1
|
Liang JY, Yuan XL, Jiang JM, Zhang P, Tan K. Targeting the NLRP3 inflammasome in Parkinson's disease: From molecular mechanism to therapeutic strategy. Exp Neurol 2025; 386:115167. [PMID: 39884329 DOI: 10.1016/j.expneurol.2025.115167] [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: 12/07/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Parkinson's disease is the second most common neurodegenerative disease, characterized by substantial loss of dopaminergic (DA) neurons, the formation of Lewy bodies (LBs) in the substantia nigra, and pronounced neuroinflammation. The nucleotide-binding domain like leucine-rich repeat- and pyrin domain-containing protein 3 (NLRP3) inflammasome is one of the pattern recognition receptors (PRRs) that function as intracellular sensors in response to both pathogenic microbes and sterile triggers associated with Parkinson's disease. These triggers include reactive oxygen species (ROS), misfolding protein aggregation, and potassium ion (K+) efflux. Upon activation, it recruits and activates caspase-1, then processes the pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18, which mediate neuroinflammation in Parkinson's disease. In this review, we provide a comprehensive overview of NLRP3 inflammasome, detailing its structure, activation pathways, and the factors that trigger its activation. We also explore the pathological mechanisms by which NLRP3 contributes to Parkinson's disease and discuss potential strategies for targeting NLRP3 as a therapeutic approach.
Collapse
Affiliation(s)
- Jin-Yu Liang
- Department of Clinical Laboratory Medicine, Zhuzhou Kind Cardiovascular Disease Hospital, Hunan Province, China
| | - Xiao-Lei Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jia-Mei Jiang
- Institute of Neurology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan, PR China
| | - Ping Zhang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan, PR China
| | - Kuang Tan
- Department of Clinical Laboratory Medicine, Zhuzhou Kind Cardiovascular Disease Hospital, Hunan Province, China.
| |
Collapse
|
2
|
Pislyagin EA, Tarbeeva DV, Yurchenko EA, Menchinskaya ES, Gorpenchenko TY, Pokhilo ND, Kalinovskiy AI, Aminin DL, Fedoreyev SA. Neuroprotective Activity of Oligomeric Stilbenes from Alpha Grape Stems in In Vitro Models of Parkinson's Disease. Int J Mol Sci 2025; 26:2411. [PMID: 40141054 PMCID: PMC11942555 DOI: 10.3390/ijms26062411] [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: 01/27/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigated the neuroprotective activity of oligomeric stilbenes (OSs) derived from Alpha grape stems in various in vitro models of Parkinson's disease (PD). Using neurotoxin-induced cellular models, including 1-methyl-4-phenylpyridine (MPP+), paraquat (PQ), 6-hydroxydopamine (6-OHDA), and rotenone, we screened the cytoprotective effects of ampelopsin A (1), ε-viniferin (2), vitisin D (3), vitisin A (4), α-viniferin (5), trans-vitisin B (6), cis-vitisin B (7), and melanoxylin A (8). The results demonstrate that certain stilbenes significantly enhanced cell viability and reduced reactive oxygen species (ROS) levels in neurotoxin-treated Neuro-2a cells. Notably, vitisin A and trans-vitisin B exhibited promising neuroprotective properties by decreasing mitochondrial ROS and cardiolipin peroxidation. This study highlights the potential of these compounds in mitigating oxidative stress and inflammation associated with PD. Additionally, we provided new insights into the antioxidant mechanisms of these stilbenes, including their direct ROS-scavenging abilities. Our findings contribute to the understanding of oligomeric stilbenes as potential therapeutic agents for the prevention and treatment of neurodegenerative diseases, particularly those associated with oxidative damage. Further research is warranted to explore its clinical applications and underlying mechanisms of action.
Collapse
Affiliation(s)
- Evgeny A. Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Darya V. Tarbeeva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Ekaterina A. Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Ekaterina S. Menchinskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Tatiana Y. Gorpenchenko
- Federal Scientific Center of the East Asia Terrestrial Biodiversity, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia;
| | - Natalya D. Pokhilo
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Anatoly I. Kalinovskiy
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| | - Dmitry L. Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shin-Chuan 1st Road, Sanmin District, Kaohsiung City 80708, Taiwan
| | - Sergey A. Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690022, Russia; (D.V.T.); (E.A.Y.); (E.S.M.); (N.D.P.); (A.I.K.); (D.L.A.); (S.A.F.)
| |
Collapse
|
3
|
Sabnam S, Kumar R, Pranav. Biofunctionalized nanomaterials for Parkinson's disease theranostics: potential for efficient PD biomarker detection and effective therapy. Biomater Sci 2025. [PMID: 40036044 DOI: 10.1039/d5bm00179j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
α-Synuclein (α-Syn) is a primary pathological indicator for Parkinson's disease (PD). The α-Syn oligomer is even more toxic and is responsible for PD. Hence, identifying α-Syn and its oligomers is an interesting approach to diagnosing PD. The prevention strategies for oligomer formation could be therapeutic in treating PD. Various conventional strategies have been developed for the management of PD. However, their clinical applications are limited due to toxicity, off-targeting, side effects, and poor bioavailability. Recently, nanomaterials have gained significant attention due to unique physicochemical characteristics such as nanoscale size, large surface area, flexibility of functionalization, and ability to protect and control a loaded payload. Functionalizing the surface of nanoparticles with a desired targeting agent could offer targeted delivery of the payload at the site of action due to specificity and selectivity against complementary molecules. Among various functionalization approaches, biomolecule-functionalized nanomaterials offer benefits such as enhanced bioavailability, improved internalization into target cells through receptor-mediated endocytosis, and delivery of therapeutics across the BBB (blood-brain barrier). In this review, we initially discussed the major milestones related to PD and highlighted the therapeutic strategies focused on clinical trials. The strategies of biomolecule functionalization of nanomaterials and their application in detecting and preventing α-Syn oligomer for the diagnosis and therapy of PD, respectively, have been reviewed comprehensively. Ultimately, we have outlined the conclusions, highlighted the limitations and challenges, and provided insight into future perspectives and alternative approaches that must be investigated.
Collapse
Affiliation(s)
- Saheli Sabnam
- Centre for Nanosciences, Indian Institute of Technology Kanpur, India-208016
| | - Raj Kumar
- University Center for Research and Development, Chandigarh University, Gharuan, Punjab-140413, India.
| | - Pranav
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore Campus, India-632014.
| |
Collapse
|
4
|
Begley E, Thomas JM, Hind W, Senior C. Can behavioural science be used to understand factors that influence the prescription choice for Parkinson's disease? A pan-European focus group study of clinicians' prescribing practice. BMJ Open 2025; 15:e090018. [PMID: 39971601 PMCID: PMC11840894 DOI: 10.1136/bmjopen-2024-090018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/02/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES This study aimed to establish a consensus on key factors that influence medication choices for Parkinson's disease and to identify the behavioural determinants of these factors using behavioural change theory as a theoretical lens. DESIGN This qualitative study used the nominal group technique to conduct structured online focus group meetings. A facilitator guided participants to (1) individually generate a list of factors that influence their decision to prescribe, (2) collectively share these factors, (3) refine and clarify factors and (4) rank the most important factors. Subsequently, the most important factors identified were mapped to the Theoretical Domains Framework (TDF) and the Capability, Opportunity, Motivation-Behaviour (COM-B) model to identify the behavioural determinants that influence medication choice. PARTICIPANTS Eighteen healthcare professionals, including neurologists, consultants and specialist nurses/practitioners who prescribe medication, were recruited across Europe and participated in one of seven focus groups. RESULTS There was good consensus among the participants about which factors influence their prescribing decisions. Overall, participants identified 60 unique factors that were broadly categorised into the following themes: medical or symptom concern, patient characteristics, side effects, access to treatment, clinical guidelines, social support and patient preference. Factors discussed and prioritised by the participants aligned with seven of the 14 TDF domains: knowledge; memory, attention and decision processes; beliefs about consequences; goals; social/professional role and identity; environment context and resources; and social influences. Together, these were subsequently mapped onto four of the six subdomains of the COM-B model: psychological capability, reflective motivation, physical opportunity and social opportunity. CONCLUSIONS These findings suggest that prescribing decisions for Parkinson's disease are determined by a complex range of factors linked to the COM-B components capability, motivation and opportunity. These can be further understood by specific behavioural domains, as identified by the TDF, which should be targeted to help optimise subsequent prescribing decisions.
Collapse
Affiliation(s)
- Emma Begley
- School of Psychology, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Jason Michael Thomas
- School of Psychology, College of Health and Life Sciences, Aston University, Birmingham, UK
| | | | - Carl Senior
- School of Psychology, College of Health and Life Sciences, Aston University, Birmingham, UK
| |
Collapse
|
5
|
Lou L, Xiang C, Hu Y, Yang J. Tai Chi improves balance, mobility and gait function of the lower limbs in patients with Parkinson's disease: a systematic review and meta-analysis. Eur J Med Res 2025; 30:107. [PMID: 39962570 PMCID: PMC11831811 DOI: 10.1186/s40001-024-02151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/08/2024] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVE This study aimed to systematically evaluate the effect of Tai Chi on the balance function, mobility function, and gait function (including gait endurance, gait amplitude, and gait speed) of the lower limbs in patients with Parkinson's disease. METHODS The "PICOS" principle was used to search seven English and six Chinese databases. Search for relevant studies published up to September 11, 2023, from the date of database creation. Two authors independently screened all eligible studies. The included studies were assessed for risk of bias based on the criteria outlined in Cochrane Handbook 5.1.0. Quantitative statistics and meta-analyses were conducted using Review Manager 5.4 software. RESULTS Th 18 studies that met the inclusion criteria, 16 of which were included in the meta-analysis, included a sample of 963 cases from four countries. The meta-analysis revealed that Tai Chi was effective compared with conventional medication in improving balance function (MD = 2.06, 95% CI [1.35, 2.78], P < 0.00001, I2 =0%), mobility function (MD = - 1.59, 95% CI [- 2.28, - 0.91], P < 0.00001, I2 =14%), and gait speed (SMD =0.59, 95% [0.28, 0.91], P = 0.0002, I2 =29%). However, there was no significant improvement in gait endurance (SMD = 0.14, 95% CI [-0.41, 0.68], P =0.62, I2 = 0%) or gait amplitude (SMD =0.30 , 95% CI [- 0.00, 0.61], P = 0.05, I2 =0%). Compared with other exercise therapies, Tai Chi was significantly superior in improving balance function (MD = 3.05, 95% CI [1.94, 4.16], P <0.00001, I2 = 38%), mobility function (MD =-0.70, 95%CI [- 1.23, - 0.17], P = 0.01, I2 = 0%), and gait range (SMD = 0.36, 95% CI [0.14, 0.58], P = 0.002, I2 = 45%). However, the advantage of improving gait speed (SMD =0.00, 95% CI [- 0.28, 0.29], P = 0.98, I2 =0%) was uncertain. CONCLUSION Tai chi is a safe and effective rehabilitation intervention that can enhance the balance and mobility functions of the lower limbs in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Lijie Lou
- Graduate School, Guangzhou Sport University, Guangzhou, 510500, China
- Martial Arts Research Institute, Guangzhou Sport University, Guangzhou, 510500, China
| | - Chunyu Xiang
- School of Sports and Health, Guangdong Institute of Science and Technology, Zhuhai, 519090, China
| | - Yingliang Hu
- Graduate School, Guangzhou Sport University, Guangzhou, 510500, China
| | - Jiapeng Yang
- Graduate School, Guangzhou Sport University, Guangzhou, 510500, China.
- Martial Arts Research Institute, Guangzhou Sport University, Guangzhou, 510500, China.
| |
Collapse
|
6
|
Wang J, Liu M, Zhao J, Hu P, Gao L, Tian S, Zhang J, Liu H, Xu X, He Z. Oxidative stress and dysregulated long noncoding RNAs in the pathogenesis of Parkinson's disease. Biol Res 2025; 58:7. [PMID: 39871377 PMCID: PMC11770960 DOI: 10.1186/s40659-025-00585-7] [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: 06/15/2023] [Accepted: 01/07/2025] [Indexed: 01/29/2025] Open
Abstract
Parkinson's disease (PD) is a progressive age-related neurodegenerative disease whose annual incidence is increasing as populations continue to age. Although its pathogenesis has not been fully elucidated, oxidative stress has been shown to play an important role in promoting the occurrence and development of the disease. Long noncoding RNAs (lncRNAs), which are more than 200 nucleotides in length, are also involved in the pathogenesis of PD at the transcriptional level via epigenetic regulation, or at the post-transcriptional level by participating in physiological processes, including aggregation of the α-synuclein, mitochondrial dysfunction, oxidative stress, calcium stabilization, and neuroinflammation. LncRNAs and oxidative stress are correlated during neurodegenerative processes: oxidative stress affects the expression of multiple lncRNAs, while lncRNAs regulate many genes involved in oxidative stress responses. Oxidative stress and lncRNAs also affect other processes associated with neurodegeneration, including mitochondrial dysfunction and increased neuroinflammation that lead to neuronal death. Therefore, modulating the levels of specific lncRNAs may alleviate pathological oxidative damage and have neuroprotective effects. This review discusses the general mechanisms of oxidative stress, pathological mechanism underlying the role of oxidative stress in the pathogenesis of PD, and teases out the mechanisms through which lncRNAs regulate oxidative stress during PD pathogenesis, as well as identifies the possible neuroprotective mechanisms of lncRNAs. Reviewing published studies will help us further understand the mechanisms underlying the role of lncRNAs in the oxidative stress process in PD and to identify potential therapeutic strategies for PD.
