1
|
Alshimemeri S, Alhayssoni A, Hazazi R. Clinical predictors of freezing of gait in patients with Parkinson's disease: A systematic review. Clin Neurol Neurosurg 2025; 252:108848. [PMID: 40101322 DOI: 10.1016/j.clineuro.2025.108848] [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: 10/24/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Freezing of gait (FOG) is a debilitating motor symptom of Parkinson's disease (PD) that markedly impacts patients' quality of life. This review aims to identify clinical predictors of FOG to facilitate early prediction and future interventions. METHODS A systematic review adhering to PRISMA guidelines was conducted. Comprehensive searches in PubMed, EBSCO, and Web of Science yielded 1761 records. After removing duplicates, 1558 records were screened by title and abstract, and 92 full-text articles were assessed. Nine studies met the inclusion criteria and were qualitatively synthesized. RESULTS Our systematic review indicates that higher baseline MDS-UPDRS scores, which reflect greater disease severity, alongside elevated doses and early use of levodopa, are predictive of FOG in patients with PD. Additionally, higher Postural Instability and Gait Disorder (PIGD) scores, motor fluctuations, and lower limb disease onset further increase the risk of FOG. Other factors associated with an increased risk of FOG include older age, longer disease duration, anxiety, hyposmia, cognitive deficits, and sleep disorders. Furthermore, decreased step initiation duration when using visual cues serves as a predictor for the development of FOG. Early treatment with amantadine, selegiline, and dopamine agonists may help reduce the risk of developing FOG. CONCLUSION A combination of motor and non-motor factors predicts the development of FOG. Understanding FOG predictors is crucial for developing future therapeutics and personalized management plans, enabling targeted interventions and improved outcomes.
Collapse
Affiliation(s)
- Sohaila Alshimemeri
- Neurology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Abdulrahman Alhayssoni
- Neurology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Reema Hazazi
- Neurology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Xiang H. The interplay between α-synuclein aggregation and necroptosis in Parkinson's disease: a spatiotemporal perspective. Front Neurosci 2025; 19:1567445. [PMID: 40264913 PMCID: PMC12011736 DOI: 10.3389/fnins.2025.1567445] [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: 01/27/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the death of dopaminergic neurons and the aggregation of alpha-synuclein (α-Syn). It presents with prominent motor symptoms, and by the time of diagnosis, a significant number of neurons have already been lost. Current medications can only alleviate symptoms but cannot halt disease progression. Studies have confirmed that both dopaminergic neuronal loss and α-Syn aggregation are associated with necroptosis mechanisms. Necroptosis, a regulated form of cell death, has been recognized as an underexplored hotspot in PD pathogenesis research. In this review, we propose a spatiotemporal model of PD progression, highlighting the interactions between α-Syn aggregation, mitochondrial dysfunction, oxidative stress, neuroinflammation and necroptosis. These processes not only drive motor symptoms but also contribute to early non-motor symptoms, offering insights into potential diagnostic markers. Finally, we touch upon the therapeutic potential of necroptosis inhibition in enhancing current PD treatments, such as L-Dopa. This review aims to provide a new perspective on the pathogenesis of PD and to identify avenues for the development of more effective therapeutic strategies.
Collapse
Affiliation(s)
- Haoran Xiang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Department of Neurology, Yichang Central People’s Hospital, Yichang, Hubei, China
| |
Collapse
|
3
|
Li L, Gao W, Ren N, Chen L. IL-2/anti-IL-2 complexes attenuates neuroinflammation and neurodegeneration in mice of experimental Parkinson's disease. Brain Res Bull 2025; 223:111273. [PMID: 39999936 DOI: 10.1016/j.brainresbull.2025.111273] [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: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 02/27/2025]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, with motor and non-motor symptoms being its main clinical manifestations. Neuroinflammation has been shown to involve in pathogenesis of PD. Regulatory T cells (Tregs) in PD exhibited reduction in number and suppressive activity. Existing methods to increase the Tregs remains challenging for clinical application because of the difficulty in Tregs expanding or serious side-effects. Therefore, new approaches still need to be explored to balance the amount and activity of Tregs. In this study, we assessed the protective effects of IL-2/anti-IL-2 complexes (IL-2C) on mouse models of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). And the results showed that IL-2C significantly increased the number of Tregs both in spleen and brain, accompanied by reduced nigral dopaminergic neuron loss and behavioral defects. Besides, IL-2C also attenuated neuroinflammation as observed by diminished glial activation, fewer infiltration of CD4+ and CD8+ T cells and reduced pro-inflammatory cytokines releasing in the nigral region. Moreover, the protective effects of IL-2C were abolished by pre-treatment of anti-CD25 antibody (PC61), which was used to delete the Tregs. In summary, our results demonstrate that IL-2C-induced Tregs expansion attenuates the dopaminergic neurons loss and the neuroinflammatory response in vivo, suggesting that IL-2C maybe a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Lanxin Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China
| | - Weiwei Gao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China.
