1
|
Gul G. In silico screening of peptide inhibitors targeting α-synuclein for Parkinson's disease. J Mol Graph Model 2025; 139:109079. [PMID: 40381333 DOI: 10.1016/j.jmgm.2025.109079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Parkinson's disease affects cognitive, motor, and autonomic functions due to nervous system degeneration. Though no cure exists, medications and therapies can help alleviate symptoms, but their effectiveness diminishes as the disease progresses, ultimately increasing the need for alternative treatments. α-Synuclein has long been one of the main targets in addressing Parkinson's through drug design studies, but no drugs are yet approved against α-Synuclein aggregation. Therefore, this study aims to develop potential inhibitors of fibrillization by screening thousands of peptides in terms of their binding abilities via Molecular Docking and Molecular Dynamics simulations. Our results show that peptides with Lysine and Arginine at terminal groups result in higher binding affinities to the C-terminal domain. Among the heptapeptides examined, RWRRKRL shows the highest binding free energy to the protein while KKRHKWR exhibits superior stabilizing effect, interacting with both N- and C-terminal regions of α-Synuclein. The inhibitory potential of peptides on the fibrillar structure of protein varies with concentration, and RWRRKRL at 1:3 protein-peptide monomer ratio shows promise as an inhibitor by reducing the internal H-bonds of the protein and increasing RMSD values. These results reveal that short-chain peptides can be designed against α-Synuclein oligomerization offering a potential therapeutic approach for preventing Parkinson's.
Collapse
Affiliation(s)
- Gulsah Gul
- Department of Chemical and Biological Engineering, Koç University, İstanbul, Turkey.
| |
Collapse
|
2
|
Ostrakhovitch EA, Ono K, Yamasaki TR. Metabolomics in Parkinson's Disease and Correlation with Disease State. Metabolites 2025; 15:208. [PMID: 40137172 PMCID: PMC11944848 DOI: 10.3390/metabo15030208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Changes in the level of metabolites, small molecules that are intermediates produced by metabolism or catabolism, are associated with developing diseases. Metabolite signatures in body fluids such as plasma, cerebrospinal fluid, urine, and saliva are associated with Parkinson's disease. Here, we discuss alteration of metabolites in the TCA cycle, pentose phosphate pathway, kynurenic network, and redox system. We also summarize the efforts of many research groups to differentiate between metabolite profiles that characterize PD motor progression and dyskinesia, gait and balance, and non-motor symptoms such as depression and cognitive decline. Understanding how changes in metabolites lead to progression in PD may allow for the identification of individuals at the earliest stage of the disease and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Elena A. Ostrakhovitch
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA;
- Lexington VA Medical Center, Department of Neurology, Lexington, KY 40502, USA
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan;
| | - Tritia R. Yamasaki
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA;
- Lexington VA Medical Center, Department of Neurology, Lexington, KY 40502, USA
| |
Collapse
|
3
|
Liu X, Li T, Tu X, Xu M, Wang J. Mitochondrial fission and fusion in neurodegenerative diseases:Ca 2+ signalling. Mol Cell Neurosci 2025; 132:103992. [PMID: 39863029 DOI: 10.1016/j.mcn.2025.103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Neurodegenerative diseases (NDs) are a group of disorders characterized by the progressive loss of neuronal structure and function. The pathogenesis is intricate and involves a network of interactions among multiple causes and systems. Mitochondria and Ca2+ signaling have long been considered to play important roles in the development of various NDs. Mitochondrial fission and fusion dynamics are important processes of mitochondrial quality control, ensuring the stability of mitochondrial structure and function. Mitochondrial fission and fusion imbalance and Ca2+ signaling disorders can aggravate the disease progression of NDs. In this review, we explore the relationship between mitochondrial dynamics and Ca2+ signaling in AD, PD, ALS, and HD, focusing on the roles of key regulatory proteins (Drp1, Fis1, Mfn1/2, and Opa1) and the association structures between mitochondria and the endoplasmic reticulum (MERCs/MAMs). We provide a detailed analysis of their involvement in the pathogenesis of these four NDs. By integrating these mechanisms, we aim to clarify their contributions to disease progression and offer insights into the development of therapeutic strategies that target mitochondrial dynamics and Ca2+ signaling. We also examine the progress in drug research targeting these pathways, highlighting their potential as therapeutic targets in the treatment of NDs.
Collapse
Affiliation(s)
- Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Xinya Tu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
4
|
Kim JH, Yang S, Kim H, Vo DK, Maeng HJ, Jo A, Shin JH, Shin JH, Baek HM, Lee GH, Kim SH, Lim KH, Dawson VL, Dawson TM, Joo JY, Lee Y. Preclinical studies and transcriptome analysis in a model of Parkinson's disease with dopaminergic ZNF746 expression. Mol Neurodegener 2025; 20:24. [PMID: 40022229 PMCID: PMC11871723 DOI: 10.1186/s13024-025-00814-3] [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: 03/08/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND The parkin-interacting substrate (PARIS, also known as ZNF746) is a transcriptional repressor, whose accumulation and phosphorylation play central pathological roles in Parkinson's disease (PD). PARIS-induced transcriptional repression of PGC-1α or MDM4 contributes to mitochondrial dysfunction and p53-dependent neuron loss in PD. Despite the important role of PARIS in PD pathogenesis, unbiased transcriptomic profiles influenced by PARIS accumulation in dopaminergic neurons remain unexplored. METHODS We engineered Tet-Off conditional transgenic mice expressing PARIS in dopaminergic neurons, driven by DAT-PF-tTA driver mice. The conditional PARIS transgenic mice were characterized by PD-associated pathologies, including progressive dopamine cell loss, neuroinflammation, PGC-1α repression, and mitochondrial proteome alteration. Motor impairment was assessed using pole and rotarod tests. L-DOPA and c-Abl inhibitors were administered to PARIS transgenic mice to evaluate their therapeutic efficacy. The transcriptomic profiles and gene ontology clusters were analyzed by bulk and single-nucleus RNA-seq for the ventral midbrains from PARIS transgenic and age-matched controls. RESULTS Conditional dopaminergic PARIS expression in mice led to the robust and selective dopaminergic neuron degeneration, neuroinflammation, and striatal dopamine deficits, resulting in L-DOPA-responsive motor impairments. Consistent with the results of previous reports, PARIS suppressed dopaminergic PGC-1α expression, disturbed mitochondrial marker protein expression, and reduced COXIV-labeled mitochondria in dopamine neurons. Pharmacological inhibition of c-Abl activity in PARIS transgenic mice largely prevents PD-associated pathological features. Unbiased transcriptomic analysis revealed PARIS-regulated differentially expressed genes (DEGs), both collectively and in a cell-type-specific manner, along with enriched biological pathways linked to PD pathogenesis. Single-cell resolution transcriptomic analysis confirmed repression of PGC-1α and several mitochondria-related target genes in dopaminergic cells. Additionally, we identified distinct glial cell subpopulations and DEGs associated with PD pathogenesis. CONCLUSIONS Conditional PARIS transgenic mice recapitulate robust and dopaminergic neuron-selective pathological features of PD, allowing the preclinical evaluation of antisymptomatic and disease-modifying therapeutic strategies within a couple of months. Based on this new PD mouse model, we provide unbiased bulk and single-nucleus transcriptomic profiles that are regulated by PARIS and potentially contribute to PD pathogenesis. A PD mouse model with flexible pathology induction capacity and a whole transcriptome could serve as a useful resource for translational PD research.
Collapse
Affiliation(s)
- Ji Hun Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute (SBRI), Suwon, 16419, Republic of Korea
| | - Sumin Yang
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea
| | - Hyojung Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute (SBRI), Suwon, 16419, Republic of Korea
| | - Dang-Khoa Vo
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Areum Jo
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute (SBRI), Suwon, 16419, Republic of Korea
| | - Joo-Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Joo-Ho Shin
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute (SBRI), Suwon, 16419, Republic of Korea
| | - Hyeon-Man Baek
- Department of Health Sciences & Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, 21999, Republic of Korea
| | - Gum Hwa Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sung-Hyun Kim
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea
| | - Key-Hwan Lim
- Department of Pharmacy, College of Pharmacy, Chungbuk National University, Cheongju-Si, 28160, Republic of Korea
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jae-Yeol Joo
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea.
| | - Yunjong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute (SBRI), Suwon, 16419, Republic of Korea.
| |
Collapse
|
5
|
Jamal M, Takei S, Tsukamoto I, Miki T, Ohta KI, Hossain MZ, Kinoshita H. Restoration of MPTP-induced Dopamine and Tyrosine Hydroxylase Depletion in the Mouse Brain Through Ethanol and Nicotine. Neurotox Res 2025; 43:9. [PMID: 39937382 DOI: 10.1007/s12640-025-00732-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/27/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025]
Abstract
Dopamine (DA) has long been considered a major factor in the development of Parkinson's disease (PD). Ethanol (EtOH) and nicotine (Nic), either alone or in combination, have been shown to affect nigrostriatal dopaminergic neuronal activity. Here, we investigate whether EtOH and Nic alone or in co-exposure can restore the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced depletion of dopamine (DA), DA metabolites, and tyrosine hydroxylase (TH) in the striatum and hippocampus of C57BL/6N mice. MPTP-treated mice were treated intraperitoneally with saline (control), EtOH (1.0-3.0 g/kg), Nic (0.5-2.0 mg/kg), or a combination of EtOH and Nic. Brain samples were collected 1 h after treatment. DA and its metabolites 3,4-dihydroxyphenylacetic acid (DOPAC), 3-methoxytyramine (3-MT), and homovanillic acid (HVA) were measured by HPLC-ECD, while TH protein content and TH phosphorylation at Ser31 (pSer31 TH) were quantified by Western blot. EtOH (2.0 and 3.0 g/kg) alone reversed the effects of MPTP treatment in both studied brain regions, as evidenced by an increase in DA, DOPAC, and HVA contents, TH protein, and pSer31 TH compared to the MPTP group, indicating restorative effects on DA neurons in the MPTP model. Likewise, Nic (1.0 and 2.0 mg/kg) alone reversed MPTP treatment effects, with treated mice showing increased DA, DOPAC, and HVA contents, TH protein, and pSer31 TH compared to MPTP mice. Co-administration of EtOH (2.0 g/kg) and Nic (1.0 mg/kg) further increased DA, DOPAC and HVA tissue contents, TH protein, and pSer31 TH, indicating an additive effect. These results show that moderate to high doses of EtOH and Nic induce similar increases in brain DA and TH via TH phosphorylation activation in MPTP model mice. EtOH and Nic showed an additive effect in combination, suggesting that their co-application could be a potent therapeutic strategy for treating PD.
Collapse
Affiliation(s)
- Mostofa Jamal
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan.
| | - Sella Takei
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Ikuko Tsukamoto
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University, Kita-Gun, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kita-Gun, Japan
| | - Ken-Ichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kita-Gun, Japan
| | - Md Zakir Hossain
- Division of Intelligent Mechanical Systems Engineering, Faculty of Engineering and Design, Kagawa University, Kita-Gun, Japan
| | - Hiroshi Kinoshita
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| |
Collapse
|
6
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Targeting Ferroptosis in Parkinson's: Repurposing Diabetes Drugs as a Promising Treatment. Int J Mol Sci 2025; 26:1516. [PMID: 40003982 PMCID: PMC11855881 DOI: 10.3390/ijms26041516] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
This review explores the promising potential of repurposing type 2 diabetes (T2D) medications for the treatment of Parkinson's disease (PD), highlighting the shared pathophysiological mechanisms between these two age-related conditions, such as oxidative stress, mitochondrial dysfunction, and ferroptosis. The overlap suggests that existing diabetes drugs could target the common pathways involved in both conditions. Specifically, the review discusses how T2D medications, including metformin (Met), peroxisome-proliferator-activated receptor gamma (PPAR-γ) agonists, sodium-glucose cotransporter-2 (SGLT2) inhibitors, incretins, and dipeptidyl-peptidase 4 (DPP-4) inhibitors, can improve mitochondrial function, reduce neuroinflammation and oxidative stress, and potentially inhibit ferroptosis. The connection between ferroptosis and existing treatments, including diabetes medication, are only beginning to be explored. The limited data can be attributed also to the complexity of mechanisms involved in ferroptosis and Parkinson's disease and to the fact that the specific role of ferroptosis in Parkinson's disease pathogenesis has not been a primary focus until recent. Despite the promising preclinical evidence, clinical findings are mixed, underscoring the need for further research to elucidate these drugs' roles in neurodegeneration. Repurposing existing diabetes medications that have well-established safety profiles for Parkinson's disease treatment could significantly reduce the time and cost associated with drug development and could offer a more comprehensive approach to managing Parkinson's disease compared to treatments targeting a single mechanism.