Collapse
Affiliation(s)
- Jialu Wang
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Meitong Liu
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jiuhan Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Pan Hu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Lianbo Gao
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Shen Tian
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jin Zhang
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Huayan Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiaoxue Xu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| | - Zhenwei He
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
7
|
Goto S. Functional pathology of neuroleptic-induced dystonia based on the striatal striosome-matrix dopamine system in humans. J Neurol Neurosurg Psychiatry 2025; 96:177-183. [PMID: 39631787 DOI: 10.1136/jnnp-2024-334545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Neuroleptic-induced dystonia is a source of great concern in clinical practice because of its iatrogenic nature which can potentially lead to life-threatening conditions. Since all neuroleptics (antipsychotics) share the ability to block the dopamine D2-type receptors (D2Rs) that are highly enriched in the striatum, this drug-induced dystonia is thought to be caused by decreased striatal D2R activity. However, how associations of striatal D2R inactivation with dystonia are formed remains elusive.A growing body of evidence suggests that imbalanced activities between D1R-expressing medium spiny neurons and D2R-expressing medium spiny neurons (D1-MSNs and D2-MSNs) in the striatal striosome-matrix system underlie the pathophysiology of various basal ganglia disorders including dystonia. Given the specificity of the striatal dopamine D1 system in 'humans', this article highlights the striatal striosome hypothesis in causing 'repetitive' and 'stereotyped' motor symptoms which are key clinical features of dystonia. It is suggested that exposure to neuroleptics may reduce striosomal D1-MSN activity and thereby cause dystonia symptoms. This may occur through an increase in the striatal cholinergic activity and the collateral inhibitory action of D2-MSNs onto neighbouring D1-MSNs within the striosome subfields. The article proposes a functional pathology of the striosome-matrix dopamine system for neuroleptic-induced acute dystonia or neuroleptic-withdrawal dystonia. A rationale for the effectiveness of dopaminergic or cholinergic pharmacotherapy is also provided for treating dystonias. This narrative review covers various aspects of the relevant field and provides a detailed discussion of the mechanisms of neuroleptic-induced dystonia.
Collapse
Affiliation(s)
- Satoshi Goto
- Research Organization of Science and Technology, Ritsumeikan University, Kyoto, Japan
| |
Collapse
|
8
|
Alotaibi S, Alfayez L, Alkhudhair M. Parkinson's Disease: Current Treatment Modalities and Emerging Therapies. Cureus 2024; 16:e75647. [PMID: 39803037 PMCID: PMC11725288 DOI: 10.7759/cureus.75647] [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] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Herein, we review the literature on Parkinson's disease (PD) management and summarize the progress in medical, surgical, and assisted therapeutic modalities for motor and non-motor symptoms. A thorough search strategy was implemented to retrieve all relevant articles and identify the best evidence from different databases including Scopus, PubMed, Google Scholar, the Cochrane Database of Systematic Reviews, and Evidence-Based Medicine reviews from the International Parkinson and Movement Disorder Society. Multiple terms, such as Parkinson, tremor, predominant, Parkinson management, deep brain stimulation, LCIG, ablative surgery for PD, medical management of PD, and assistive devices for PD, were used for screening. A total of 160 articles were gathered; irrelevant papers and older articles were excluded. After initial exclusion, we had 140 articles to review from 1980 to 2022. Five articles were found to be duplicated, and another five articles were excluded as they did not have additional information on management that could be used in this research paper. We found that management options and assistive devices for PD are improving, with new therapeutic options emerging every year. Medical therapy is the most common therapy as it corrects dopamine deficiency which is the main factor implicated in PD; other surgical treatment options are available in cases of chronic progressive disease course. This article adds significant value to the literature as it includes the history and the role of most Parkinson's disease management options.
Collapse
Affiliation(s)
- Shabab Alotaibi
- Neurology, Movement Disorder and Neuromodulation, King Saud Medical City, Riyadh, SAU
| | - Lujain Alfayez
- Neurology, Neurology Center, Prince Sultan Military Medical City, Riyadh, SAU
| | | |
Collapse
|
9
|
Huang CH, Lin SK, Lin MC, Hung SY. Reducing Parkinson's disease incidence in patients with insomnia through acupuncture: A cohort study. Integr Med Res 2024; 13:101083. [PMID: 39635074 PMCID: PMC11616590 DOI: 10.1016/j.imr.2024.101083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 12/07/2024] Open
Abstract
Background Parkinson's disease (PD) is a prevalent neurodegenerative condition characterized primarily by motor symptoms, often accompanied by non-motor manifestations such as insomnia. Acupuncture, an increasingly popular alternative therapy, has shown promise in the prevention and alleviation of PD motor symptoms. However, the specific impact of acupuncture on the risk of developing PD in insomnia patients remains uncertain. Methods In this retrospective study, we identified and matched 152,585 newly diagnosed insomnia patients from Taiwan's National Health Insurance Research Database (NHIRD) from 2000 to 2010. Using a 1:1 propensity score matching method, we ensured the comparability of two groups: patients who received acupuncture treatment and those who did not. It resulted in a final cohort of 20,112 patients in both the acupuncture and non-acupuncture groups. Results Our analysis revealed that insomnia patients who underwent acupuncture treatment exhibited a significantly reduced risk of developing PD. The adjusted hazard ratio (aHR) was 0.44 (95 % confidence interval = 0.39-0.50) compared to those who did not receive acupuncture. Furthermore, the cumulative incidence of PD in the acupuncture group was significantly lower, as evidenced by the log-rank test (p < 0.001). Conclusion In conclusion, our study provides evidence suggesting that acupuncture treatment is associated with a decreased risk of PD in patients with insomnia. However, further research is warranted to strengthen the evidence supporting these findings.
Collapse
Affiliation(s)
- Cheng-Hao Huang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shun-Ku Lin
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Chinese Medicine, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- General Education Center, University of Taipei, Taipei, Taiwan
- The Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Chen Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Division of Surgery, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
10
|
Liang Y, Feng L, Zheng Y, Gao Y, Shi R, Zhang Z, Ying X, Zeng Y. Targeted Liposomal Co-Delivery Dopamine with 3-n-Butylphthalide for Effective Against Parkinson's Disease in Mice Model. Int J Nanomedicine 2024; 19:12851-12870. [PMID: 39640048 PMCID: PMC11618862 DOI: 10.2147/ijn.s483595] [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: 06/24/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Parkinson's disease (PD) is a multifactor-induced neurodegenerative disease with high incidence in the elderly population. We found for the first time that the combination of dopamine (DA) and 3-n-butylphthalide (NBP) has great potential for the synergistic treatment of PD. To further improve the therapeutic performance of the drugs, a brain-targeting liposomal co-delivery system encapsulating NBP and DA ((NBP+DA)-Lips-RVG29) was designed using a rabies virus polypeptide with 29 amino acids (RVG29) as the targeting ligand. Methods The synergistic neuroprotective effects of NBP and DA were assessed in 6-OHDA-induced PC12 cells. Then, (NBP+DA)-Lips-RVG29 loading with NBP and DA at an optimal ratio was prepared using the thin-film hydration and sonication method. The physicochemical and biological characterization of (NBP+DA)-Lips-RVG29 were systemically investigated, and the therapeutic efficiency and underlying mechanisms of (NBP+DA)-Lips-RVG29 were also explored in vitro and in vivo. Finally, the safety of (NBP+DA)-Lips-RVG29 was evaluated. Results The synergistic effects of NBP and DA peaked at 1:1 (NBP/DA, mol/mol). The functionalized liposomes showed significantly higher uptake efficiency and blood-brain barrier (BBB) penetration efficiency in vitro. After systemic administration, (NBP+DA)-Lips-RVG29 prolonged the blood circulation of drugs, enhanced BBB penetration and increased drug accumulation in the striatum, substantia nigra and hippocampus. Moreover, (NBP+DA)-Lips-RVG29 showed excellent neuroprotective effects in a cellular PD model of PC12 cells and improved therapeutic efficacy in a PD mouse model. Furthermore, the safety evaluation of (NBP+DA)-Lips-RVG29 revealed no systemic toxicity. Conclusion NBP and DA exhibited the synergistic anti-PD effects. The RVG29-modified liposomes encapsulating NBP and DA contributed to the accumulation of drugs in the brain lesions area of PD and further improved treatment efficacy. Therefore, (NBP+DA)-Lips-RVG29 represents a promising strategy for the treatment of PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Liang
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Liping Feng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yue Zheng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yunzhen Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Rongying Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xue Ying
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Yingchun Zeng
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| |
Collapse
|
11
|
De Jesus S, Daya A, Blumberger L, Lewis MM, Leslie D, Tabbal SD, Dokholyan R, Snyder AM, Mailman RB, Huang X. Prevalence of Late-Stage Parkinson's Disease in the US Healthcare System: Insights from TriNetX. Mov Disord 2024; 39:1592-1601. [PMID: 38962960 DOI: 10.1002/mds.29900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Patients in late-stage Parkinson's disease (PDLS) are caregiver-dependent, have low quality of life, and higher healthcare costs. OBJECTIVE To estimate the prevalence of PDLS patients in the current US healthcare system. METHODS We downloaded the 2010-2022 data from the TriNetX Diamond claims network that consists of 92 US healthcare sites. PD was identified using standard diagnosis codes, and PDLS was identified by the usage of wheelchair dependence, personal care assistance, and/or presence of diagnoses of dementia. Age of PDLS identification and survival information were obtained and stratified by demographic and the disability subgroups. RESULTS We identified 1,031,377 PD patients in the TriNetX database. Of these, 18.8% fitted our definition of PDLS (n = 194,297), and 10.2% met two or more late-stage criteria. Among all PDLS, the mean age of PDLS identification was 78.1 (±7.7) years, and 49% were already reported as deceased. PDLS patients were predominantly male (58.5%) with similar distribution across PDLS subgroups. The majority did not have race (71%) or ethnicity (69%) information, but for the available information >90% (n = 53,162) were White, 8.2% (n = 5121) Hispanic/Latino, 7.8% (n = 4557) Black, and <0.01% (n = 408) Asian. Of the PDLS cohort, 71.6% identified with dementia, 12.9% had personal care assistance, and 4.8% were wheelchair-bound. CONCLUSIONS Late-stage patients are a significant part of the PD landscape in the current US healthcare system, and largely missed by traditional motor-based disability staging. It is imperative to include this population as a clinical, social, and research priority. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sol De Jesus
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Annika Daya
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Liba Blumberger
- Department of Public Health Science, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mechelle M Lewis
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Doug Leslie
- Department of Public Health Science, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Samer D Tabbal
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Rachel Dokholyan
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Amanda M Snyder
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Richard B Mailman
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Translational Brain Research Center, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
12
|
Ali A, Manzoor S, Ali T, Asim M, Muhammad G, Ahmad A, Jamaludin MI, Devaraj S, Munawar N. Innovative aspects and applications of single cell technology for different diseases. Am J Cancer Res 2024; 14:4028-4048. [PMID: 39267684 PMCID: PMC11387862 DOI: 10.62347/vufu1836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Recent developments in single-cell technologies have provided valuable insights from cancer genomics to complex microbial communities. Single-cell technologies including the RNA-seq, next-generation sequencing (NGS), epigenomics, genomics, and transcriptomics can be used to uncover the single cell nature and molecular characterization of individual cells. These technologies also reveal the cellular transition states, evolutionary relationships between genes, the complex structure of single-cell populations, cell-to-cell interaction leading to biological discoveries and more reliable than traditional bulk technologies. These technologies are becoming the first choice for the early detection of inflammatory biomarkers affecting the proliferation and progression of tumor cells in the tumor microenvironment and improving the clinical efficacy of patients undergoing immunotherapy. These technologies also hold a central position in the detection of checkpoint inhibitors and thus determining the signaling pathways evoked by tumor invasion. This review addressed the emerging approaches of single cell-based technologies in cancer immunotherapies and different human diseases at cellular and molecular levels and the emerging role of sequencing technologies leading to drug discovery. Advancements in these technologies paved for discovering novel diagnostic markers for better understanding the pathological and biochemical mechanisms also for controlling the rate of different diseases.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College Shantou 515041, Guangdong, China
| | - Saba Manzoor
- Department of Zoology, University of Sialkot Sialkot 51310, Pakistan
| | - Tayyab Ali
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture Faisalabad 38000, Pakistan
| | - Muhammad Asim
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture Faisalabad 38000, Pakistan
| | - Ghulam Muhammad
- Jinnah Burn and Reconstructive Surgery Centre, Jinnah Hospital, Allama Iqbal Medical College Lahore 54000, Pakistan
| | - Aftab Ahmad
- Biochemistry/Center for Advanced Studies in Agriculture and Food Security (CAS-AFS), University of Agriculture Faisalabad 38040, Pakistan
| | - Mohamad Ikhwan Jamaludin
- BioInspired Device and Tissue Engineering Research Group (BioInspira), Department of Biomedical Engineering and Health Sciences, Faculty of Electrical Engineering, Universiti Teknologi Malaysia Johor Bahru 81310, Johor, Malaysia
| | - Sutha Devaraj
- Graduate School of Medicine, Perdana University Wisma Chase Perdana, Changkat Semantan, Damansara Heights, Kuala Lumpur 50490, Malaysia
| | - Nayla Munawar
- Department of Chemistry, College of Science, United Arab Emirates University Al-Ain 15551, United Arab Emirates
| |
Collapse
|
13
|
Dong L, Gao L. SP1-Driven FOXM1 Upregulation Induces Dopaminergic Neuron Injury in Parkinson's Disease. Mol Neurobiol 2024; 61:5510-5524. [PMID: 38200349 DOI: 10.1007/s12035-023-03854-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024]
Abstract
The aberrant expression of Forkhead box M1 (FOXM1) has been associated with the pathological processes of Parkinson's disease (PD), but the upstream and downstream regulators remain poorly understood. This study sought to examine the underlying mechanism of FOXM1 in dopaminergic neuron injury in PD. Bioinformatics analysis was conducted to pinpoint the differential expression of FOXM1, which was verified in the nigral tissues of rotenone-lesioned mice and dopaminergic neuron MN9D cells. Interactions among SP1, FOXM1, SNAI2, and CXCL12 were analyzed. To evaluate their effects on dopaminergic neuron injury, the lentiviral vector-mediated manipulation of FOXM1, SP1, and CXCL12 was introduced in rotenone-lesioned mice and MN9D cells. SP1, FOXM1, SNAI2, and CXCL12 abundant expression occurred in rotenone-lesioned mice and MN9D cells. Silencing of FOXM1 delayed the rotenone-induced dopaminergic neuron injury in vitro. Mechanistically, SP1 was an upstream transcription factor of FOXM1 and upregulated FOXM1 expression, leading to increased SNAI2 and CXCL12 expression. In vivo, data confirmed that SP1 promoted dopaminergic neuron injury by activating the FOXM1/SNAI2/CXCL12 axis. Our data indicate that SP1 silencing has neuroprotective effects on dopaminergic neurons, which is dependent upon the inactivated FOXM1/SNAI2/CXCL12 axis.