| | - Ning Ren
- Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China
| | - Lei Chen
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China.
| |
Collapse
|
4
|
Alshammari QA. Redox modulatory role of DJ-1 in Parkinson's disease. Biogerontology 2025; 26:81. [PMID: 40159591 DOI: 10.1007/s10522-025-10227-w] [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: 02/09/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
In particular, oxidative stress, generated by excessive reactive oxygen species (ROS), plays a major role in the neurodegenerative component of Parkinson's disease (PD) in aged neurons. DJ-1 (PARK7) is a key factor for maintaining redox homeostasis and modulation of mitochondrial function to preserve the cellular survival pathways. DJ-1 also plays a role in redox signaling independently of its antioxidant capacity by preventing the redox chain disulfide formation and stabilizing the master regulator of cellular antioxidant defense, Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2). In the DJ-1 or Nrf2 axis, expression of key antioxidant enzymes (glutathione peroxidase (GPx), superoxide dismutase (SOD), and heme oxygenase-1 (HO-1) in response to oxidative stress is increased, and decreased neuronal damage resulting from oxidative stress is achieved. It has been demonstrated that DJ-1 functions as an oxidative stress sensor, and mutations like L166P cause loss of antioxidant activity and increased Reactive Oxygen Species (ROS) accumulation with subsequent mitochondrial dysfunction in dopaminergic neurons. The highly conserved cysteine residue at position 106 (Cys106) of DJ-1 becomes stepwise oxidized (Cys-SOH → Cys-SO₂H → Cys-SO3H), functioning as a redox sensor as well as redox modulator of cellular stress responses. Furthermore, by protecting against α-synuclein aggregation, DJ-1 also protects in models lacking DJ-1, whereby DJ-1 deficiency promotes protein misfolding and neurotoxicity. In addition, DJ-1 participates in regulating neuroinflammation since its diminution provokes NF-κB-mediated exacerbation of proinflammatory cytokine production, leading to neuronal death. Oxidized DJ-1 (OxiDJ-1) is generated in aging brains, particularly in the substantia nigra (SN), and is correlated with PD progression both as a biomarker for disease monitoring and diagnosis of PD early in its course. The therapeutic strategies aimed at DJ-1 include small molecular activators, protein supplementation (Tat-DJ-1, ND-13), and gene therapy aiming to restore the neuroprotective function of DJ-1. Since DJ-1 is multitasking to protect neurons from oxidative damage, mitochondrial dysfunction, and even inflammation, it remains a promising therapeutic target. This review highlights the molecular mechanisms through which DJ-1 can protect from PD and aging-related neurodegeneration and has potential utility as a biomarker or therapeutic target.
Collapse
Affiliation(s)
- Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
5
|
Soni U, Singh K, Jain D, Pujari R. Exploring Alzheimer's disease treatment: Established therapies and novel strategies for future care. Eur J Pharmacol 2025; 998:177520. [PMID: 40097131 DOI: 10.1016/j.ejphar.2025.177520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by a gradual decline in cognitive function, memory impairment, and alterations in behavior. As the predominant etiology of dementia, AD affects millions of individuals worldwide, with its hallmark pathological feature being the accumulation of amyloid beta (Aβ) plaques, which disrupt neuronal function and progressively compromise brain structure. Early clinical manifestations often include forgetfulness, disorientation, and social withdrawal. Primarily impacting the elderly population, AD significantly impairs daily functioning and diminishes overall quality of life. Current therapeutic approaches for AD mainly focus on symptomatic relief and decelerating the disease's progression. Cholinesterase inhibitors, such as donepezil and rivastigmine, increase acetylcholine (ACh) levels to enhance cognitive function in individuals with mild to moderate AD. For individuals in more advanced stages of the disease, NMDA receptor antagonists modulate glutamate activity to mitigate excitotoxicity. In addition to pharmacological interventions, lifestyle modifications such as adherence to a balanced diet, regular physical activity, and cognitive engagement are advocated to support brain health. Novel therapeutic avenues are being explored to address underlying pathophysiological mechanisms, such as metal ion dysregulation within the brain. Furthermore, non-pharmacological approaches, including cognitive-behavioral therapy and patient support groups, provide essential behavioral and emotional support. Cutting-edge research continues to investigate innovative treatments, such as immunotherapies targeting amyloid plaques and tau tangles and neuroprotective compounds derived from natural sources. The goal of these multifaceted strategies is to alleviate symptoms, enhance quality of life, and offer hope for individuals and families affected by AD. This review provides a comprehensive summary of both established and emerging therapeutic interventions for the management of AD.
