1
|
Yang ZF, Jiang XC, Gao JQ. Present insights into the progress in gene therapy delivery systems for central nervous system diseases. Int J Pharm 2024; 669:125069. [PMID: 39662855 DOI: 10.1016/j.ijpharm.2024.125069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Central nervous system (CNS) diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), spinal cord injury (SCI), and ischemic strokes and certain rare diseases, such as amyotrophic lateral sclerosis (ALS) and ataxia, present significant obstacles to treatment using conventional molecular pharmaceuticals. Gene therapy, with its ability to target previously "undruggable" proteins with high specificity and safety, is increasingly utilized in both preclinical and clinical research for CNS ailments. As our comprehension of the pathophysiology of these conditions deepens, gene therapy stands out as a versatile and promising strategy with the potential to both prevent and treat these diseases. Despite the remarkable progress in refining and enhancing the structural design of gene therapy agents, substantial obstacles persist in their effective and safe delivery within living systems. To surmount these obstacles, a diverse array of gene delivery systems has been devised and continuously improved. Notably, Adeno-Associated Virus (AAVs)-based viral gene vectors and lipid-based nanocarriers have each advanced the in vivo delivery of gene therapies to various extents. This review aims to concisely summarize the pathophysiological foundations of CNS diseases and to shed light on the latest advancements in gene delivery vector technologies. It discusses the primary categories of these vectors, their respective advantages and limitations, and their specialized uses in the context of gene therapy delivery.
Collapse
Affiliation(s)
- Ze-Feng Yang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Chi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| | - Jian-Qing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| |
Collapse
|
2
|
Knez D, Wang F, Duan WX, Hrast Rambaher M, Gobec S, Cheng XY, Wang XB, Mao CJ, Liu CF, Frlan R. Development of novel aza-stilbenes as a new class of selective MAO-B inhibitors for the treatment of Parkinson's disease. Bioorg Chem 2024; 153:107877. [PMID: 39396452 DOI: 10.1016/j.bioorg.2024.107877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/21/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of nigrostriatal dopaminergic neurons. Inhibitors of monoamine oxidase B (MAO-B) have shown promise in alleviating motor symptoms and reducing oxidative stress associated with PD. In this study, we report the novel use of an azastilbene-based compound library for screening human (h)MAO-B, followed by optimization of initial hits to obtain compounds with low nanomolar inhibitory potencies (compound 9, IC50 = 42 nM) against hMAO-B. To ensure specificity and minimize false positives due to non-specific hydrophobic interactions, we performed comprehensive selectivity profiling against hMAO-A, butyrylcholinesterase (hBChE) and acetylcholinesterase (hAChE) - enzymes with hydrophobic active sites that are structurally distinct from hMAO-B. Docking analysis with Glide provided valuable insights into the binding interactions between the inhibitors and hMAO-B and also explained the selectivity against hMAO-A. In the cell-based model of Parkinson's disease, one of the compounds significantly reduced rotenone-induced accumulation of reactive oxygen species. In addition, these compounds showed a protective effect against acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced motor dysfunction in PD model mice and reduced MPTP-induced loss of striatal tyrosine hydroxylase-positive neurons in the substantia nigra. These results make azastilbene-based compounds a promising new class of hMAO-B inhibitors with potential therapeutic applications in Parkinson's disease and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Wen-Xiang Duan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Martina Hrast Rambaher
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Xiao-Yu Cheng
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xiao-Bo Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Rok Frlan
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
3
|
Kale MB, Wankhede NL, Bishoyi AK, Ballal S, Kalia R, Arya R, Kumar S, Khalid M, Gulati M, Umare M, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Fareed M, Koppula S. Emerging biophysical techniques for probing synaptic transmission in neurodegenerative disorders. Neuroscience 2024:S0306-4522(24)00643-2. [PMID: 39608699 DOI: 10.1016/j.neuroscience.2024.11.055] [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: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Plethora of research has shed light on the critical role of synaptic dysfunction in various neurodegenerative disorders (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Synapses, the fundamental units for neural communication in the brain, are highly vulnerable to pathological conditions and are central to the progression of neurological diseases. The presynaptic terminal, a key component of synapses responsible for neurotransmitter release and synaptic communication, undergoes structural and functional alterations in these disorders. Understanding synaptic transmission abnormalities is crucial for unravelling the pathophysiological mechanisms underlying neurodegeneration. In the quest to probe synaptic transmission in NDDs, emerging biophysical techniques play a pivotal role. These advanced methods offer insights into the structural and functional changes occurring at nerve terminals in conditions like AD, PD, HD & ALS. By investigating synaptic plasticity and alterations in neurotransmitter release dynamics, researchers can uncover valuable information about disease progression and potential therapeutic targets. The review articles highlighted provide a comprehensive overview of how synaptic vulnerability and pathology are shared mechanisms across a spectrum of neurological disorders. In major neurodegenerative diseases, synaptic dysfunction is a common thread linking these conditions. The intricate molecular machinery involved in neurotransmitter release, synaptic vesicle dynamics, and presynaptic protein regulation are key areas of focus for understanding synaptic alterations in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Mohit Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11597, Saudi Arabia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
4
|
N M P, Fullard JF, Clarence T, Mathur D, Casey C, Hennigan E, Alvia M, Krause-Massaguer J, Barreda A, Davis DA, Vontell RT, Garamszegi SP, Vance JM, Sang L, Chatigny M, Vismer D, Landin B, Burstein D, Lee D, Voloudakis G, Berretta S, Haroutunian V, Scott WK, Bendl J, Roussos P. A multi-region single nucleus transcriptomic atlas of Parkinson's disease. Sci Data 2024; 11:1274. [PMID: 39580497 PMCID: PMC11585549 DOI: 10.1038/s41597-024-04117-y] [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: 05/10/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
Parkinson's Disease (PD) is a debilitating neurodegenerative disorder, characterized by motor and cognitive impairments, that affects >1% of the population over the age of 60. The pathogenesis of PD is complex and remains largely unknown. Due to the cellular heterogeneity of the human brain and changes in cell type composition with disease progression, this complexity cannot be fully captured with bulk tissue studies. To address this, we generated single-nucleus RNA sequencing and whole-genome sequencing data from 100 postmortem cases and controls, carefully selected to represent the entire spectrum of PD neuropathological severity and diverse clinical symptoms. The single nucleus data were generated from five brain regions, capturing the subcortical and cortical spread of PD pathology. Rigorous preprocessing and quality control were applied to ensure data reliability. Committed to collaborative research and open science, this dataset is available on the AMP PD Knowledge Platform, offering researchers a valuable tool to explore the molecular bases of PD and accelerate advances in understanding and treating the disease.
Collapse
Affiliation(s)
- Prashant N M
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepika Mathur
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Clara Casey
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evelyn Hennigan
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcela Alvia
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joana Krause-Massaguer
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ayled Barreda
- Brain Endowment Bank, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David A Davis
- Brain Endowment Bank, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Regina T Vontell
- Brain Endowment Bank, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Susanna P Garamszegi
- Brain Endowment Bank, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | - David Burstein
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Sabina Berretta
- McLean Hospital, Belmont, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William K Scott
- Brain Endowment Bank, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
5
|
Kim NH, Lee YA. The Effects of Nanoplastics on the Dopamine System of Cerebrocortical Neurons. Int J Toxicol 2024:10915818241293993. [PMID: 39486087 DOI: 10.1177/10915818241293993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Nanoplastics (NPx) can enter living organisms, including humans, through ecosystems, inhalation, and dermal contact and can be found from the intestine to the brain. However, it is unclear whether NPx accumulates and affects the dopamine system. In this study, we investigated the effects of NPx on the dopamine system in cultured murine cerebral cortex neurons. Cultured cerebrocortical neurons were treated with 100 nm NPx at the following concentrations for 24 h: 1.896 × 105, 3.791 × 106, 7.583 × 107, 1.571 × 109, 3.033 × 1010, and 3.033 × 1011 particles/mL. Dopamine-associated proteins were analyzed using immunofluorescence staining. NPx treatment induced its accumulation in neurons in a dose-dependent manner and increased the levels of dopamine receptors D1 and D2 and their co-expression. However, NPx treatment did not affect the levels of other dopamine receptors, dopamine transporters, tyrosine hydroxylase, and microtubule-associated protein 2, or synaptophysin in neuronal structures. This study demonstrated that NPx is a potential modulator of the dopamine system via its receptors rather than its synthesis and reuptake in neurons and may be associated with dopamine-based psychiatric disorders.
Collapse
Affiliation(s)
- Na-Hyun Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Young-A Lee
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, Republic of Korea
| |
Collapse
|
6
|
Farhangian M, Azarafrouz F, Valian N, Dargahi L. The role of interferon beta in neurological diseases and its potential therapeutic relevance. Eur J Pharmacol 2024; 981:176882. [PMID: 39128808 DOI: 10.1016/j.ejphar.2024.176882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Interferon beta (IFNβ) is a member of the type-1 interferon family and has various immunomodulatory functions in neuropathological conditions. Although the level of IFNβ is low under healthy conditions, it is increased during inflammatory processes to protect the central nervous system (CNS). In particular, microglia and astrocytes are the main sources of IFNβ upon inflammatory insult in the CNS. The protective effects of IFNβ are well characterized in reducing the progression of multiple sclerosis (MS); however, little is understood about its effects in other neurological/neurodegenerative diseases. In this review, different types of IFNs and their signaling pathways will be described. Then we will focus on the potential role and therapeutic effect of IFNβ in several CNS-related diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury, prion disease and spinocerebellar ataxia 7.
Collapse
Affiliation(s)
- Mohsen Farhangian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forouzan Azarafrouz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Vahidinia Z, Barati S, Azami Tameh A, Bagheri-Mohammadi S, Garshasebi A. Bee venom as a promising therapeutic strategy in central nervous system diseases. Neuropeptides 2024; 107:102451. [PMID: 38936137 DOI: 10.1016/j.npep.2024.102451] [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: 05/17/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Central nervous system (CNS) disorders are one of the leading health problems today, accounting for a large proportion of global morbidity and mortality. Most these disorders are characterized by high levels of oxidative stress and intense inflammatory responses in degenerated neuronal tissues. While extensive research has been conducted on CNS diseases, but few breakthroughs have been made in treatment methods. To date, there are no disease-modifying drugs available for CNS treatment, underscoring the urgent need for finding effective medications. Bee venom (BV), which is produced by honeybee workers' stingers, has been a subject of interest and study across various cultures. Over the past few decades, extensive research has focused on BV and its therapeutic potentials. BV consists a variety of substances, mainly proteins and peptides like melittin and phospholipase A2 (PLA2). Research has proven that BV is effective in various medical conditions, including pain, arthritis and inflammation and CNS disorders such as Multiple sclerosis, Alzheimer's disease and Parkinson's disease. This review provides a comprehensive overview of the existing knowledge concerning the therapeutic effects of BV and its primary compounds on various CNS diseases. Additionally, we aim to shed light on the potential cellular and molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeid Bagheri-Mohammadi
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran.; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Garshasebi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Fisher RMA, Torrente MP. Histone post-translational modification and heterochromatin alterations in neurodegeneration: revealing novel disease pathways and potential therapeutics. Front Mol Neurosci 2024; 17:1456052. [PMID: 39346681 PMCID: PMC11427407 DOI: 10.3389/fnmol.2024.1456052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), Frontotemporal Dementia (FTD), and Amyotrophic lateral sclerosis (ALS) are complex and fatal neurodegenerative diseases. While current treatments for these diseases do alleviate some symptoms, there is an imperative need for novel treatments able to stop their progression. For all of these ailments, most cases occur sporadically and have no known genetic cause. Only a small percentage of patients bear known mutations which occur in a multitude of genes. Hence, it is clear that genetic factors alone do not explain disease occurrence. Chromatin, a DNA-histone complex whose basic unit is the nucleosome, is divided into euchromatin, an open form accessible to the transcriptional machinery, and heterochromatin, which is closed and transcriptionally inactive. Protruding out of the nucleosome, histone tails undergo post-translational modifications (PTMs) including methylation, acetylation, and phosphorylation which occur at specific residues and are connected to different chromatin structural states and regulate access to transcriptional machinery. Epigenetic mechanisms, including histone PTMs and changes in chromatin structure, could help explain neurodegenerative disease processes and illuminate novel treatment targets. Recent research has revealed that changes in histone PTMs and heterochromatin loss or gain are connected to neurodegeneration. Here, we review evidence for epigenetic changes occurring in AD, PD, and FTD/ALS. We focus specifically on alterations in the histone PTMs landscape, changes in the expression of histone modifying enzymes and chromatin remodelers as well as the consequences of these changes in heterochromatin structure. We also highlight the potential for epigenetic therapies in neurodegenerative disease treatment. Given their reversibility and pharmacological accessibility, epigenetic mechanisms provide a promising avenue for novel treatments. Altogether, these findings underscore the need for thorough characterization of epigenetic mechanisms and chromatin structure in neurodegeneration.