Collapse
Affiliation(s)
| | | | - Carmen Beatrice Dogaru
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.M.); (I.S.)
| | | |
Collapse
|
7
|
Ahanger IA, Hajam IB, Wani OH. Modulation of conformational integrity and aggregation propensity of α-synuclein by osmolytes: Implications in therapeutic intervention of Parkinson's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 211:63-87. [PMID: 39947754 DOI: 10.1016/bs.pmbts.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Understanding the factors capable of modulation of conformational stability and aggregation propensity of α-synuclein (α-Syn), a hallmark of Parkinson's disease (PD), is crucial for developing future therapeutic interventions for this disease. This chapter aims at exploring the roles of osmolytes in affecting the structural dynamics of α-Syn as well as focuses on how these osmolytes impact folding, stability, and aggregation behavior of this important intrinsically disordered protein. A number of potent osmolytes, including trimethylamine N-oxide (TMAO), trehalose, myo-inositol, taurine, glycine, glutamate, and glycerol were discussed along with their overall effect on α-Syn. These osmolytes can stabilize native conformations or promote alternative folding pathways, thereby influencing α-Syn aggregation. The chapter highlights the dual role of osmolytes in either preventing or exacerbating aggregation, depending on their concentration and interaction mechanism with α-Syn. Moreover, by integrating current research results, the chapter provides insights into how osmolytes might be utilized for therapeutic interventions with potential avenues for managing PD. Overall, the chapter underscores the significance of osmolyte-induced modulation of α-Syn aggregation in the context of PD and highlights future research areas in this direction.
Collapse
Affiliation(s)
| | | | - Owais Hassan Wani
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| |
Collapse
|
8
|
Liu F, Ran Q, Zhang H, Chen J. The Systemic Immune-Inflammation Index and the Risk of Parkinson's Disease in the U.S.: A Cross-Sectional Study. J Clin Med 2025; 14:403. [PMID: 39860410 PMCID: PMC11765590 DOI: 10.3390/jcm14020403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Inflammation is reportedly related to Parkinson's disease (PD). However, the relationship between the systemic immune-inflammation index (SII) and PD remains unexplored. This study aimed to explore the potential relationship between the SII and PD. Methods: This retrospective cross-sectional study analyzed data from the National Health and Nutrition Examination Survey (NHANES) covering the years 2003 to 2020. We analyzed patients over 40 years of age after excluding those with missing SII, PD and covariate data. Logistic regression, subgroup analysis, and restricted cubic spline models were subsequently conducted to evaluate the associations between the SII and PD. Results: Finally, 30,638 participants were included in this study, of whom 416 (1.36%) were identified as having PD. Weighted multivariate regression analysis, adjusted for all covariates, revealed that participants with elevated in-transform (SII) values had a higher likelihood of PD [OR 1.39; 95% CI (1.02, 1.91), p = 0.039] compared to those with lower SII values. The fully adjusted restricted cubic spline curve revealed that the SII/100 was positively and linearly associated with the incidence of PD (p for nonlinearity > 0.05). Additionally, subgroup analysis revealed a stronger correlation between the SII and PD in female participants [OR = 1.06, 95% CI (1.03, 1.08)] compared to male participants [OR = 1.02, 95% CI (1.00, 1.03)] (p for interaction = 0.01). Conclusions: The SII showed a positive correlation with the incidence of PD, particularly in females. Further large-scale prospective studies are necessary to confirm these findings and explore the causal factors that may contribute to the early prevention of PD.
Collapse
Affiliation(s)
- Fujun Liu
- State Key Laboratory of Biotherapy Center, West China Hospital, Sichuan University, Chengdu 610041, China; (F.L.); (H.Z.)
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qibo Ran
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huajin Zhang
- State Key Laboratory of Biotherapy Center, West China Hospital, Sichuan University, Chengdu 610041, China; (F.L.); (H.Z.)
| | - Jing Chen
- Department of Neurosurgery and Neuromodulation Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Tsuboi T, Tatsumi H, Kobayasi K, Hashimoto R, Aiba I. Cognitive impairment in PSP compared with PD: assessment by clinical subtype and longitudinal change. BMJ Neurol Open 2025; 7:e000946. [PMID: 39850796 PMCID: PMC11751889 DOI: 10.1136/bmjno-2024-000946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/21/2024] [Indexed: 01/25/2025] Open
Abstract
Background Longitudinal studies investigating cognitive function changes in patients with progressive supranuclear palsy (PSP) are limited. The variability of cognitive impairment across clinical subtypes of PSP remains unclear. Objective This study aimed to compare the longitudinal changes in cognitive function between patients with PSP and Parkinson's disease (PD) and to assess differences in cognitive impairment among PSP subtypes. Methods A retrospective observational study was conducted using neuropsychological testing data from patients with PSP and PD admitted to our hospital. Results The study included 38 patients with PD and 41 patients with PSP (23 PSP-Richardson's syndrome, 14 PSP-progressive gait freezing (PSP-PGF), 3 PSP-Parkinsonism and 1 PSP-predominant corticobasal syndrome). At baseline, cognitive function was significantly lower in the PSP group than in the PD group. Over 12 months, patients with PSP exhibited significant declines in multiple cognitive domains, whereas no significant changes were observed in the PD group. Among PSP subtypes, PSP-RS showed a faster rate of cognitive decline than PD, while PSP-PGF demonstrated a lower progression than PSP-RS. Conclusion PSP is associated with progressive cognitive impairment, with rates of decline varying by subtype. PSP-PGF exhibited a slower progression than PSP-RS. Clinical management should consider subtype-specific differences in cognitive prognosis to tailor treatment and care.
Collapse
Affiliation(s)
- Takeharu Tsuboi
- Department of Rehabilitation, National Hospital Organization Higashinagoya National Hospital, Nagoya, Japan
| | | | - Kosuke Kobayasi
- Department of Rehabilitation, National Hospital Organization Higashinagoya National Hospital, Nagoya, Japan
| | - Rina Hashimoto
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Japan
| | - Ikuko Aiba
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Japan
| |
Collapse
|
10
|
Kumari S, Kamiya A, Karnik SS, Rohilla S, Dubey SK, Taliyan R. Novel Gene Therapy Approaches for Targeting Neurodegenerative Disorders: Focusing on Delivering Neurotrophic Genes. Mol Neurobiol 2025; 62:386-411. [PMID: 38856793 DOI: 10.1007/s12035-024-04260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
Neurodegenerative illnesses (NDDs) like Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, spinal muscular atrophy, and Huntington's disease have demonstrated considerable potential for gene therapy as a viable therapeutic intervention. NDDs are marked by the decline of neurons, resulting in changes in both behavior and pathology within the body. Strikingly, only symptomatic management is available without a cure for the NDDs. There is an unmet need for a permanent therapeutic approach. Many studies have been going on to target the newer therapeutic molecular targets for NDDs including gene-based therapy. Gene therapy has the potential to provide therapeutic benefits to a large number of patients with NDDs by offering mechanisms including neuroprotection, neuro-restoration, and rectification of pathogenic pathways. Gene therapy is a medical approach that aims to modify the biological characteristics of living cells by controlling the expression of specific genes in certain neurological disorders. Despite being the most complex and well-protected organ in the human body, there is clinical evidence to show that it is possible to specifically target the central nervous system (CNS). This provides hope for the prospective application of gene therapy in treating NDDs in the future. There are several advanced techniques available for using viral or non-viral vectors to deliver the therapeutic gene to the afflicted region. Neurotrophic factors (NTF) in the brain are crucial for the development, differentiation, and survival of neurons in the CNS, making them important in the context of various neurological illnesses. Gene delivery of NTF has the potential to be used as a therapeutic approach for the treatment of neurological problems in the brain. This review primarily focuses on the methodologies employed for delivering the genes of different NTFs to treat neurological disorders. These techniques are currently being explored as a viable therapeutic approach for neurodegenerative diseases. The article exclusively addresses gene delivery approaches and does not cover additional therapy strategies for NDDs. Gene therapy offers a promising alternative treatment for NDDs by stimulating neuronal growth instead of solely relying on symptom relief from drugs and their associated adverse effects. It can serve as a long-lasting and advantageous treatment choice for the management of NDDs. The likelihood of developing NDDs increases with age as a result of neuronal degradation in the brain. Gene therapy is an optimal approach for promoting neuronal growth through the introduction of nerve growth factor genes.
Collapse
Affiliation(s)
- Shobha Kumari
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Aayush Kamiya
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sanika Sanjay Karnik
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sumedha Rohilla
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | | | - Rajeev Taliyan
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India.
| |
Collapse
|
11
|
Böck D, Wilhelm M, Mumenthaler J, Carpanese DF, Kulcsár PI, d'Aquin S, Cremonesi A, Rassi A, Häberle J, Patriarchi T, Schwank G. Base editing of Ptbp1 in neurons alleviates symptoms in a mouse model of Parkinson's disease. eLife 2024; 13:RP97180. [PMID: 39714464 DOI: 10.7554/elife.97180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Parkinson's disease (PD) is a multifactorial disease caused by irreversible progressive loss of dopaminergic neurons (DANs). Recent studies have reported the successful conversion of astrocytes into DANs by repressing polypyrimidine tract binding protein 1 (PTBP1), which led to the rescue of motor symptoms in a chemically-induced mouse model of PD. However, follow-up studies have questioned the validity of this astrocyte-to-DAN conversion model. Here, we devised an adenine base editing strategy to downregulate PTBP1 in astrocytes and neurons in a chemically-induced PD mouse model. While PTBP1 downregulation in astrocytes had no effect, PTBP1 downregulation in neurons of the striatum resulted in the expression of the DAN marker tyrosine hydroxylase (TH) in non-dividing neurons, which was associated with an increase in striatal dopamine concentrations and a rescue of forelimb akinesia and spontaneous rotations. Phenotypic analysis using multiplexed iterative immunofluorescence imaging further revealed that most of these TH-positive cells co-expressed the dopaminergic marker DAT and the pan-neuronal marker NEUN, with the majority of these triple-positive cells being classified as mature GABAergic neurons. Additional research is needed to fully elucidate the molecular mechanisms underlying the expression of the observed markers and understand how the formation of these cells contributes to the rescue of spontaneous motor behaviors. Nevertheless, our findings support a model where downregulation of neuronal, but not astrocytic, PTBP1 can mitigate symptoms in PD mice.
Collapse
Affiliation(s)
- Desiree Böck
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Jonas Mumenthaler
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | | | - Peter I Kulcsár
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Simon d'Aquin
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anahita Rassi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Byrne MD, Petramfar P, Lee JK, Smeyne RJ. Templating of Monomeric Alpha-Synuclein Induces Inflammation and SNpc Dopamine Neuron Death in a Genetic Mouse Model of Synucleinopathy. RESEARCH SQUARE 2024:rs.3.rs-5269499. [PMID: 39606453 PMCID: PMC11601858 DOI: 10.21203/rs.3.rs-5269499/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
While the etiology of most cases of Parkinson's disease (PD) are idiopathic, it has been estimated that 5-10% of PD arise from known genetic mutations. The first mutations described that leads to the development of an autosomal dominant form of PD are in the SNCA gene that codes for the protein alpha-synuclein (α-syn). α-syn is an abundant presynaptic protein that is natively disordered and whose function is still unclear. In PD, α-syn misfolds into multimeric b-pleated sheets that aggregate in neurons (Lewy Bodies/neurites) and spread throughout the neuraxis in a pattern that aligns with disease progression. Here, using IHC, HC, HPLC, and cytokine analysis, we examined the sequelae of intraparenchymal brain seeding of pre-formed fibrils (PFFs) and monomeric α-syn in C57BL/6J (WT) and A53T SNCA mutant mice. We found that injection of PFFs, but not monomeric α-syn, into the striatum of C57BL/6J mice induced spread of aggregated α-syn, loss of SNpc DA neurons and increased neuroinflammation. However, in A53T SNCA mice, we found that both PFFs and monomeric α-syn induced this pathology. This suggests that the conformation changes in α-syn seen in the A53T strain can recruit wild-type α-syn to a pathological misfolded conformation which may provide a mechanism for the induction of PD in humans with SNCA duplication/triplication.