Collapse
Affiliation(s)
- Urvashi Soni
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Kothrud, Pune, 411023, Maharashtra, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Divya Jain
- Department of Microbiology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, 248007, Uttarakhand, India
| | - Rohini Pujari
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Kothrud, Pune, 411023, Maharashtra, India.
| |
Collapse
|
6
|
Zhao R, Jia N, Wu S, Wen J, Huang Y, Zhao C, Chen W. Therapeutic potential and limitation of condensed and hydrolyzed tannins in Parkinson's disease. Int J Biol Macromol 2025; 307:141814. [PMID: 40057098 DOI: 10.1016/j.ijbiomac.2025.141814] [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: 08/29/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
Parkinson's disease is a complex neurodegenerative disorder characterized by neuroinflammation, mitochondrial dysfunction, and the accumulation of misfolded proteins such as α-synuclein. This review explores the therapeutic potential of tannins, particularly proanthocyanidins and hydrolyzable tannins from grape seeds, in alleviating Parkinson's disease pathology. Condensed tannins exhibit significant antioxidant properties, can cross the blood-brain barrier, reduce oxidative stress, upregulate antioxidant proteins, and prevent neuronal apoptosis. Hydrolyzable tannins, through their unique chemical structure, further help reduce neuroinflammation and improve mitochondrial function. Both types of tannins can modulate inflammatory responses and enhance mitochondrial integrity, addressing key aspects of Parkinson's disease pathogenesis. Tannins possess excellent neuroprotective effects, representing a promising therapeutic approach. However, due to their chemical nature and structural characteristics, the bioavailability of tannins in the human body remains low. Current methods to enhance their bioavailability are limited. Further exploration is needed to improve their bioavailability and strengthen their potential clinical applications. Based on this, new Parkinson's disease treatment strategies can be developed, warranting in-depth research and clinical validation.
Collapse
Affiliation(s)
- Runfan Zhao
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Nan Jia
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shuyang Wu
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jiahui Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yajun Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Weichao Chen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
7
|
Elbatrawy A, Ademoye TA, Alnakhala H, Tripathi A, Zhu X, Plascencia-Villa G, Perry G, Dettmer U, Fortin JS. Inspecting the Triazole Scaffold as Powerful Antifibril Agents against 2N4R Tau and α-Synuclein Aggregates. ACS OMEGA 2025; 10:6721-6734. [PMID: 40028124 PMCID: PMC11866179 DOI: 10.1021/acsomega.4c08385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/22/2025] [Accepted: 01/29/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's (AD) and Parkinson's (PD) disease are neurodegenerative disorders that are considered to be a significant global health challenge due to their increasing prevalence and profound impact on both individuals and society. These disorders are characterized by the progressive loss of neuronal function, leading to cognitive and motor impairments. A key pathological feature of AD and PD is the abnormal accumulation of misfolded proteins within the brain. In AD, amyloid-beta aggregates into plaques, while tau proteins form neurofibrillary tangles (NFTs). Parkinson's disease, on the other hand, is marked by the accumulation of α-synuclein (α-syn) in the form of Lewy bodies (LBs). These protein aggregates are involved in neuronal dysfunction and neurodegeneration, contributing to disease progression. Research efforts are increasingly focused on identifying small molecules that can simultaneously target multiple pathological processes, offering the potential to not only alleviate symptoms but also modify the progression of neurodegeneration. Herein, a novel group of triazole-based compounds was designed and synthesized to curtail the aggregation of α-syn and tau proteins, which are closely linked to the physiopathology of PD and AD, respectively. A thioflavin T (ThT) fluorescence assay was used to measure fibril formation and assess the antiaggregation effects of various compounds. To further validate these findings, transmission electron microscopy (TEM) was employed as a direct method to visualize the impact of these compounds on fibril morphology. Inhibition of oligomer formation was evaluated using photoinduced cross-linking of unmodified proteins (PICUP), enabling the detection of early protein aggregation events. During fibril formation assays, three compounds (3e, 4b, 4d) demonstrated superior inhibitory activity as assessed by ThT fluorescence and TEM imaging. Subsequent evaluations, which included tests for antioligomer, anti-inclusion, and disaggregation effects identified compound 4d as the most promising candidate overall.