Collapse
Affiliation(s)
- Raven M. A. Fisher
- PhD. Program in Biochemistry, City University of New York - The Graduate Center, New York, NY, United States
| | - Mariana P. Torrente
- Department of Chemistry and Biochemistry, Brooklyn College, Brooklyn, NY, United States
- PhD. Programs in Chemistry, Biochemistry, and Biology, City University of New York - The Graduate Center, New York, NY, United States
| |
Collapse
|
9
|
Asuku AO, Ayinla MT, Olajide TS, Oyerinde TO, Yusuf JA, Bayo-Olugbami AA, Fajemidagba GA. Coffee and Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2024; 289:1-19. [PMID: 39168575 DOI: 10.1016/bs.pbr.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease marked by dopaminergic neuronal loss and misfolded alpha-synuclein (α-syn) accumulation, which results in both motor and cognitive symptoms. Its occurrence grows with age, with a larger prevalence among males. Despite substantial study, effective medicines to reduce or stop the progression of diseases remain elusive. Interest has grown in examining dietary components, such as caffeine present in coffee, for potential medicinal effects. Epidemiological studies imply a lower incidence of PD with coffee drinking, attributable to caffeine's neuroprotective abilities. Beyond caffeine, coffee constituent like chlorogenic acid and cafestol have anti-Parkinsonian benefits. Moreover, coffee use has been related with variations in gut microbiota composition, which may reduce intestinal inflammation and prevent protein misfolding in enteric nerves, perhaps through the microbiota-gut-brain axis. This review gives a summary of the neuroprotective effects of coffee, investigating both its motor and non-motor advantages in individuals with PD as well as in experimental models of PD. We reviewed some bioactive constituents of coffee, their respective interactions with misfolded α-syn accumulation, and its emerging mechanisms associated to the gut microbiome.
Collapse
Affiliation(s)
- Abraham Olufemi Asuku
- Bioresources Development Centre, National Biotechnology Research and Development Agency, Ogbomoso, Oyo, Nigeria; Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria.
| | - Maryam Tayo Ayinla
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| | - Tobiloba Samuel Olajide
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Toheeb O Oyerinde
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Joshua Ayodele Yusuf
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | | | | |
Collapse
|
10
|
Khan A, Ezeugwa J, Ezeugwu VE. A systematic review of the associations between sedentary behavior, physical inactivity, and non-motor symptoms of Parkinson's disease. PLoS One 2024; 19:e0293382. [PMID: 38551932 PMCID: PMC10980241 DOI: 10.1371/journal.pone.0293382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 02/19/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD), known for motor symptoms, often presents early non-motor issues that significantly affect patients' quality of life. While effective treatments are limited, physical activity and exercise offer potential benefits. However, an overlooked aspect of the movement intensity continuum is prolonged sitting or sedentary behavior, and physical inactivity. Thus, this study aimed to conduct a systematic review investigating the associations between sedentary behavior, physical inactivity, and non-motor symptoms, specifically cognitive impairment, depression, and poor sleep in PD. METHODS Conforming to PRISMA guidelines, a systematic search of the literature was conducted via electronic databases including MEDLINE, CINAHL, Scopus, PubMed and PsycINFO up to February 28, 2023. Studies were included if they investigated associations between sedentary behavior or physical inactivity and at least one non-motor symptom such as depression, poor sleep, and/or cognitive impairment, in adults aged 18 years or older with PD. Quality assessment of the studies was performed using the Newcastle-Ottawa scale for cross-sectional and cohort studies. RESULTS Of the 463 publications found, 7 studies met the inclusion criteria (n = 980 unique participants). Sample sizes ranged from 17 to 487 participants, and all studies were observational, conducted in home or community settings. Collectively, these studies show that higher amounts of both objectively-measured and self-reported sedentary time are associated with worse scores on standardized measures of cognition and the Parkinson's Disease Questionnaire (PDQ) summary index and its subscales, such as cognition (memory and concentration). Additionally, longitudinal cohort studies suggest that physical inactivity and higher sedentary behavior are associated with depression and cognitive impairment in PD. Less sleep was associated with higher sedentary behavior. CONCLUSION Associations observed between physical inactivity, sedentary behavior, and non-motor symptoms in PD underscore the need to address these factors for enhanced well-being. Further well-designed studies are essential to assess the impact of reducing sedentary behavior and physical inactivity on non-motor symptoms in PD. Prospero registration number: PROSPERO (ID: CRD42023405422) on April 11, 2023.
Collapse
Affiliation(s)
- Aiza Khan
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Joy Ezeugwa
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Victor E. Ezeugwu
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Pandey M, Karmakar V, Majie A, Dwivedi M, Md S, Gorain B. The SH-SY5Y cell line: a valuable tool for Parkinson's disease drug discovery. Expert Opin Drug Discov 2024; 19:303-316. [PMID: 38112196 DOI: 10.1080/17460441.2023.2293158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Owing to limited efficient treatment strategies for highly prevalent and distressing Parkinson's disease (PD), an impending need emerged for deciphering new modes and mechanisms for effective management. SH-SY5Y-based in vitro neuronal models have emerged as a new possibility for the elucidation of cellular and molecular processes in the pathogenesis of PD. SH-SY5Y cells are of human origin, adhered to catecholaminergic neuronal attributes, which consequences in imparting wide acceptance and significance to this model over conventional in vitro PD models for high-throughput screening of therapeutics. AREAS COVERED Herein, the authors review the SH-SY5Y cell line and its value to PD research. The authors also provide the reader with their expert perspectives on how these developments can lead to the development of new impactful therapeutics. EXPERT OPINION Encouraged by recent research on SH-SY5Y cell lines, it was envisaged that this in vitro model can serve as a primary model for assessing efficacy and toxicity of new therapeutics as well as for nanocarriers' capacity in delivering therapeutic agents across BBB. Considering the proximity with human neuronal environment as in pathogenic PD conditions, SH-SY5Y cell lines vindicated consistency and reproducibility in experimental results. Accordingly, exploitation of this standardized SH-SY5Y cell line can fast-track the drug discovery and development path for novel therapeutics.
Collapse
Affiliation(s)
- Manisha Pandey
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Monika Dwivedi
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| |
Collapse
|
12
|
Tan JB, Müller EJ, Orlando IF, Taylor NL, Margulies DS, Szeto J, Lewis SJG, Shine JM, O'Callaghan C. Abnormal higher-order network interactions in Parkinson's disease visual hallucinations. Brain 2024; 147:458-471. [PMID: 37677056 DOI: 10.1093/brain/awad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/14/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
Visual hallucinations in Parkinson's disease can be viewed from a systems-level perspective, whereby dysfunctional communication between brain networks responsible for perception predisposes a person to hallucinate. To this end, abnormal functional interactions between higher-order and primary sensory networks have been implicated in the pathophysiology of visual hallucinations in Parkinson's disease, however the precise signatures remain to be determined. Dimensionality reduction techniques offer a novel means for simplifying the interpretation of multidimensional brain imaging data, identifying hierarchical patterns in the data that are driven by both within- and between-functional network changes. Here, we applied two complementary non-linear dimensionality reduction techniques-diffusion-map embedding and t-distributed stochastic neighbour embedding (t-SNE)-to resting state functional MRI data, in order to characterize the altered functional hierarchy associated with susceptibility to visual hallucinations. Our study involved 77 people with Parkinson's disease (31 with hallucinations; 46 without hallucinations) and 19 age-matched healthy control subjects. In patients with visual hallucinations, we found compression of the unimodal-heteromodal gradient consistent with increased functional integration between sensory and higher order networks. This was mirrored in a traditional functional connectivity analysis, which showed increased connectivity between the visual and default mode networks in the hallucinating group. Together, these results suggest a route by which higher-order regions may have excessive influence over earlier sensory processes, as proposed by theoretical models of hallucinations across disorders. By contrast, the t-SNE analysis identified distinct alterations in prefrontal regions, suggesting an additional layer of complexity in the functional brain network abnormalities implicated in hallucinations, which was not apparent in traditional functional connectivity analyses. Together, the results confirm abnormal brain organization associated with the hallucinating phenotype in Parkinson's disease and highlight the utility of applying convergent dimensionality reduction techniques to investigate complex clinical symptoms. In addition, the patterns we describe in Parkinson's disease converge with those seen in other conditions, suggesting that reduced hierarchical differentiation across sensory-perceptual systems may be a common transdiagnostic vulnerability in neuropsychiatric disorders with perceptual disturbances.
Collapse
Affiliation(s)
- Joshua B Tan
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| | - Eli J Müller
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
- Centre for Complex Systems, School of Physics, University of Sydney, Sydney 2050, Australia
| | - Isabella F Orlando
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| | - Natasha L Taylor
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| | - Daniel S Margulies
- Integrative Neuroscience and Cognition Center, Center National de la Recherche Scientifique (CNRS) and Université de Paris, 75006 Paris, France
| | - Jennifer Szeto
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| | - Simon J G Lewis
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| | - James M Shine
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
- Centre for Complex Systems, School of Physics, University of Sydney, Sydney 2050, Australia
| | - Claire O'Callaghan
- Brain and Mind Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2050, Australia
| |
Collapse
|
13
|
Skibska A, Perlikowska R. Natural Plant Materials as a Source of Neuroprotective Peptides. Curr Med Chem 2024; 31:5027-5045. [PMID: 37403392 DOI: 10.2174/0929867331666230703145043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
In many circumstances, some crucial elements of the neuronal defense system fail, slowly leading to neurodegenerative diseases. Activating this natural process by administering exogenous agents to counteract unfavourable changes seems promising. Therefore, looking for neuroprotective therapeutics, we have to focus on compounds that inhibit the primary mechanisms leading to neuronal injuries, e.g., apoptosis, excitotoxicity, oxidative stress, and inflammation. Among many compounds considered neuroprotective agents, protein hydrolysates and peptides derived from natural materials or their synthetic analogues are good candidates. They have several advantages, such as high selectivity and biological activity, a broad range of targets, and high safety profile. This review aims to provide biological activities, the mechanism of action and the functional properties of plant-derived protein hydrolysates and peptides. We focused on their significant role in human health by affecting the nervous system and having neuroprotective and brain-boosting properties, leading to memory and cognitive improving activities. We hope our observation may guide the evaluation of novel peptides with potential neuroprotective effects. Research into neuroprotective peptides may find application in different sectors as ingredients in functional foods or pharmaceuticals to improve human health and prevent diseases.