Collapse
Affiliation(s)
- Li Dong
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning Province, People's Republic of China.
| | - Lianbo Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning Province, People's Republic of China
| |
Collapse
|
14
|
Al Meslamani AZ. The long-term clinical impact of digital endpoints and biomarkers in data collection. Expert Rev Pharmacoecon Outcomes Res 2024; 24:697-699. [PMID: 38362754 DOI: 10.1080/14737167.2024.2320233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/14/2024] [Indexed: 02/17/2024]
Affiliation(s)
- Ahmad Z Al Meslamani
- College of Pharmacy, Al Ain University, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research quality of care Center, Al Ain University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
15
|
Hull BT, Miller KM, Corban C, Backer G, Sheehan S, Korstanje R, Sutphin GL. 3-Hydroxyanthranilic Acid Delays Paralysis in Caenorhabditis elegans Models of Amyloid-Beta and Polyglutamine Proteotoxicity. Biomolecules 2024; 14:599. [PMID: 38786006 PMCID: PMC11117628 DOI: 10.3390/biom14050599] [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: 05/01/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Age is the primary risk factor for neurodegenerative diseases such as Alzheimer's and Huntington's disease. Alzheimer's disease is the most common form of dementia and a leading cause of death in the elderly population of the United States. No effective treatments for these diseases currently exist. Identifying effective treatments for Alzheimer's, Huntington's, and other neurodegenerative diseases is a major current focus of national scientific resources, and there is a critical need for novel therapeutic strategies. Here, we investigate the potential for targeting the kynurenine pathway metabolite 3-hydroxyanthranilic acid (3HAA) using Caenorhabditis elegans expressing amyloid-beta or a polyglutamine peptide in body wall muscle, modeling the proteotoxicity in Alzheimer's and Huntington's disease, respectively. We show that knocking down the enzyme that degrades 3HAA, 3HAA dioxygenase (HAAO), delays the age-associated paralysis in both models. This effect on paralysis was independent of the protein aggregation in the polyglutamine model. We also show that the mechanism of protection against proteotoxicity from HAAO knockdown is mimicked by 3HAA supplementation, supporting elevated 3HAA as the mediating event linking HAAO knockdown to delayed paralysis. This work demonstrates the potential for 3HAA as a targeted therapeutic in neurodegenerative disease, though the mechanism is yet to be explored.
Collapse
Affiliation(s)
- Bradford T. Hull
- Molecular and Cellular Biology Department, University of Arizona, Tucson, AZ 85721, USA
| | - Kayla M. Miller
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA
| | | | - Grant Backer
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | - George L. Sutphin
- Molecular and Cellular Biology Department, University of Arizona, Tucson, AZ 85721, USA
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
16
|
Riasi A, Delrobaei M, Salari M. A decision support system based on recurrent neural networks to predict medication dosage for patients with Parkinson's disease. Sci Rep 2024; 14:8424. [PMID: 38600209 PMCID: PMC11006681 DOI: 10.1038/s41598-024-59179-0] [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: 09/12/2023] [Accepted: 04/08/2024] [Indexed: 04/12/2024] Open
Abstract
Using deep learning has demonstrated significant potential in making informed decisions based on clinical evidence. In this study, we deal with optimizing medication and quantitatively present the role of deep learning in predicting the medication dosage for patients with Parkinson's disease (PD). The proposed method is based on recurrent neural networks (RNNs) and tries to predict the dosage of five critical medication types for PD, including levodopa, dopamine agonists, monoamine oxidase-B inhibitors, catechol-O-methyltransferase inhibitors, and amantadine. Recurrent neural networks have memory blocks that retain crucial information from previous patient visits. This feature is helpful for patients with PD, as the neurologist can refer to the patient's previous state and the prescribed medication to make informed decisions. We employed data from the Parkinson's Progression Markers Initiative. The dataset included information on the Unified Parkinson's Disease Rating Scale, Activities of Daily Living, Hoehn and Yahr scale, demographic details, and medication use logs for each patient. We evaluated several models, such as multi-layer perceptron (MLP), Simple-RNN, long short-term memory (LSTM), and gated recurrent units (GRU). Our analysis found that recurrent neural networks (LSTM and GRU) performed the best. More specifically, when using LSTM, we were able to predict levodopa and dopamine agonist dosage with a mean squared error of 0.009 and 0.003, mean absolute error of 0.062 and 0.030, root mean square error of 0.099 and 0.053, and R-squared of 0.514 and 0.711, respectively.
Collapse
Affiliation(s)
- Atiye Riasi
- Department of Biomedical Engineering, Faculty of Electrical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mehdi Delrobaei
- Department of Mechatronics, Faculty of Electrical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, Western University, London, ON, Canada.
| | - Mehri Salari
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Wu X, Cheng D, Lu Y, Rong R, Kong Y, Wang X, Niu B. A liquid crystal in situ gel based on rotigotine for the treatment of Parkinson's disease. Drug Deliv Transl Res 2024; 14:1048-1062. [PMID: 37875660 DOI: 10.1007/s13346-023-01449-x] [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] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
One of the most common neurodegenerative illnesses is Parkinson's disease (PD). Rotigotine (RTG) is a dopamine agonist that exerts anti-Parkinsonian effects through dopamine receptor agonism to improve motor symptoms and overall performance in PD patients. In this study, an in situ liquid crystal gel called rotigotine-gel (RTG-gel) was developed using soya phosphatidyl choline (SPC) and glycerol dioleate (GDO) to provide long-acting slow-release benefits of rotigotine while minimizing side effects. This study prepared the RTG-gel precursor solution using SPC, GDO, and ethanol (in the ratio of 54:36:10, w/w/w). The internal structures of the gel were confirmed by crossed-polarized light microscopy (PLM), small-angle X-ray scattering (SAXS), and differential scanning calorimetry (DSC). The rheological properties of the RTG-gel precursor solution indicate a favorable combination of low viscosity and excellent flowability. The gel that produced during water absorption was also highly viscous and structurally stable, which helped to maintain the drug delayed release at the injection site. In vitro release assays showed that the in vitro release of RTG-gel followed Ritger-Peppas. The RTG-gel precursor solution was administered by subcutaneous injection, and the results of in vivo pharmacokinetic tests in SD rats showed that the plasma elimination half-life (t1/2) was 59.28 ± 16.08 h; the time to peak blood concentration (Tmax) was 12.00 ± 10.32 h, and the peak concentration (Cmax) was 29.9 ± 10.10 ng/mL. The blood concentration remained above 0.1 ng/mL for 20 days after administration and was still detectable after 31 days of administration, and the bioavailability of RTG can reach 72.59%. The results of in vitro solvent exchange tests showed that the RTG-gel precursor solution undergoes rapid exchange upon contact with PBS, and the diffusion of ethanol can reach 48.1% within 60 min and 80% within 8 h. The results of cytotoxicity test showed 89.27 ± 4.32% cell survival after administration of the drug using RTG-gel. The results of tissue extraction at the administration site showed that healing of the injection site without redness and hemorrhage could be observed after 14 days of injection. The results of tissue section of the administered site showed that the inflammatory cells decreased and granulation tissue appeared after 14 days of administration, and there was basically no inflammatory cell infiltration after 35 days of administration, and the inflammatory reaction was basically eliminated. It shows that RTG-gel has some irritation to the injection site, but it can be recovered by itself in the later stage, and it has good biocompatibility. In summary, RTG-gel might be a potential RTG extended-release formulation for treating PD.
Collapse
Affiliation(s)
- Xiaxia Wu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China
- School of Pharmacy, Yantai University, Yantai, 264005, People's Republic of China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, People's Republic of China.
| | - Yue Lu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China
- School of Pharmacy, Yantai University, Yantai, 264005, People's Republic of China
| | - Rong Rong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, People's Republic of China
| | - Ying Kong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, People's Republic of China
| | - Xiuzhi Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, People's Republic of China
| | - Baohua Niu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, Shandong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, People's Republic of China
| |
Collapse
|
18
|
Otzen DE. Antibodies and α-synuclein: What to target against Parkinson's Disease? BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:140943. [PMID: 37783321 DOI: 10.1016/j.bbapap.2023.140943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 10/04/2023]
Abstract
Parkinson's Disease (PD) is strongly linked to the aggregation of the protein α-synuclein (α-syn), an intrinsically disordered protein. However, strategies to combat PD by targeting the aggregation of α-syn are challenged by the multiple types of aggregates formed both in vivo and in vitro, the potential influence of chemical modifications and the as yet unresolved question of which aggregate types (oligomeric or fibrillar) are most cytotoxic. Here I briefly review the social history of α-syn, the many efforts to raise antibodies against α-syn and the disappointing results of clinical trials based on such antibodies. Ultimately a thorough understanding of the molecular and mechanistic properties of mAbs towards aggregated species of α-syn is an essential prerequisite for any clinical trial, but this is missing in most cases. I highlight new microfluidic techniques which may address this need and call for a more concerted effort to standardize antibody studies as the basis to allow us to link molecular insights to clinical efficacy.