Collapse
Affiliation(s)
- Ahmed
A. Elbatrawy
- Department
of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907, United States
| | - Taiwo A. Ademoye
- Department
of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907, United States
| | - Heba Alnakhala
- Ann
Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Arati Tripathi
- Ann
Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Xiongwei Zhu
- Department
of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Germán Plascencia-Villa
- Department
of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - George Perry
- Department
of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Ulf Dettmer
- Ann
Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Jessica S. Fortin
- Department
of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
8
|
Liu N, Zhang T, Zhao W, Zhao X, Xue Y, Deng Q. Current trends in blood biomarkers detection and neuroimaging for Parkinson's disease. Ageing Res Rev 2025; 104:102658. [PMID: 39793764 DOI: 10.1016/j.arr.2025.102658] [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: 10/19/2024] [Revised: 12/01/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by both motor and cognitive impairments. A significant challenge in managing PD is the variability of symptoms and disease progression rates. This variability is primarily attributed to unclear biomarkers associated with the disease and the lack of early diagnostic technologies and effective imaging methods. PD-specific biomarkers are essential for developing practical tools that facilitate accurate diagnosis, patient stratification, and monitoring of disease progression. Hence, creating valuable tools for detecting and diagnosing PD based on specific biomarkers is imperative. Blood testing, less invasive than obtaining cerebrospinal fluid through a lumbar puncture, is an ideal source for these biomarkers. Although such biomarkers were previously lacking, recent advancements in various detection techniques related to PD biomarkers and new imaging methods have emerged. However, basic research requires more detailed guidelines on effectively implementing these biomarkers in diagnostic procedures to enhance the diagnostic accuracy of PD blood testing in clinical practice. This review discusses the developmental trends of PD-related blood biomarker detection technologies, including optical analysis platforms. Despite the progress in developing various biomarkers for PD, their specificity and sensitivity remain suboptimal. Therefore, the integration of multimodal biomarkers along with optical and imaging technologies is likely to significantly improve diagnostic accuracy and facilitate the implementation of personalized medicine. This review forms valid research hypotheses for PD research and guides future empirical studies.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Tianjiao Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Wei Zhao
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Xuechao Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Yuan Xue
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Qihong Deng
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
9
|
Ajala A, Asipita OH, Michael AT, Tajudeen MT, Abdulganiyyu IA, Ramu R. Therapeutic exploration potential of adenosine receptor antagonists through pharmacophore ligand-based modelling and pharmacokinetics studies against Parkinson disease. In Silico Pharmacol 2025; 13:17. [PMID: 39872470 PMCID: PMC11762050 DOI: 10.1007/s40203-025-00305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder that primarily affects persons aged 65 and older. It leads to a decline in motor function as a result of the buildup of abnormal protein deposits called Lewy bodies in the brain. Existing therapies exhibit restricted effectiveness and undesirable side effects. The objective was to discover potent medications that have demonstrated effectiveness in treating PD by employing computational methods. This work employed a comprehensive approach to evaluate 70 pyrimidine derivatives for their potential in treating PD. The evaluation involved the use of QSAR modelling, virtual screening, molecular docking, MD simulation, ADMET analysis, and antagonist inhibitor creation. Six compounds passed all the evaluation, while for MD simulation, carried out between the compound with best docking score and the reference drug, compound 57 was discovered to possess more stability compared to theophylline which is the reference drug, and it functions as a primary inhibitor of the adenosine A2A receptor. Additionally, the study determined that compound 57 satisfied the Rule of Five (Ro5) standards and exhibited robust physicochemical characteristics. The compound exhibited moderate to low levels of hERG inhibition. The conducted investigations highlighted promising outcomes of natural compounds that can orient towards the rational development of effective Parkinson's disease inhibitors. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00305-9.
Collapse
Affiliation(s)
- Abduljelil Ajala
- Department of Chemistry, Faculty of Physical Sciences, Ahmad Bello University, Zaria, Nigeria
| | - Otaru Habiba Asipita
- Department of Chemistry, Faculty of Physical Science, Nigerian Defence Academy Kaduna, Kaduna, Nigeria
| | | | - Murtala Taiwo Tajudeen
- Chemistry Department, School of Physical Science, Federal University of Technology, Minna, Niger, Nigeria
| | | | - Ramith Ramu
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysore, Karnataka 570015 India
| |
Collapse
|
10
|
Kim S, Jung UJ, Kim SR. Role of Oxidative Stress in Blood-Brain Barrier Disruption and Neurodegenerative Diseases. Antioxidants (Basel) 2024; 13:1462. [PMID: 39765790 PMCID: PMC11673141 DOI: 10.3390/antiox13121462] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Upregulation of reactive oxygen species (ROS) levels is a principal feature observed in the brains of neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). In these diseases, oxidative stress can disrupt the blood-brain barrier (BBB). This disruption allows neurotoxic plasma components, blood cells, and pathogens to enter the brain, leading to increased ROS production, mitochondrial dysfunction, and inflammation. Collectively, these factors result in protein modification, lipid peroxidation, DNA damage, and, ultimately, neural cell damage. In this review article, we present the mechanisms by which oxidative damage leads to BBB breakdown in brain diseases. Additionally, we summarize potential therapeutic approaches aimed at reducing oxidative damage that contributes to BBB disruption in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
11
|