Collapse
Affiliation(s)
- Agnieszka Skibska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| | - Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University, Lodz, Poland
| |
Collapse
|
14
|
Herkes G, McGee C, Liebert A, Bicknell B, Isaac V, Kiat H, McLachlan CS. A novel transcranial photobiomodulation device to address motor signs of Parkinson's disease: a parallel randomised feasibility study. EClinicalMedicine 2023; 66:102338. [PMID: 38094162 PMCID: PMC10716000 DOI: 10.1016/j.eclinm.2023.102338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Parkinson's disease is a progressive neurological disease with limited treatment options. Animal models and a proof-of-concept case series have suggested that photobiomodulation may be an effective adjunct treatment for the symptoms of Parkinson's disease. The aim was to determine the safety and feasibility of transcranial photobiomodulation (tPBM) to reduce the motor signs of Parkinson's disease. METHODS In this double-blind, randomised, sham-controlled feasibility trial, patients (aged 59-85 years) with idiopathic Parkinson's disease were treated with a tPBM helmet for 12 weeks (72 treatments with either active or sham therapy; stage 1). Treatment was delivered in the participants' homes, monitored by internet video conferencing (Zoom). Stage 1 was followed by 12 weeks of no treatment for those on active therapy (active-to-no-treatment group), and 12 weeks of active treatment for those on sham (sham-to-active group), for participants who chose to continue (stage 2). The active helmet device delivered red and infrared light to the head for 24 min, 6 days per week. The primary endpoints were safety and motor signs, as assessed by a modified Movement Disorders Society revision of the Unified Parkinson's Disease Rating Scale Part III (MDS-UPDRS-III)-motor scale. This trial is registered with ANZCTR, ACTRN 12621001722886. FINDINGS Between Dec 6, 2021, and Aug 12, 2022, 20 participants were randomly allocated to each of the two groups (10 females plus 10 males per group). All participants in the active group and 18 in the sham group completed 12 weeks of treatment. 14 participants in the sham group chose to continue to active treatment and 12 completed the full 12 weeks of active treatment. Treatment was well tolerated and feasible to deliver, with only minor, temporary adverse events. Of the nine suspected adverse events that were identified, two minor reactions may have been attributable to the device in the sham-to-active group during the active treatment weeks of the trial. One participant experienced temporary leg weakness. A second participant reported decreased fine motor function in the right hand. Both participants continued the trial. The mean modified MDS-UPDRS-III scores for the sham-to-active group at baseline, after 12 weeks of sham treatment, and after 12 weeks of active treatment were 26.8 (sd 14.6), 20.4 (sd 12.8), and 12.2 (sd 8.9), respectively, and for the active-to-no-treatment group these values were 21.3 (sd 9.4), 16.5 (sd 9.4), and 15.3 (sd 10.8), respectively. There was no significant difference between groups at any assessment point. The mean difference between groups at baseline was 5.5 (95% confidence interval (CI) -2.4 to 13.4), after stage 1 was 3.9 (95% CI -3.5 to 11.3 and after stage 2 was -3.1 (95% CI 2.7 to -10.6). INTERPRETATION Our findings add to the evidence base to suggest that tPBM is a safe, tolerable, and feasible non-pharmaceutical adjunct therapy for Parkinson's disease. While future work is needed our results lay the foundations for an adequately powered randomised placebo-controlled clinical trial. FUNDING SYMBYX Pty Ltd.
Collapse
Affiliation(s)
- Geoffrey Herkes
- Department of Neurology, Sydney Adventist Hospital, Wahroonga, NSW, 2076, Australia
| | - Claire McGee
- Faculty of Health Sciences, Torrens University Australia, Sydney, NSW, 2000, Australia
| | - Ann Liebert
- Sydney Adventist Hospital, Wahroonga, NSW, 2076, Australia
- School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, 2145, Australia
| | - Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, 2145, Australia
| | - Vivian Isaac
- School of Allied Health, Exercise & Sports Sciences, Charles Sturt University, Albury, NSW, 2640, Australia
| | - Hosen Kiat
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
- College of Health and Medicine, Australian National University, Canberra, ACT, 2601, Australia
- Cardiac Health Institute, Sydney, NSW, 2010, Australia
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, 2000, Australia
| | - Craig S. McLachlan
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, 2000, Australia
| |
Collapse
|
15
|
Slézia A, Hegedüs P, Rusina E, Lengyel K, Solari N, Kaszas A, Balázsfi D, Botzanowski B, Acerbo E, Missey F, Williamson A, Hangya B. Behavioral, neural and ultrastructural alterations in a graded-dose 6-OHDA mouse model of early-stage Parkinson's disease. Sci Rep 2023; 13:19478. [PMID: 37945922 PMCID: PMC10636184 DOI: 10.1038/s41598-023-46576-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Studying animal models furthers our understanding of Parkinson's disease (PD) pathophysiology by providing tools to investigate detailed molecular, cellular and circuit functions. Different versions of the neurotoxin-based 6-hydroxydopamine (6-OHDA) model of PD have been widely used in rats. However, these models typically assess the result of extensive and definitive dopaminergic lesions that reflect a late stage of PD, leading to a paucity of studies and a consequential gap of knowledge regarding initial stages, in which early interventions would be possible. Additionally, the better availability of genetic tools increasingly shifts the focus of research from rats to mice, but few mouse PD models are available yet. To address these, we characterize here the behavioral, neuronal and ultrastructural features of a graded-dose unilateral, single-injection, striatal 6-OHDA model in mice, focusing on early-stage changes within the first two weeks of lesion induction. We observed early onset, dose-dependent impairments of overall locomotion without substantial deterioration of motor coordination. In accordance, histological evaluation demonstrated a partial, dose-dependent loss of dopaminergic neurons of substantia nigra pars compacta (SNc). Furthermore, electron microscopic analysis revealed degenerative ultrastructural changes in SNc dopaminergic neurons. Our results show that mild ultrastructural and cellular degradation of dopaminergic neurons of the SNc can lead to certain motor deficits shortly after unilateral striatal lesions, suggesting that a unilateral dose-dependent intrastriatal 6-OHDA lesion protocol can serve as a successful model of the early stages of Parkinson's disease in mice.
Collapse
Affiliation(s)
- Andrea Slézia
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary.
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France.
- Institute of Cognitive Neuroscience and Psychology, Eotvos Lorand Research Network, Budapest, Hungary.
- Institut de Neurosciences de la Timone, CNRS UMR 7289, Aix-Marseille Université, Marseille, France.
| | - Panna Hegedüs
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Evgeniia Rusina
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France
| | - Katalin Lengyel
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary
| | - Nicola Solari
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary
| | - Attila Kaszas
- Institut de Neurosciences de la Timone, CNRS UMR 7289, Aix-Marseille Université, Marseille, France
| | - Diána Balázsfi
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary
| | - Boris Botzanowski
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France
| | - Emma Acerbo
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France
| | - Florian Missey
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France
| | - Adam Williamson
- Institut de Neurosciences Des Systèmes, INSERM UMR S 1106, Aix-Marseille Université, Marseille, France.
- International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czech Republic.
| | - Balázs Hangya
- Institute of Experimental Medicine, Lendület Laboratory of Systems Neuroscience, Budapest, Hungary.
| |
Collapse
|
16
|
Ni B, Liu Y, Dai M, Zhao J, Liang Y, Yang X, Han B, Jiang M. The role of cannabidiol in aging. Biomed Pharmacother 2023; 165:115074. [PMID: 37418976 DOI: 10.1016/j.biopha.2023.115074] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/09/2023] Open
Abstract
Aging is usually considered a key risk factor associated with multiple diseases, such as neurodegenerative diseases, cardiovascular diseases and cancer. Furthermore, the burden of age-related diseases has become a global challenge. It is of great significance to search for drugs to extend lifespan and healthspan. Cannabidiol (CBD), a natural nontoxic phytocannabinoid, has been regarded as a potential candidate drug for antiaging. An increasing number of studies have suggested that CBD could benefit healthy longevity. Herein, we summarized the effect of CBD on aging and analyzed the possible mechanism. All these conclusions may provide a perspective for further study of CBD on aging.
Collapse
Affiliation(s)
- Beibei Ni
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yanying Liu
- Department of Basic Medical, Qingdao Huanghai University, Qingdao 266427, China
| | - Meng Dai
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jun Zhao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yu Liang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xue Yang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Bing Han
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Man Jiang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
17
|
Mohammad, Khan UA, Warsi MH, Alkreathy HM, Karim S, Jain GK, Ali A. Intranasal cerium oxide nanoparticles improves locomotor activity and reduces oxidative stress and neuroinflammation in haloperidol-induced parkinsonism in rats. Front Pharmacol 2023; 14:1188470. [PMID: 37324485 PMCID: PMC10267740 DOI: 10.3389/fphar.2023.1188470] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/09/2023] [Indexed: 08/26/2023] Open
Abstract
Introduction: Cerium oxide nanoparticles (CONPs) have been investigated for their therapeutic potential in Parkinson's disease (PD) due to their potent and regenerative antioxidant activity. In the present study, CONPs were used to ameliorate the oxidative stress caused by free radicals in haloperidol-induced PD in rats following intranasal administration. Method: The antioxidant potential of the CONPs was evaluated in vitro using ferric reducing antioxidant power (FRAP) assay. The penetration and local toxicity of the CONPs was evaluated ex-vivo using goat nasal mucosa. The acute local toxicity of intranasal CONPs was also studied in rat. Gamma scintigraphy was used to assess the targeted brain delivery of CONPs. Acute toxicity studies were performed in rats to demonstrate safety of intranasal CONPs. Further, open field test, pole test, biochemical estimations and brain histopathology was performed to evaluate efficacy of intranasal CONPs in haloperidol-induced PD rat model. Results: The FRAP assay revealed highest antioxidant activity of prepared CONPs at a concentration of 25 μg/mL. Confocal microscopy showed deep and homogenous distribution of CONPs in the goat nasal mucus layers. No signs of irritation or injury were seen in goat nasal membrane when treated with optimized CONPs. Scintigraphy studies in rats showed targeted brain delivery of intranasal CONPs and acute toxicity study demonstrated safety. The results of open field and pole test showed highly significant (p < 0.001) improvement in locomotor activity of rats treated with intranasal CONPs compared to untreated rats. Further, brain histopathology of treatment group rats showed reduced neurodegeneration with presence of more live cells. The amount of thiobarbituric acid reactive substances (TBARS) was reduced significantly, whereas the levels of catalase (CAT), superoxide dismutase (SOD), and GSH were increased significantly, while amounts of interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α) showed significant reduction after intranasal administration of CONPs. Also, the intranasal CONPs, significantly high (p < 0.001) dopamine concentration (13.93 ± 0.85 ng/mg protein) as compared to haloperidol-induced control rats (5.76 ± 0.70 ng/mg protein). Conclusion: The overall results concluded that the intranasal CONPs could be safe and effective therapeutics for the management of PD.
Collapse
Affiliation(s)
- Mohammad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Urooj Ahmed Khan
- Department of Pharmaceutics, Dr. Ram Manohar Lohia College of Pharmacy, Ghaziabad, Uttar Pradesh, India
| | - Musarrat Husain Warsi
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Huda Mohammed Alkreathy
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahid Karim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
- Center for Advanced Formulation Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Asgar Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| |
Collapse
|
18
|
Senevirathne DKL, Mahboob A, Zhai K, Paul P, Kammen A, Lee DJ, Yousef MS, Chaari A. Deep Brain Stimulation beyond the Clinic: Navigating the Future of Parkinson's and Alzheimer's Disease Therapy. Cells 2023; 12:1478. [PMID: 37296599 PMCID: PMC10252401 DOI: 10.3390/cells12111478] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/30/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
Deep brain stimulation (DBS) is a surgical procedure that uses electrical neuromodulation to target specific regions of the brain, showing potential in the treatment of neurodegenerative disorders such as Parkinson's disease (PD) and Alzheimer's disease (AD). Despite similarities in disease pathology, DBS is currently only approved for use in PD patients, with limited literature on its effectiveness in AD. While DBS has shown promise in ameliorating brain circuits in PD, further research is needed to determine the optimal parameters for DBS and address any potential side effects. This review emphasizes the need for foundational and clinical research on DBS in different brain regions to treat AD and recommends the development of a classification system for adverse effects. Furthermore, this review suggests the use of either a low-frequency system (LFS) or high-frequency system (HFS) depending on the specific symptoms of the patient for both PD and AD.
Collapse
Affiliation(s)
| | - Anns Mahboob
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Kevin Zhai
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Alexandra Kammen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Darrin Jason Lee
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Neurorestoration Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Mohammad S. Yousef
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| |
Collapse
|
19
|
Echeverria V, Mendoza C, Iarkov A. Nicotinic acetylcholine receptors and learning and memory deficits in Neuroinflammatory diseases. Front Neurosci 2023; 17:1179611. [PMID: 37255751 PMCID: PMC10225599 DOI: 10.3389/fnins.2023.1179611] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/07/2023] [Indexed: 06/01/2023] Open
Abstract
Animal survival depends on cognitive abilities such as learning and memory to adapt to environmental changes. Memory functions require an enhanced activity and connectivity of a particular arrangement of engram neurons, supported by the concerted action of neurons, glia, and vascular cells. The deterioration of the cholinergic system is a common occurrence in neurological conditions exacerbated by aging such as traumatic brain injury (TBI), posttraumatic stress disorder (PTSD), Alzheimer's disease (AD), and Parkinson's disease (PD). Cotinine is a cholinergic modulator with neuroprotective, antidepressant, anti-inflammatory, antioxidant, and memory-enhancing effects. Current evidence suggests Cotinine's beneficial effects on cognition results from the positive modulation of the α7-nicotinic acetylcholine receptors (nAChRs) and the inhibition of the toll-like receptors (TLRs). The α7nAChR affects brain functions by modulating the function of neurons, glia, endothelial, immune, and dendritic cells and regulates inhibitory and excitatory neurotransmission throughout the GABA interneurons. In addition, Cotinine acting on the α7 nAChRs and TLR reduces neuroinflammation by inhibiting the release of pro-inflammatory cytokines by the immune cells. Also, α7nAChRs stimulate signaling pathways supporting structural, biochemical, electrochemical, and cellular changes in the Central nervous system during the cognitive processes, including Neurogenesis. Here, the mechanisms of memory formation as well as potential mechanisms of action of Cotinine on memory preservation in aging and neurological diseases are discussed.