Collapse
Affiliation(s)
- Daniel E Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK - 8000, Aarhus, Denmark.
| |
Collapse
|
19
|
Shejul PP, Doshi GM. Glutamate Receptors and C-ABL Inhibitors: A New Therapeutic Approach for Parkinson's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:22-44. [PMID: 38273763 DOI: 10.2174/0118715249268627231206115942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 01/27/2024]
Abstract
Parkinson's disease (PD) is the second-most prevalent central nervous system (CNS) neurodegenerative condition. Over the past few decades, suppression of BCR-Abelson tyrosine kinase (c-Abl), which serves as a marker of -synuclein aggregation and oxidative stress, has shown promise as a potential therapy target in PD. c-Abl inhibition has the potential to provide neuroprotection against PD, as shown by experimental results and the first-in-human trial, which supports the strategy in bigger clinical trials. Furthermore, glutamate receptors have also been proposed as potential therapeutic targets for the treatment of PD since they facilitate and regulate synaptic neurotransmission throughout the basal ganglia motor system. It has been noticed that pharmacological manipulation of the receptors can change normal as well as abnormal neurotransmission in the Parkinsonian brain. The review study contributes to a comprehensive understanding of the approach toward the role of c-Abl and glutamate receptors in Parkinson's disease by highlighting the significance and urgent necessity to investigate new pharmacotherapeutic targets. The article covers an extensive insight into the concept of targeting, pathophysiology, and c-Abl interaction with α-synuclein, parkin, and cyclin-dependent kinase 5 (Cdk5). Furthermore, the concepts of Nmethyl- D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPA) receptor, and glutamate receptors are discussed briefly. Conclusion: This review article focuses on in-depth literature findings supported by an evidence-based discussion on pre-clinical trials and clinical trials related to c-Abl and glutamate receptors that act as potential therapeutic targets for PD.
Collapse
Affiliation(s)
- Priya P Shejul
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
20
|
Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther 2024; 9:1. [PMID: 38161204 PMCID: PMC10758001 DOI: 10.1038/s41392-023-01668-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024] Open
Abstract
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Xiuju Peng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Makhloufi Zoulikha
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, PR China
| | - Kosheli Thapa Magar
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Yanmin Ju
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
21
|
Tran KK, Lee PY, Finkelstein DI, McKendrick AM, Nguyen BN, Bui BV, Nguyen CT. Altered Outer Retinal Structure, Electrophysiology and Visual Perception in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:167-180. [PMID: 38189711 PMCID: PMC10836541 DOI: 10.3233/jpd-230293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Visual biomarkers of Parkinson's disease (PD) are attractive as the retina is an outpouching of the brain. Although inner retinal neurodegeneration in PD is well-established this has overlap with other neurodegenerative diseases and thus outer retinal (photoreceptor) measures warrant further investigation. OBJECTIVE To examine in a cross-sectional study whether clinically implementable measures targeting outer retinal function and structure can differentiate PD from healthy ageing and whether these are sensitive to intraday levodopa (L-DOPA) dosing. METHODS Centre-surround perceptual contrast suppression, macular visual field sensitivity, colour discrimination, light-adapted electroretinography and optical coherence tomography (OCT) were tested in PD participants (n = 16) and controls (n = 21). Electroretinography and OCT were conducted before and after midday L-DOPA in PD participants, or repeated after ∼2 hours in controls. RESULTS PD participants had decreased center-surround contrast suppression (p < 0.01), reduced macular visual field sensitivity (p < 0.05), color vision impairment (p < 0.01) photoreceptor dysfunction (a-wave, p < 0.01) and photoreceptor neurodegeneration (outer nuclear layer thinning, p < 0.05), relative to controls. Effect size comparison between inner and outer retinal parameters showed that photoreceptor metrics were similarly robust in differentiating the PD group from age-matched controls as inner retinal changes. Electroretinography and OCT were unaffected by L-DOPA treatment or time. CONCLUSIONS We show that outer retinal outcomes of photoreceptoral dysfunction (decreased cone function and impaired color vision) and degeneration (i.e., outer nuclear layer thinning) were equivalent to inner retinal metrics at differentiating PD from healthy age-matched adults. These findings suggest outer retinal metrics may serve as useful biomarkers for PD.
Collapse
Affiliation(s)
- Katie K.N. Tran
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Pei Ying Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Allison M. McKendrick
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
- Division of Optometry, School of Allied Health, The University of Western Australia, Crawley, WA, Australia
- Lions Eye Institute, Nedlands, WA, Australia
| | - Bao N. Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Carrick FR, Hernandez LSAV, Sugaya K. Amelioration of Motor Performance and Nigrostriatal Dopamine Cell Volume Using a Novel Far-Infrared Ceramic Blanket in an A53T Alpha-Synuclein Transgenic Parkinson's Disease Mouse Model. Curr Issues Mol Biol 2023; 45:9823-9837. [PMID: 38132459 PMCID: PMC10742635 DOI: 10.3390/cimb45120613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
We had attended a Parkinson's Disease (PD) patient for a non-healing wound who reported a marked decrease in his hand tremor and freezing of gait when his wound was exposed to a ceramic far-field infrared (cFIR) blanket. PD is the most frequent motor disorder and the second most frequent neurodegenerative disease after Alzheimer's Disease (AD). The tremor, rigidity, and slowness of movement associated with Parkinson's disease (PD) affect up to 10 million people throughout the world, and the major contributing factor to the pathogenesis of PD is the accumulation and propagation of pathological α-synuclein (α-Syn) and the death of dopaminergic cells in the Nigrostriatal system. Efforts to slow or stop its spreading have resulted in the development and use of dopaminergic drug replacement therapy. Unfortunately, there is a loss of about 70-80% of substantia nigral dopaminergic neurons in patients by the time they are diagnosed with PD, and various dopaminergic drugs provide only temporary relief of their motor symptoms. There are limitations in treating PD with many conventional medications, necessitating a combination of pharmaceutical and non-pharmacological therapy as an essential adjunct to better address the health and welfare of PD patients. We used male adult A53T alpha-synuclein transgenic mice exposed to a ceramic far-infrared blanket. Motor activity was assessed using the rotarod apparatus, and mouse brains were examined to quantify the fluorescence intensities of the immunostained samples. A53T alpha-synuclein transgenic mice had a significantly shorter time stay on the rotating bar than the wild-type mice (B6C3H). The rotarod performance was significantly improved in A53T alpha-synuclein transgenic mice exposed to cFIR as well as B6C3H healthy wild mice exposed to cFIR. There was a significant statistical and substantive increase in the cellular composition of the Striatum and substantia nigra of cFIR-treated mice. Improvement in motor performance is seen in PD mice and wild mice and is associated with increases in cell volume in the substantia nigra and striatum after treatment.
Collapse
Affiliation(s)
- Frederick Robert Carrick
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
- Burnett School of Biomedical Science, University of Central Florida, Orlando, FL 32827, USA;
- MGH Institute for Health Professions, Boston, MA 02129, USA
- Centre for Mental Health Research in Association, University of Cambridge, Cambridge CB2 1TN, UK
- Department of Neurology, Carrick Institute, Cape Canaveral, FL 32920, USA
| | | | - Kiminobu Sugaya
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
- Burnett School of Biomedical Science, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
23
|
Kawahata I, Fukunaga K. Pathogenic Impact of Fatty Acid-Binding Proteins in Parkinson's Disease-Potential Biomarkers and Therapeutic Targets. Int J Mol Sci 2023; 24:17037. [PMID: 38069360 PMCID: PMC10707307 DOI: 10.3390/ijms242317037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease is a neurodegenerative condition characterized by motor dysfunction resulting from the degeneration of dopamine-producing neurons in the midbrain. This dopamine deficiency gives rise to a spectrum of movement-related symptoms, including tremors, rigidity, and bradykinesia. While the precise etiology of Parkinson's disease remains elusive, genetic mutations, protein aggregation, inflammatory processes, and oxidative stress are believed to contribute to its development. In this context, fatty acid-binding proteins (FABPs) in the central nervous system, FABP3, FABP5, and FABP7, impact α-synuclein aggregation, neurotoxicity, and neuroinflammation. These FABPs accumulate in mitochondria during neurodegeneration, disrupting their membrane potential and homeostasis. In particular, FABP3, abundant in nigrostriatal dopaminergic neurons, is responsible for α-synuclein propagation into neurons and intracellular accumulation, affecting the loss of mesencephalic tyrosine hydroxylase protein, a rate-limiting enzyme of dopamine biosynthesis. This review summarizes the characteristics of FABP family proteins and delves into the pathogenic significance of FABPs in the pathogenesis of Parkinson's disease. Furthermore, it examines potential novel therapeutic targets and early diagnostic biomarkers for Parkinson's disease and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
- BRI Pharma Inc., Sendai 982-0804, Japan
| |
Collapse
|
24
|
Burns J, Buck AC, D’ Souza S, Dube A, Bardien S. Nanophytomedicines as Therapeutic Agents for Parkinson's Disease. ACS OMEGA 2023; 8:42045-42061. [PMID: 38024675 PMCID: PMC10652730 DOI: 10.1021/acsomega.3c04862] [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: 07/06/2023] [Revised: 09/11/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023]
Abstract
Phytochemicals are promising therapeutics for various neurodegenerative disorders, including Parkinson's disease (PD). However, their efficacy, pharmacokinetic properties, and penetration across the blood-brain barrier can be improved using delivery systems such as nanoparticles. We reviewed recently published work in which nanoparticles were used to deliver phytochemicals toward PD treatment. The studies show that nanoparticles not only improve the pharmacological effect of the phytochemicals but also enable targeting to the brain and crossing of the blood-brain barrier. Various ligands were added to the nanoparticles to improve blood-brain barrier transportation. The promising findings from the published studies reveal that more research into nanophytomedicine approaches as therapeutic targets for PD is warranted, especially since they have the potential to protect against key features of PD, including α-synuclein aggregation, mitochondrial dysfunction, and dopaminergic neuronal death. Furthermore, future directions should involve smart designs to tailor nanoparticles for improved therapeutic delivery by modifying their features, such as architecture, surface and material properties, targeting ligands, and responsiveness.
Collapse
Affiliation(s)
- Jessica Burns
- Division
of Molecular Biology and Human Genetics, Faculty of Medicine and Health
Sciences, Stellenbosch University, Stellenbosch, Cape Town 7600, South Africa
| | - Amy Claire Buck
- Division
of Molecular Biology and Human Genetics, Faculty of Medicine and Health
Sciences, Stellenbosch University, Stellenbosch, Cape Town 7600, South Africa
| | - Sarah D’ Souza
- School
of Pharmacy, University of the Western Cape, Bellville, Cape Town 7535, South Africa
| | - Admire Dube
- School
of Pharmacy, University of the Western Cape, Bellville, Cape Town 7535, South Africa
| | - Soraya Bardien
- Division
of Molecular Biology and Human Genetics, Faculty of Medicine and Health
Sciences, Stellenbosch University, Stellenbosch, Cape Town 7600, South Africa
- South
African Medical Research Council/Stellenbosch University Genomics
of Brain Disorders Research Unit, Stellenbosch
University, Stellenbosch, Cape Town 7600, South Africa
| |
Collapse
|
25
|
Podturkina AV, Ardashov OV, Volcho KP, Salakhutdinov NF. A New Stereoselective Approach to the Substitution of Allyl Hydroxy Group in para-Mentha-1,2-diol in the Search for New Antiparkinsonian Agents. Molecules 2023; 28:7303. [PMID: 37959723 PMCID: PMC10650740 DOI: 10.3390/molecules28217303] [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: 09/13/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Two approaches to the synthesis of para-menthene epoxide ((1S,5S,6R)-4) are developed. The first approach includes a reaction between chlorohydrin 7 and NaH in THF. The second involves the formation of epoxide in the reaction of corresponding diacetate 6 with sodium tert-butoxide. One possible mechanism of this reaction is proposed to explain unexpected outcomes in the regio- and stereospecificity of epoxide (1S,5S,6R)-4 formation. The epoxide ring in (1S,5S,6R)-4 is then opened by various S- and O-nucleophiles. This series of reactions allows for the stereoselective synthesis of diverse derivatives of the monoterpenoid Prottremine 1, a compound known for its antiparkinsonian activity, including promising antiparkinsonian properties.