Kuang Y, Mao H, Huang X, Chen M, Dai W, Gan T, Wang J, Sun H, Lin H, Liu Q, Yang X, Xu PY. α-Synuclein seeding amplification assays for diagnosing synucleinopathies: an innovative tool in clinical implementation. Transl Neurodegener 2024; 13:56. [PMID: 39574205 PMCID: PMC11580393 DOI: 10.1186/s40035-024-00449-2] [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: 07/26/2024] [Accepted: 10/25/2024] [Indexed: 11/25/2024] Open
Abstract
The spectrum of synucleinopathies, including Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), is characterized by α-synuclein (αSyn) pathology, which serves as the definitive diagnostic marker. However, current diagnostic methods primarily rely on motor symptoms that manifest years after the initial neuropathological changes, thereby delaying potential treatment. The symptomatic overlap between PD and MSA further complicates the diagnosis, highlighting the need for precise and differential diagnostic methods for these overlapping neurodegenerative diseases. αSyn misfolding and aggregation occur before clinical symptoms appear, suggesting that detection of pathological αSyn could enable early molecular diagnosis of synucleinopathies. Recent advances in seed amplification assay (SAA) offer a tool for detecting neurodegenerative diseases by identifying αSyn misfolding in fluid and tissue samples, even at preclinical stages. Extensive research has validated the effectiveness and reproducibility of SAAs for diagnosing synucleinopathies, with ongoing efforts focusing on optimizing conditions for detecting pathological αSyn in more accessible samples and identifying specific αSyn species to differentiate between various synucleinopathies. This review offers a thorough overview of SAA technology, exploring its applications for diagnosing synucleinopathies, addressing the current challenges, and outlining future directions for its clinical use.
Collapse
Affiliation(s)
- Yaoyun Kuang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hengxu Mao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaoyun Huang
- Houjie Hospital of Dongguan, Dongguan, 523000, China
| | - Minshan Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei Dai
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, 830054, Xinjiang, China
| | - Tingting Gan
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Jiaqi Wang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Sun
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hao Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qin Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xinling Yang
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
12
|
Sechi GP, Sechi MM. Small Molecules, α-Synuclein Pathology, and the Search for Effective Treatments in Parkinson's Disease. Int J Mol Sci 2024; 25:11198. [PMID: 39456980 PMCID: PMC11508228 DOI: 10.3390/ijms252011198] [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: 08/07/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive age-related neurodegenerative disorder affecting millions of people worldwide. Essentially, it is characterised by selective degeneration of dopamine neurons of the nigro-striatal pathway and intraneuronal aggregation of misfolded α-synuclein with formation of Lewy bodies and Lewy neurites. Moreover, specific small molecules of intermediary metabolism may have a definite pathophysiological role in PD. These include dopamine, levodopa, reduced glutathione, glutathione disulfide/oxidised glutathione, and the micronutrients thiamine and ß-Hydroxybutyrate. Recent research indicates that these small molecules can interact with α-synuclein and regulate its folding and potential aggregation. In this review, we discuss the current knowledge on interactions between α-synuclein and both the small molecules of intermediary metabolism in the brain relevant to PD, and many other natural and synthetic small molecules that regulate α-synuclein aggregation. Additionally, we analyse some of the relevant molecular mechanisms potentially involved. A better understanding of these interactions may have relevance for the development of rational future therapies. In particular, our observations suggest that the micronutrients ß-Hydroxybutyrate and thiamine might have a synergistic therapeutic role in halting or reversing the progression of PD and other neuronal α-synuclein disorders.
Collapse
Affiliation(s)
- Gian Pietro Sechi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | | |
Collapse
|
13
|
Kumar A, Gupta AK, Singh PK. Novel perspective of therapeutic modules to overcome motor and nonmotor symptoms in Parkinson's disease. AIMS Neurosci 2024; 11:312-340. [PMID: 39431269 PMCID: PMC11486614 DOI: 10.3934/neuroscience.2024020] [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: 05/18/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 10/22/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that involves the loss of dopaminergic neurons, which leads to motor and non-motor symptoms that have a significant impact. The pathophysiology of PD is complex and involves environmental and genetic factors that contribute to alpha-synuclein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. The current treatments of PD primarily focus on symptom management and have limitations in addressing disease progression and non-motor symptoms. Epidemiological data indicates a rise in PD cases worldwide, which highlights the need for effective treatments. Pathophysiological insights point out the involvement of various factors in PD progression, such as dopamine dysregulation, genetic mutations, oxidative stress, mitochondrial damage, alpha-synuclein aggregation, and neuroinflammation. Although current treatments, which include dopamine precursors, monoamine oxidase (MAO) inhibitors, and non-dopaminergic drugs, can alleviate motor symptoms, they are not effective in preventing disease progression or managing non-motor symptoms. Additionally, they can lead to adverse effects and become less effective over time. Novel therapeutic approaches, including cell-based therapies, gene therapies, targeted drug delivery therapies, and magnetic field therapies, are promising in improving symptom management and providing personalized treatment. Additionally, emerging therapies that target alpha-synuclein aggregation, mitochondrial dysfunction, and neuroinflammation may have potential disease-modifying effects. To sum up, for dealing with the multiple aspects of PD, there is a great need to come up with new and creative therapeutic approaches that not only relieve symptoms, but also prevent the progression of disease and non-motor symptoms. The progress made in comprehending the underlying mechanisms of PD provides optimism for developing successful treatments that can enhance the outcomes and quality of life.