Collapse
Affiliation(s)
- Valentina Echeverria
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
- Research and Development Department, Bay Pines VAHCS, Bay Pines, FL, United States
| | - Cristhian Mendoza
- Facultad de Odontologia y Ciencias de la Rehabilitacion, Universidad San Sebastián, Concepción, Chile
| | - Alex Iarkov
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Concepción, Chile
| |
Collapse
|
20
|
Zamanian MY, Parra RMR, Soltani A, Kujawska M, Mustafa YF, Raheem G, Al-Awsi L, Lafta HA, Taheri N, Heidari M, Golmohammadi M, Bazmandegan G. Targeting Nrf2 signaling pathway and oxidative stress by resveratrol for Parkinson's disease: an overview and update on new developments. Mol Biol Rep 2023; 50:5455-5464. [PMID: 37155008 DOI: 10.1007/s11033-023-08409-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/24/2023] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) as a prevalent neurodegenerative condition impairs motor function and is caused by the progressive deterioration of nigrostriatal dopaminergic (DAergic) neurons. The current therapy solutions for PD are ineffective because they could not inhibit the disease's progression and they even have adverse effects. Natural polyphenols, a group of phytochemicals, have been found to offer various health benefits, including neuroprotection against PD. Among these, resveratrol (RES) has neuroprotective properties owing to its capacity to protect mitochondria and act as an antioxidant. An increase in the formation of reactive oxygen species (ROS) leads to oxidative stress (OS), which is responsible for cellular damage resulting in lipid peroxidation, oxidative protein alteration, and DNA damage. In PD models, it's been discovered that RES pretreatment can diminish oxidative stress by boosting endogenous antioxidant status and directly scavenging ROS. Several studies have examined the involvement of RES in the modulation of the transcriptional factor Nrf2 in PD models because this protein recognizes oxidants and controls the antioxidant defense. In this review, we have examined the molecular mechanisms underlying the RES activity and reviewed its effects in both in vitro and in vivo models of PD. The gathered evidence herein showed that RES treatment provides neuroprotection against PD by reducing OS and upregulation of Nrf2. Moreover, in the present study, scientific proof of the neuroprotective properties of RES against PD and the mechanism supporting clinical development consideration has been described.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | | | - Afsaneh Soltani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, Poznan, 60-631, Poland
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Ghaidaa Raheem
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Lateef Al-Awsi
- Department of Radiological Techniques, Al-Mustaqbal University College, Babylon, Iraq
| | - Holya A Lafta
- Department of Pharmacy, Al-Nisour University College, Baghdad, Iraq
| | - Niloofar Taheri
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahsa Heidari
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamreza Bazmandegan
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
21
|
Elango R, Banaganapalli B, Mujalli A, AlRayes N, Almaghrabi S, Almansouri M, Sahly A, Jadkarim GA, Malik MZ, Kutbi HI, Shaik NA, Alefishat E. Potential Biomarkers for Parkinson Disease from Functional Enrichment and Bioinformatic Analysis of Global Gene Expression Patterns of Blood and Substantia Nigra Tissues. Bioinform Biol Insights 2023; 17:11779322231166214. [PMID: 37153842 PMCID: PMC10155030 DOI: 10.1177/11779322231166214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 05/10/2023] Open
Abstract
The Parkinson disease (PD) is the second most common neurodegenerative disorder affecting the central nervous system and motor functions. The biological complexity of PD is yet to reveal potential targets for intervention or to slow the disease severity. Therefore, this study aimed to compare the fidelity of blood to substantia nigra (SN) tissue gene expression from PD patients to provide a systematic approach to predict role of the key genes of PD pathobiology. Differentially expressed genes (DEGs) from multiple microarray data sets of PD blood and SN tissue from GEO database are identified. Using the theoretical network approach and variety of bioinformatic tools, we prioritized the key genes from DEGs. A total of 540 and 1024 DEGs were identified in blood and SN tissue samples, respectively. Functional pathways closely related to PD such as ERK1 and ERK2 cascades, mitogen-activated protein kinase (MAPK) signaling, Wnt, nuclear factor-κB (NF-κB), and PI3K-Akt signaling were observed by enrichment analysis. Expression patterns of 13 DEGs were similar in both blood and SN tissues. Comprehensive network topological analysis and gene regulatory networks identified additional 10 DEGs functionally connected with molecular mechanisms of PD through the mammalian target of rapamycin (mTOR), autophagy, and AMP-activated protein kinase (AMPK) signaling pathways. Potential drug molecules were identified by chemical-protein network and drug prediction analysis. These potential candidates can be further validated in vitro/in vivo to be used as biomarkers and/or novel drug targets for the PD pathology and/or to arrest or delay the neurodegeneration over the years, respectively.
Collapse
Affiliation(s)
- Ramu Elango
- Department of Genetic Medicine, Faculty
of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of
Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah,
Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty
of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of
Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah,
Saudi Arabia
| | - Abdulrahman Mujalli
- Department of Laboratory Medicine,
Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi
Arabia
| | - Nuha AlRayes
- Princess Al-Jawhara Al-Brahim Center of
Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah,
Saudi Arabia
- Department of Medical Laboratory
Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah,
Saudi Arabia
| | - Sarah Almaghrabi
- Department of Medical Laboratory
Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah,
Saudi Arabia
- Center of Innovation in Personalized
Medicine (CIPM), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majid Almansouri
- Department of Clinical Biochemistry,
Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Sahly
- Princess Al-Jawhara Al-Brahim Center of
Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah,
Saudi Arabia
| | - Gada Ali Jadkarim
- Department of Genetic Medicine, Faculty
of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Md Zubbair Malik
- School of Computational and Integrative
Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Hussam Ibrahim Kutbi
- Department of Pharmacy Practice,
Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor Ahmad Shaik
- Department of Genetic Medicine, Faculty
of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of
Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah,
Saudi Arabia
| | - Eman Alefishat
- Department of Clinical Pharmacology,
College of Medicine, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
22
|
Tryphena KP, Singh G, Jain N, Famta P, Srivastava S, Singh SB, Khatri DK. Integration of miRNA's Theranostic Potential with Nanotechnology: Promises and Challenges for Parkinson's Disease Therapeutics. Mech Ageing Dev 2023; 211:111800. [PMID: 36958539 DOI: 10.1016/j.mad.2023.111800] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023]
Abstract
Despite the wide research going on in Parkinson's disease (PD), the burden of PD still remains high and continues to increase. The current drugs available for the treatment of PD are only aimed at symptomatic control. Hence, research is mainly focused on identifying the novel therapeutic targets that can be effectively targeted in order to slow down or culminate the disease progression. Recently the role of microRNAs (miRNAs) in the regulation of various pathological mechanisms of PD has been thoroughly explored and many of them were found to be dysregulated in the biological samples of PD patients. These miRNAs can be used as diagnostic markers and novel therapeutic options to manage PD. The delivery of miRNAs to the target site in brain is a challenging job owing to their nature of degradability by endonucleases as well as poor blood brain barrier (BBB) permeability. Nanoparticles appear to be the best solution to effectively encase the miRNA in their core as well as cross the BBB to deliver them into brain. Functionalisation of these nanoparticles further enhances the site-specific delivery.
Collapse
Affiliation(s)
- Kamatham Pushpa Tryphena
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Gurpreet Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Naitik Jain
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Paras Famta
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Saurabh Srivastava
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad.
| | - Shashi Bala Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Dharmendra Kumar Khatri
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad.
| |
Collapse
|
23
|
Prabhakaran P, Nadig A, M S, Tuladhar S, Raju RM, Chidambaram SB, Kempaiah BB, Raghavendra NM, Kumar BR P. Design and Development of Novel Glitazones for Activation of PGC-1α Signaling Via PPAR-γ Agonism: A Promising Therapeutic Approach against Parkinson's Disease. ACS OMEGA 2023; 8:6825-6837. [PMID: 36844520 PMCID: PMC9948211 DOI: 10.1021/acsomega.2c07521] [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: 11/24/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Herein, we rationally designed and developed two novel glitazones (G1 and G2) to target peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) signaling through peroxisome proliferator-activated receptors (PPAR)-γ agonism as a therapeutic for Parkinson's disease (PD). The synthesized molecules were analyzed by mass spectrometry and NMR spectroscopy. The neuroprotective functionality of the synthesized molecules was assessed by a cell viability assay in lipopolysaccharide-intoxicated SHSY5Y neuroblastoma cell lines. The ability of these new glitazones to scavenge free radicals was further ascertained via a lipid peroxide assay, and pharmacokinetic properties were verified using in silico absorption, distribution, metabolism, excretion, and toxicity analyses. The molecular docking reports recognized the mode of interaction of the glitazones with PPAR-γ. The G1 and G2 exhibited a noticeable neuroprotective effect in lipopolysaccharide-intoxicated SHSY5Y neuroblastoma cells with the half-maximal inhibitory concentration value of 2.247 and 4.509 μM, respectively. Both test compounds prevented 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced motor impairment in mice, as demonstrated by the beam walk test. Further, treating the diseased mice with G1 and G2 resulted in significant restoration of antioxidant enzymes glutathione and superoxide and reduced the intensity of lipid peroxidation inside the brain tissues. Histopathological analysis of the glitazones-treated mice brain revealed a reduced apoptotic region and a rise in the number of viable pyramidal neurons and oligodendrocytes. The study concluded that G1 and G2 showed promising results in treating PD by activating PGC-1α signaling in brain via PPAR-γ agonism. However, more extensive research is necessary for a better understanding of functional targets and signaling pathways.
Collapse
Affiliation(s)
- Prabitha Prabhakaran
- Department
of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Abhishek Nadig
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Sahyadri M
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Sunanda Tuladhar
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Ruby Mariam Raju
- Department
of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | | | | | - Prashantha Kumar BR
- Department
of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| |
Collapse
|
24
|
Tryphena KP, Anuradha U, Kumar R, Rajan S, Srivastava S, Singh SB, Khatri DK. Understanding the Involvement of microRNAs in Mitochondrial Dysfunction and Their Role as Potential Biomarkers and Therapeutic Targets in Parkinson's Disease. J Alzheimers Dis 2023; 94:S187-S202. [PMID: 35848027 PMCID: PMC10473154 DOI: 10.3233/jad-220449] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting the elderly worldwide and causing significant movement impairments. The goal of PD treatment is to restore dopamine levels in the striatum and regulate movement symptoms. The lack of specific biomarkers for early diagnosis, as well as medication aimed at addressing the pathogenic mechanisms to decelerate the progression of dopaminergic neurodegeneration, are key roadblocks in the management of PD. Various pathogenic processes have been identified to be involved in the progression of PD, with mitochondrial dysfunction being a major contributor to the disease's pathogenesis. The regulation of mitochondrial functions is influenced by a variety of factors, including epigenetics. microRNAs (miRNAs) are epigenetic modulators involved in the regulation of gene expression and regulate a variety of proteins that essential for proper mitochondrial functioning. They are found to be dysregulated in PD, as evidenced by biological samples from PD patients and in vitro and in vivo research. In this article, we attempt to provide an overview of several miRNAs linked to mitochondrial dysfunction and their potential as diagnostic biomarkers and therapeutic targets in PD.