Collapse
|
13
|
Ye Q, Li X, Gao W, Gao J, Zheng L, Zhang M, Yang F, Li H. Role of Rho-associated kinases and their inhibitor fasudil in neurodegenerative diseases. Front Neurosci 2024; 18:1481983. [PMID: 39628659 PMCID: PMC11613983 DOI: 10.3389/fnins.2024.1481983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/01/2024] [Indexed: 12/06/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are prevalent in the elderly. The pathogenesis of NDDs is complex, and currently, there is no cure available. With the increase in aging population, over 20 million people are affected by common NDDs alone (Alzheimer's disease and Parkinson's disease). Therefore, NDDs have profound negative impacts on patients, their families, and society, making them a major global health concern. Rho-associated kinases (ROCKs) belong to the serine/threonine protein kinases family, which modulate diverse cellular processes (e.g., apoptosis). ROCKs may elevate the risk of various NDDs (including Huntington's disease, Parkinson's disease, and Alzheimer's disease) by disrupting synaptic plasticity and promoting inflammatory responses. Therefore, ROCK inhibitors have been regarded as ideal therapies for NDDs in recent years. Fasudil, one of the classic ROCK inhibitor, is a potential drug for treating NDDs, as it repairs nerve damage and promotes axonal regeneration. Thus, the current review summarizes the relationship between ROCKs and NDDs and the mechanism by which fasudil inhibits ROCKs to provide new ideas for the treatment of NDDs.
Collapse
Affiliation(s)
- Qiuyan Ye
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xue Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- Jiangsu College of Nursing, Huaian, China
| | - Jiayue Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liping Zheng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Miaomiao Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fengge Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Honglin Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
14
|
Matsuda K, Mita Y, Saigoh K, Saito Y, Noguchi N. Modifications of DJ-1 in which pI shifts to acidic in red blood cells a potential biomarker for Parkinson's disease at early stages. Free Radic Res 2024; 58:748-757. [PMID: 39576630 DOI: 10.1080/10715762.2024.2430536] [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: 09/06/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 12/28/2024]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases, the incidence of which increases with age. However, since there is no fundamental treatment or methods for early diagnosis, new methods of treatment and diagnosis are urgently needed. We focused on post-translational modifications of DJ-1, which is encoded by the familial PD-causative gene PARK7 in red blood cells (RBCs). DJ-1 has three cysteines (Cys46, Cys53, and Cys106), with Cys106 being preferentially oxidized. We previously reported that sulfinated/sulfonated Cys106 DJ-1 (oxDJ-1) is increased in the RBCs of PD patients. In this study, we analyzed RBC-derived DJ-1 from PD patients and control subjects by 2-dimensional electrophoresis. We found that the ratio of the spot of DJ-1 with a more acidic isoelectric point than oxDJ-1 was increased more significantly than that of oxDJ-1 in RBCs from patients at the early stage of unmedicated PD and decreased with the progression of PD stage and treatment. Furthermore, we revealed that this acidic spot of DJ-1 increased upon exposure to H2O2. However, when either Cys53 or Cys106 of DJ-1 was replaced with serine, there was no significant increase in the acidic spot caused by H2O2. In this study, we propose a new biomarker for early diagnosis of PD using both the ratios of oxDJ-1 to total DJ-1 and the acidic spot of DJ-1 to total DJ-1.
Collapse
Affiliation(s)
- Kohei Matsuda
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Yuichiro Mita
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Kazumasa Saigoh
- Department of Clinical Genetics, Kindai University Hospital, Osaka, Japan
| | - Yoshiro Saito
- Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Noriko Noguchi
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| |
Collapse
|
15
|
Yang K, Lv Z, Zhao W, Lai G, Zheng C, Qi F, Zhao C, Hu K, Chen X, Fu F, Li J, Xie G, Wang H, Wu X, Zheng W. The potential of natural products to inhibit abnormal aggregation of α-Synuclein in the treatment of Parkinson's disease. Front Pharmacol 2024; 15:1468850. [PMID: 39508052 PMCID: PMC11537895 DOI: 10.3389/fphar.2024.1468850] [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: 07/26/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Parkinson's disease (PD), as a refractory neurological disorder with complex etiology, currently lacks effective therapeutic agents. Natural products (NPs), derived from plants, animals, or microbes, have shown promising effects in PD models through their antioxidative and anti-inflammatory properties, as well as the enhancement of mitochondrial homeostasis and autophagy. The misfolding and deposition of α-Synuclein (α-Syn), due to abnormal overproduction and impaired clearance, being central to the death of dopamine (DA) neurons. Thus, inhibiting α-Syn misfolding and aggregation has become a critical focus in PD discovery. This review highlights NPs that can reduce α-Syn aggregation by preventing its overproduction and misfolding, emphasizing their potential as novel drugs or adjunctive therapies for PD treatment, thereby providing further insights for clinical translation.
Collapse
Affiliation(s)
- Kaixia Yang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zhongyue Lv
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wen Zhao
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guogang Lai
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Cheng Zheng
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Feiteng Qi
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Cui Zhao
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaikai Hu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiao Chen
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Fan Fu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiayi Li
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guomin Xie
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Haifeng Wang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiping Wu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wu Zheng
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Neuroscience Medical Center, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
16
|
Singh P, Akhtar A, Admane N, Grover A. The antiviral drug Ribavirin effectively modulates the amyloid transformation of α-Synuclein protein. Comput Biol Chem 2024; 112:108155. [PMID: 39084146 DOI: 10.1016/j.compbiolchem.2024.108155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
α-Synuclein (α-syn) is an intrinsically disordered protein, linked genetically and neuropathologically to Parkinson's disease where this protein aggregates within the brain. Hence, identifying compounds capable of impeding α-syn aggregation puts forward a promising approach for the development of disease-modifying therapies. Herein, we investigated the efficacy of Ribavirin, an FDA-approved compound, in curtailing α-syn amyloid transformation, employing an array of bioinformatic tools and systematic analysis using biophysical techniques. Ribavirin shows a dose dependent anti-aggregation propensity where it effectively subdued the formation of mature fibrillar aggregates of α-syn, where even at the lowest concentration there was a 69 % reduction in the ThT maxima. Ribavirin averts the formation of mature fibrillar aggregates by interacting with the NAC domain of α-syn. Ribavirin redirects the amyloid transformation of α-syn by emanating aggregates of lower order with reduced cross β-sheet signature and revokes the formation of on-pathway amyloids. Collectively, our study puts forward the novel potency of Ribavirin as a promising molecule for therapeutic intervention in Parkinson's disease.
Collapse
Affiliation(s)
- Payal Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Almas Akhtar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nikita Admane
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
17
|
Sung CC, Lam WY, Chung KKK. The role of polo-like kinases 2 in the proteasomal and lysosomal degradation of alpha-synuclein in neurons. FASEB J 2024; 38:e70121. [PMID: 39436202 PMCID: PMC11580719 DOI: 10.1096/fj.202401035r] [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: 05/08/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by the degeneration of dopaminergic neurons in the brain stem. PD is mostly sporadic, but familial PD (FPD) cases are recorded in different studies. The first gene mutation that is linked to FPD is α-synuclein (α-syn). It was then found that α-syn is also accumulated in Lewy body (LB), a classical pathological hallmark in PD patients. Different studies have shown that α-syn accumulation and aggregation can be a crucial factor contributing to the degeneration of dopaminergic neurons in PD. α-syn has been found to be degraded by the ubiquitin proteasomal system (UPS) and autophagy-lysosomal pathway (ALP). In this study, we initially explored how α-syn phosphorylation by GRK6, PLK2 and CK2α would facilitate its degradation in relation to the UPS or ALP. Unexpectedly, we found that the degradation of α-syn through PLK2 phosphorylation could be modulated by UPS and ALP in a novel mechanism. Specially, attenuation of UPS could increase the amount of PLK2 and then could facilitate the phosphorylation and degradation of α-syn through ALP. To test this further in vivo, we attenuate the proteasomal activity in a well-established A53T α-syn transgenic PD mouse model. We found that attenuation of proteasomal activity in the A53T α-syn transgenic mice could reduce the accumulation of α-syn in the striatum and midbrain. Based on our results, this study provides a new insight into how α-syn is degraded through the UPS and ALP.
Collapse
Affiliation(s)
- Chun Chau Sung
- Division of Life Science, State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Wai Yun Lam
- Division of Life Science, State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Kenny K. K. Chung
- Division of Life Science, State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| |
Collapse
|
18
|
Shahid A, Bhatia M. Hydrogen Sulfide: A Versatile Molecule and Therapeutic Target in Health and Diseases. Biomolecules 2024; 14:1145. [PMID: 39334911 PMCID: PMC11430449 DOI: 10.3390/biom14091145] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, research has unveiled the significant role of hydrogen sulfide (H2S) in many physiological and pathological processes. The role of endogenous H2S, H2S donors, and inhibitors has been the subject of studies that have aimed to investigate this intriguing molecule. The mechanisms by which H2S contributes to different diseases, including inflammatory conditions, cardiovascular disease, viral infections, and neurological disorders, are complex. Despite noteworthy progress, several questions remain unanswered. H2S donors and inhibitors have shown significant therapeutic potential for various diseases. This review summarizes our current understanding of H2S-based therapeutics in inflammatory conditions, cardiovascular diseases, viral infections, and neurological disorders.
Collapse
Affiliation(s)
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| |
Collapse
|
19
|
Kim E, Yang W, Cha DS, Han YT. Synthesis and Dopaminergic Neuroprotective Activity of the Proposed Structure of Bassiamide A, a Caffeamide Alkaloid Derived from Bassia Indica Wight. Chem Biodivers 2024; 21:e202400836. [PMID: 38693058 DOI: 10.1002/cbdv.202400836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/03/2024]
Abstract
Herein, we describe the synthesis of the proposed structure of the caffeamide alkaloid bassiamide A. The amide moiety of bassiamide A was readily formed via an amide coupling reaction between caffeic acid and the known N-(3-aminopropyl)-3-methylbutanamide. However, the spectral data of the synthesized bassiamide A did not agree with that of a previous study. The structure of the synthesized bassiamide A was confirmed using combined two-dimensional NMR analysis. Extended analyses of the bioactivity of the synthesized bassiamide A revealed its efficacy in protecting dopaminergic neurons from MPP+-induced neurotoxicity in Caenorhabditis elegans. Additionally, treatment with bassiamide A notably ameliorated the impaired food-sensing ability and locomotion of Caenorhabditis elegans, suggesting a protective effect on the functionality of dopaminergic neurons.