Collapse
Affiliation(s)
- Anmol Kumar
- School of Pharmaceutical Science (formerly University Institute of Pharmacy), Chhatrapati Shahu Ji Maharaj University (formerly Kanpur University), Kanpur 208024, India
| | - Ajay Kumar Gupta
- School of Pharmaceutical Science (formerly University Institute of Pharmacy), Chhatrapati Shahu Ji Maharaj University (formerly Kanpur University), Kanpur 208024, India
| | | |
Collapse
|
14
|
Bankapalli K, Thomas RE, Vincow ES, Milstein G, Fisher LV, Pallanck LJ. A Drosophila model for mechanistic investigation of tau protein spread. Dis Model Mech 2024; 17:dmm050858. [PMID: 39350752 PMCID: PMC11463956 DOI: 10.1242/dmm.050858] [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: 04/22/2024] [Accepted: 08/09/2024] [Indexed: 10/09/2024] Open
Abstract
Brain protein aggregates are a hallmark of neurodegenerative disease. Previous work indicates that specific protein components of these aggregates are toxic, including tau (encoded by MAPT) in Alzheimer's disease and related tauopathies. Increasing evidence also indicates that these toxic proteins traffic between cells in a prion-like fashion, thereby spreading pathology from one brain region to another. However, the mechanisms involved in trafficking are poorly understood. We therefore developed a transgenic Drosophila model to facilitate rapid evaluation of candidate tau trafficking modifiers. Our model uses the bipartite Q system to drive co-expression of tau and GFP in the fly eye. We found age-dependent spread of tau into the brain, represented by detection of tau, but not of GFP. We also found that tau trafficking was attenuated upon inhibition of the endocytic factor dynamin (encoded by shi) or knockdown of glycogen synthase kinase-3β (GSK-3β, encoded by sgg). Further work revealed that dynamin promoted tau uptake in recipient tissues, whereas GSK-3β appeared to promote tau spread via direct phosphorylation of tau. Our robust and flexible system will promote the identification of tau-trafficking components involved in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Kondalarao Bankapalli
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| | - Ruth E. Thomas
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| | - Evelyn S. Vincow
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| | - Gillian Milstein
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| | - Laura V. Fisher
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| | - Leo J. Pallanck
- Department of Genome Sciences, University of Washington, 3720 15th Avenue NE, Seattle, WA98195, USA
| |
Collapse
|
15
|
He G, Huang X, Sun H, Xing Y, Gu S, Ren J, Liu W, Lu M. Gray matter volume alterations in de novo Parkinson's disease: A mediational role in the interplay between sleep quality and anxiety. CNS Neurosci Ther 2024; 30:e14867. [PMID: 39031989 PMCID: PMC11259571 DOI: 10.1111/cns.14867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/24/2024] [Accepted: 07/07/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is increasingly recognized for its non-motor symptoms, among which emotional disturbances and sleep disorders frequently co-occur. The commonality of neuroanatomical underpinnings for these symptoms is not fully understood. This study is intended to investigate the differences in gray matter volume (GMV) between PD patients with anxiety (A-PD) and those without anxiety (NA-PD). Additionally, it seeks to uncover the interplay between GMV variations and the manifestations of anxiety and sleep quality. METHODS A total of 37 A-PD patients, 43 NA-PD patients, and 36 healthy controls (HCs) were recruited, all of whom underwent voxel-based morphometry (VBM) analysis. Group differences in GMV were assessed using analysis of covariance (ANCOVA). Partial correlation between GMV, anxiety symptom, and sleep quality were analyzed. Mediation analysis explored the mediating role of the volume of GMV-distinct brain regions on the relationship between sleep quality and anxiety within the PD patient cohort. RESULTS A-PD patients showed significantly lower GMV in the fusiform gyrus (FG) and right inferior temporal gyrus (ITG) compared to HCs and NA-PD patients. GMV in these regions correlated negatively with Hamilton Anxiety Rating Scale (HAMA) scores (right ITG: r = -0.690, p < 0.001; left FG: r = -0.509, p < 0.001; right FG: r = -0.576, p < 0.001) and positively with sleep quality in PD patients (right ITG: r = 0.592, p < 0.001; left FG: r = 0.356, p = 0.001; right FG: r = 0.470, p < 0.001). Mediation analysis revealed that GMV in the FG and right ITG mediated the relationship between sleep quality and anxiety symptoms, with substantial effect sizes accounted for by the right ITG (25.74%) and FG (left: 11.90%, right: 15.59%). CONCLUSION This study has shed further light on the relationship between sleep disturbances and anxiety symptoms in PD patients. Given the pivotal roles of the FG and the ITG in facial recognition and the recognition of emotion-related facial expressions, our findings indicate that compromised sleep quality, under the pathological conditions of PD, may exacerbate the reduction in GMV within these regions, impairing the recognition of emotional facial expressions and thereby intensifying anxiety symptoms.