Collapse
Affiliation(s)
- Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Urati Anuradha
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Rohith Kumar
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shruti Rajan
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
25
|
Adam H, Gopinath SC, Kumarevel T, Arshad MM, Tijjani A, Sauli Z, Subramaniam S, Hashim U, Chen Y. Selective Detection of Amyloid Fibrils by a Dipole Moment Mechanism on Dielectrode – Structural Insights by in silico Analysis. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
26
|
Razali K, Algantri K, Loh SP, Cheng SH, Mohamed W. Integrating nutriepigenomics in Parkinson's disease management: New promising strategy in the omics era. IBRO Neurosci Rep 2022; 13:364-372. [PMID: 36590101 PMCID: PMC9795299 DOI: 10.1016/j.ibneur.2022.10.003] [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: 06/20/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is the most prevalent brain motor disorder and is frequently regarded as an idiopathic and sporadic disease due to its unclear etiology. Although the pathological mechanisms of PD have already been investigated at various omics levels, no disease-modifying drugs are currently available. At the moment, treatments can only provide symptomatic relief to control or improve motor symptoms. Parkinson's disease is a multifactorial disease, the development and progression of which are influenced by multiple factors, including the genetic markups and the environment. As an indispensable component of our daily life, nutrition is considered one of the most robust environmental factors affecting our health. Consequently, depending on our dietary habits, nutrition can either induce or reduce our susceptibility to PD. Epigenetic mechanisms regulate gene expression through DNA methylation, histone modifications, and non-coding RNAs (ncRNAs) activity. Accumulating evidence from nutriepigenomics studies has reported altered epigenetic mechanisms in clinical and pre-clinical PD models, and the potential role of nutrition in modifying the changes. In addition, through nutrigenetics and nutrigenomics studies, the diet-gene, and gene-diet interactions concerning PD development and progression have been investigated. Herein, current findings on the roles of nutrition in epigenetic mechanisms underpinning PD development and progression are discussed. Recent advancements in the multi-omics approach in PD nutrition research are also underlined. The ability of nutrients to influence epigenetic mechanisms and the availability of multi-omics applications compel the immediate use of personalized nutrition as adjuvant therapy for PD.
Collapse
Affiliation(s)
- Khairiah Razali
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), 25200 Kuantan, Pahang, Malaysia
| | - Khaled Algantri
- Faculty of Medicine, Anatomy Department, Widad University College, BIM Point, Bandar Indera Mahkota, 25200 Kuantan, Pahang, Malaysia
| | - Su Peng Loh
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Shi-Hui Cheng
- Faculty of Science and Engineering, School of Biosciences, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Wael Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), 25200 Kuantan, Pahang, Malaysia
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Menoufia, Egypt
| |
Collapse
|
27
|
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
|
28
|
Alov P, Stoimenov H, Lessigiarska I, Pencheva T, Tzvetkov NT, Pajeva I, Tsakovska I. In Silico Identification of Multi-Target Ligands as Promising Hit Compounds for Neurodegenerative Diseases Drug Development. Int J Mol Sci 2022; 23:13650. [PMID: 36362434 PMCID: PMC9655539 DOI: 10.3390/ijms232113650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 09/29/2023] Open
Abstract
The conventional treatment of neurodegenerative diseases (NDDs) is based on the "one molecule-one target" paradigm. To combat the multifactorial nature of NDDs, the focus is now shifted toward the development of small-molecule-based compounds that can modulate more than one protein target, known as "multi-target-directed ligands" (MTDLs), while having low affinity for proteins that are irrelevant for the therapy. The in silico approaches have demonstrated a potential to be a suitable tool for the identification of MTDLs as promising drug candidates with reduction in cost and time for research and development. In this study more than 650,000 compounds were screened by a series of in silico approaches to identify drug-like compounds with predicted activity simultaneously towards three important proteins in the NDDs symptomatic treatment: acetylcholinesterase (AChE), histone deacetylase 2 (HDAC2), and monoamine oxidase B (MAO-B). The compounds with affinities below 5.0 µM for all studied targets were additionally filtered to remove known non-specifically binding or unstable compounds. The selected four hits underwent subsequent refinement through in silico blood-brain barrier penetration estimation, safety evaluation, and molecular dynamics simulations resulting in two hit compounds that constitute a rational basis for further development of multi-target active compounds against NDDs.
Collapse
Affiliation(s)
- Petko Alov
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Hristo Stoimenov
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Iglika Lessigiarska
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Tania Pencheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Nikolay T. Tzvetkov
- Institute of Molecular Biology “Acad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria
| | - Ilza Pajeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Ivanka Tsakovska
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| |
Collapse
|
29
|
Rana A, Dumka A, Singh R, Panda MK, Priyadarshi N. A Computerized Analysis with Machine Learning Techniques for the Diagnosis of Parkinson's Disease: Past Studies and Future Perspectives. Diagnostics (Basel) 2022; 12:2708. [PMID: 36359550 PMCID: PMC9689408 DOI: 10.3390/diagnostics12112708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 08/03/2023] Open
Abstract
According to the World Health Organization (WHO), Parkinson's disease (PD) is a neurodegenerative disease of the brain that causes motor symptoms including slower movement, rigidity, tremor, and imbalance in addition to other problems like Alzheimer's disease (AD), psychiatric problems, insomnia, anxiety, and sensory abnormalities. Techniques including artificial intelligence (AI), machine learning (ML), and deep learning (DL) have been established for the classification of PD and normal controls (NC) with similar therapeutic appearances in order to address these problems and improve the diagnostic procedure for PD. In this article, we examine a literature survey of research articles published up to September 2022 in order to present an in-depth analysis of the use of datasets, various modalities, experimental setups, and architectures that have been applied in the diagnosis of subjective disease. This analysis includes a total of 217 research publications with a list of the various datasets, methodologies, and features. These findings suggest that ML/DL methods and novel biomarkers hold promising results for application in medical decision-making, leading to a more methodical and thorough detection of PD. Finally, we highlight the challenges and provide appropriate recommendations on selecting approaches that might be used for subgrouping and connection analysis with structural magnetic resonance imaging (sMRI), DaTSCAN, and single-photon emission computerized tomography (SPECT) data for future Parkinson's research.
Collapse
Affiliation(s)
- Arti Rana
- Computer Science & Engineering, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun 248007, Uttarakhand, India
| | - Ankur Dumka
- Department of Computer Science and Engineering, Women Institute of Technology, Dehradun 248007, Uttarakhand, India
- Department of Computer Science & Engineering, Graphic Era Deemed to be University, Dehradun 248001, Uttarakhand, India
| | - Rajesh Singh
- Division of Research and Innovation, Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, Uttarakhand, India
- Department of Project Management, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Manoj Kumar Panda
- Department of Electrical Engineering, G.B. Pant Institute of Engineering and Technology, Pauri 246194, Uttarakhand, India
| | - Neeraj Priyadarshi
- Department of Electrical Engineering, JIS College of Engineering, Kolkata 741235, West Bengal, India
| |
Collapse
|
30
|
Torres-Ortega PV, Del Campo-Montoya R, Plano D, Paredes J, Aldazabal J, Luquin MR, Santamaría E, Sanmartín C, Blanco-Prieto MJ, Garbayo E. Encapsulation of MSCs and GDNF in an Injectable Nanoreinforced Supramolecular Hydrogel for Brain Tissue Engineering. Biomacromolecules 2022; 23:4629-4644. [DOI: 10.1021/acs.biomac.2c00853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Pablo Vicente Torres-Ortega
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| | - Rubén Del Campo-Montoya
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| | - Jacobo Paredes
- Tecnun, School of Engineering, University of Navarra, C/Manuel de Lardizábal 15, 20018San Sebastián, Spain
| | - Javier Aldazabal
- Tecnun, School of Engineering, University of Navarra, C/Manuel de Lardizábal 15, 20018San Sebastián, Spain
| | - María-Rosario Luquin
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
- Department of Neurology and Neurosciences, Clínica Universidad de Navarra, Pamplona, C/Pío XII 36, 31008Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdisNa), 31008Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| | - María J. Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, C/Irunlarrea 3, 31008Pamplona, Spain
| |
Collapse
|
31
|
Uddin A, Malla JA, Kumar H, Kumari M, Sinha S, Sharma VK, Kumar Y, Talukdar P, Lahiri M, Maiti TK, Hazra P. Development of a Systematic Strategy toward Promotion of α-Synuclein Aggregation Using 2-Hydroxyisophthalamide-Based Systems. Biochemistry 2022; 61:2267-2279. [PMID: 36219819 DOI: 10.1021/acs.biochem.2c00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Establishing a potent scheme against α-synuclein aggregation involved in Parkinson's disease has been evaluated as a promising route to identify compounds that either inhibit or promote the aggregation process of α-synuclein. In the last two decades, this perspective has guided a dramatic increase in the efforts, focused on developing potent drugs either for retardation or promotion of the self-assembly process of α-synuclein. To address this issue, using a chemical kinetics platform, we developed a strategy that enabled a progressively detailed analysis of the molecular events leading to protein aggregation at the microscopic level in the presence of a recently synthesized 2-hydroxyisophthalamide class of small organic molecules based on their binding affinity. Furthermore, qualitatively, we have developed a strategy of disintegration of α-synuclein fibrils in the presence of these organic molecules. Finally, we have shown that these organic molecules effectively suppress the toxicity of α-synuclein oligomers in neuron cells.
Collapse
Affiliation(s)
- Aslam Uddin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Javid Ahmad Malla
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Harish Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru560065, India
| | - Manisha Kumari
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad121001, India
| | - Suman Sinha
- Institute of Pharmaceutical Research, GLA University, Mathura281406, India
| | - Virender Kumar Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Yashwant Kumar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India.,National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru560065, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Mayurika Lahiri
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| | - Tushar Kanti Maiti
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad121001, India
| | - Partha Hazra
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune411008, Maharashtra, India
| |
Collapse
|
32
|
Nayeri Z, Aliakbari F, Afzali F, Parsafar S, Gharib E, Otzen DE, Morshedi D. Characterization of exogenous αSN response genes and their relation to Parkinson’s disease using network analyses. Front Pharmacol 2022; 13:966760. [PMID: 36249814 PMCID: PMC9563388 DOI: 10.3389/fphar.2022.966760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Despite extensive research, the molecular mechanisms underlying the toxicity of αSN in Parkinson’s disease (PD) pathology are still poorly understood. To address this, we used a microarray dataset to identify genes that are induced and differentially expressed after exposure to toxic αSN aggregates, which we call exogenous αSN response (EASR) genes. Using systems biology approaches, we then determined, at multiple levels of analysis, how these EASR genes could be related to PD pathology. A key result was the identification of functional connections between EASR genes and previously identified PD-related genes by employing the proteins’ interactions networks and 9 brain region-specific co-expression networks. In each brain region, co-expression modules of EASR genes were enriched for gene sets whose expression are altered by SARS-CoV-2 infection, leading to the hypothesis that EASR co-expression genes may explain the observed links between COVID-19 and PD. An examination of the expression pattern of EASR genes in different non-neurological healthy brain regions revealed that regions with lower mean expression of the upregulated EASR genes, such as substantia nigra, are more vulnerable to αSN aggregates and lose their neurological functions during PD progression. Gene Set Enrichment Analysis of healthy and PD samples from substantia nigra revealed that a specific co-expression network, “TNF-α signaling via NF-κB”, is an upregulated pathway associated with the PD phenotype. Inhibitors of the “TNF-α signaling via NF-κB” pathway may, therefore, decrease the activity level of this pathway and thereby provide therapeutic benefits for PD patients. We virtually screened FDA-approved drugs against these upregulated genes (NR4A1, DUSP1, and FOS) using docking-based drug discovery and identified several promising drugs. Altogether, our study provides a better understanding of αSN toxicity mechanisms in PD and identifies potential therapeutic targets and small molecules for treatment of PD.