Collapse
Affiliation(s)
- Eunyeong Kim
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Korea
| | - Wooin Yang
- College of Pharmacy, Woosuk University, 443 Samnye-ro, Wanju-gun, Jeonbuk, Korea
| | - Dong Seok Cha
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Korea
| | - Young Taek Han
- College of Pharmacy, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Korea
| |
Collapse
|
20
|
Li J, Ng KW, Sung CC, Chung KKK. The role of age-associated alpha-synuclein aggregation in a conditional transgenic mouse model of Parkinson's disease: Implications for Lewy body formation. J Neurochem 2024; 168:1215-1236. [PMID: 38693066 DOI: 10.1111/jnc.16122] [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: 11/08/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that is affecting an increasing number of older adults. Although PD is mostly sporadic, genetic mutations have been found in cohorts of families with a history of familial PD (FPD). The first such mutation linked to FPD causes a point mutation (A53T) in α-synuclein (α-syn), a major component of Lewy bodies, which are a classical pathological hallmark of PD. These findings suggest that α-syn is an important contributor to the development of PD. In our previous study, we developed an adenoviral mouse model of PD and showed that the expression of wild-type (WT) α-syn or a mutant form with an increased propensity to aggregate, designated as WT-CL1 α-syn, could be used to study how α-syn aggregation contributes to PD. In this study, we established a transgenic mouse model that conditionally expresses WT or WT-CL1 α-syn in dopaminergic (DA) neurons and found that the expression of either WT or WT-CL1 α-syn was associated with an age-dependent degeneration of DA neurons and movement dysfunction. Using this model, we were able to monitor the process of α-syn aggregate formation and found a correlation between age and the number and sizes of α-syn aggregates formed. These results provide a potential mechanism by which age-dependent α-syn aggregation may lead to the formation of Lewy bodies in PD pathogenesis.
Collapse
Affiliation(s)
- Jiahua Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ka Wai Ng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chun Chau Sung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kenny K K Chung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
21
|
Kotsyuba E, Dyachuk V. Effects of Chronic Exposure to Low Doses of Rotenone on Dopaminergic and Cholinergic Neurons in the CNS of Hemigrapsus sanguineus. Int J Mol Sci 2024; 25:7159. [PMID: 39000265 PMCID: PMC11241242 DOI: 10.3390/ijms25137159] [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: 05/22/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Rotenone, as a common pesticide and insecticide frequently found in environmental samples, may be present in aquatic habitats worldwide. Exposure to low concentrations of this compound may cause alterations in the nervous system, thus contributing to Parkinsonian motor symptoms in both vertebrates and invertebrates. However, the effects of chronic exposure to low doses of rotenone on the activity of neurotransmitters that govern motor functions and on the specific molecular mechanisms leading to movement morbidity remain largely unknown for many aquatic invertebrates. In this study, we analyzed the effects that rotenone poisoning exerts on the activity of dopamine (DA) and acetylcholine (ACh) synthesis enzymes in the central nervous system (CNS) of Asian shore crab, Hemigrapsus sanguineus (de Haan, 1835), and elucidated the association of its locomotor behavior with Parkinson's-like symptoms. An immunocytochemistry analysis showed a reduction in tyrosine hydroxylase (TH) in the median brain and the ventral nerve cord (VNC), which correlated with the subsequent decrease in the locomotor activity of shore crabs. We also observed a variation in cholinergic neurons' activity, mostly in the ventral regions of the VNC. Moreover, the rotenone-treated crabs showed signs of damage to ChAT-lir neurons in the VNC. These data suggest that chronic treatment with low doses of rotenone decreases the DA level in the VNC and the ACh level in the brain and leads to progressive and irreversible reductions in the crab's locomotor activity, life span, and changes in behavior.
Collapse
Affiliation(s)
- Elena Kotsyuba
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| |
Collapse
|
22
|
Amiri B, Yazdani Tabrizi M, Naziri M, Moradi F, Arzaghi M, Archin I, Behaein F, Bagheri Pour A, Ghannadikhosh P, Imanparvar S, Akhtari Kohneshahri A, Sanaye Abbasi A, Zerangian N, Alijanzadeh D, Ghayyem H, Azizinezhad A, Ahmadpour Youshanlui M, Poudineh M. Neuroprotective effects of flavonoids: endoplasmic reticulum as the target. Front Neurosci 2024; 18:1348151. [PMID: 38957188 PMCID: PMC11218733 DOI: 10.3389/fnins.2024.1348151] [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: 12/01/2023] [Accepted: 03/28/2024] [Indexed: 07/04/2024] Open
Abstract
The incidence of neurological disorders, particularly age-related neurodegenerative pathologies, exhibits an alarming upward trend, while current pharmacological interventions seldom achieve curative outcomes. Despite their diverse clinical presentations, neurological diseases often share a common pathological thread: the aberrant accumulation of misfolded proteins within the endoplasmic reticulum (ER). This phenomenon, known as ER stress, arises when the cell's intrinsic quality control mechanisms fail to cope with the protein-folding burden. Consequently, misfolded proteins accumulate in the ER lumen, triggering a cascade of cellular stress responses. Recognizing this challenge, researchers have intensified their efforts over the past two decades to explore natural compounds that could potentially slow or even reverse these devastating pathologies. Flavonoids constitute a vast and heterogeneous class of plant polyphenols, with over 10,000 identified from diverse natural sources such as wines, vegetables, medicinal plants, and organic products. Flavonoids are generally divided into six different subclasses: anthocyanidins, flavanones, flavones, flavonols, isoflavones, and flavonols. The diverse family of flavonoids, featuring a common phenolic ring backbone adorned with varying hydroxyl groups and additional modifications, exerts its antioxidant activity by inhibiting the formation of ROS, as evidenced by research. Also, studies suggest that polyphenols such as flavonoids can regulate ER stress through apoptosis and autophagy. By understanding these mechanisms, we can unlock the potential of flavonoids as novel therapeutic agents for neurodegenerative disorders. Therefore, this review critically examines the literature exploring the modulatory effects of flavonoids on various steps of the ER stress in neurological disorders.
Collapse
Affiliation(s)
- Bita Amiri
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Yazdani Tabrizi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdyieh Naziri
- Student Research Committee, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Moradi
- Student Research Committee, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Arzaghi
- Department of Physical Education and Sports Science-Nutrition, Branch Islamic Azad University, Tehran, Iran
| | - Iman Archin
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Parna Ghannadikhosh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Imanparvar
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ata Akhtari Kohneshahri
- Student Research Committee, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Ali Sanaye Abbasi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasibeh Zerangian
- PhD Student in Health Education and Health Promotion, Department of Health Education and Health Promotion, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dorsa Alijanzadeh
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hani Ghayyem
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Azizinezhad
- Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | | | - Mohadeseh Poudineh
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
23
|
Papandreou A, Singh N, Gianfrancesco L, Budinger D, Barwick K, Agrotis A, Luft C, Shao Y, Lenaerts AS, Gregory A, Jeong SY, Hogarth P, Hayflick S, Barral S, Kriston-Vizi J, Gissen P, Kurian MA, Ketteler R. Cardiac glycosides restore autophagy flux in an iPSC-derived neuronal model of WDR45 deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.13.556416. [PMID: 37745522 PMCID: PMC10515824 DOI: 10.1101/2023.09.13.556416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Beta-Propeller Protein-Associated Neurodegeneration (BPAN) is one of the commonest forms of Neurodegeneration with Brain Iron Accumulation, caused by mutations in the gene encoding the autophagy-related protein, WDR45. The mechanisms linking autophagy, iron overload and neurodegeneration in BPAN are poorly understood and, as a result, there are currently no disease-modifying treatments for this progressive disorder. We have developed a patient-derived, induced pluripotent stem cell (iPSC)-based midbrain dopaminergic neuronal cell model of BPAN (3 patient, 2 age-matched controls and 2 isogenic control lines) which shows defective autophagy and aberrant gene expression in key neurodegenerative, neurodevelopmental and collagen pathways. A high content imaging-based medium-throughput blinded drug screen using the FDA-approved Prestwick library identified 5 cardiac glycosides that both corrected disease-related defective autophagosome formation and restored BPAN-specific gene expression profiles. Our findings have clear translational potential and emphasise the utility of iPSC-based modelling in elucidating disease pathophysiology and identifying targeted therapeutics for early-onset monogenic disorders.
Collapse
Affiliation(s)
- Apostolos Papandreou
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
- Laboratory for Molecular Cell Biology, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Nivedita Singh
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Lorita Gianfrancesco
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Katy Barwick
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Alexander Agrotis
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Christin Luft
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Ying Shao
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | | | | | | | | | | | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Janos Kriston-Vizi
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Paul Gissen
- Inborn Errors of Metabolism, Genetics & Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- These authors contributed equally
| | - Robin Ketteler
- Laboratory for Molecular Cell Biology, University College London, London, UK
- Department of Human Medicine, Medical School Berlin, Berlin, Germany
- These authors contributed equally
| |
Collapse
|
24
|
Wang Z, Huang PE, Wang N, Zhang Q, Kang J, Fang Y, Ning B, Li L. β-asarone inhibits autophagy by activating the PI3K/Akt/mTOR pathway in a rat model of depression in Parkinson's disease. Behav Brain Res 2024; 465:114966. [PMID: 38518853 DOI: 10.1016/j.bbr.2024.114966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE It is unclear whether β-asarone has a good antidepressant effect and what is the main mechanism in Depression in Parkinson's disease (DPD) model rats. METHODS In this study, DPD model rats were screened from 6-OHDA induced rats by sucrose preference test (SPT) and forced swimming test (FST). DPD model rats were divided into eight groups: model group, pramipexole group, β-asarone low-dose group (β-asarone 7.5 group), β-asarone medium-dose group (β-asarone 15 group), β-asarone high-dose group (β-asarone 30 group), 3-MA group, rapamycin group, and PI3K inhibitor group. 28 days after the end of treatment, open field test (OFT), SPT and FST were conducted in rats. The level of α-synuclein (α-syn) in the striatum was determined by enzyme-linked immunosorbent assay (ELISA). The expression of Beclin-1, p62 in the striatum was determined by western blot. The expression of PI3K, p-PI3K, Akt, p-Akt, mTOR, p-mTOR, Beclin-1, and p62 in the hippocampus was determined by western blot. The spine density of neurons in the hippocampus was detected by golgi staining. RESULTS The results showed that 4-week oral administration of β-asarone improve the motor and depressive symptoms of DPD model rats, and decrease the content of α-syn in the striatum. β-asarone inhibited the expression of autophagy in the striatum of DPD model rats. Furthermore, β-asarone decreased the levels of Beclin-1 protein, increased the expression of p62, p-PI3K, p-AKT, and p-mTOR, and improved the density of neuron dendritic spine in the hippocampus. CONCLUSIONS We concluded that β-asarone might improve the behavior of DPD model rats by activating the PI3K/Akt/mTOR pathway, inhibiting autophagy and protecting neuron.
Collapse
Affiliation(s)
- Zhifang Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping-E Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nanbu Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Jian Kang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongqi Fang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baile Ning
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Ling Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Sharma M, Aggarwal N, Mishra J, Panda JJ. Neuroglia targeting nano-therapeutic approaches to rescue aging and neurodegenerating brain. Int J Pharm 2024; 654:123950. [PMID: 38430951 DOI: 10.1016/j.ijpharm.2024.123950] [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: 09/11/2023] [Revised: 02/12/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Despite intense efforts at the bench, the development of successful brain-targeting therapeutics to relieve malicious neural diseases remains primitive. The brain, being a beautifully intricate organ, consists of heterogeneous arrays of neuronal and glial cells. Primarily acting as the support system for neuronal functioning and maturation, glial cells have been observed to be engaged more apparently in the progression and worsening of various neural pathologies. The diseased state is often related to metabolic alterations in glial cells, thereby modulating their physiological homeostasis in conjunction with neuronal dysfunction. A plethora of data indicates the effect of oxidative stress, protein aggregation, and DNA damage in neuroglia impairments. Still, a deeper insight is needed to gain a conflict-free understanding in this arena. As a consequence, glial cells hold the potential to be identified as promising targets for novel therapeutic approaches aimed at brain protection. In this review, we describe the recent strides taken in the direction of understanding the impact of oxidative stress, protein aggregation, and DNA damage on neuroglia impairment and neuroglia-directed nanotherapeutic approaches to mitigate the burden of various neural disorders.
Collapse
Affiliation(s)
- Manju Sharma
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | - Nidhi Aggarwal
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | - Jibanananda Mishra
- School of Biosciences, RIMT University, Mandi Gobindgarh, Punjab 147301, India.