Collapse
Affiliation(s)
- Guixiang He
- Department of NeurologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
- The Yancheng School of Clinical Medicine of Nanjing Medical UniversityYancheng Third People's HospitalYanchengChina
- Jiangsu Key Laboratory of Neurodegeneration, Department of PharmacologyNanjing Medical UniversityNanjingChina
| | - Xiaofang Huang
- Department of NeurologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Haihua Sun
- The Yancheng School of Clinical Medicine of Nanjing Medical UniversityYancheng Third People's HospitalYanchengChina
| | - Yi Xing
- Department of NeurologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Siyu Gu
- The Yancheng School of Clinical Medicine of Nanjing Medical UniversityYancheng Third People's HospitalYanchengChina
| | - Jingru Ren
- Department of NeurologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Weiguo Liu
- Department of NeurologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of PharmacologyNanjing Medical UniversityNanjingChina
| |
Collapse
|
16
|
Al-Musawi I, Dennis BH, Clowry GJ, LeBeau FEN. Evidence for prodromal changes in neuronal excitability and neuroinflammation in the hippocampus in young alpha-synuclein (A30P) transgenic mice. FRONTIERS IN DEMENTIA 2024; 3:1404841. [PMID: 39081599 PMCID: PMC11285622 DOI: 10.3389/frdem.2024.1404841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/20/2024] [Indexed: 08/02/2024]
Abstract
Introduction Neuronal hyperexcitability and neuroinflammation are thought to occur at early stages in a range of neurodegenerative diseases. Neuroinflammation, notably activation of microglia, has been identified as a potential prodromal marker of dementia with Lewy bodies (DLB). Using a transgenic mouse model of DLB that over-expresses human mutant (A30P) alpha-synuclein (hα-syn) we have investigated whether early neuroinflammation is evident in the hippocampus in young pre-symptomatic animals. Methods Previous studies have shown early hyperexcitability in the hippocampal CA3 region in male A30P mice at 2-4 months of age, therefore, in the current study we have immunostained this region for markers of neuronal activity (c-Fos), reactive astrocytes (glial fibrillary acidic protein, GFAP), microglia (ionizing calcium binding adapter protein 1, Iba-1) and reactive microglia (inducible nitric oxide synthase, iNOS). Results We found an interesting biphasic change in the expression of c-Fos in A30P mice with high expression at 1 month, consistent with early onset of hyperexcitability, but lower expression from 2-4 months in male A30P mice compared to wild-type (WT) controls, possibly indicating chronic hyperexcitability. Neuroinflammation was indicated by significant increases in the % area of GFAP and the number of Iba-1+ cells that expressed iNOS immunoreactivity in the CA3 region in 2-4 months A30P male mice compared to WT controls. A similar increase in % area of GFAP was observed in female A30P mice, however, the Iba-1 count was not different between female WT and A30P mice. In WT mice aged 2-4 months only 4.6% of Iba-1+ cells co-expressed iNOS. In contrast, in age matched A30P mice 87% of cells co-expressed Iba-1 and iNOS. Although there was no difference in GFAP immunoreactivity at 1 month, Iba-1/iNOS co-expression was also increased in a cohort of 1 month old A30P mice. Discussion Abnormal hα-syn expression in A30P mice caused early changes in network excitability, as indicated by c-Fos expression, and neuroinflammation which might contribute to disease progression.
