hippocampus, ventral tegmental area, and amygdala. Increased pAMPK/total AMPK and PGC-1α levels, functions of oxidative phosphorylation, and mitochondrial biogenesis were also observed after NAc-DBS treatment. Our findings demonstrate that NAc-DBS can promote BDNF expression, which alters FC and metabolic profile in the dopaminergic pathway, suggesting a potential strategy for ameliorating emotional processes in individuals with MDD.
Collapse
Affiliation(s)
- Ssu-Ju Li
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Yu-Chun Lo
- The Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Xing St., Taipei, 11031, Taiwan, ROC
| | - Hsin-Yi Tseng
- The Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, 11031, Taiwan, ROC
| | - Sheng-Huang Lin
- Department of Neurology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Zhongyang Rd., Hualien, 97002, Taiwan, ROC
- Department of Neurology, School of Medicine, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien, 97004, Taiwan, ROC
| | - Chao-Hung Kuo
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei, 11217, Taiwan, ROC
| | - Ting-Chieh Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Ching-Wen Chang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Rd., Taipei, 115024, Taiwan
| | - Yao-Wen Liang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Yi-Chen Lin
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Chih-Yu Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Tsai-Yu Cho
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Mu-Hua Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
| | - Ching-Te Chen
- Abbott Medical Taiwan Co, 5/F No. 407, Ruei-Guang Rd., Taipei, 11492, Taiwan, ROC
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, No.155, Sec. 2, Linong St., Taipei, 112304, Taiwan, ROC
- The Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Xing St., Taipei, 11031, Taiwan, ROC
| |
Collapse
|
38
|
Sobrado P, Neira JL. Paul F. Fitzpatrick: A life of editorial duties and elucidating the mechanism of enzyme action. Arch Biochem Biophys 2023; 742:109635. [PMID: 37209767 DOI: 10.1016/j.abb.2023.109635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023]
Affiliation(s)
- Pablo Sobrado
- Department of Biochemistry, Virginia Tech, 360 West Campus Drive, Blacksburg, VA, 24061, USA.
| | - José Luis Neira
- IDIBE, Universidad Miguel Hernández, 03202, Elche, Alicante, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) - Unidad Mixta GBsC-CSIC-BIFI, Universidad de Zaragoza, 50018, Zaragoza, Spain.
| |
Collapse
|
39
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
40
|
Li Y, Zhang L, Mao M, He L, Wang T, Pan Y, Zhao X, Li Z, Mu X, Qian Y, Qiu J. Multi-omics analysis of a drug-induced model of bipolar disorder in zebrafish. iScience 2023; 26:106744. [PMID: 37207274 PMCID: PMC10189518 DOI: 10.1016/j.isci.2023.106744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/16/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Emerging studies demonstrate that inflammation plays a crucial role in the pathogenesis of bipolar disorder (BD), but the underlying mechanism remains largely unclear. Given the complexity of BD pathogenesis, we performed high-throughput multi-omic profiling (metabolomics, lipidomics, and transcriptomics) of the BD zebrafish brain to comprehensively unravel the molecular mechanism. Our research proved that in BD zebrafish, JNK-mediated neuroinflammation altered metabolic pathways involved in neurotransmission. On one hand, disturbed metabolism of tryptophan and tyrosine limited the participation of the monoamine neurotransmitters serotonin and dopamine in synaptic vesicle recycling. On the other hand, dysregulated metabolism of the membrane lipids sphingomyelin and glycerophospholipids altered the synaptic membrane structure and neurotransmitter receptors (chrnα7, htr1b, drd5b, and gabra1) activity. Our findings revealed that disturbance of serotonergic and dopaminergic synaptic transmission mediated by the JNK inflammatory cascade was the key pathogenic mechanism in a zebrafish model of BD, provides critical biological insights into the pathogenesis of BD.