Collapse
Affiliation(s)
- Zahra Nayeri
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farhang Aliakbari
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Molecular Medicine Research Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Farzaneh Afzali
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Soha Parsafar
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ehsan Gharib
- Department of Chemistry and Biochemistry, University de Moncton, Moncton, ON, Canada
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dina Morshedi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- *Correspondence: Dina Morshedi,
| |
Collapse
|
33
|
Nwogo RO, Kammermeier S, Singh A. Abnormal neural oscillations during gait and dual-task in Parkinson’s disease. Front Syst Neurosci 2022; 16:995375. [PMID: 36185822 PMCID: PMC9522469 DOI: 10.3389/fnsys.2022.995375] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Gait dysfunctions are debilitating motor symptoms of Parkinson’s disease (PD) and may result in frequent falling with health complications. The contribution of the motor-cognitive network to gait disturbance can be studied more thoroughly by challenging motor-cognitive dual-task gait performances. Gait is a complex motor task that requires an appropriate contribution from motor and cognitive networks, reflected in frequency modulations among several cortical and subcortical networks. Electrophysiological recordings by scalp electroencephalography and implanted deep brain stimulation (DBS) electrodes have unveiled modulations of specific oscillatory patterns in the cortical-subcortical circuits in PD. In this review, we summarize oscillatory contributions of the cortical, basal ganglia, mesencephalic locomotor, and cerebellar regions during gait and dual-task activities in PD. We detail the involvement of the cognitive network in dual-task settings and compare how abnormal oscillations in the specific frequency bands in the cortical and subcortical regions correlate with gait deficits in PD, particularly freezing of gait (FOG). We suggest that altered neural oscillations in different frequencies can cause derangements in broader brain networks, so neuromodulation and pharmacological therapies should be considered to normalize those network oscillations to improve challenged gait and dual-task motor functions in PD. Specifically, the theta and beta bands in premotor cortical areas, subthalamic nucleus, as well as alpha band activity in the brainstem prepontine nucleus, modulate under clinically effective levodopa and DBS therapies, improving gait and dual-task performance in PD with FOG, compared to PD without FOG and age-matched healthy control groups.
Collapse
Affiliation(s)
- Rachel O. Nwogo
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | | | - Arun Singh
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- *Correspondence: Arun Singh,
| |
Collapse
|
34
|
The Anti-Inflammatory Effect of Preventive Intervention with Ketogenic Diet Mediated by the Histone Acetylation of mGluR5 Promotor Region in Rat Parkinson’s Disease Model: A Dual-Tracer PET Study. PARKINSON'S DISEASE 2022; 2022:3506213. [PMID: 36105302 PMCID: PMC9467749 DOI: 10.1155/2022/3506213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
Abstract
Materials and Methods The neuroprotective effect of ketosis state prior to the onset of PD (preventive KD, KDp) was compared with that receiving KD after the onset (therapeutic KD, KDt) in the lipopolysaccharide- (LPS-) induced rat PD model. A total of 100 rats were randomly assigned to the following 4 groups: sham, LPS, LPS + KDp, and LPS + KDt groups. Results Significant dopamine deficient behaviors (rotational behavior and contralateral forelimb akinesia), upregulation of proinflammatory mediators (TNF-α, IL-1β, and IL-6), loss of dopaminergic neurons, reduction of mGluR5+ microglia cells, increase of TSPO+ microglia cells, reduction of H3K9 acetylation in the mGluR5 promoter region and mGluR5 mRNA expression, and decline in the phosphorylation levels of Akt/GSK-3β/CREB pathway were observed after the intervention of LPS (P < 0.01). TSPO and DAT PET imaging revealed the increased uptake of 18F-DPA-714 in substantia nigra and decreased uptake of 18F-FP-CIT in substantia nigra and striatum in LPS-treated rats (P < 0.001). These impairments were alleviated by the dietary intervention of KD, especially with the strategy of KDp (P < 0.05). Conclusions The anti-inflammatory effect of KD on PD was supposed to be related to the modulation of Akt/GSK-3β/CREB signaling pathway mediated by the histone acetylation of mGluR5 promotor region. The KD intervention should be initiated prior to the PD onset in high-risk population to achieve a more favorable outcome.
Collapse
|
35
|
Zhang RQ, Kuo K, Liu FT, Chen SD, Yang YX, Guo Y, Dong Q, Tan L, Zhang C, Yu JT. Shared polygenic risk and causal inferences in Parkinson's disease. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2022.100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
36
|
Luo M, Lee LKC, Peng B, Choi CHJ, Tong WY, Voelcker NH. Delivering the Promise of Gene Therapy with Nanomedicines in Treating Central Nervous System Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201740. [PMID: 35851766 PMCID: PMC9475540 DOI: 10.1002/advs.202201740] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/19/2022] [Indexed: 06/01/2023]
Abstract
Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Australian Institute for Bioengineering and Nanotechnologythe University of QueenslandSt LuciaQLD4072Australia
| | - Leo Kit Cheung Lee
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Bo Peng
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Frontiers Science Center for Flexible ElectronicsXi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical materials & EngineeringNorthwestern Polytechnical UniversityXi'an710072China
| | - Chung Hang Jonathan Choi
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatinNew TerritoriesHong Kong
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutics ScienceMonash UniversityParkville Campus, 381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO)ClaytonVIC3168Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClaytonVIC3168Australia
- Materials Science and EngineeringMonash University14 Alliance LaneClaytonVIC3800Australia
| |
Collapse
|
37
|
Wu X, Nagasawa S, Muto K, Ueda M, Suzuki C, Abe T, Higashitani A. Mitochonic Acid 5 Improves Duchenne Muscular Dystrophy and Parkinson's Disease Model of Caenorhabditis elegans. Int J Mol Sci 2022; 23:9572. [PMID: 36076995 PMCID: PMC9455831 DOI: 10.3390/ijms23179572] [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: 07/18/2022] [Revised: 08/09/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Mitochonic Acid 5 (MA-5) enhances mitochondrial ATP production, restores fibroblasts from mitochondrial disease patients and extends the lifespan of the disease model "Mitomouse". Additionally, MA-5 interacts with mitofilin and modulates the mitochondrial inner membrane organizing system (MINOS) in mammalian cultured cells. Here, we used the nematode Caenorhabditis elegans to investigate whether MA-5 improves the Duchenne muscular dystrophy (DMD) model. Firstly, we confirmed the efficient penetration of MA-5 in the mitochondria of C. elegans. MA-5 also alleviated symptoms such as movement decline, muscular tone, mitochondrial fragmentation and Ca2+ accumulation of the DMD model. To assess the effect of MA-5 on mitochondria perturbation, we employed a low concentration of rotenone with or without MA-5. MA-5 significantly suppressed rotenone-induced mitochondria reactive oxygen species (ROS) increase, mitochondrial network fragmentation and nuclear destruction in body wall muscles as well as endogenous ATP levels decline. In addition, MA-5 suppressed rotenone-induced degeneration of dopaminergic cephalic (CEP) neurons seen in the Parkinson's disease (PD) model. Furthermore, the application of MA-5 reduced mitochondrial swelling due to the immt-1 null mutation. These results indicate that MA-5 has broad mitochondrial homing and MINOS stabilizing activity in metazoans and may be a therapeutic agent for these by ameliorating mitochondrial dysfunction in DMD and PD.
Collapse
Affiliation(s)
- Xintong Wu
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Satoi Nagasawa
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Kasumi Muto
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Maiko Ueda
- Biomedical Research Core, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | - Chitose Suzuki
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan
| | | |
Collapse
|
38
|
Rana A, Dumka A, Singh R, Panda MK, Priyadarshi N, Twala B. Imperative Role of Machine Learning Algorithm for Detection of Parkinson’s Disease: Review, Challenges and Recommendations. Diagnostics (Basel) 2022; 12:diagnostics12082003. [PMID: 36010353 PMCID: PMC9407112 DOI: 10.3390/diagnostics12082003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease that affects the neural, behavioral, and physiological systems of the brain. This disease is also known as tremor. The common symptoms of this disease are a slowness of movement known as ‘bradykinesia’, loss of automatic movements, speech/writing changes, and difficulty with walking at early stages. To solve these issues and to enhance the diagnostic process of PD, machine learning (ML) algorithms have been implemented for the categorization of subjective disease and healthy controls (HC) with comparable medical appearances. To provide a far-reaching outline of data modalities and artificial intelligence techniques that have been utilized in the analysis and diagnosis of PD, we conducted a literature analysis of research papers published up until 2022. A total of 112 research papers were included in this study, with an examination of their targets, data sources and different types of datasets, ML algorithms, and associated outcomes. The results showed that ML approaches and new biomarkers have a lot of promise for being used in clinical decision-making, resulting in a more systematic and informed diagnosis of PD. In this study, some major challenges were addressed along with a future recommendation.
Collapse
Affiliation(s)
- Arti Rana
- Computer Science & Engineering, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun 248007, Uttarakhand, India
| | - Ankur Dumka
- Department of Computer Science and Engineering, Women Institute of Technology, Uttarakhand Technical University (UTU), Dehradun 248007, Uttarakhand, India
| | - Rajesh Singh
- Division of Research and Innovation, Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, Uttarakhand, India
- Department of Project Management, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Manoj Kumar Panda
- Department of Electrical Engineering, G.B. Pant Institute of Engineering and Technology, Pauri 246194, Uttarakhand, India
| | - Neeraj Priyadarshi
- Department of Electrical Engineering, JIS College of Engineering, Kolkata 741235, West Bengal, India
| | - Bhekisipho Twala
- Digital Transformation Portfolio, Tshwane University of Technology, Staatsartillerie Rd, Pretoria West, Pretoria 0183, South Africa
- Correspondence:
| |
Collapse
|
39
|
McGee C, Liebert A, Herkes G, Bicknell B, Pang V, McLachlan CS, Kiat H. Protocol for randomized controlled trial to evaluate the safety and feasibility of a novel helmet to deliver transcranial light emitting diodes photobiomodulation therapy to patients with Parkinson’s disease. Front Neurosci 2022; 16:945796. [PMID: 36061601 PMCID: PMC9428720 DOI: 10.3389/fnins.2022.945796] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/15/2022] Open
Abstract
Introduction Parkinson’s disease (PD) is the second most common, progressive, and debilitating neurodegenerative disease associated with aging and the most common movement disorder. Photobiomodulation (PBM), the use of non-thermal light for therapeutic purposes using laser or light emitting diodes (LED) is an emerging non-invasive treatment for a diverse range of neurological conditions. The main objectives of this clinical trial are to investigate the feasibility, safety, tolerability, and efficacy of a novel transcranial LED helmet device (the “PDNeuro”) in the alleviation of symptoms of PD. Methods and analysis This is a 24-week, two-arm, triple-blinded randomized placebo-controlled clinical trial of a novel transcranial “PDNeuro” LED Helmet, comparing an active helmet to a sham helmet device. In a survey, 40 PD participants with Hoehn and Yahr Stage I–III during ON periods will be enrolled and randomly assigned into two groups. Both groups will be monitored weekly for the safety and tolerability of the “PDNeuro” LED Helmet. Clinical signs and symptoms assessed will include mobility, fine motor skills and cognition, with data collected at baseline, 12 weeks, and 24 weeks. Assessment tools include the TUG, UPDRS, and MoCA all validated for use in PD patients. Patient’s adherence to the device usage and participant drop out will be monitored weekly. At 12 weeks both placebo and treatment groups will crossover and placebo participants offered the treatment. The main indicator for clinical efficacy of the “PDneuro” Helmet is evidence of sustained improvements in motor and non-motor symptoms obtained from participant self-reported changes, carer reporting of changes and objective reassessment by the investigators. The outcomes will assist in a future larger randomized trial design. Clinical Trial Registration [https://www.anzctr.org.au], identifier [12621001722886].