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India.
| |
Collapse
|
26
|
Zhang F, Chen B, Ren W, Yan Y, Zheng X, Jin S, Chang Y. Association analysis of dopaminergic degeneration and the neutrophil-to-lymphocyte ratio in Parkinson's disease. Front Aging Neurosci 2024; 16:1377994. [PMID: 38650864 PMCID: PMC11033456 DOI: 10.3389/fnagi.2024.1377994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Peripheral inflammatory responses are suggested to play a major role in the pathophysiology of Parkinson's disease (PD). The neutrophil-to-lymphocyte ratio (NLR), a new recognized biomarker, can reflect peripheral inflammation in PD. However, the association between the NLR and dopaminergic degeneration in PD remains unclear. Methods In this retrospective study, 101 enrolled PD patients were categorized into early-stage and advanced-stage PD based on the Hoehn and Yahr (HY) scale. We evaluated the clinical characteristics, peripheral immune profile, and 11C-CFT striatal dopamine transporter (DAT) binding levels. Linear regression analyses were employed to assess the associations between NLR and striatal DAT levels at different stages in PD patients. Results Covariate-controlled regression analysis revealed that higher NLR was significantly associated with lower DAT levels in the caudate (β = -0.27, p = 0.003) and the putamen (β = -0.27, p = 0.011). Moreover, in the early-stage PD subgroup, a similar association was observed (caudate: β = -0.37, p = 0.013; putamen: β = -0.45, p = 0.005). The lymphocytes count was correlated positively with the striatal DAT levels in the Spearman correlation analysis whether in total patients (caudate: ρ = 0.25, p = 0.013; putamen: ρ = 0.22, p = 0.026) or in the early-stage subgroup (caudate: ρ = 0.31, p = 0.023, putamen: ρ = 0.34, p = 0.011). Conclusion Dopaminergic degeneration is associated with peripheral inflammation in PD. The NLR, a widely used inflammatory marker, may have the potential to reflect the degree of dopaminergic degeneration in individuals with early-stage PD.
Collapse
Affiliation(s)
- Fengjiao Zhang
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Bin Chen
- Departments of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Wenhua Ren
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yayun Yan
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiaoqi Zheng
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Shuxian Jin
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ying Chang
- Departments of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
27
|
Jeon SJ, Chung KC. The SCF-FBW7β E3 ligase mediates ubiquitination and degradation of the serine/threonine protein kinase PINK1. J Biol Chem 2024; 300:107198. [PMID: 38508312 PMCID: PMC11026729 DOI: 10.1016/j.jbc.2024.107198] [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: 10/27/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
Understanding the mechanisms that govern the stability of functionally crucial proteins is essential for various cellular processes, development, and overall cell viability. Disturbances in protein homeostasis are linked to the pathogenesis of neurodegenerative diseases. PTEN-induced kinase 1 (PINK1), a protein kinase, plays a significant role in mitochondrial quality control and cellular stress response, and its mutated forms lead to early-onset Parkinson's disease. Despite its importance, the specific mechanisms regulating PINK1 protein stability have remained unclear. This study reveals a cytoplasmic interaction between PINK1 and F-box and WD repeat domain-containing 7β (FBW7β) in mammalian cells. FBW7β, a component of the Skp1-Cullin-1-F-box protein complex-type ubiquitin ligase, is instrumental in recognizing substrates. Our findings demonstrate that FBW7β regulates PINK1 stability through the Skp1-Cullin-1-F-box protein complex and the proteasome pathway. It facilitates the K48-linked polyubiquitination of PINK1, marking it for degradation. When FBW7 is absent, PINK1 accumulates, leading to heightened mitophagy triggered by carbonyl cyanide 3-chlorophenylhydrazone treatment. Moreover, exposure to the toxic compound staurosporine accelerates PINK1 degradation via FBW7β, correlating with increased cell death. This study unravels the intricate mechanisms controlling PINK1 protein stability and sheds light on the novel role of FBW7β. These findings deepen our understanding of PINK1-related pathologies and potentially pave the way for therapeutic interventions.
Collapse
Affiliation(s)
- Seo Jeong Jeon
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| |
Collapse
|
28
|
Tavi L, Penttilä N. Functional data analysis of prosodic prominence in Parkinson's disease: a pilot study. CLINICAL LINGUISTICS & PHONETICS 2024; 38:64-81. [PMID: 36636014 DOI: 10.1080/02699206.2022.2158372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/08/2022] [Accepted: 12/08/2022] [Indexed: 06/17/2023]
Abstract
This study aims to reveal dynamic changes in prosodic prominence patterns associated with Parkinson's disease (PD). To fulfill this purpose, the study proposes an exploratory methodology involving measuring a novel syllable-based prosody index (SPI) and performing functional principal component analyses (fPCAs) in a semi-automatic manner. First, SPI trajectories were collected from 31 speakers with PD before and after speech therapy and from 36 healthy controls. Then, the SPI trajectories were converted to continuous functions using B-splines. Finally, the functional SPIs were examined using fPCAs. The results showed that PD was associated with an increase of overall prominence for male speakers. The findings regarding higher prominence patterns in PD were supported by traditional phonetic measurements. For female speakers, however, there were no significant differences in prosodic prominence between speakers with PD and healthy controls. The results encourage to explore the proposed methodology also in analyses of other forms of atypical speech.
Collapse
Affiliation(s)
- Lauri Tavi
- School of Humanities, University of Eastern Finland, Joensuu, Finland
| | - Nelly Penttilä
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| |
Collapse
|
29
|
Unnithan D, Sartaj A, Iqubal MK, Ali J, Baboota S. A neoteric annotation on the advances in combination therapy for Parkinson's disease: nanocarrier-based combination approach and future anticipation. Part I: exploring theoretical insights and pharmacological advances. Expert Opin Drug Deliv 2024; 21:423-435. [PMID: 38481172 DOI: 10.1080/17425247.2024.2331214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is a neurological condition defined by a substantial reduction in dopamine-containing cells in the substantia nigra. Levodopa (L-Dopa) is considered the gold standard in treatment. Recent research has clearly shown that resistance to existing therapies can develop. Moreover, the involvement of multiple pathways in the nigrostriatal dopaminergic neuronal loss suggests that modifying the treatment strategy could effectively reduce this degeneration. AREAS COVERED This review summarizes the key concerns with treating PD patients and the combinations, aimed at effectively managing PD. Part I focuses on the clinical diagnosis at every stage of the disease as well as the pharmacological treatment strategies that are applied throughout its course. It methodically elucidates the potency of multifactorial interventions in attenuating the disease trajectory, substantiating the rationale for co-administration of dual or multiple therapeutic agents. Significant emphasis is laid on evidence-based pharmacological combinations for PD management. EXPERT OPINION By utilizing multiple drugs in a combination fashion, this approach can leverage the additive or synergistic effects of these agents, amplify the spectrum of treatment, and curtail the risk of side effects by reducing the dose of each drug, demonstrating significantly greater efficacy.
Collapse
Affiliation(s)
- Devika Unnithan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Kashif Iqubal
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Texas A&M University, College Station, TX, USA
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
30
|
Garg P, Würtz F, Hobbie F, Buttgereit K, Aich A, Leite K, Rehling P, Kügler S, Bähr M. Human serum-derived α-synuclein auto-antibodies mediate NMDA receptor-dependent degeneration of CNS neurons. J Neuroinflammation 2024; 21:62. [PMID: 38419079 PMCID: PMC10902935 DOI: 10.1186/s12974-024-03050-6] [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: 11/21/2023] [Accepted: 02/18/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Presence of autoantibodies against α-synuclein (α-syn AAb) in serum of the general population has been widely reported. That such peripheral factors may be involved in central nervous system pathophysiology was demonstrated by detection of immunoglobulins (IgGs) in cerebrospinal fluid and brain of Parkinson's disease (PD) patients. Thus, blood-borne IgGs may reach the brain parenchyma through an impaired blood-brain barrier (BBB). FINDINGS The present study aims to evaluate the patho-physiological impact of α-syn AAbs on primary brain cells, i.e., on spontaneously active neurons and on astrocytes. Exposure of neuron-astrocyte co-cultures to human serum containing α-syn AAbs mediated a dose-dependent reduction of spontaneous neuronal activity, and subsequent neurodegeneration. Removal specifically of α-syn AAbs from the serum prevented neurotoxicity, while purified, commercial antibodies against α-syn mimicked the neurodegenerative effect. Mechanistically, we found a strong calcium flux into neurons preceding α-syn AAbs-induced cell death, specifically through NMDA receptors. NMDA receptor antagonists prevented neurodegeneration upon treatment with α-syn (auto)antibodies. α-syn (auto)antibodies did not affect astrocyte survival. However, in presence of α-syn, astrocytes reacted to α-syn antibodies by secretion of the chemokine RANTES. CONCLUSION These findings provide a novel basis to explain how a combination of BBB impairment and infiltration of IgGs targeting synuclein may contribute to neurodegeneration in PD and argue for caution with α-syn immunization therapies for treatment of PD.
Collapse
Affiliation(s)
- Pretty Garg
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Göttingen, Germany
| | - Franziska Würtz
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Fabian Hobbie
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Klemens Buttgereit
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Abhishek Aich
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Göttingen, Germany
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Kristian Leite
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Peter Rehling
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Göttingen, Germany
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany.
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Göttingen, Germany
| |
Collapse
|
31
|
Marchesan E, Nardin A, Mauri S, Bernardo G, Chander V, Di Paola S, Chinellato M, von Stockum S, Chakraborty J, Herkenne S, Basso V, Schrepfer E, Marin O, Cendron L, Medina DL, Scorrano L, Ziviani E. Activation of Ca 2+ phosphatase Calcineurin regulates Parkin translocation to mitochondria and mitophagy in flies. Cell Death Differ 2024; 31:217-238. [PMID: 38238520 PMCID: PMC10850161 DOI: 10.1038/s41418-023-01251-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 02/09/2024] Open
Abstract
Selective removal of dysfunctional mitochondria via autophagy is crucial for the maintenance of cellular homeostasis. This event is initiated by the translocation of the E3 ubiquitin ligase Parkin to damaged mitochondria, and it requires the Serine/Threonine-protein kinase PINK1. In a coordinated set of events, PINK1 operates upstream of Parkin in a linear pathway that leads to the phosphorylation of Parkin, Ubiquitin, and Parkin mitochondrial substrates, to promote ubiquitination of outer mitochondrial membrane proteins. Ubiquitin-decorated mitochondria are selectively recruiting autophagy receptors, which are required to terminate the organelle via autophagy. In this work, we show a previously uncharacterized molecular pathway that correlates the activation of the Ca2+-dependent phosphatase Calcineurin to Parkin translocation and Parkin-dependent mitophagy. Calcineurin downregulation or genetic inhibition prevents Parkin translocation to CCCP-treated mitochondria and impairs stress-induced mitophagy, whereas Calcineurin activation promotes Parkin mitochondrial recruitment and basal mitophagy. Calcineurin interacts with Parkin, and promotes Parkin translocation in the absence of PINK1, but requires PINK1 expression to execute mitophagy in MEF cells. Genetic activation of Calcineurin in vivo boosts basal mitophagy in neurons and corrects locomotor dysfunction and mitochondrial respiratory defects of a Drosophila model of impaired mitochondrial functions. Our study identifies Calcineurin as a novel key player in the regulation of Parkin translocation and mitophagy.
Collapse
Affiliation(s)
| | - Alice Nardin
- Department of Biology, University of Padova, Padova, Italy
| | - Sofia Mauri
- Department of Biology, University of Padova, Padova, Italy
| | - Greta Bernardo
- Department of Biology, University of Padova, Padova, Italy
| | - Vivek Chander
- Department of Biology, University of Padova, Padova, Italy
| | - Simone Di Paola
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Napoli, Italy
| | | | | | | | | | | | - Emilie Schrepfer
- Department of Biology, University of Padova, Padova, Italy
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Oriano Marin
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Laura Cendron
- Department of Biology, University of Padova, Padova, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Elena Ziviani
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
32
|
Eser P, Kocabicak E, Bekar A, Temel Y. The interplay between neuroinflammatory pathways and Parkinson's disease. Exp Neurol 2024; 372:114644. [PMID: 38061555 DOI: 10.1016/j.expneurol.2023.114644] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Parkinson's disease, a progressive neurodegenerative disorder predominantly affecting elderly, is marked by the gradual degeneration of the nigrostriatal dopaminergic pathway, culminating in neuronal loss within the substantia nigra pars compacta (SNpc) and dopamine depletion. At the molecular level, neuronal loss in the SNpc has been attributed to factors including neuroinflammation, impaired protein homeostasis, as well as mitochondrial dysfunction and the resulting oxidative stress. This review focuses on the interplay between neuroinflammatory pathways and Parkinson's disease, drawing insights from current literature.