Collapse
Affiliation(s)
- Yameng Li
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Lin Zhang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Mingcai Mao
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Linjuan He
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Tiancai Wang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yecan Pan
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaoyu Zhao
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zishu Li
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiyan Mu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yongzhong Qian
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Corresponding author
| | - Jing Qiu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Corresponding author
| |
Collapse
|
41
|
Nagatsu T, Nakashima A, Watanabe H, Ito S, Wakamatsu K, Zucca FA, Zecca L, Youdim M, Wulf M, Riederer P, Dijkstra JM. The role of tyrosine hydroxylase as a key player in neuromelanin synthesis and the association of neuromelanin with Parkinson's disease. J Neural Transm (Vienna) 2023; 130:611-625. [PMID: 36939908 PMCID: PMC10121510 DOI: 10.1007/s00702-023-02617-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/28/2023] [Indexed: 03/21/2023]
Abstract
The dark pigment neuromelanin (NM) is abundant in cell bodies of dopamine (DA) neurons in the substantia nigra (SN) and norepinephrine (NE) neurons in the locus coeruleus (LC) in the human brain. During the progression of Parkinson's disease (PD), together with the degeneration of the respective catecholamine (CA) neurons, the NM levels in the SN and LC markedly decrease. However, questions remain among others on how NM is associated with PD and how it is synthesized. The biosynthesis pathway of NM in the human brain has been controversial because the presence of tyrosinase in CA neurons in the SN and LC has been elusive. We propose the following NM synthesis pathway in these CA neurons: (1) Tyrosine is converted by tyrosine hydroxylase (TH) to L-3,4-dihydroxyphenylalanine (L-DOPA), which is converted by aromatic L-amino acid decarboxylase to DA, which in LC neurons is converted by dopamine β-hydroxylase to NE; (2) DA or NE is autoxidized to dopamine quinone (DAQ) or norepinephrine quinone (NEQ); and (3) DAQ or NEQ is converted to eumelanic NM (euNM) and pheomelanic NM (pheoNM) in the absence and presence of cysteine, respectively. This process involves proteins as cysteine source and iron. We also discuss whether the NM amounts per neuromelanin-positive (NM+) CA neuron are higher in PD brain, whether NM quantitatively correlates with neurodegeneration, and whether an active lifestyle may reduce NM formation.
Collapse
Affiliation(s)
- Toshiharu Nagatsu
- Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi, Japan.
| | - Akira Nakashima
- Department of Physiological Chemistry, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Hirohisa Watanabe
- Department of Neurology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Aichi, Japan
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Aichi, Japan
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milan), Italy
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milan), Italy
| | - Moussa Youdim
- Technion-Rappaport Family Faculty of Medicine, Haifa, Israel
- Department of Biology, Yonsey World Central University, Seoul, South Korea
| | - Maximilian Wulf
- Medical Proteome-Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Medizinisches Proteom‑Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Peter Riederer
- Clinic and Polyclinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Germany
- Department and Research Unit of Psychiatry, Syddansk University, Odense, Denmark
| | | |
Collapse
|
42
|
Vedel IM, Prestel A, Zhang Z, Skawinska NT, Stark H, Harris P, Kragelund BB, Peters GHJ. Structural characterization of human tryptophan hydroxylase 2 reveals that L-Phe is superior to L-Trp as the regulatory domain ligand. Structure 2023:S0969-2126(23)00127-2. [PMID: 37119821 DOI: 10.1016/j.str.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/03/2023] [Accepted: 04/04/2023] [Indexed: 05/01/2023]
Abstract
Tryptophan hydroxylase 2 (TPH2) catalyzes the rate-limiting step in serotonin biosynthesis in the brain. Consequently, regulation of TPH2 is relevant for serotonin-related diseases, yet the regulatory mechanism of TPH2 is poorly understood and structural and dynamical insights are missing. We use NMR spectroscopy to determine the structure of a 47 N-terminally truncated variant of the regulatory domain (RD) dimer of human TPH2 in complex with L-Phe, and show that L-Phe is the superior RD ligand compared with the natural substrate, L-Trp. Using cryo-EM, we obtain a low-resolution structure of a similarly truncated variant of the complete tetrameric enzyme with dimerized RDs. The cryo-EM two-dimensional (2D) class averages additionally indicate that the RDs are dynamic in the tetramer and likely exist in a monomer-dimer equilibrium. Our results provide structural information on the RD as an isolated domain and in the TPH2 tetramer, which will facilitate future elucidation of TPH2's regulatory mechanism.
Collapse
Affiliation(s)
- Ida M Vedel
- Department of Chemistry, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Andreas Prestel
- Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Zhenwei Zhang
- Department of Structural Dynamics, Max Planck Institute for Multidisciplinary Sciences, Am Faßberg 11, 37077 Göttingen, Germany
| | - Natalia T Skawinska
- Department of Chemistry, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Holger Stark
- Department of Structural Dynamics, Max Planck Institute for Multidisciplinary Sciences, Am Faßberg 11, 37077 Göttingen, Germany
| | - Pernille Harris
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen Ø, Denmark
| | - Birthe B Kragelund
- Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark.
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
43
|
Fitzpatrick PF. The aromatic amino acid hydroxylases: Structures, catalysis, and regulation of phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase. Arch Biochem Biophys 2023; 735:109518. [PMID: 36639008 DOI: 10.1016/j.abb.2023.109518] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023]
Abstract
The aromatic amino acid hydroxylases phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase are non-heme iron enzymes that catalyze key physiological reactions. This review discusses the present understanding of the common catalytic mechanism of these enzymes and recent advances in understanding the relationship between their structures and their regulation.