Collapse
Affiliation(s)
| | | | - Konstantin P. Volcho
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave. 9, 630090 Novosibirsk, Russia; (A.V.P.); (O.V.A.); (N.F.S.)
| | | |
Collapse
|
26
|
Ibrahim A, Ipinloju N, Atasie NH, Babalola RM, Muhammad SA, Oyeneyin OE. Discovery of Small Molecule PARKIN Activator from Antipsychotic/Anti-neuropsychiatric Drugs as Therapeutics for PD: an In Silico Repurposing Approach. Appl Biochem Biotechnol 2023; 195:5980-6002. [PMID: 36735144 DOI: 10.1007/s12010-023-04376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Although there is presently no cure for Parkinson's disease (PD), the available therapies are only able to lessen symptoms and preserve the quality of life. Around 10 million people globally had PD as of 2020. The widely used standard drug has recently been revealed to have several negative effects. Additionally, there is a dearth of innovative compounds entering the market as a result of subpar ADMET characteristics. Drug repurposing provides a chance to reenergize the sluggish drug discovery process by identifying new applications for already-approved medications. As this strategy offers a practical way to speed up the process of developing alternative medications for PD. This study used a computer-aided technique to select therapeutic agent(s) from FDA-approved neuropsychiatric/psychotic drugs that can be adopted in the treatment of Parkinson's disease. In the current work, a computational approach via molecular docking, density functional theory (DFT), and pharmacokinetics were used to identify possible (anti)neuropsychiatric/psychotic medications for the treatment of PD. By using molecular docking, about eight (anti)neuropsychiatric/psychotic medications were tested against PARKIN, a key protein in PD. Based on the docking score, the best ligand in the trial was determined. The top hits were compared to the reference ligand levodopa (L-DOPA). A large proportion of the drugs displayed binding affinity that was relatively higher than L-DOPA. Also, DFT analysis confirms the ligand-receptor interactions and the molecular charge transfer. All the compounds were found to obey Lipinski's rule with acceptable pharmacokinetic properties. The current study has revealed the effectiveness of antineuropsychiatric/antipsychotic drugs against PARKIN in the treatment of PD and lumateperone was revealed to be the most promising candidate interacting with PARKIN.
Collapse
Affiliation(s)
- Abdulwasiu Ibrahim
- Drosophila Laboratory, Department of Biochemistry, University of Ibadan, Ibadan, Oyo State, Nigeria.
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria.
- Drosophila Research and Training Centre, Ibadan, Oyo State, Nigeria.
| | - Nureni Ipinloju
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.
| | | | | | | | - Oluwatoba Emmanuel Oyeneyin
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| |
Collapse
|
27
|
Das D, Bharadwaz P, Mattaparthi VSK. Computational investigation on the effect of the peptidomimetic inhibitors (NPT100-18A and NPT200-11) on the α-synuclein and lipid membrane interactions. J Biomol Struct Dyn 2023; 42:11471-11482. [PMID: 37768058 DOI: 10.1080/07391102.2023.2262599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Parkinson's disease (PD) is associated with α-synuclein (α-Syn), a presynaptic protein that binds to cell membranes. The molecular pathophysiology of PD most likely begins with the binding of α-Syn to membranes. Recently, two peptidomimetic inhibitors (NPT100-18A and NPT200-11) were identified to potentially interact with α-Syn and affect the interaction of α-Syn with the membrane. In this study, the effect of the two peptidomimetic inhibitors on the α-Syn-membrane interaction was demonstrated. DFT calculations were performed for optimization of the two inhibitors, and the nucleophilicity (N) and electrophilicity (ω) of NPT100-18A and NPT200-11 were calculated to be 3.90 and 3.86 (N); 1.06 and 1.04 (ω), respectively. Using the docking tool (CB-dock2), the two α-Syn-peptidomimetic inhibitor complexes (α-Syn-NPT100-18A and α-Syn-NPT200-11) have been prepared. Then all-atom molecular dynamics (MD) simulation was carried out on the α-Syn (control), α-Syn-NPT100-18A and α-Syn-NPT200-11 complex systems in presence of DOPE: DOPS: DOPC (5:3:2) lipid bilayer. From the conformational dynamics analysis, the 3-D structure of α-Syn was found to be stable, and the helices present in the regions (1-37) and (45-95) of α-Syn were found to be retained in the presence of the two peptidomimetic inhibitors. The electron density profile analysis revealed the binding modes of NAC and C-terminal region of α-Syn (in the presence of NPT200-11 inhibitor) with lipid membrane are in the close vicinity from the lipid bilayer centre. Our findings in this study on α-Syn-membrane interactions may be useful for developing a new therapeutic approach for treating PD and other neurodegenerative disorders.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dorothy Das
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Priyam Bharadwaz
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Venkata Satish Kumar Mattaparthi
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| |
Collapse
|
28
|
Khanna A, Jones G. Toward Personalized Medicine Approaches for Parkinson Disease Using Digital Technologies. JMIR Form Res 2023; 7:e47486. [PMID: 37756050 PMCID: PMC10568402 DOI: 10.2196/47486] [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: 03/21/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder that afflicts over 10 million people worldwide, resulting in debilitating motor and cognitive impairment. In the United States alone (with approximately 1 million cases), the economic burden for treating and caring for persons with PD exceeds US $50 billion and myriad therapeutic approaches are under development, including both symptomatic- and disease-modifying agents. The challenges presented in addressing PD are compounded by observations that numerous, statistically distinct patient phenotypes present with a wide variety of motor and nonmotor symptomatic profiles, varying responses to current standard-of-care symptom-alleviating medications (L-DOPA and dopaminergic agonists), and different disease trajectories. The existence of these differing phenotypes highlights the opportunities in personalized approaches to symptom management and disease control. The prodromal period of PD can span across several decades, allowing the potential to leverage the unique array of composite symptoms presented to trigger early interventions. This may be especially beneficial as disease progression in PD (alongside Alzheimer disease and Huntington disease) may be influenced by biological processes such as oxidative stress, offering the potential for individual lifestyle factors to be tailored to delay disease onset. In this viewpoint, we offer potential scenarios where emerging diagnostic and monitoring strategies might be tailored to the individual patient under the tenets of P4 medicine (predict, prevent, personalize, and participate). These approaches may be especially relevant as the causative factors and biochemical pathways responsible for the observed neurodegeneration in patients with PD remain areas of fluid debate. The numerous observational patient cohorts established globally offer an excellent opportunity to test and refine approaches to detect, characterize, control, modify the course, and ultimately stop progression of this debilitating disease. Such approaches may also help development of parallel interventive strategies in other diseases such as Alzheimer disease and Huntington disease, which share common traits and etiologies with PD. In this overview, we highlight near-term opportunities to apply P4 medicine principles for patients with PD and introduce the concept of composite orthogonal patient monitoring.
Collapse
Affiliation(s)
- Amit Khanna
- Neuroscience Global Drug Development, Novartis Pharma AG, Basel, Switzerland
| | - Graham Jones
- GDD Connected Health and Innovation Group, Novartis Pharmaceuticals, East Hanover, NJ, United States
- Clinical and Translational Science Institute, Tufts University Medical Center, Boston, MA, United States
| |
Collapse
|
29
|
He S, Fang W, Wu J, Lv H, Zhang J, Wang T, Huang Y, Li G, Li M. Whether mindfulness-guided therapy can be a new direction for the rehabilitation of patients with Parkinson's disease: a network meta-analysis of non-pharmacological alternative motor-/sensory-based interventions. Front Psychol 2023; 14:1162574. [PMID: 37780170 PMCID: PMC10540073 DOI: 10.3389/fpsyg.2023.1162574] [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: 02/11/2023] [Accepted: 07/20/2023] [Indexed: 10/03/2023] Open
Abstract
Background The treatment for Parkinson's disease (PD) consumes a lot of manpower and financial resources. Non-pharmacological alternative motor-/sensory-based interventions are optimized for the rehabilitation of PD patients. Mindfulness-based therapy shows ideal efficacy, but the diversity of the therapy brings difficulties to the selection of clinicians and patients. Methods Network meta-analysis in the Bayesian framework was used to evaluate the efficacy of non-pharmacological alternative motor-/sensory-based interventions in improving motor and non-motor symptoms in PD patients. Results A total of 58 studies (2,227 patients) were included. Compared with the non-intervention group, qigong was associated with improved outcomes in the Timed Up and Go (TUG) test (mean difference (MD) -5.54, 95% confidence interval (CI) -8.28 to -2.77), and UPDRS-I (MD -15.50, 95% CI -19.93 to -7.63). Differences between non-pharmacological alternative motor-/sensory-based interventions were not significant for PDQ-39, UPDRS-I, or UPDRS-II; however, qigong was superior to dance (MD -3.91, 95% CI -6.90 to -0.95), Tai Chi (MD -3.54, 95% CI -6.53 to -0.69), acupuncture (MD -6.75, 95% CI -10.86 to -2.70), music (MD -3.91, 95% CI -7.49 to -0.48), and exercise (MD -3.91, 95% CI -6.49 to -1.33) in the TUG test. Conclusion This network meta-analysis supports mindfulness-based therapy (e.g., qigong, yoga, and Tai Chi) as a preferred non-pharmacological alternative motor-/sensory-based intervention for PD rehabilitation. Systematic review registration https://inplasy.com/inplasy-2022-10-0109/, INPLASY2022100109.
Collapse
Affiliation(s)
- Shenglan He
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanyi Fang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaoyang Wu
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hang Lv
- The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jueyu Zhang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Second Traditional Chinese Medicine Hospital (Fifth Clinical Medical College of Guangzhou University of Chinese Medicine), Guangzhou, China
| | - Tunyi Wang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingjie Huang
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangyao Li
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Li
- Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
30
|
Janssen B, Tian G, Lengyel-Zhand Z, Hsieh CJ, Lougee MG, Riad A, Xu K, Hou C, Weng CC, Lopresti BJ, Kim HJ, Pagar VV, Ferrie JJ, Garcia BA, Mathis CA, Luk K, Petersson EJ, Mach RH. Identification of a Putative α-synuclein Radioligand Using an in silico Similarity Search. Mol Imaging Biol 2023; 25:704-719. [PMID: 36991273 PMCID: PMC10527666 DOI: 10.1007/s11307-023-01814-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
PURPOSE Previous studies from our lab utilized an ultra-high throughput screening method to identify compound 1 as a small molecule that binds to alpha-synuclein (α-synuclein) fibrils. The goal of the current study was to conduct a similarity search of 1 to identify structural analogs having improved in vitro binding properties for this target that could be labeled with radionuclides for both in vitro and in vivo studies for measuring α-synuclein aggregates. METHODS Using 1 as a lead compound in a similarity search, isoxazole derivative 15 was identified to bind to α-synuclein fibrils with high affinity in competition binding assays. A photocrosslinkable version was used to confirm binding site preference. Derivative 21, the iodo-analog of 15, was synthesized, and subsequently radiolabeled isotopologs [125I]21 and [11C]21 were successfully synthesized for use in in vitro and in vivo studies, respectively. [125I]21 was used in radioligand binding studies in post-mortem Parkinson's disease (PD) and Alzheimer's disease (AD) brain homogenates. In vivo imaging of an α-synuclein mouse model and non-human primates was performed with [11C]21. RESULTS In silico molecular docking and molecular dynamic simulation studies for a panel of compounds identified through a similarity search, were shown to correlate with Ki values obtained from in vitro binding studies. Improved affinity of isoxazole derivative 15 for α-synuclein binding site 9 was indicated by photocrosslinking studies with CLX10. Design and successful (radio)synthesis of iodo-analog 21 of isoxazole derivative 15 enabled further in vitro and in vivo evaluation. Kd values obtained in vitro with [125I]21 for α-synuclein and Aβ42 fibrils were 0.48 ± 0.08 nM and 2.47 ± 1.30 nM, respectively. [125I]21 showed higher binding in human postmortem PD brain tissue compared with AD tissue, and low binding in control brain tissue. Lastly, in vivo preclinical PET imaging showed elevated retention of [11C]21 in PFF-injected mouse brain. However, in PBS-injected control mouse brain, slow washout of the tracer indicates high non-specific binding. [11C]21 showed high initial brain uptake in a healthy non-human primate, followed by fast washout that may be caused by rapid metabolic rate (21% intact [11C]21 in blood at 5 min p.i.). CONCLUSION Through a relatively simple ligand-based similarity search, we identified a new radioligand that binds with high affinity (<10 nM) to α-synuclein fibrils and PD tissue. Although the radioligand has suboptimal selectivity for α-synuclein towards Aβ and high non-specific binding, we show here that a simple in silico approach is a promising strategy to identify novel ligands for target proteins in the CNS with the potential to be radiolabeled for PET neuroimaging studies.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guilong Tian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zsofia Lengyel-Zhand
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marshall G Lougee
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Aladdin Riad
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kuiying Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Chi-Chang Weng
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Hee Jong Kim
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vinayak V Pagar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John J Ferrie
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kelvin Luk
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
31
|
Hsu WT, Hsu CM, Hung SC, Hung SY. Acupuncture Improves Sleep Disorders and Depression among Patients with Parkinson's Disease: A Meta-Analysis. Healthcare (Basel) 2023; 11:2042. [PMID: 37510483 PMCID: PMC10379076 DOI: 10.3390/healthcare11142042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Parkinson's disease (PD) is associated with a range of non-motor symptoms that lack effective treatments. Acupuncture is a popular alternative therapy for PD patients that has been shown to improve motor symptoms. However, the efficacy of acupuncture in treating non-motor symptoms has remained controversial. The goal of our study was to systematically assess the existing evidence for acupuncture's efficacy in treating PD non-motor symptoms of sleep disorders, depression, anxiety, and fatigue. We conducted a meta-analysis of clinical trials by searching Pubmed, Embase, CINAHL, and Web of Science as electronic databases to evaluate acupuncture treatment for PD non-motor symptoms. Thirteen clinical trials met our inclusion criteria, and their methodological quality was assessed using the modified Jadad scale, indicating a moderate overall quality. Our results showed that acupuncture improved PD-related sleep disorders and depression but had no effect on anxiety and fatigue. Our meta-analysis suggests that acupuncture can be used as a complementary treatment for sleep disturbances and depression in PD patients and may exhibit a dual therapeutic effect on motor and non-motor symptoms. However, further well-designed clinical trials with larger sample sizes are needed to confirm these findings. Overall, our study highlights the potential of acupuncture as a viable complementary therapy for the treatment of PD non-motor symptoms of sleep disorders and depression, which can improve the quality of life of PD patients.