Collapse
Affiliation(s)
- Pinar Eser
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey.
| | - Ersoy Kocabicak
- Ondokuz Mayis University, Health Practise and Research Hospital, Neuromodulation Center, Samsun, Turkey
| | - Ahmet Bekar
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
33
|
Kang S, Noh Y, Oh SJ, Yoon HJ, Im S, Kwon HT, Pak YK. Neuroprotective Effects of Aldehyde-Reducing Composition in an LPS-Induced Neuroinflammation Model of Parkinson's Disease. Molecules 2023; 28:7988. [PMID: 38138478 PMCID: PMC10745824 DOI: 10.3390/molecules28247988] [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: 11/02/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease in which neuroinflammation and oxidative stress interact to contribute to pathogenesis. This study investigates the in vivo neuroprotective effects of a patented yeast extract lysate in a lipopolysaccharide (LPS)-induced neuroinflammation model. The yeast extract lysate, named aldehyde-reducing composition (ARC), exhibited potent antioxidant and anti-aldehyde activities in vitro. Oral administration of ARC at 10 or 20 units/kg/day for 3 days prior to intraperitoneal injection of LPS (10 mg/kg) effectively preserved dopaminergic neurons in the substantia nigra (SN) and striatum by preventing LPS-induced cell death. ARC also normalized the activation of microglia and astrocytes in the SN, providing further evidence for its neuroprotective properties. In the liver, ARC downregulated the LPS-induced increase in inflammatory cytokines and reversed the LPS-induced decrease in antioxidant-related genes. These findings indicate that ARC exerts potent antioxidant, anti-aldehyde, and anti-inflammatory effects in vivo, suggesting its potential as a disease-modifying agent for the prevention and treatment of neuroinflammation-related diseases, including Parkinson's disease.
Collapse
Affiliation(s)
- Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngjin Noh
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hung Taeck Kwon
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
34
|
Sands LP, Jiang A, Liebenow B, DiMarco E, Laxton AW, Tatter SB, Montague PR, Kishida KT. Subsecond fluctuations in extracellular dopamine encode reward and punishment prediction errors in humans. SCIENCE ADVANCES 2023; 9:eadi4927. [PMID: 38039368 PMCID: PMC10691773 DOI: 10.1126/sciadv.adi4927] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023]
Abstract
In the mammalian brain, midbrain dopamine neuron activity is hypothesized to encode reward prediction errors that promote learning and guide behavior by causing rapid changes in dopamine levels in target brain regions. This hypothesis (and alternatives regarding dopamine's role in punishment-learning) has limited direct evidence in humans. We report intracranial, subsecond measurements of dopamine release in human striatum measured, while volunteers (i.e., patients undergoing deep brain stimulation surgery) performed a probabilistic reward and punishment learning choice task designed to test whether dopamine release encodes only reward prediction errors or whether dopamine release may also encode adaptive punishment learning signals. Results demonstrate that extracellular dopamine levels can encode both reward and punishment prediction errors within distinct time intervals via independent valence-specific pathways in the human brain.
Collapse
Affiliation(s)
- L. Paul Sands
- Neuroscience Graduate Program, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Angela Jiang
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Brittany Liebenow
- Neuroscience Graduate Program, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Emily DiMarco
- Neuroscience Graduate Program, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Adrian W. Laxton
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Stephen B. Tatter
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - P. Read Montague
- Wellcome Centre for Human Neuroimaging, University College London, WC1N 3BG London, UK
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA
- Department of Physics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kenneth T. Kishida
- Neuroscience Graduate Program, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
35
|
Garmendia-Berges M, Sola-Sevilla N, Mera-Delgado MC, Puerta E. Age-Associated Changes of Sirtuin 2 Expression in CNS and the Periphery. BIOLOGY 2023; 12:1476. [PMID: 38132302 PMCID: PMC10741187 DOI: 10.3390/biology12121476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Sirtuin 2 (SIRT2), one of the seven members of the sirtuin family, has emerged as a potential regulator of aging and age-related pathologies since several studies have demonstrated that it shows age-related changes in humans and different animal models. A detailed analysis of the relevant works published to date addressing this topic shows that the changes that occur in SIRT2 with aging seem to be opposite in the brain and in the periphery. On the one hand, aging induces an increase in SIRT2 levels in the brain, which supports the notion that its pharmacological inhibition is beneficial in different neurodegenerative diseases. However, on the other hand, in the periphery, SIRT2 levels are reduced with aging while keeping its expression is protective against age-related peripheral inflammation, insulin resistance, and cardiovascular diseases. Thus, systemic administration of any known modulator of this enzyme would have conflicting outcomes. This review summarizes the currently available information on changes in SIRT2 expression in aging and the underlying mechanisms affected, with the aim of providing evidence to determine whether its pharmacological modulation could be an effective and safe pharmacological strategy for the treatment of age-related diseases.
Collapse
Affiliation(s)
- Maider Garmendia-Berges
- Pharmaceutical Sciences Department, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (M.G.-B.); (N.S.-S.); (M.M.-D.)
| | - Noemi Sola-Sevilla
- Pharmaceutical Sciences Department, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (M.G.-B.); (N.S.-S.); (M.M.-D.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - MCarmen Mera-Delgado
- Pharmaceutical Sciences Department, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (M.G.-B.); (N.S.-S.); (M.M.-D.)
| | - Elena Puerta
- Pharmaceutical Sciences Department, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (M.G.-B.); (N.S.-S.); (M.M.-D.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
36
|
Xu Y, Hong H, Lin X, Tong T, Zhang J, He H, Yang L, Mao G, Hao R, Deng P, Yu Z, Pi H, Cheng Y, Zhou Z. Chronic cadmium exposure induces Parkinson-like syndrome by eliciting sphingolipid disturbance and neuroinflammation in the midbrain of C57BL/6J mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122606. [PMID: 37742865 DOI: 10.1016/j.envpol.2023.122606] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Cadmium (Cd) is known as a widespread environmental neurotoxic pollutant. Cd exposure is recently recognized as an etiological factor of Parkinson's disease (PD) in humans. However, the mechanism underlying Cd neurotoxicity in relation to Parkinsonism pathogenesis is unclear. In our present study, C57BL/6 J mice were exposed to 100 mg/L CdCl2 in drinking water for 8 weeks. It was found Cd exposure caused motor deficits, decreased DA neurons and induced neuropathological changes in the midbrain. Non-targeted lipidomic analysis uncovered that Cd exposure altered lipid profile, increased the content of proinflammatory sphingolipid ceramides (Cer), sphingomyelin (SM) and ganglioside (GM3) in the midbrain. In consistency with increased proinflammatory lipids, the mRNA levels of genes encoding sphingolipids biosynthesis in the midbrain were dysregulated by Cd exposure. Neuroinflammation in the midbrain was evinced by the up-regulation of proinflammatory cytokines at mRNA and protein levels. Blood Cd contents and lipid metabolites in Parkinsonism patients by ICP-MS and LC-MS/MS analyses demonstrated that elevated blood Cd concentration and proinflammatory lipid metabolites were positively associated with the score of Unified Parkinson's Disease Rating Scale (UPDRS). 3 ceramide metabolites in the blood showed good specificity as the candidate biomarkers to predict and monitor Parkinsonism and Cd neurotoxicity (AUC>0.7, p < 0.01). In summary, our present study uncovered that perturbed sphingomyelin lipid metabolism is related to the Parkinsonism pathogenesis and Cd neurotoxicity, partially compensated for the deficiency in particular metabolic biomarkers for Parkinsonism in relation to Cd exposure, and emphasized the necessity of reducing Cd exposure at population level.
Collapse
Affiliation(s)
- Yudong Xu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Huihui Hong
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Xiqin Lin
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Tong
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Zhang
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haotian He
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Gaofeng Mao
- Neurology Department, General Hospital of Center Theater Command, Wuhan, China
| | - Rongrong Hao
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yong Cheng
- Neurology Department, General Hospital of Center Theater Command, Wuhan, China
| | - Zhou Zhou
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
37
|
Heo JY, Park AH, Lee MJ, Ryu MJ, Kim YK, Jang YS, Kim SJ, Shin SY, Son HJ, Stein TD, Huh YH, Chung SK, Choi SY, Kim JM, Hwang O, Shong M, Hyeon SJ, Lee J, Ryu H, Kim D, Kweon GR. Crif1 deficiency in dopamine neurons triggers early-onset parkinsonism. Mol Psychiatry 2023; 28:4474-4484. [PMID: 37648779 DOI: 10.1038/s41380-023-02234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.
Collapse
Affiliation(s)
- Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Ah Hyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yun Seon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - So Yeon Shin
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
- VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Yang Hoon Huh
- Electron Microscopy Research center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Sookja K Chung
- Faculty of Medicine & Dr Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Song Yi Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jin Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Minho Shong
- Graduate School of Medical Science and Education, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
38
|
Zhang Z, Shi M, Li Z, Ling Y, Zhai L, Yuan Y, Ma H, Hao L, Li Z, Zhang Z, Hölscher C. A Dual GLP-1/GIP Receptor Agonist Is More Effective than Liraglutide in the A53T Mouse Model of Parkinson's Disease. PARKINSON'S DISEASE 2023; 2023:7427136. [PMID: 37791037 PMCID: PMC10545468 DOI: 10.1155/2023/7427136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/07/2023] [Accepted: 09/02/2023] [Indexed: 10/05/2023]
Abstract
Parkinson's disease (PD) is a complex syndrome with many elements, such as chronic inflammation, oxidative stress, mitochondrial dysfunction, loss of dopaminergic neurons, build-up of alpha-synuclein (α-syn) in cells, and energy depletion in neurons, that drive the disease. We and others have shown that treatment with mimetics of the growth factor glucagon-like peptide 1 (GLP-1) can normalize energy utilization, neuronal survival, and dopamine levels and reduce inflammation. Liraglutide is a GLP-1 analogue that recently showed protective effects in phase 2 clinical trials in PD patients and in Alzheimer disease patients. We have developed a novel dual GLP-1/GIP receptor agonist that can cross the blood-brain barrier and showed good protective effects in animal models of PD. Here, we test liraglutide against the dual GLP-1/GIP agonist DA5-CH (KP405) in the A53T tg mouse model of PD which expresses a human-mutated gene of α-synuclein. Drug treatment reduced impairments in three different motor tests, reduced levels of α-syn in the substantia nigra, reduced the inflammation response and proinflammatory cytokine levels in the substantia nigra and striatum, and normalized biomarker levels of autophagy and mitochondrial activities in A53T mice. DA5-CH was superior in almost all parameters measured and therefore may be a better drug treatment for PD than liraglutide.