Collapse
Affiliation(s)
- Paul F Fitzpatrick
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
44
|
Molecular Mechanisms, Genotype-Phenotype Correlations and Patient-Specific Treatments in Inherited Metabolic Diseases. J Pers Med 2023; 13:jpm13010117. [PMID: 36675778 PMCID: PMC9864038 DOI: 10.3390/jpm13010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Advances in DNA sequencing technologies are revealing a vast genetic heterogeneity in human population, which may predispose to metabolic alterations if the activity of metabolic enzymes is affected [...].
Collapse
|
45
|
Neuroprotective Effects of Nicotinamide against MPTP-Induced Parkinson's Disease in Mice: Impact on Oxidative Stress, Neuroinflammation, Nrf2/HO-1 and TLR4 Signaling Pathways. Biomedicines 2022; 10:biomedicines10112929. [PMID: 36428497 PMCID: PMC9687839 DOI: 10.3390/biomedicines10112929] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Nicotinamide (NAM) is the amide form of niacin and an important precursor of nicotinamide adenine dinucleotide (NAD), which is needed for energy metabolism and cellular functions. Additionally, it has shown neuroprotective properties in several neurodegenerative diseases. Herein, we sought to investigate the potential protective mechanisms of NAM in an intraperitoneal (i.p) 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) mouse model (wild-type mice (C57BL/6N), eight weeks old, average body weight 25-30 g). The study had four groups (n = 10 per group): control, MPTP (30 mg/kg i.p. for 5 days), MPTP treated with NAM (500 mg/kg, i.p for 10 days) and control treated with NAM. Our study showed that MPTP increased the expression of α-synuclein 2.5-fold, decreased tyrosine hydroxylase (TH) 0.5-fold and dopamine transporters (DAT) levels up to 0.5-fold in the striatum and substantia nigra pars compacta (SNpc), and impaired motor function. However, NAM treatment significantly reversed these PD-like pathologies. Furthermore, NAM treatment reduced oxidative stress by increasing the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) between 0.5- and 1.0-fold. Lastly, NAM treatment regulated neuroinflammation by reducing Toll-like receptor 4 (TLR-4), phosphorylated nuclear factor-κB, tumor (p-NFκB), and cyclooxygenase-2 (COX-2) levels by 0.5- to 2-fold in the PD mouse brain. Overall, these findings suggest that NAM exhibits neuroprotective properties and may be an effective therapeutic agent for PD.
Collapse
|
46
|
Lehrer S, Rheinstein PH. α-synuclein enfolds tyrosine hydroxylase and dopamine ß-hydroxylase, potentially reducing dopamine and norepinephrine synthesis. JOURNAL OF PROTEINS AND PROTEOMICS 2022; 13:109-115. [PMID: 36277464 PMCID: PMC9585989 DOI: 10.1007/s42485-022-00088-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Parkinson's disease (PD) results from degeneration of dopamine and norepinephrine neurons due to α-synuclein aggregates that likely have their origin in the gut. Tyrosine hydroxylase (TH) catalyses the formation of L-DOPA, the rate-limiting step in the biosynthesis of dopamine. A second enzyme, DOPA decarboxylase (DDC), catalyzes the conversion of L-DOPA to dopamine. A third enzyme, dopamine ß-hydroxylase (DBH), catalyzes the conversion of dopamine to norepinephrine. To analyze possible interactions of α-synuclein with TH, DDC and DBH, we performed in silico protein-protein docking. METHODS Protein data bank (pdb) entries were searched on the RCSB Protein Data Bank. We identified four structures that allowed us to examine the relationship of α-synuclein with TH, DDC, and DBH: (1) Human micelle-bound alpha-synuclein, (2) solution structure of the regulatory domain of tyrosine hydroxylase (Rattus norvegicus), (3) crystal structure of human aromatic L-amino acid decarboxylase (DOPA decarboxylase) in the apo form and (4) crystal structure of human dopamine ß-hydroxylase at 2.9 angstrom resolution. We used the ClusPro server (https://cluspro.org) for protein-protein docking. The protein structures were visualized with PyMOL v 2.3.4. RESULTS α-synuclein partially enfolds tyrosine hydroxylase and dopamine ß-hydroxylase, potentially reducing dopamine and norepinephrine synthesis. α-synuclein may dock too far away from DOPA decarboxylase to affect its function directly. CONCLUSIONS Our in silico finding of α-synuclein partly enfolding tyrosine hydroxylase and dopamine ß-hydroxylase suggests that α-synuclein docking inhibition could increase dopamine and norepinephrine biosynthesis, ameliorating PD symptoms. Small molecules that bind to α-synuclein have already been identified. Further studies may lead to new small molecule drugs that block α-synuclein enfolding of tyrosine hydroxylase and dopamine ß-hydroxylase.
Collapse
Affiliation(s)
- Steven Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, Mount Sinai Medical Center, 1 Gustave L. Levy Place, Box 1236, New York, NY 10029, USA
| | | |
Collapse
|