Collapse
Affiliation(s)
- Wei-Ti Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chieh-Min Hsu
- Department of Anesthesiology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shao-Chi Hung
- Department of Anesthesiology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
- Division of Surgery, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| |
Collapse
|
32
|
Amo-Aparicio J, Daly J, Højen JF, Dinarello CA. Pharmacologic inhibition of NLRP3 reduces the levels of α-synuclein and protects dopaminergic neurons in a model of Parkinson's disease. J Neuroinflammation 2023; 20:147. [PMID: 37349821 PMCID: PMC10286423 DOI: 10.1186/s12974-023-02830-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by a progressive degeneration of dopaminergic neurons, which leads to irreversible loss of peripheral motor functions. Death of dopaminergic neurons induces an inflammatory response in microglial cells, which further exacerbates neuronal loss. Reducing inflammation is expected to ameliorate neuronal loss and arrest motor dysfunctions. Because of the contribution of the NLRP3 inflammasome to the inflammatory response in PD, we targeted NLRP3 using the specific inhibitor OLT1177®. METHODS We evaluated the effectiveness of OLT1177® in reducing the inflammatory response in an MPTP neurotoxic model of PD. Using a combination of in vitro and in vivo studies, we analyzed the effects of NLRP3 inhibition on pro-inflammatory markers in the brain, α-synuclein aggregation, and dopaminergic neuron survival. We also determined the effects of OLT1177® on locomotor deficits associated with MPTP and brain penetrance. RESULTS Treatment with OLT1177® prevented the loss of motor function, reduced the levels of α-synuclein, modulated pro-inflammatory markers in the nigrostriatal areas of the brain, and protected dopaminergic neurons from degeneration in the MPTP model of PD. We also demonstrated that OLT1177® crosses the blood-brain barrier and reaches therapeutic concentrations in the brain. CONCLUSIONS These data suggest that targeting the NLRP3 inflammasome by OLT1177® may be a safe and novel therapeutic approach to arrest neuroinflammation and protect against neurological deficits of Parkinson's disease in humans.
Collapse
Affiliation(s)
- Jesus Amo-Aparicio
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA.
| | - Jonathan Daly
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
| | - Jesper Falkesgaard Højen
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus, Denmark
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, 12700 E 19th Ave, Aurora, CO, 80045, USA
| |
Collapse
|
33
|
Song LZX, Li Y, Qian X, Xu N, Yu Z, Dai JW, Xu B, Hu XM. Parkinson's disease constipation effect of electroacupuncture at ST25 through colonic motility and enteric neuropathology. Front Neurol 2023; 13:1092127. [PMID: 36733445 PMCID: PMC9888431 DOI: 10.3389/fneur.2022.1092127] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Background The enteric nervous system (ENS) plays a central role in developing Parkinson's disease (PD) constipation, and the regulation of the ENS may be a key component in treating PD constipation. Electroacupuncture (EA) can effectively treat constipation symptoms in PD, but research on its specific mechanisms, especially in terms of ENS, is relatively lacking. Therefore, we investigated whether EA at ST25 promotes the restoration of ENS structure and colonic motor function in the rotenone-induced PD constipation rat model. Methods In this study, we evaluated constipation symptoms by stool characteristics, excretion and water volume, and whole gut transit time and observed colonic motility regulation through colonic motion detection and pathological changes in the colonic myenteric nervous plexus by transmission electron microscopy and immunofluorescence staining. Results EA significantly improved the constipation symptoms and positively adjusted the colonic motility in rotenone-induced PD constipation rats. At the same time, EA reversed the rotenone-induced colonic myenteric nervous plexus injury and regulated the ratio of inhibitory and excitatory neurotransmitters. Conclusion Our results indicate that EA treatment of PD constipation may be mediated through the adjustment of ENS.
Collapse
Affiliation(s)
- Li-zhe-xiong Song
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xu Qian
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Na Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-wen Dai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China,School of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China,*Correspondence: Bin Xu ✉
| | - Xuan-ming Hu
- Acupuncture Department, Nanjing Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, China,Xuan-ming Hu ✉
| |
Collapse
|
34
|
Soraci L, Gambuzza ME, Biscetti L, Laganà P, Lo Russo C, Buda A, Barresi G, Corsonello A, Lattanzio F, Lorello G, Filippelli G, Marino S. Toll-like receptors and NLRP3 inflammasome-dependent pathways in Parkinson's disease: mechanisms and therapeutic implications. J Neurol 2023; 270:1346-1360. [PMID: 36460875 PMCID: PMC9971082 DOI: 10.1007/s00415-022-11491-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/10/2022] [Accepted: 11/13/2022] [Indexed: 12/05/2022]
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder characterized by motor and non-motor disturbances as a result of a complex and not fully understood pathogenesis, probably including neuroinflammation, oxidative stress, and formation of alpha-synuclein (α-syn) aggregates. As age is the main risk factor for several neurodegenerative disorders including PD, progressive aging of the immune system leading to inflammaging and immunosenescence may contribute to neuroinflammation leading to PD onset and progression; abnormal α-syn aggregation in the context of immune dysfunction may favor activation of nucleotide-binding oligomerization domain-like receptor (NOD) family pyrin domain containing 3 (NLRP3) inflammasome within microglial cells through interaction with toll-like receptors (TLRs). This process would further lead to activation of Caspase (Cas)-1, and increased production of pro-inflammatory cytokines (PC), with subsequent impairment of mitochondria and damage to dopaminergic neurons. All these phenomena are mediated by the translocation of nuclear factor kappa-B (NF-κB) and enhanced by reactive oxygen species (ROS). To date, drugs to treat PD are mainly aimed at relieving clinical symptoms and there are no disease-modifying options to reverse or stop disease progression. This review outlines the role of the TLR/NLRP3/Cas-1 pathway in PD-related immune dysfunction, also focusing on specific therapeutic options that might be used since the early stages of the disease to counteract neuroinflammation and immune dysfunction.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center on Aging (INRCA-IRCCS), 87100 Cosenza, Italy
| | - Maria Elsa Gambuzza
- Territorial Office of Messina, Italian Ministry of Health, 98122 Messina, Italy
| | - Leonardo Biscetti
- Section of Neurology, Italian National Research Center on Aging (INRCA-IRCCS), 60121, Ancona, Italy.
| | - Pasqualina Laganà
- Biomedical, Dental, Morphological and Functional Imaging Department, University of Messina, 98124 Messina, Italy
| | - Carmela Lo Russo
- Unit of Geriatric Medicine, Italian National Research Center on Aging (INRCA-IRCCS), 87100 Cosenza, Italy
| | - Annamaria Buda
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy
| | - Giada Barresi
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center on Aging (INRCA-IRCCS), 87100 Cosenza, Italy
| | - Fabrizia Lattanzio
- Scientific Direction, Italian National Research Center on Aging (INRCA-IRCCS), 60121 Ancona, Italy
| | - Giuseppe Lorello
- Unit of Internal Medicine, Polyclinic G Martino Hospital, 98125 Messina, Italy
| | | | - Silvia Marino
- IRCCS Centro Neurolesi Bonino-Pulejo, 98124 Messina, Italy
| |
Collapse
|
35
|
Affiliation(s)
- Sanjeev Kharel
- Maharajgunj Medical Campus, Tribhuvan University Institute of Medicine, Maharajgunj, Kathmandu, Nepal
| | - Rajeev Ojha
- Department of Neurology, Tribhuvan University Teaching Hospital, Maharajgunj, Kathmandu, Nepal
| |
Collapse
|
36
|
Delila L, Nebie O, Le NTN, Barro L, Chou M, Wu Y, Watanabe N, Takahara M, Buée L, Blum D, Devos D, Burnouf T. Neuroprotective activity of a virus-safe nanofiltered human platelet lysate depleted of extracellular vesicles in Parkinson's disease and traumatic brain injury models. Bioeng Transl Med 2023; 8:e10360. [PMID: 36684076 PMCID: PMC9842020 DOI: 10.1002/btm2.10360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/15/2022] [Accepted: 06/07/2022] [Indexed: 01/25/2023] Open
Abstract
Brain administration of human platelet lysates (HPL) is a potential emerging biotherapy of neurodegenerative and traumatic diseases of the central nervous system. HPLs being prepared from pooled platelet concentrates, thereby increasing viral risks, manufacturing processes should incorporate robust virus-reduction treatments. We evaluated a 19 ± 2-nm virus removal nanofiltration process using hydrophilic regenerated cellulose hollow fibers on the properties of a neuroprotective heat-treated HPL (HPPL). Spiking experiments demonstrated >5.30 log removal of 20-22-nm non-enveloped minute virus of mice-mock particles using an immuno-quantitative polymerase chain reaction assay. The nanofiltered HPPL (NHPPL) contained a range of neurotrophic factors like HPPL. There was >2 log removal of extracellular vesicles (EVs), associated with decreased expression of pro-thrombogenic phosphatidylserine and procoagulant activity. LC-MS/MS proteomics showed that ca. 80% of HPPL proteins, including neurotrophins, cytokines, and antioxidants, were still found in NHPPL, whereas proteins associated with some infections and cancer-associated pathways, pro-coagulation and EVs, were removed. NHPPL maintained intact neuroprotective activity in Lund human mesencephalic dopaminergic neuron model of Parkinson's disease (PD), stimulated the differentiation of SH-SY5Y neuronal cells and showed preserved anti-inflammatory function upon intranasal administration in a mouse model of traumatic brain injury (TBI). Therefore, nanofiltration of HPL is feasible, lowers the viral, prothrombotic and procoagulant risks, and preserves the neuroprotective and anti-inflammatory properties in neuronal pre-clinical models of PD and TBI.
Collapse
Affiliation(s)
- Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
| | - Nhi Thao Ngoc Le
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Present address:
National Center of Blood TransfusionOuagadougouBurkina Faso
| | - Ming‐Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Present address:
Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yu‐Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | | | | | - Luc Buée
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - David Blum
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - David Devos
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Biomedical Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- NeuroTMULilleTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Cell Therapy and Regeneration MedicineTaipei Medical UniversityTaipeiTaiwan
- PhD Program in Graduate Institute of Mind Brain and Consciousness, College of Humanities and Social SciencesTaipei Medical UniversityTaipeiTaiwan
- Neuroscience Research CenterTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
37
|
Chopade P, Chopade N, Zhao Z, Mitragotri S, Liao R, Chandran Suja V. Alzheimer's and Parkinson's disease therapies in the clinic. Bioeng Transl Med 2023; 8:e10367. [PMID: 36684083 PMCID: PMC9842041 DOI: 10.1002/btm2.10367] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most prevalent neurodegenerative diseases, affecting millions and costing billions each year in the United States alone. Despite tremendous progress in developing therapeutics that manage the symptoms of these two diseases, the scientific community has yet to develop a treatment that effectively slows down, inhibits, or cures neurodegeneration. To gain a better understanding of the current therapeutic frontier for the treatment of AD and PD, we provide a review on past and present therapeutic strategies for these two major neurodegenerative disorders in the clinical trial process. We briefly recap currently US Food and Drug Administration-approved therapies, and then explore trends in clinical trials across the variables of therapy mechanism of disease intervention, administration route, use of delivery vehicle, and outcome measures, across the clinical phases over time for "Drug" and "Biologic" therapeutics. We then present the success rate of past clinical trials and analyze the intersections in therapeutic approaches for AD and PD, revealing the shift in clinical trials away from therapies targeting neurotransmitter systems that provide symptomatic relief, and towards anti-aggregation, anti-inflammatory, anti-oxidant, and regeneration strategies that aim to inhibit the root causes of disease progression. We also highlight the evolving distribution of the types of "Biologic" therapies investigated, and the slowly increasing yet still severe under-utilization of delivery vehicles for AD and PD therapeutics. We then briefly discuss novel preclinical strategies for treating AD and PD. Overall, this review aims to provide a succinct overview of the clinical landscape of AD and PD therapies to better understand the field's therapeutic strategy in the past and the field's evolution in approach to the present, to better inform how to effectively treat AD and PD in the future.