Collapse
Affiliation(s)
- Zijuan Zhang
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Ming Shi
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Zhengmin Li
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Yuan Ling
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Luke Zhai
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Ye Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - He Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Li Hao
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Zhonghua Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan, China
| |
Collapse
|
39
|
Toader C, Tataru CP, Florian IA, Covache-Busuioc RA, Dumitrascu DI, Glavan LA, Costin HP, Bratu BG, Ciurea AV. From Homeostasis to Pathology: Decoding the Multifaceted Impact of Aquaporins in the Central Nervous System. Int J Mol Sci 2023; 24:14340. [PMID: 37762642 PMCID: PMC10531540 DOI: 10.3390/ijms241814340] [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: 09/02/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Aquaporins (AQPs), integral membrane proteins facilitating selective water and solute transport across cell membranes, have been the focus of extensive research over the past few decades. Particularly noteworthy is their role in maintaining cellular homeostasis and fluid balance in neural compartments, as dysregulated AQP expression is implicated in various degenerative and acute brain pathologies. This article provides an exhaustive review on the evolutionary history, molecular classification, and physiological relevance of aquaporins, emphasizing their significance in the central nervous system (CNS). The paper journeys through the early studies of water transport to the groundbreaking discovery of Aquaporin 1, charting the molecular intricacies that make AQPs unique. It delves into AQP distribution in mammalian systems, detailing their selective permeability through permeability assays. The article provides an in-depth exploration of AQP4 and AQP1 in the brain, examining their contribution to fluid homeostasis. Furthermore, it elucidates the interplay between AQPs and the glymphatic system, a critical framework for waste clearance and fluid balance in the brain. The dysregulation of AQP-mediated processes in this system hints at a strong association with neurodegenerative disorders such as Parkinson's Disease, idiopathic normal pressure hydrocephalus, and Alzheimer's Disease. This relationship is further explored in the context of acute cerebral events such as stroke and autoimmune conditions such as neuromyelitis optica (NMO). Moreover, the article scrutinizes AQPs at the intersection of oncology and neurology, exploring their role in tumorigenesis, cell migration, invasiveness, and angiogenesis. Lastly, the article outlines emerging aquaporin-targeted therapies, offering a glimpse into future directions in combatting CNS malignancies and neurodegenerative diseases.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Calin Petru Tataru
- Department of Opthamology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Central Military Emergency Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan-Alexandru Florian
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (R.-A.C.-B.); (D.-I.D.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
40
|
Medeiros TB, Cosendey P, Gerin DR, de Sousa GF, Portal TM, Monteiro-de-Barros C. The effect of the sulfation patterns of dermatan and chondroitin sulfate from vertebrates and ascidians on their neuritogenic and neuroprotective properties. Int J Biol Macromol 2023; 247:125830. [PMID: 37454999 DOI: 10.1016/j.ijbiomac.2023.125830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Neurodegeneration is caused by the progressive loss of the structure and function of neurons, leading to cell death, and it is the main cause of many neurodegenerative diseases. Many molecules, such as glycosaminoglycans (GAGs), have been studied for their potential to prevent or treat these diseases. They are widespread in nature and perform an important role in neuritogenesis and neuroprotection. Here we investigated the neuritogenic and neuroprotective role of Phallusia nigra dermatan sulfate (PnD2,6S) and compared it with two distinct structures of chondroitin sulfate (C6S) and dermatan sulfate (D4S). For this study, a neuro 2A murine neuroblastoma cell line was used, and a chemical lesion was induced by the pesticide rotenone (ROT). We observed that PnD2,6S + ROT had a better neuritogenic effect than either C6S + ROT or D4S + ROT at a lower concentration (0.05 μg/mL). When evaluating the mitochondrial membrane potential, PnD2,6S showed a neuroprotective effect at a concentration of 0.4 μg/mL. These data indicate different mechanisms underlying this neuronal potential, in which the sulfation pattern is important for neuritogenic activity, while for neuroprotection all DS/CS structures had similar effects. This finding leads to a better understanding the chemical structures of PnD2,6S, C6S, and D4S and their therapeutic potential.
Collapse
Affiliation(s)
- Taiane Barreto Medeiros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Paloma Cosendey
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Diovana Ramos Gerin
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil
| | - Graziele Fonseca de Sousa
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil
| | - Taynan Motta Portal
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil
| | - Cintia Monteiro-de-Barros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade, NUPEM, Av. São José do Barreto, 764, Universidade Federal do Rio de Janeiro, Macaé CEP: 27910-970, RJ, Brazil; Programa de Pós-Graduação em Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, UENF, Av. Alberto Lamego, 2000, Campos dos Goytacazes CEP: 28013-602, RJ, Brazil.
| |
Collapse
|
41
|
Bellamri M, Brandt K, Cammerrer K, Syeda T, Turesky RJ, Cannon JR. Nuclear DNA and Mitochondrial Damage of the Cooked Meat Carcinogen 2-Amino-1-methyl-6-phenylimidazo[4,5- b]pyridine in Human Neuroblastoma Cells. Chem Res Toxicol 2023; 36:1361-1373. [PMID: 37421305 PMCID: PMC10626466 DOI: 10.1021/acs.chemrestox.3c00109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Animal fat and iron-rich diets are risk factors for Parkinson's disease (PD). The heterocyclic aromatic amines (HAAs) harman and norharman are neurotoxicants formed in many foods and beverages, including cooked meats, suggesting a role for red meat in PD. The structurally related carcinogenic HAAs 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), 2-amino-3,8-dimethylmidazo[4,5-f]quinoxaline (MeIQx), and 2-amino-9H-pyrido[2,3-b]indole (AαC) also form in cooked meats. We investigated the cytotoxicity, DNA-damaging potential, and mitochondrial damage of HAAs and their genotoxic HONH-HAA metabolites in galactose-dependent SH-SY5Y cells, a human neuroblastoma cell line relevant for PD-related neurotoxicity. All HAAs and HONH-HAAs induced weak toxicity except HONH-PhIP, which was 1000-fold more potent than the other chemicals. HONH-PhIP DNA adduct formation occurred at 300-fold higher levels than adducts formed with HONH-MeIQx and HONH-AαC, assuming similar cellular uptake rates. PhIP-DNA adduct levels occurred at concentrations as low as 1 nM and were threefold or higher and more persistent in mitochondrial DNA than nuclear DNA. N-Acetyltransferases (NATs), sulfotransferases, and kinases catalyzed PhIP-DNA binding and converted HONH-PhIP to highly reactive ester intermediates. DNA binding assays with cytosolic, mitochondrial, and nuclear fractions of SH-SY5Y fortified with cofactors revealed that cytosolic AcCoA-dependent enzymes, including NAT1, mainly carried out HONH-PhIP bioactivation to form N-acetoxy-PhIP, which binds to DNA. Furthermore, HONH-PHIP and N-acetoxy-PhIP inhibited mitochondrial complex-I, -II, and -III activities in isolated SH-SY5Y mitochondria. Mitochondrial respiratory chain complex dysfunction and DNA damage are major mechanisms in PD pathogenesis. Our data support the possible role of PhIP in PD etiology.
Collapse
Affiliation(s)
- Medjda Bellamri
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, Minnesota 55455, United States
| | - Kyle Brandt
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, Minnesota 55455, United States
| | - Kari Cammerrer
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, Minnesota 55455, United States
| | - Tauqeerunnisa Syeda
- School of Health Sciences, Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
| | - Robert J Turesky
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, Minnesota 55455, United States
| | - Jason R Cannon
- School of Health Sciences, Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
42
|
Cunha GM, Corso G, Lima MMS, Dos Santos Lima GZ. Electrophysiological damage to neuronal membrane alters ephaptic entrainment. Sci Rep 2023; 13:11974. [PMID: 37488148 PMCID: PMC10366241 DOI: 10.1038/s41598-023-38738-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023] Open
Abstract
The brain is commonly understood as a complex network system with a particular organization and topology that can result in specific electrophysiological patterns. Among all the dynamic elements resulting from the circuits of the brain's network, ephapticity is a cellular communication mechanism that has received little attention. To understand the network's properties of ephaptic entrainment, we start investigating the ephaptic effect on a single neuron. In this study, we used numerical simulations to examine the relationship between alterations in ephaptic neuronal entrainment and impaired electrophysiological properties of the neuronal membrane, which can occur via spike field coherence (SFC). This change in frequency band amplitude is observed in some neurodegenerative diseases, such as Parkinson's or Alzheimer's. To further investigate these phenomena, we proposed a damaged model based on the impairment of both the resistance of the ion channels and the capacitance of the lipid membrane. Therefore, we simulated ephaptic entrainment with the hybrid neural model quadratic integrate-and-fire ephaptic (QIF-E), which mimics an ephaptic entrainment generated by an LFP (simulate a neuronal group). Our results indicate a link between peak entrainment (ephapticity) preference and a shift in frequency band when damage occurs mainly in ion channels. Finally, we discuss possible relationships between ephaptic entrainment and neurodegenerative diseases associated with aging factors.
Collapse
Affiliation(s)
- Gabriel Moreno Cunha
- Departamento de Física Teórica e Experimental, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil
| | - Gilberto Corso
- Departamento de Física Teórica e Experimental, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil
| | - Marcelo M S Lima
- Departamento de Fisiologia, Universidade Federal do Paraná, Curitiba, PR, 81531-980, Brazil
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Gustavo Zampier Dos Santos Lima
- Departamento de Física Teórica e Experimental, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil.
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil.
- Escola de Ciências e Tecnologia, Universidade Federal do Rio Grande do Norte, Natal, RN, 59078-970, Brazil.
| |
Collapse
|
43
|
Kavungal D, Magalhães P, Kumar ST, Kolla R, Lashuel HA, Altug H. Artificial intelligence-coupled plasmonic infrared sensor for detection of structural protein biomarkers in neurodegenerative diseases. SCIENCE ADVANCES 2023; 9:eadg9644. [PMID: 37436975 PMCID: PMC10337894 DOI: 10.1126/sciadv.adg9644] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
Diagnosis of neurodegenerative disorders (NDDs) including Parkinson's disease and Alzheimer's disease is challenging owing to the lack of tools to detect preclinical biomarkers. The misfolding of proteins into oligomeric and fibrillar aggregates plays an important role in the development and progression of NDDs, thus underscoring the need for structural biomarker-based diagnostics. We developed an immunoassay-coupled nanoplasmonic infrared metasurface sensor that detects proteins linked to NDDs, such as alpha-synuclein, with specificity and differentiates the distinct structural species using their unique absorption signatures. We augmented the sensor with an artificial neural network enabling unprecedented quantitative prediction of oligomeric and fibrillar protein aggregates in their mixture. The microfluidic integrated sensor can retrieve time-resolved absorbance fingerprints in the presence of a complex biomatrix and is capable of multiplexing for the simultaneous monitoring of multiple pathology-associated biomarkers. Thus, our sensor is a promising candidate for the clinical diagnosis of NDDs, disease monitoring, and evaluation of novel therapies.
Collapse
Affiliation(s)
- Deepthy Kavungal
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Pedro Magalhães
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Senthil T. Kumar
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Rajasekhar Kolla
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Hatice Altug
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
44
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
45
|
Ma C, Feng Y, Li X, Sun L, He Z, Gan J, He M, Zhang X, Chen X. Potential Therapeutic Effects of Policosanol from Insect Wax on Caenorhabditis elegans Models of Parkinson's Disease. J Neuroimmune Pharmacol 2023; 18:127-144. [PMID: 36637699 DOI: 10.1007/s11481-022-10057-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 11/17/2022] [Indexed: 01/14/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. The standard treatments for PD focus on symptom relief rather than attempting to address the underlying degenerative processes completely. This study aimed to evaluate the potential therapeutic effects of policosanol derived from insect wax (PIW) by investigating improvements in disease symptoms represented in Caenorhabditis elegans models of PD. For our assessments, we used the following three models: NL5901, which is a transgenic model for α-synuclein aggregation; wild-type N2 induced with 6-hydroxydopamine (6-OHDA); and 6-OHDA-induced BZ555 as a model for loss of dopaminergic neurons (DNs). Specifically, we examined the effects of PIW treatment on α-synuclein aggregation, the loss of DNs, lipid abundance, and the lifespan of treated organisms. Further, we examined treatment-related changes in the levels of reactive oxygen species (ROS), malondialdehyde (MDA), adenosine triphosphate (ATP), glutathione S-transferase (GST), and superoxide dismutase (SOD), as well as the mRNA production profiles of relevant genes. A 10 µg/mL dose of PIW reduced the aggregation of α-synuclein in NL5901 and suppressed the loss of DNs in 6-OHDA-induced BZ555. Overall, PIW treatment decreased ROS and MDA levels, restored lipid abundance, and prolonged the lifespans of worms in all the three models, which may be associated with changes in the expression profiles of genes related to cell survival and oxidative stress response pathways. Our findings show that PIW alleviated the symptoms of PD in these models, possibly by regulating the stress responses initiated by injuries such as α-synuclein aggregation or 6-OHDA treatment.