Collapse
Affiliation(s)
- Claire McGee
- Faculty of Health Sciences, Torrens University, Sydney, NSW, Australia
| | - Ann Liebert
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- Department of Research and Governance, San Hospital, Wahroonga, NSW, Australia
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
- *Correspondence: Ann Liebert,
| | - Geoffrey Herkes
- Department of Neurology, San Hospital, Wahroonga, NSW, Australia
- Australian National University, Canberra, ACT, Australia
| | - Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
| | - Vincent Pang
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
| | | | - Hosen Kiat
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, Australia
- College of Health and Medicine, Australian National University, Canberra, ACT, Australia
- Cardiac Health Institute, Sydney, NSW, Australia
| |
Collapse
|
40
|
Design, Synthesis and Pharmacological Evaluation of Novel Conformationally Restricted N-arylpiperazine Derivatives Characterized as D 2/D 3 Receptor Ligands, Candidates for the Treatment of Neurodegenerative Diseases. Biomolecules 2022; 12:biom12081112. [PMID: 36009006 PMCID: PMC9405847 DOI: 10.3390/biom12081112] [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: 07/12/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Most neurodegenerative diseases are multifactorial, and the discovery of several molecular mechanisms related to their pathogenesis is constantly advancing. Dopamine and dopaminergic receptor subtypes are involved in the pathophysiology of several neurological disorders, such as schizophrenia, depression and drug addiction. For this reason, the dopaminergic system and dopamine receptor ligands play a key role in the treatment of such disorders. In this context, a novel series of conformationally restricted N-arylpiperazine derivatives (5a–f) with a good affinity for D2/D3 dopamine receptors is reported herein. Compounds were designed as interphenylene analogs of the drugs aripiprazole (2) and cariprazine (3), presenting a 1,3-benzodioxolyl subunit as a ligand of the secondary binding site of these receptors. The six new N-arylpiperazine compounds were synthesized in good yields by using classical methodologies, and binding and guanosine triphosphate (GTP)-shift studies were performed. Affinity values below 1 μM for both target receptors and distinct profiles of intrinsic efficacy were found. Docking studies revealed that Compounds 5a–f present a different binding mode with dopamine D2 and D3 receptors, mainly as a consequence of the conformational restriction imposed on the flexible spacer groups of 2 and 3.
Collapse
|
41
|
Prahl J, Coetzee GA. Genetic Elements at the Alpha-Synuclein Locus. Front Neurosci 2022; 16:889802. [PMID: 35898413 PMCID: PMC9309432 DOI: 10.3389/fnins.2022.889802] [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: 03/04/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Genome-wide association studies have consistently shown that the alpha-synuclein locus is significantly associated with Parkinson's disease. The mechanism by which this locus modulates the disease pathology and etiology remains largely under-investigated. This is due to the assumption that SNCA is the only driver of the functional aspects of several single nucleotide polymorphism (SNP) risk-signals at this locus. Recent evidence has shown that the risk associated with the top GWAS-identified variant within this locus is independent of SNCA expression, calling into question the validity of assigning function to the nearest gene, SNCA. In this review, we examine additional genes and risk variants present at the SNCA locus and how they may contribute to Parkinson's disease. Using the SNCA locus as an example, we hope to demonstrate that deeper and detailed functional validations are required for high impact disease-linked variants.
Collapse
Affiliation(s)
- Jordan Prahl
- Department of Neurodegenerative Disease, Van Andel Institute, Grand Rapids, MI, United States
| | | |
Collapse
|
42
|
Behl T, Madaan P, Sehgal A, Singh S, Makeen HA, Albratty M, Alhazmi HA, Meraya AM, Bungau S. Demystifying the Neuroprotective Role of Neuropeptides in Parkinson's Disease: A Newfangled and Eloquent Therapeutic Perspective. Int J Mol Sci 2022; 23:4565. [PMID: 35562956 PMCID: PMC9099669 DOI: 10.3390/ijms23094565] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) refers to one of the eminently grievous, preponderant, tortuous nerve-cell-devastating ailments that markedly impacts the dopaminergic (DArgic) nerve cells of the midbrain region, namely the substantia nigra pars compacta (SN-PC). Even though the exact etiopathology of the ailment is yet indefinite, the existing corroborations have suggested that aging, genetic predisposition, and environmental toxins tremendously influence the PD advancement. Additionally, pathophysiological mechanisms entailed in PD advancement encompass the clumping of α-synuclein inside the lewy bodies (LBs) and lewy neurites, oxidative stress, apoptosis, neuronal-inflammation, and abnormalities in the operation of mitochondria, autophagy lysosomal pathway (ALP), and ubiquitin-proteasome system (UPS). The ongoing therapeutic approaches can merely mitigate the PD-associated manifestations, but until now, no therapeutic candidate has been depicted to fully arrest the disease advancement. Neuropeptides (NPs) are little, protein-comprehending additional messenger substances that are typically produced and liberated by nerve cells within the entire nervous system. Numerous NPs, for instance, substance P (SP), ghrelin, neuropeptide Y (NPY), neurotensin, pituitary adenylate cyclase-activating polypeptide (PACAP), nesfatin-1, and somatostatin, have been displayed to exhibit consequential neuroprotection in both in vivo and in vitro PD models via suppressing apoptosis, cytotoxicity, oxidative stress, inflammation, autophagy, neuronal toxicity, microglia stimulation, attenuating disease-associated manifestations, and stimulating chondriosomal bioenergetics. The current scrutiny is an effort to illuminate the neuroprotective action of NPs in various PD-experiencing models. The authors carried out a methodical inspection of the published work procured through reputable online portals like PubMed, MEDLINE, EMBASE, and Frontier, by employing specific keywords in the subject of our article. Additionally, the manuscript concentrates on representing the pathways concerned in bringing neuroprotective action of NPs in PD. In sum, NPs exert substantial neuroprotection through regulating paramount pathways indulged in PD advancement, and consequently, might be a newfangled and eloquent perspective in PD therapy.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Piyush Madaan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (H.A.M.); (A.M.M.)
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.); (H.A.A.)
| | - Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.); (H.A.A.)
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulkarim M. Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (H.A.M.); (A.M.M.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biomedical Sciences, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
43
|
Hatano T, Kano O, Sengoku R, Yoritaka A, Suzuki K, Nishikawa N, Mukai Y, Nomura K, Yoshida N, Seki M, Matsukawa MK, Terashi H, Kimura K, Tashiro J, Hirano S, Murakami H, Joki H, Uchiyama T, Shimura H, Ogaki K, Fukae J, Tsuboi Y, Takahashi K, Yamamoto T, Yanagisawa N, Nagayama H. Evaluating the impact of adjunctive istradefylline on the cumulative dose of levodopa-containing medications in Parkinson's disease: study protocol for the ISTRA ADJUST PD randomized, controlled study. BMC Neurol 2022; 22:71. [PMID: 35241003 PMCID: PMC8892732 DOI: 10.1186/s12883-022-02600-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/23/2022] [Indexed: 12/02/2022] Open
Abstract
Background Levodopa remains the most effective symptomatic treatment for Parkinson’s disease (PD) more than 50 years after its clinical introduction. However, the onset of motor complications can limit pharmacological intervention with levodopa, which can be a challenge when treating PD patients. Clinical data suggest using the lowest possible levodopa dose to balance the risk/benefit. Istradefylline, an adenosine A2A receptor antagonist indicated as an adjunctive treatment to levodopa-containing preparations in PD patients experiencing wearing off, is currently available in Japan and the US. Preclinical and preliminary clinical data suggested that adjunctive istradefylline may provide sustained antiparkinsonian benefits without a levodopa dose increase; however, available data on the impact of istradefylline on levodopa dose titration are limited. The ISTRA ADJUST PD study will evaluate the effect of adjunctive istradefylline on levodopa dosage titration in PD patients. Methods This 37-week, multicenter, randomized, open-label, parallel-group controlled study in PD patients aged 30–84 years who are experiencing the wearing-off phenomenon despite receiving levodopa-containing medications ≥ 3 times daily (daily dose 300–400 mg) began in February 2019 and will continue until February 2022. Enrollment is planned to attain 100 evaluable patients for the efficacy analyses. Patients will receive adjunctive istradefylline (20 mg/day, increasing to 40 mg/day) or the control in a 1:1 ratio, stratified by age, levodopa equivalent dose, and presence/absence of dyskinesia. During the study, the levodopa dose will be increased according to symptom severity. The primary study endpoint is the comparison of the cumulative additional dose of levodopa-containing medications during the treatment period between the adjunctive istradefylline and control groups. Secondary endpoints include changes in efficacy rating scales and safety outcomes. Discussion This study aims to clarify whether adjunctive istradefylline can reduce the cumulative additional dose of levodopa-containing medications in PD patients experiencing the wearing-off phenomenon, and lower the risk of levodopa-associated complications. It is anticipated that data from ISTRA ADJUST PD will help inform future clinical decision-making for patients with PD in the real-world setting. Trial registration Japan Registry of Clinical Trials, jRCTs031180248; registered 12 March 2019.
Collapse
Affiliation(s)
- Taku Hatano
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, 113-8421, Tokyo, Japan.
| | - Osamu Kano
- Department of Neurology, Faculty of Medicine, Toho University, 6-11-1 Omorinishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Renpei Sengoku
- Department of Neurology, Jikei University Daisan Hospital, 4-11-1 Izumihoncho, Komae, Tokyo, 201-0003, Japan
| | - Asako Yoritaka
- Department of Neurology, Juntendo University Koshigaya Hospital, 560 Fukuroyama, Koshigaya-shi, Saitama, 343-0032, Japan
| | - Keisuke Suzuki
- Department of Neurology, Dokkyo Medical University Hospital, 880 Oaza Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, 321-0293, Japan
| | - Noriko Nishikawa
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, 113-8421, Tokyo, Japan.,Department of Neurology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira-shi, Tokyo, 187-8551, Japan
| | - Yohei Mukai
- Department of Neurology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira-shi, Tokyo, 187-8551, Japan
| | - Kyoichi Nomura
- Department of Neurology, Saitama Medical Center, Kawagoe-shi, Saitama, 350-8550, Japan
| | - Norihito Yoshida
- Department of Neurology, Saitama Medical Center, Kawagoe-shi, Saitama, 350-8550, Japan
| | - Morinobu Seki
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Miho Kawabe Matsukawa
- Department of Neurology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Hiroo Terashi
- Department of Neurology, Tokyo Medical University Hospital, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Katsuo Kimura
- Department of Neurology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama-shi, Kanagawa, 232-0024, Japan
| | - Jun Tashiro
- Sapporo Parkinson MS Neurological Clinic, Dai 27 Big Sapporo-kita Sky Building 12F, 7-6 Kita-7 jo Nishi-5 chome, Kita-ku, Sapporo-shi, Hokkaido, 060-0807, Japan
| | - Shigeki Hirano
- Department of Neurology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Hidetomo Murakami
- Department of Neurology, The Jikei University Hospital, 3-19-18 Nishishinbashi, Minato-ku, Tokyo, 105-8471, Japan
| | - Hideto Joki
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tsuyoshi Uchiyama
- Department of Neurology, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Naka-ku, Hamamatsu-shi, Shizuoka, 430-8558, Japan
| | - Hideki Shimura
- Department of Neurology, Juntendo Tokyo Koto Geriatric Medical Center, 3-3-20 Shinsuna, Koto-ku, Tokyo, 136-0075, Japan
| | - Kotaro Ogaki
- Department of Neurology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, Chiba, 279-0021, Japan
| | - Jiro Fukae
- Department of Neurology, Juntendo University Nerima Hospital, 3-1-10 Takano-dai, Nerima-ku, Tokyo, 177-8521, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Johnan-ku, Fukuoka, 814-0180, Japan
| | - Kazushi Takahashi
- Department of Neurology, Tokyo Metropolitan Neurological Hospital, Musashidai 2-6-1, Fuchu-shi, Tokyo, 183-0042, Japan
| | - Toshimasa Yamamoto
- Department of Neurology, Saitama Medical University Hospital, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Naotake Yanagisawa
- Medical Technology Innovation Center, Juntendo University and Juntendo Clinical Research and Trial Center, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroshi Nagayama
- Department of Neurology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| |
Collapse
|
44
|
El-Kattan MM, Rashed LA, Shazly SR, Ismail RS. Relation of serum level of tumor necrosis factor-alpha to cognitive functions in patients with Parkinson’s disease. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2022. [DOI: 10.1186/s41983-022-00460-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Inflammation is suggested to play a role in the development of non-motor Parkinson’s disease (PD) symptoms. We aimed to investigate the association between serum tumor necrosis factor-alpha (TNF-α) levels and cognition in PD patients. Thirty patients with PD and 30 healthy controls were included. Evaluation and staging of PD were done using Unified PD Rating Scale. Cognitive assessment was done using Addenbrooke’s Cognitive Examination (ACE-III) and trail making B tests. Measurement of serum levels of TNF-α was done.
Results
Patients had significantly worser cognitive scores than controls except for language subclass of ACE score. Mean serum TNF-α level was significantly greater in PD patients as compared to controls. TNF-α serum level was significantly negatively correlated with ACE visuospatial function. Sensitivity and specificity of TNF-α to detect cognitive dysfunction in PD using ACE III and trail making B tests were (73.1, 75%), (57.1, 56.2%), respectively, whereas sensitivity and specificity of TNF-α to detect severity of PD using H&Y staging in PD were 50%.