Collapse
Affiliation(s)
| | | | | | - Fiona E. N. LeBeau
- Biosciences Institute and Centre for Transformative Neuroscience, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
17
|
Bankapalli K, Thomas RE, Vincow ES, Milstein G, Fisher LV, Pallanck LJ. A Drosophila model for mechanistic investigation of tau protein spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.21.590466. [PMID: 38712083 PMCID: PMC11071371 DOI: 10.1101/2024.04.21.590466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Brain protein aggregates are a hallmark of neurodegenerative disease. Previous work indicates that specific protein components of these aggregates are toxic, including tau in Alzheimer's disease and related tauopathies. Increasing evidence also indicates that these toxic proteins traffic between cells in a prion-like fashion, thereby spreading pathology from one brain region to another. However, the mechanisms involved in trafficking are poorly understood. We therefore developed a transgenic Drosophila model to facilitate rapid evaluation of candidate tau trafficking modifiers. Our model uses the bipartite Q system to drive co-expression of tau and GFP in the fly eye. We find age-dependent tau spread into the brain, represented by detection of tau, but not GFP in the brain. We also found that tau trafficking was attenuated upon inhibition of the endocytic factor dynamin or the kinase glycogen synthase kinase-3β ( GSK-3β ). Further work revealed that dynamin promotes tau uptake in recipient tissues, whereas GSK-3β appears to promote tau spread via direct phosphorylation of tau. Our robust and flexible system will promote the identification of tau trafficking components involved in the pathogenesis of neurodegenerative diseases. SUMMARY STATEMENT The trafficking of toxic proteins in neurodegenerative disease is well-known but poorly understood. Our model will allow rapid and new insight into molecular mechanisms underlying this process.
Collapse
|
18
|
Soares ÉN, Costa ACDS, Ferrolho GDJ, Ureshino RP, Getachew B, Costa SL, da Silva VDA, Tizabi Y. Nicotinic Acetylcholine Receptors in Glial Cells as Molecular Target for Parkinson's Disease. Cells 2024; 13:474. [PMID: 38534318 PMCID: PMC10969434 DOI: 10.3390/cells13060474] [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: 01/30/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by resting tremor, bradykinesia, rigidity, and postural instability that also includes non-motor symptoms such as mood dysregulation. Dopamine (DA) is the primary neurotransmitter involved in this disease, but cholinergic imbalance has also been implicated. Current intervention in PD is focused on replenishing central DA, which provides remarkable temporary symptomatic relief but does not address neuronal loss and the progression of the disease. It has been well established that neuronal nicotinic cholinergic receptors (nAChRs) can regulate DA release and that nicotine itself may have neuroprotective effects. Recent studies identified nAChRs in nonneuronal cell types, including glial cells, where they may regulate inflammatory responses. Given the crucial role of neuroinflammation in dopaminergic degeneration and the involvement of microglia and astrocytes in this response, glial nAChRs may provide a novel therapeutic target in the prevention and/or treatment of PD. In this review, following a brief discussion of PD, we focus on the role of glial cells and, specifically, their nAChRs in PD pathology and/or treatment.
Collapse
Affiliation(s)
- Érica Novaes Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Ana Carla dos Santos Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Gabriel de Jesus Ferrolho
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
- Laboratory of Neurosciences, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Rodrigo Portes Ureshino
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema 09961-400, SP, Brazil
- Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil
| | - Bruk Getachew
- Department of Pharmacology, College of Medicine, Howard University, 520 W Street NW, Washington, DC 20059, USA
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
- Laboratory of Neurosciences, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, BA, Brazil
| | - Yousef Tizabi
- Department of Pharmacology, College of Medicine, Howard University, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
19
|
Iwaniak P, Owe-Larsson M, Urbańska EM. Microbiota, Tryptophan and Aryl Hydrocarbon Receptors as the Target Triad in Parkinson's Disease-A Narrative Review. Int J Mol Sci 2024; 25:2915. [PMID: 38474162 DOI: 10.3390/ijms25052915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
In the era of a steadily increasing lifespan, neurodegenerative diseases among the elderly present a significant therapeutic and socio-economic challenge. A properly balanced diet and microbiome diversity have been receiving increasing attention as targets for therapeutic interventions in neurodegeneration. Microbiota may affect cognitive function, neuronal survival and death, and gut dysbiosis was identified in Parkinson's disease (PD). Tryptophan (Trp), an essential amino acid, is degraded by microbiota and hosts numerous compounds with immune- and neuromodulating properties. This broad narrative review presents data supporting the concept that microbiota, the Trp-kynurenine (KYN) pathway and aryl hydrocarbon receptors (AhRs) form a triad involved in PD. A disturbed gut-brain axis allows the bidirectional spread of pro-inflammatory molecules and α-synuclein, which may contribute to the development/progression of the disease. We suggest that the peripheral levels of kynurenines and AhR ligands are strongly linked to the Trp metabolism in the gut and should be studied together with the composition of the microbiota. Such an approach can clearly delineate the sub-populations of PD patients manifesting with a disturbed microbiota-Trp-KYN-brain triad, who would benefit from modifications in the Trp metabolism. Analyses of the microbiome, Trp-KYN pathway metabolites and AhR signaling may shed light on the mechanisms of intestinal distress and identify new targets for the diagnosis and treatment in early-stage PD. Therapeutic interventions based on the combination of a well-defined food regimen, Trp and probiotics seem of potential benefit and require further experimental and clinical research.
Collapse
Affiliation(s)
- Paulina Iwaniak
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Maja Owe-Larsson
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland
- Laboratory of Center for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Ewa M Urbańska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|