Collapse
Affiliation(s)
- Chenjing Ma
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Ying Feng
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Xian Li
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Long Sun
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Zhao He
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Jin Gan
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| | - Minjie He
- Health Management Center, The First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan Province, China
| | - Xin Zhang
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China.
| | - Xiaoming Chen
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Panlong District, Kunming, 650224, Yunnan Province, China
| |
Collapse
|
46
|
Paccosi E, Proietti-De-Santis L. Parkinson's Disease: From Genetics and Epigenetics to Treatment, a miRNA-Based Strategy. Int J Mol Sci 2023; 24:ijms24119547. [PMID: 37298496 DOI: 10.3390/ijms24119547] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, characterized by an initial and progressive loss of dopaminergic neurons of the substantia nigra pars compacta via a potentially substantial contribution from protein aggregates, the Lewy bodies, mainly composed of α-Synuclein among other factors. Distinguishing symptoms of PD are bradykinesia, muscular rigidity, unstable posture and gait, hypokinetic movement disorder and resting tremor. Currently, there is no cure for PD, and palliative treatments, such as Levodopa administration, are directed to relieve the motor symptoms but induce severe side effects over time. Therefore, there is an urgency for discovering new drugs in order to design more effective therapeutic approaches. The evidence of epigenetic alterations, such as the dysregulation of different miRNAs that may stimulate many aspects of PD pathogenesis, opened a new scenario in the research for a successful treatment. Along this line, a promising strategy for PD treatment comes from the potential exploitation of modified exosomes, which can be loaded with bioactive molecules, such as therapeutic compounds and RNAs, and can allow their delivery to the appropriate location in the brain, overcoming the blood-brain barrier. In this regard, the transfer of miRNAs within Mesenchymal stem cell (MSC)-derived exosomes has yet to demonstrate successful results both in vitro and in vivo. This review, besides providing a systematic overview of both the genetic and epigenetic basis of the disease, aims to explore the exosomes/miRNAs network and its clinical potential for PD treatment.
Collapse
Affiliation(s)
- Elena Paccosi
- Unit of Molecular Genetics of Aging, Department of Ecology and Biology (DEB), University of Tuscia, 01100 Viterbo, Italy
| | - Luca Proietti-De-Santis
- Unit of Molecular Genetics of Aging, Department of Ecology and Biology (DEB), University of Tuscia, 01100 Viterbo, Italy
| |
Collapse
|
47
|
Brembati V, Faustini G, Longhena F, Bellucci A. Alpha synuclein post translational modifications: potential targets for Parkinson's disease therapy? Front Mol Neurosci 2023; 16:1197853. [PMID: 37305556 PMCID: PMC10248004 DOI: 10.3389/fnmol.2023.1197853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative disorder with motor symptoms. The neuropathological alterations characterizing the brain of patients with PD include the loss of dopaminergic neurons of the nigrostriatal system and the presence of Lewy bodies (LB), intraneuronal inclusions that are mainly composed of alpha-synuclein (α-Syn) fibrils. The accumulation of α-Syn in insoluble aggregates is a main neuropathological feature in PD and in other neurodegenerative diseases, including LB dementia (LBD) and multiple system atrophy (MSA), which are therefore defined as synucleinopathies. Compelling evidence supports that α-Syn post translational modifications (PTMs) such as phosphorylation, nitration, acetylation, O-GlcNAcylation, glycation, SUMOylation, ubiquitination and C-terminal cleavage, play important roles in the modulation α-Syn aggregation, solubility, turnover and membrane binding. In particular, PTMs can impact on α-Syn conformational state, thus supporting that their modulation can in turn affect α-Syn aggregation and its ability to seed further soluble α-Syn fibrillation. This review focuses on the importance of α-Syn PTMs in PD pathophysiology but also aims at highlighting their general relevance as possible biomarkers and, more importantly, as innovative therapeutic targets for synucleinopathies. In addition, we call attention to the multiple challenges that we still need to face to enable the development of novel therapeutic approaches modulating α-Syn PTMs.
Collapse
Affiliation(s)
| | | | | | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
48
|
Liu Z, Shen C, Li H, Tong J, Wu Y, Ma Y, Wang J, Wang Z, Li Q, Zhang X, Dong H, Yang Y, Yu M, Wang J, Zhou R, Fei J, Huang F. NOD-like receptor NLRC5 promotes neuroinflammation and inhibits neuronal survival in Parkinson's disease models. J Neuroinflammation 2023; 20:96. [PMID: 37072793 PMCID: PMC10111753 DOI: 10.1186/s12974-023-02755-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 03/02/2023] [Indexed: 04/20/2023] Open
Abstract
Parkinson's disease (PD) is mainly characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neuroinflammation mediated by overactivated microglia and astrocytes. NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) has been reported to participate in various immune disorders, but its role in neurodegenerative diseases remains unclear. In the current study, we found that the expression of NLRC5 was increased in the nigrostriatal axis of mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD, as well as in primary astrocytes, microglia and neurons exposed to different neurotoxic stimuli. In an acute MPTP-induced PD model, NLRC5 deficiency significantly reduced dopaminergic system degeneration and ameliorated motor deficits and striatal inflammation. Furthermore, we found that NLRC5 deficiency decreased the expression of the proinflammatory genes IL-1β, IL-6, TNF-α and COX2 in primary microglia and primary astrocytes treated with neuroinflammatory stimuli and reduced the inflammatory response in mixed glial cells in response to LPS treatment. Moreover, NLRC5 deficiency suppressed activation of the NF-κB and MAPK signaling pathways and enhanced the activation of AKT-GSK-3β and AMPK signaling in mixed glial cells. Furthermore, NLRC5 deficiency increased the survival of primary neurons treated with MPP+ or conditioned medium from LPS-stimulated mixed glial cells and promoted activation of the NF-κB and AKT signaling pathways. Moreover, the mRNA expression of NLRC5 was decreased in the blood of PD patients compared to healthy subjects. Therefore, we suggest that NLRC5 promotes neuroinflammation and dopaminergic degeneration in PD and may serve as a marker of glial activation.
Collapse
Affiliation(s)
- Zhaolin Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Heng Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jiabin Tong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yufei Wu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zishan Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Qing Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yufang Yang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Renyuan Zhou
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai, 201203, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
49
|
de Jager L, Vidigal CB, de Campos BH, Reginato GS, Fernandes LM, Ariza D, Higashi-Mckeown CM, Bertozzi MM, Rasquel de Oliveira FS, Verri Junior WA, Ceravolo GS, Crestani CC, Pinge-Filho P, Martins-Pinge MC. Role of the iNOS isoform in the cardiovascular dysfunctions of male rats with 6-OHDA-induced Parkinsonism. Nitric Oxide 2023; 134-135:49-60. [PMID: 37054808 DOI: 10.1016/j.niox.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
INTRODUCTION Available studies have shown the involvement of nitric oxide (NO) in the processes that lead to neurodegeneration in PD. Also, the use of inhibitors of the inducible isoform of NO-synthase (iNOS) promotes neuroprotection and attenuates dopamine (DA) loss in experimental models of Parkinsonism. In addition, NO also appears to be involved in cardiovascular changes in 6-hydroxydopamine (6-OHDA)-induced Parkinsonism. The current study aimed to evaluate the effects of iNOS inhibition on cardiovascular and autonomic function in animals that were subjected to Parkinsonism by the administration of 6-OHDA. MATERIALS AND METHODS The animals underwent stereotaxic surgery for bilateral microinfusion of the neurotoxin 6-OHDA (6 mg/mL in 0.2% ascorbic acid in sterile saline solution) or vehicle solution for the Sham group. From the day of stereotaxis until the day of femoral artery catheterization, the animals were treated with the iNOS inhibitor, S-methylisothiourea (SMT; 10 mg/kg; i.p.) or saline solution (0.9%; i.p.) for 7 days. The animals were divided into four groups: Sham-Saline, Sham-SMT, 6-OHDA-Saline, and 6-OHDA-SMT. Subsequent analyses were performed on these four groups. After 6 days, they underwent catheterization of the femoral artery, and 24 hours later, mean arterial pressure (MAP) and heart rate (HR) were recorded. Another group of animals (the 6-OHDA and Sham groups) was assessed for aortic vascular reactivity after 7 days of bilateral infusion of 6-OHDA or vehicle, in which cumulative concentration-effect curves (CCEC) were made for phenylephrine (Phenyl), acetylcholine and sodium nitroprusside (NPS). Also, CCEC in the presence of Nw-nitro-arginine-methyl-ester (l-NAME) (10-5 M), SMT (10-6 M), and indomethacin (10-5M) blockers were made. RESULTS The effectiveness of the 6-OHDA lesion was confirmed with the reduction of DA in 6-OHDA animals. However, treatment with SMT could not reverse the loss of DA. Concerning the baseline parameters, SBP and MAP values were lower in 6-OHDA animals compared to their Sham control, with no effect of treatment with SMT. In the analysis of SBP variability, a decrease in variance, the VLFabs component, and the LFabs component were observed in the 6-OHDA groups when compared to their controls, regardless of treatment with SMT. It was also observed that intravenous injections of SMT resulted in an increase in BP and a decrease in HR. However, the response was not different between the Sham and 6-OHDA groups. In vascular function, there was a hyporeactivity to Phenyl in the 6-OHDA group, and when investigating the mechanisms of this hyporeactivity, it was seen that the Rmax to Phenyl increased with incubation with SMT, indicating that iNOS could be involved in the vascular hyporeactivity of animals with Parkinsonism. CONCLUSION Thus, the set of results presented in this study suggests that part of the cardiovascular dysfunction in animals subjected to 6-OHDA Parkinsonism may be peripheral and involve the participation of endothelial iNOS.
Collapse
Affiliation(s)
- Lorena de Jager
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | - Camila Borecki Vidigal
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | - Blenda Hyedra de Campos
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | - Gabriela Souza Reginato
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | - Lorena Maria Fernandes
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | - Deborah Ariza
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | | | - Mariana Marques Bertozzi
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | | | | | | | - Carlos César Crestani
- Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Princípios Ativos Naturais e Toxicologia, Universidade Estadual Paulista, UNESP Araraquara, Brazil
| | - Phileno Pinge-Filho
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina - UEL, Londrina, PR, Brazil
| | | |
Collapse
|
50
|
The BDNF-TrkB signaling pathway is partially involved in the neuroprotective effects of hydrogen sulfide in Parkinson's disease. Eur J Pharmacol 2023; 944:175595. [PMID: 36804547 DOI: 10.1016/j.ejphar.2023.175595] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Recent studies have demonstrated that hydrogen sulfide (H2S) has a neuroprotective effect in neurodegenerative diseases. It is possible that this effect is supported by brain-derived neurotrophic factor (BDNF). Our aim is to examine the effects of H2S on neural damage in Parkinson's disease (PD) and to reveal the role of the BDNF-TrkB pathway in its possible effect. PD model was created with 1-methyl-phenyl-1,2,3,6-tetrahydropyridine (MPTP). C57BL/6 breed male mice were randomly divided into six groups: control, K252a, MPTP, MPTP + K252a, MPTP + NaHS, and MPTP + NaHS + K252a. TrkB receptor antagonist K252a and sodium hydrosulfide (NaHS) as a H2S donor were administered intraperitoneally. An increase was observed in the motor behavior tests in MPTP group, but NaHS treatment shortened the time spent on the balance beam and pole tests. It was also noticed that the BDNF-pathway played a role in the shortening of this period. Mice that received NaHS were found to have less MPTP-induced cellular damage. A positive effect of BDNF was also detected in the protection of these neurons. BDNF levels in the SN were significantly increased in MPTP group, compared to control group. Tissue CBS levels decreased in the groups that received K252a, compared to MPTP group. The findings of the present study display that the BDNF-TrkB pathway partially plays a role in the protective effect of H2S in the experimental mouse model of PD. This effect is probably due to changes in intracellular signaling pathways, rather than TrkB receptor expression.
Collapse
|