Collapse
Affiliation(s)
| | | | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of PharmacyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| | - Rick Liao
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| | - Vineeth Chandran Suja
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringCambridgeMassachusettsUSA
| |
Collapse
|
38
|
Tuunainen J, Sjöstedt N, Vahteristo M, Ellmén J, Kuoppamäki M, Rouru J, Yliperttula M. Effect of Carbidopa Dose on Levodopa Pharmacokinetics With and Without Catechol-O-Methyltransferase Inhibition in Healthy Subjects. Eur J Drug Metab Pharmacokinet 2023; 48:23-34. [PMID: 36309950 DOI: 10.1007/s13318-022-00800-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVES The treatment of Parkinson's disease (PD) is still symptomatic since disease-modifying treatments for PD are not available. Oral levodopa is the gold standard for the treatment of PD motor symptoms. However, incomplete and fluctuating plasma exposure of levodopa leads to suboptimal treatment of the symptoms. The main objective of this study was to investigate to what extent increased carbidopa doses (50 and 100 mg) increase the plasma levels of 100-mg immediate-release (IR) levodopa compared to a 25-mg carbidopa dose with and without co-administration of 200 mg entacapone. METHODS A double-blind, placebo-controlled, randomized, crossover, phase I, pharmacokinetic study with 25 healthy volunteers was conducted. In addition, a semi-mechanistic pharmacokinetic model was built to theoretically evaluate the effect of inhibiting aromatic amino acid decarboxylase (AADC) and catechol-O-methyltransferase (COMT) mediated metabolism of levodopa on the exposure of levodopa. RESULTS The effect of increased carbidopa doses 50 and 100 mg on the total exposure (AUC) of 100 mg IR levodopa was +29% and +36%, respectively, when entacapone was co-administered. Without entacapone, the corresponding increases were +13% and +17%. With entacapone co-administration, the increased carbidopa dose also clearly increased levodopa trough concentration. There was no significant effect on the peak concentrations of levodopa. CONCLUSIONS Increasing carbidopa doses significantly increased the exposure and reduced the fluctuation of IR levodopa in plasma during simultaneous COMT inhibition with entacapone. Theoretical pharmacokinetic simulations suggested that the plasma profile of oral IR levodopa can be even further improved by optimizing AADC and COMT inhibition.
Collapse
Affiliation(s)
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Juha Ellmén
- Orion Corporation Orion Pharma, Turku, Finland
| | | | - Juha Rouru
- Orion Corporation Orion Pharma, Turku, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Physiologic Functions and Therapeutic Applications of α7 Nicotinic Acetylcholine Receptor in Brain Disorders. Pharmaceutics 2022; 15:pharmaceutics15010031. [PMID: 36678660 PMCID: PMC9865019 DOI: 10.3390/pharmaceutics15010031] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Accumulating data suggest that α7 nicotinic acetylcholine receptors (α7nAChRs) are an important therapeutic target for the treatment of Alzheimer's disease (AD) and schizophrenia. The homopentameric ligand-gated ion channel α7nAChR consists of five identical α7 subunits that are encoded by the CHRNA7 (cholinergic receptor nicotinic alpha7 subunit) gene. Moreover, α7nAChRs are densely distributed throughout the hippocampus, cortex, and thalamus brain regions, but sparsely in the striatum, forebrain, and medulla. Compared with other nAChRs, α7nAChR binds with low affinity to the naturally occurring neurotransmitter acetylcholine and the non-specific exogenous agonist nicotine, and with high affinity to the specific antagonists α-bungarotoxin and methyllycaconitine. Reports indicate that α7nAChR plays important roles in neurotransmitter release, cognitive functioning, and the cholinergic anti-inflammatory response. Genetic variations that alter CHRNA7 mRNA and protein expression or cause α7nAChR dysfunction are associated with many brain disorders. Our previous studies revealed that α7nAChR exerts neuroprotection in AD by acting as a cargo receptor for binding the autophagosomal marker protein LC3 and engulfing extracellular neurotoxic Aβ1-42 during autophagic degradation of the α7nAChR-Aβ1-42 complex. However, the role of α7nAChRs in other diseases remains unknown. Here, we review and summarize the essential characteristics and current findings concerning α7nAChRs in four common brain diseases (AD, Parkinson's disease, schizophrenia, and depression), which may elucidate the role of α7nAChRs and inform innovative research and novel treatments that target α7nAChRs in brain disease.
Collapse
|
40
|
Targeting G Protein-Coupled Receptors in the Treatment of Parkinson's Disease. J Mol Biol 2022:167927. [PMID: 36563742 DOI: 10.1016/j.jmb.2022.167927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized in part by the deterioration of dopaminergic neurons which leads to motor impairment. Although there is no cure for PD, the motor symptoms can be treated using dopamine replacement therapies including the dopamine precursor L-DOPA, which has been in use since the 1960s. However, neurodegeneration in PD is not limited to dopaminergic neurons, and many patients experience non-motor symptoms including cognitive impairment or neuropsychiatric disturbances, for which there are limited treatment options. Moreover, there are currently no treatments able to alter the progression of neurodegeneration. There are many therapeutic strategies being investigated for PD, including alternatives to L-DOPA for the treatment of motor impairment, symptomatic treatments for non-motor symptoms, and neuroprotective or disease-modifying agents. G protein-coupled receptors (GPCRs), which include the dopamine receptors, are highly druggable cell surface proteins which can regulate numerous intracellular signaling pathways and thereby modulate the function of neuronal circuits affected by PD. This review will describe the treatment strategies being investigated for PD that target GPCRs and their downstream signaling mechanisms. First, we discuss new developments in dopaminergic agents for alleviating PD motor impairment, the role of dopamine receptors in L-DOPA induced dyskinesia, as well as agents targeting non-dopamine GPCRs which could augment or replace traditional dopaminergic treatments. We then discuss GPCRs as prospective treatments for neuropsychiatric and cognitive symptoms in PD. Finally, we discuss the evidence pertaining to ghrelin receptors, β-adrenergic receptors, angiotensin receptors and glucagon-like peptide 1 receptors, which have been proposed as disease modifying targets with potential neuroprotective effects in PD.
Collapse
|
41
|
Lu C, Qu S, Zhong Z, Luo H, Lei SS, Zhong HJ, Su H, Wang Y, Chong CM. The effects of bioactive components from the rhizome of gastrodia elata blume (Tianma) on the characteristics of Parkinson's disease. Front Pharmacol 2022; 13:963327. [PMID: 36532787 PMCID: PMC9748092 DOI: 10.3389/fphar.2022.963327] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/07/2022] [Indexed: 08/04/2023] Open
Abstract
Parkinson's disease (PD) is an age-related chronic neurodegenerative disease caused by the death and degeneration of dopaminergic neurons in the substantia nigra of the midbrain. The decrease of the neurotransmitter dopamine in the patient's brain leads to various motor symptoms. PD drugs mainly enhance dopamine levels but cannot prevent or slow down the loss of dopaminergic neurons. In addition, they exhibit significant side effects and addiction issues during long-term use. Therefore, it is particularly urgent to develop novel drugs that have fewer side effects, can improve PD symptoms, and prevent the death of dopaminergic neurons. The rhizome of Gastrodia elata Blume (Tianma) is a well-known medicinal herb and has long been used as a treatment of nervous system-related diseases in China. Several clinical studies showed that formula comprising Tianma could be used as an add-on therapy for PD patients. Pharmacological studies indicated that Tianma and its bioactive components can reduce the death of dopaminergic neurons, α-synuclein accumulation, and neuroinflammation in various PD models. In this review, we briefly summarize studies regarding the effects of Tianma and its bioactive components' effects on major PD features and explore the potential use of Tianma components for the treatment of PD.
Collapse
Affiliation(s)
- Changcheng Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shuhui Qu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hua Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Si San Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hai-Jing Zhong
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
42
|
Kotliarova A, Podturkina AV, Pavlova AV, Gorina DS, Lastovka AV, Ardashov OV, Rogachev AD, Izyurov AE, Arefieva AB, Kulikov AV, Tolstikova TG, Volcho KP, Salakhutdinov NF, Sidorova Y. A Newly Identified Monoterpenoid-Based Small Molecule Able to Support the Survival of Primary Cultured Dopamine Neurons and Alleviate MPTP-Induced Toxicity In Vivo. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238286. [PMID: 36500381 PMCID: PMC9738927 DOI: 10.3390/molecules27238286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is the most common age-related movement disorder characterized by the progressive loss of nigrostriatal dopaminergic neurons. To date, PD treatment strategies are mostly based on dopamine replacement medicines, which can alleviate motor symptoms but do not slow down the progression of neurodegeneration. Thus, there is a need for disease-modifying PD therapies. The aim of this work was to evaluate the neuroprotective effects of the novel compound PA96 on dopamine neurons in vivo and in vitro, assess its ability to alleviate motor deficits in MPTP- and haloperidol-based PD models, as well as PK profile and BBB penetration. PA96 was synthesized from (1R,2R,6S)-3-methyl-6-(prop-1-en-2-yl) cyclohex-3-ene-1,2-diol (Prottremin) using the original three-step stereoselective procedure. We found that PA96: (1) supported the survival of cultured näive dopamine neurons; (2) supported the survival of MPP+-challenged dopamine neurons in vitro and in vivo; (3) had chemically appropriate properties (synthesis, solubility, etc.); (4) alleviated motor deficits in MPTP- and haloperidol-based models of PD; (5) penetrated the blood-brain barrier in vivo; and (6) was eliminated from the bloodstream relative rapidly. In conclusion, the present article demonstrates the identification of PA96 as a lead compound for the future development of this compound into a clinically used drug.
Collapse
Affiliation(s)
- Anastasiia Kotliarova
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFe, Viikinkaari 5D, University of Helsinki, 00014 Helsinki, Finland
| | - Alexandra V. Podturkina
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Alla V. Pavlova
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Daria S. Gorina
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova, 2, 630090 Novosibirsk, Russia
| | - Anastasiya V. Lastovka
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova, 2, 630090 Novosibirsk, Russia
| | - Oleg V. Ardashov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Artem D. Rogachev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogova, 2, 630090 Novosibirsk, Russia
| | - Arseniy E. Izyurov
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alla B. Arefieva
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexander V. Kulikov
- Department of Genetic Collections of Neural Disorders, Federal Research Center Institute of Cytology and Genetic, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Tatyana G. Tolstikova
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Konstantin P. Volcho
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
- Correspondence: (K.P.V.); (Y.S.)
| | - Nariman F. Salakhutdinov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia
| | - Yulia Sidorova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFe, Viikinkaari 5D, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: (K.P.V.); (Y.S.)
| |
Collapse
|
43
|
Cavitation Feedback Control of Focused Ultrasound Blood-Brain Barrier Opening for Drug Delivery in Patients with Parkinson's Disease. Pharmaceutics 2022; 14:pharmaceutics14122607. [PMID: 36559101 PMCID: PMC9781334 DOI: 10.3390/pharmaceutics14122607] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Magnetic resonance-guided focused ultrasound (MRgFUS), in conjunction with circulating microbubbles, is an emerging technology that can transiently enhance the permeability of the blood-brain barrier (BBB) locally and non-invasively to facilitate targeted drug delivery to the brain. In this clinical trial, the feasibility and safety of BBB modulation in the putamen were evaluated for biweekly therapeutic agent delivery in patients with Parkinson's disease. The performance of the clinical MRgFUS system's cavitation feedback controller for active power modulation throughout the exposures was examined. The putamen was targeted unilaterally by an ExAblate Neuro MRgFUS system operating at 220 kHz. Definity microbubbles were infused via a saline bag gravity drip at a rate of 4 µL/kg per 5 min. A cavitation emissions-based feedback controller was employed to modulate the acoustic power automatically according to prescribed target cavitation dose levels. BBB opening was measured by Gadolinium (Gd)-enhanced T1-weighted MR imaging, and the presence of potential micro-hemorrhages induced by the exposures was assessed via T2*-weighted MR imaging. A total of 12 treatment sessions were carried out across four patients, with target cavitation dose levels ranging from 0.20-0.40. BBB permeability in the targeted putamen was elevated successfully in all treatments, with a 14% ± 6% mean increase in Gd-enhanced T1-weighted MRI signal intensity relative to the untreated contralateral side. No indications of red blood cell extravasations were observed on MR imaging scans acquired one day following each treatment session. The cavitation emissions-based feedback controller was effective in modulating acoustic power levels to ensure BBB permeability enhancement while avoiding micro-hemorrhages, however, further technical advancements are warranted to improve its performance for use across a wide variety of brain diseases.
Collapse
|
44
|
Fang Y, Xu Y, Liu Z, Dong S, Su Y. Efficacy and safety of abdominal acupuncture in Parkinson's disease: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e31804. [PMID: 36451468 PMCID: PMC9704911 DOI: 10.1097/md.0000000000031804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Parkinson disease (PD) is a worldwide spread neurodegenerative disorder. Dopamine replacement therapy is currently the mainstream treatment, which can alleviate the symptoms but induces motor complications. Acupuncture therapy is effective for PD. As a form of acupuncture, the abdominal acupuncture has been used to relieve symptoms in patients with PD, but its effectiveness and safety have not yet reached a definitive conclusion. Therefore, this systematic review and meta-analysis protocol is planned to evaluate the efficacy and safety of abdominal acupuncture for PD patients. METHODS Six English databases (PubMed, Web of Science, MEDLINE, EMBASE, Springer Cochrane Library, and WHO International Clinical Trials Registry Platform) and 4 Chinese databases (Wan Fang Database, Chinese Scientific Journal Database, China National Knowledge Infrastructure Database, and Chinese Biomedical Literature Database) will be searched normatively according to the rule of each database from the inception to August 20, 2022. Two reviewers will independently conduct article selection, data collection, and risk of bias evaluation. Any disagreement will be resolved by discussion with the third reviewer. Either the fixed-effects or random-effects model will be used for data synthesis based on the heterogeneity test. Either the fixed-effects or random-effects model will be used for data synthesis based on the heterogeneity test. The analysis will be conducted by RevMan 5.3 software according to Cochrane Handbook. RESULTS The aim of this systematic review is to provide high-quality evidence to assess the efficacy and safety of abdominal acupuncture for patients in Parkinson's disease. The efficacy and safety of abdominal acupuncture for PD will be comprehensively assessed from the outcomes, including the effectiveness rate. The Unified Parkinson Disease Rating Scale (UPDRS) and Webster scale, Motor symptom scores utilizing UPDRS III scale, Dopamine (DA) content, and Nonmotor symptom scores employing UPDRS I scale, Activities of daily living using UDPRS II; Complications of treatment applying UPDRS IV, antioxidant ability: super oxide dismutase activity and Lipide Peroxide (LPO) content, Content of inflammatory cytokines, tumor necrosis factor-α and interleukin-1β, and adverse events as the secondary outcome. CONCLUSION This systematic review will explore whether abdominal acupuncture is an effective and safe intervention for patients in Parkinson's disease.
Collapse
Affiliation(s)
- Yuehong Fang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yilian Xu
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zhengzhong Liu
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Sihan Dong
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Su
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, China
- * Correspondence: Ying Su, Master of Medicine, School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, China (e-mail: )
| |
Collapse
|
45
|
Basile MS, Mazzon E. The Role of Cannabinoid Type 2 Receptors in Parkinson's Disease. Biomedicines 2022; 10:biomedicines10112986. [PMID: 36428554 PMCID: PMC9687889 DOI: 10.3390/biomedicines10112986] [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: 10/22/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disease and currently represents a clear unmet medical need. Therefore, novel preventive and therapeutic strategies are needed. Cannabinoid type 2 (CB2) receptors, one of the components of the endocannabinoid system, can regulate neuroinflammation in PD. Here, we review the current preclinical and clinical studies investigating the CB2 receptors in PD with the aim to clarify if these receptors could have a role in PD. Preclinical data show that CB2 receptors could have a neuroprotective action in PD and that the therapeutic targeting of CB2 receptors could be promising. Indeed, it has been shown that different CB2 receptor-selective agonists exert protective effects in different PD models. Moreover, the alterations in the expression of CB2 receptors observed in brain tissues from PD animal models and PD patients suggest the potential value of CB2 receptors as possible novel biomarkers for PD. However, to date, there is no direct evidence of the role of CB2 receptors in PD. Further studies are strongly needed in order to fully clarify the role of CB2 receptors in PD and thus pave the way to novel possible diagnostic and therapeutic opportunities for PD.
Collapse
Affiliation(s)
- Maria Sofia Basile
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
46
|
Siafaka PI, Okur ME, Erim PD, Çağlar EŞ, Özgenç E, Gündoğdu E, Köprülü REP, Karantas ID, Üstündağ Okur N. Protein and Gene Delivery Systems for Neurodegenerative Disorders: Where Do We Stand Today? Pharmaceutics 2022; 14:2425. [PMID: 36365243 PMCID: PMC9698227 DOI: 10.3390/pharmaceutics14112425] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2023] Open
Abstract
It has been estimated that every year, millions of people are affected by neurodegenerative disorders, which complicate their lives and their caregivers' lives. To date, there has not been an approved pharmacological approach to provide the complete treatment of neurodegenerative disorders. The only available drugs may only relieve the symptoms or slow down the progression of the disease. The absence of any treatment is quite rational given that neurodegeneration occurs by the progressive loss of the function or structure of the nerve cells of the brain or the peripheral nervous system, which eventually leads to their death either by apoptosis or necrotic cell death. According to a recent study, even though adult brain cells are injured, they can revert to an embryonic state, which may help to restore their function. These interesting findings might open a new path for the development of more efficient therapeutic strategies to combat devastating neurodegenerative disorders. Gene and protein therapies have emerged as a rapidly growing field for various disorders, especially neurodegenerative diseases. Despite these promising therapies, the complete treatment of neurodegenerative disorders has not yet been achieved. Therefore, the aim of this review is to address the most up-to-date data for neurodegenerative diseases, but most importantly, to summarize the available delivery systems incorporating proteins, peptides, and genes that can potentially target such diseases and pass into the blood-brain barrier. The authors highlight the advancements, at present, on delivery based on the carrier, i.e., lipid, polymeric, and inorganic, as well as the recent studies on radiopharmaceutical theranostics.
Collapse
Affiliation(s)
| | - Mehmet Evren Okur
- Department of Pharmacology, Faculty of Pharmacy, University of Health Sciences, Istanbul 34668, Turkey
| | - Pelin Dilsiz Erim
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul 34810, Turkey
- Faculty of Pharmacy, Altınbaş University, Istanbul 34217, Turkey
| | - Emre Şefik Çağlar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Health Sciences, Istanbul 34668, Turkey
| | - Emre Özgenç
- Department of Radiopharmacy, Faculty of Pharmacy, Ege University, Izmir 35040, Turkey
| | - Evren Gündoğdu
- Department of Radiopharmacy, Faculty of Pharmacy, Ege University, Izmir 35040, Turkey
| | - Rabia Edibe Parlar Köprülü
- Department of Medical Pharmacology, Institute of Health Sciences, İstanbul Medipol University, Istanbul 34810, Turkey
| | | | - Neslihan Üstündağ Okur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul 34668, Turkey
| |
Collapse
|
47
|
Faria P, Pacheco C, Moura RP, Sarmento B, Martins C. Multifunctional nanomedicine strategies to manage brain diseases. Drug Deliv Transl Res 2022; 13:1322-1342. [PMID: 36344871 DOI: 10.1007/s13346-022-01256-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Brain diseases represent a substantial social and economic burden, currently affecting one in six individuals worldwide. Brain research has been focus of great attention in order to unravel the pathogenesis and complexity of brain diseases at the cellular, molecular, and microenvironmental levels. Due to the intrinsic nature of the brain, the presence of the highly restrictive blood-brain barrier (BBB), and the pathophysiology of most diseases, therapies can hardly be considered successful purely by the administration of one drug to a patient. Apart from improving pharmacokinetic parameters, tailoring biodistribution, and reducing the number of side effects, nanomedicines are able to actively co-target the therapeutics to the brain parenchyma and brain lesions, as well as to achieve the delivery of multiple cargos with therapeutic, diagnostic, and theranostic properties. Among other multivalent effects that can be personalized according to the disease needs, this represents a promising class of novel nanosystems, termed multifunctional nanomedicines. Herein, we review the principal mechanisms of therapeutic resistance of the most prevalent brain diseases, how to overcome this therapeutic resistance through the use of multifunctional nanomedicines that tackle multiple fronts of the disease microenvironment, and the promising therapeutic responses achieved by some of the most cutting-edge multifunctional nanomedicines reported in literature.
Collapse
|
48
|
Nose-to-brain delivery of rotigotine redispersible nanosuspension: In vitro and in vivo characterization. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
49
|
Dantas CG, da Paixão AO, Nunes TLGM, Silva IJF, dos S. Lima B, Araújo AAS, de Albuquerque-Junior RLC, Gramacho KP, Padilha FF, da Costa LP, Severino P, Cardoso JC, Souto EB, Gomes MZ. Africanized Bee Venom ( Apis mellifera Linnaeus): Neuroprotective Effects in a Parkinson's Disease Mouse Model Induced by 6-hydroxydopamine. TOXICS 2022; 10:583. [PMID: 36287863 PMCID: PMC9609968 DOI: 10.3390/toxics10100583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
This study evaluated the neuroprotective effects of the Africanized bee venom (BV) and its mechanisms of action after 6-hydroxydopamine-(6-OHDA)-induced lesion in a mice model. Prior to BV treatment, mice received intrastriatal microinjections of 6-OHDA (no induced dopaminergic neuronal death) or ascorbate saline (as a control). BV was administered subcutaneously at different dosages (0.01, 0.05 or 0.1 mg·Kg-1) once every two days over a period of 3 weeks. The open field test was carried out, together with the immunohistochemical and histopathological analysis. The chemical composition of BV was also assessed, identifying the highest concentrations of apamin, phospholipase A2 and melittin. In the behavioral evaluation, the BV (0.1 mg·Kg-1) counteracted the 6-OHDA-induced decrease in crossings and rearing. 6-OHDA caused loss of dopaminergic cell bodies in the substantia nigra pars compacta and fibers in striatum (STR). Mice that received 0.01 mg·Kg-1 showed significant increase in the mean survival of dopaminergic cell bodies. Increased astrocytic infiltration occurred in the STR of 6-OHDA injected mice, differently from those of the groups treated with BV. The results suggested that Africanized BV has neuroprotective activity in an animal model of Parkinson's disease.
Collapse
Affiliation(s)
- Camila G. Dantas
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Ailma O. da Paixão
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Tássia L. G. M. Nunes
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Italo J. F. Silva
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Bruno dos S. Lima
- Department of Pharmacy, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | - Adriano A. S. Araújo
- Department of Pharmacy, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | | | - Kátia P. Gramacho
- Department of Animal Science, Rural Federal University of Semi-Árido (U.F.E.R.S.A), Av. Francisco Mota, Costa e Silva, Mossoró 49032-490, Natal, Brazil
| | - Francine F. Padilha
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Luiz P. da Costa
- Post-Graduation Program in Chemistry, Federal University of Sergipe (U.F.S.), Cidade Universitária Prof. José Aloísio de Campos, Av. Marechal Rondon, Jardim Rosa Elze, São Cristóvão 49100-000, Sergipe, Brazil
| | - Patricia Severino
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Juliana C. Cardoso
- Institute of Research and Technology, Tiradentes University, Av. Murilo Dantas, 300, Aracaju 49032-490, Sergipe, Brazil
| | - Eliana B. Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Margarete Z. Gomes
- Department of Animal Science, Rural Federal University of Semi-Árido (U.F.E.R.S.A), Av. Francisco Mota, Costa e Silva, Mossoró 49032-490, Natal, Brazil
| |
Collapse
|
50
|
Khairnar RC, Parihar N, Prabhavalkar KS, Bhatt LK. Emerging targets signaling for inflammation in Parkinson's disease drug discovery. Metab Brain Dis 2022; 37:2143-2161. [PMID: 35536461 DOI: 10.1007/s11011-022-00999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/29/2022] [Indexed: 10/18/2022]
Abstract
Parkinson's disease (PD) patients not only show motor features such as bradykinesia, tremor, and rigidity but also non-motor features such as anxiety, depression, psychosis, memory loss, attention deficits, fatigue, sexual dysfunction, gastrointestinal issues, and pain. Many pharmacological treatments are available for PD patients; however, these treatments are partially or transiently effective since they only decrease the symptoms. As these therapies are unable to restore dopaminergic neurons and stop the development of Parkinson's disease, therefore, the need for an effective therapeutic approach is required. The current review summarizes novel targets for PD, that can be utilized to identify disease-modifying treatments.
Collapse
Affiliation(s)
- Rhema Chandan Khairnar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|