Conclusion
Patients with PD frequently have cognitive impairment. Elevated serum TNF-α levels in patients with PD, and association of this cytokine to visuospatial impairment, implicate this pro-inflammatory cytokine in the neurobiology of cognitive impairment in PD.
Collapse
|
45
|
Mapping Motor Pathways in Parkinson’s Disease Patients with Subthalamic Deep Brain Stimulator: A Diffusion MRI Tractography Study. Neurol Ther 2022; 11:659-677. [PMID: 35165822 PMCID: PMC9095781 DOI: 10.1007/s40120-022-00331-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction This study assessed the safety of postoperative diffusion tensor imaging (DTI) with on-state deep brain stimulation (DBS) and the feasibility of reconstruction of the white matter tracts in the vicinity of the stimulation site of the subthalamic nucleus (STN). The association between the impact of DBS on the nigrostriatal pathway (NSP) and the treatment effect on motor symptoms in Parkinson’s disease (PD) was then evaluated. Methods Thirty-one PD patients implanted with STN-DBS (mean age: 66 years; 25 male) were scanned on a 1.5-T magnetic resonance imaging (MRI) scanner using the DTI sequence with DBS on. Twenty-three of them were scanned a second time with DBS off. The NSP, dentato-rubro-thalamic tract (DRTT), and hyperdirect pathway (HDP) were generated using both deterministic and probabilistic tractography methods. The DBS-on-state and off-state tractography results were validated and compared. Afterward, the relationships between the characteristics of the reconstructed white matter tracts and the clinical assessment of PD symptoms and the DBS effect were further examined. Results No adverse events related to DTI were identified in either the DBS-on-state or off-state. Overall, the NSP was best reconstructed, followed by the DRTT and HDP, using the probabilistic tractography method. The connection probability of the left NSP was significantly lower than that of the right side (p < 0.05), and a negative correlation (r = −0.39, p = 0.042) was identified between the preoperative symptom severity in the medication-on state and the connection probability of the left NSP in the DBS-on-state images. Furthermore, the distance from the estimated left-side volume of tissue activated (VTA) by STN-DBS to the ipsilateral NSP was significantly shorter in the DBS-responsive group compared to the DBS-non-responsive group (p = 0.046). Conclusions DTI scanning is safe and delineation of white matter pathway is feasible for PD patients implanted with the DBS device. Postoperative DTI is a useful technique to strengthen our current understanding of the therapeutic effect of DBS for PD and has the potential to refine target selection strategies for brain stimulation. Supplementary Information The online version contains supplementary material available at 10.1007/s40120-022-00331-1. For some more seriously affected Parkinson’s disease (PD) patients, drugs are no longer effective in treating their symptoms. An alternate treatment is to use deep brain stimulation (DBS), a commonly used neurosurgical therapy for PD patients. For those DBS treatments targeting the subthalamic nucleus (STN), the electrical stimulation used may impact nearby white matter tracts and alter the effectiveness of the DBS treatment. The nigrostriatal pathway (NSP), dentato-rubro-thalamic tract, and hyperdirect pathway are three white matter tracts near the STN. They are all relevant to motor symptoms in PD. This study examined whether imaging these tracts using magnetic resonance imaging (MRI) is safe and feasible in the presence of DBS leads. The relationships between the fiber-tracking characteristics and distance to the DBS leads were then evaluated. For this purpose, 31 PD patients with stimulation-on were scanned on a 1.5 T MRI scanner using a diffusion tensor imaging sequence. A total of 23 subjects underwent another scan using the same sequence with stimulation-off. No adverse events related to diffusion tensor imaging were found. Among the white matter tracts near the STN, the NSP was best delineated, followed by the dentato-rubro-thalamic tract and the hyperdirect pathway. The connection probability of the left NSP was significantly lower than that of the right side as were the subject’s motor symptoms. The closer the distance between the NSP and the stimulation location, the better the DBS outcome. These findings indicate that imaging white matter tracts with DBS on is safe and useful in mapping DBS outcomes.
Collapse
|
46
|
Sudevan S, Muto K, Higashitani N, Hashizume T, Higashibata A, Ellwood RA, Deane CS, Rahman M, Vanapalli SA, Etheridge T, Szewczyk NJ, Higashitani A. Loss of physical contact in space alters the dopamine system in C. elegans. iScience 2022; 25:103762. [PMID: 35141505 PMCID: PMC8810405 DOI: 10.1016/j.isci.2022.103762] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Affiliation(s)
- Surabhi Sudevan
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby, UK
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Kasumi Muto
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Nahoko Higashitani
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Toko Hashizume
- Advanced Engineering Services Co. Ltd, Tsukuba Mitsui Building7F,1-6-1 Takezono, Tsukuba, Ibaraki 305-0032, Japan
| | - Akira Higashibata
- Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency, 2-1-1 Sengen, Tsukuba, Ibaraki 305-8505, Japan
| | - Rebecca A. Ellwood
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby, UK
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Colleen S. Deane
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK
- Living Systems Institute, University of Exeter, StockerRoad, Exeter, UK
| | - Mizanur Rahman
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Siva A. Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Timothy Etheridge
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK
- Corresponding author
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby, UK
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Corresponding author
| | - Atsushi Higashitani
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
- Corresponding author
| |
Collapse
|
47
|
Goyal S, Seth B, Chaturvedi RK. Polyphenols and Stem Cells for Neuroregeneration in Parkinson's Disease and Amyotrophic Lateral Sclerosis. Curr Pharm Des 2021; 28:806-828. [PMID: 34781865 DOI: 10.2174/1381612827666211115154450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/02/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS) are neurological disorders, pathologically characterized by chronic degeneration of dopaminergic neurons and motor neurons respectively. There is still no cure or effective treatment against the disease progression and most of the treatments are symptomatic. The present review offers an overview of the different factors involved in the pathogenesis of these diseases. Subsequently, we focused on the recent advanced studies of dietary polyphenols and stem cell therapies, which have made it possible to slow down the progression of neurodegeneration. To date, stem cells and different polyphenols have been used for the directional induction of neural stem cells into dopaminergic neurons and motor neurons. We have also discussed their involvement in the modulation of different signal transduction pathways and growth factor levels in various in vivo and in vitro studies. Likewise stem cells, polyphenols also exhibit the potential of neuroprotection by their anti-apoptotic, anti-inflammatory, anti-oxidant properties regulating the growth factors levels and molecular signaling events. Overall this review provides a detailed insight into recent strategies that promise the use of polyphenol with stem cell therapy for the possible treatment of PD and ALS.
Collapse
Affiliation(s)
- Shweta Goyal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001. India
| |
Collapse
|
48
|
Quercetin exhibits potent antioxidant activity, restores motor and non-motor deficits induced by rotenone toxicity. PLoS One 2021; 16:e0258928. [PMID: 34767546 PMCID: PMC8589152 DOI: 10.1371/journal.pone.0258928] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/10/2021] [Indexed: 02/06/2023] Open
Abstract
The rotenone-induced animal model of Parkinson's disease (PD) has been used to investigate the pathogenesis of PD. Oxidative stress is one of the main contributors of neurodegeneration in PD. Flavonoids have the potential to modulate neuronal function and combat various neurodegenerative diseases. The pre- and post-supplementation of quercetin (50 mg/kg, p.o) was done in rats injected with rotenone (1.5 mg/kg, s.c). After the treatment, behavioral activities were monitored for motor activity, depression-like behavior, and cognitive changes. Rats were decapitated after behavioral analysis and the brain samples were dissected out for neurochemical and biochemical estimation. Results showed that supplementation of quercetin significantly (p<0.01) restored rotenone-induced motor and non-motor deficits (depression and cognitive impairments), enhanced antioxidant enzyme activities (p<0.01), and attenuated neurotransmitter alterations (p<0.01). It is suggested that quercetin supplementation improves neurotransmitter levels by mitigating oxidative stress via increasing antioxidant enzyme activity and hence improves motor activity, cognitive functions, and reduces depressive behavior. The results of the present study showed that quercetin pre-supplementation produced more significant results as compared to post-supplementation. These findings show that quercetin can be a potential therapeutic agent to reduce the risk and progression of PD.
Collapse
|
49
|
Ahmed-Farid OA, Taha M, Bakeer RM, Radwan OK, Hendawy HAM, Soliman AS, Yousef E. Effects of bee venom and dopamine-loaded nanoparticles on reserpine-induced Parkinson's disease rat model. Sci Rep 2021; 11:21141. [PMID: 34707203 PMCID: PMC8551202 DOI: 10.1038/s41598-021-00764-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a progressive chronic neurodegenerative condition characterized by the loss of dopaminergic neurons within the substantia nigra. Current PD therapeutic strategies are mainly symptomatic and can lead to motor complications overtime. As a result, alternative medicine may provide an effective adjuvant treatment for PD as an addition to or as a replacement of the conventional therapies. The aim of this work was to evaluate the effects of Bee Venom (BV) and dopamine (DA)-loaded nanoparticles in a reserpine-induced animal model of PD. After inducing PD with reserpine injection, different groups of male rats were treated with L-Dopa, BV, DA-nanoparticles. Our findings showed that BV and DA-nanoparticles administration restored monoamines, balanced glutamate/GABA levels, halted DNA fragmentation, decreased pro-inflammatory mediators (IL-1β and TNF-α), and elevated anti-inflammatory mediators (PON1) and neurotropic factor (BDNF) levels in comparison with conventional therapy of PD. Furthermore, in a reserpine-induced PD rat model, the ameliorative effects of BV were significantly superior to that of DA-nanoparticles. These findings imply that BV and DA-nanoparticles could be useful as adjuvant treatments for PD.
Collapse
Affiliation(s)
- Omar A Ahmed-Farid
- Physiology Department, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Mohamed Taha
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Giza, Egypt.
| | - Rofanda M Bakeer
- Pathology Department, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Omyma K Radwan
- Physiology Department, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | | | - Ayman S Soliman
- Medical Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Einas Yousef
- Basic Medical Sciences Department, College of Medicine, Dar Al Uloom University, Riyadh, Kingdom of Saudi Arabia
- Histology and Cell Biology Department, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| |
Collapse
|
50
|
Mendes-Pinheiro B, Soares-Cunha C, Marote A, Loureiro-Campos E, Campos J, Barata-Antunes S, Monteiro-Fernandes D, Santos D, Duarte-Silva S, Pinto L, José Salgado A. Unilateral Intrastriatal 6-Hydroxydopamine Lesion in Mice: A Closer Look into Non-Motor Phenotype and Glial Response. Int J Mol Sci 2021; 22:ijms222111530. [PMID: 34768962 PMCID: PMC8584172 DOI: 10.3390/ijms222111530] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is a prevalent movement disorder characterized by the progressive loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). The 6-hydroxydopamine (6-OHDA) lesion is still one of the most widely used techniques for modeling Parkinson’s disease (PD) in rodents. Despite commonly used in rats, it can be challenging to reproduce a similar lesion in mice. Moreover, there is a lack of characterization of the extent of behavioral deficits and of the neuronal loss/neurotransmitter system in unilateral lesion mouse models. In this study, we present an extensive behavioral and histological characterization of a unilateral intrastriatal 6-OHDA mouse model. Our results indicate significant alterations in balance and fine motor coordination, voluntary locomotion, and in the asymmetry’s degree of forelimb use in 6-OHDA lesioned animals, accompanied by a decrease in self-care and motivational behavior, common features of depressive-like symptomatology. These results were accompanied by a decrease in tyrosine hydroxylase (TH)-labelling and dopamine levels within the nigrostriatal pathway. Additionally, we also identify a marked astrocytic reaction, as well as proliferative and reactive microglia in lesioned areas. These results confirm the use of unilateral intrastriatal 6-OHDA mice for the generation of a mild model of nigrostriatal degeneration and further evidences the recapitulation of key aspects of PD, thereby being suitable for future studies beholding new therapeutical interventions for this disease.
Collapse
Affiliation(s)
- Bárbara Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Eduardo Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Diogo Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - António José Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (B.M.-P.); (C.S.-C.); (A.M.); (E.L.-C.); (J.C.); (S.B.-A.); (D.M.-F.); (D.S.); (S.D.-S.); (L.P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
- Correspondence: ; Tel.: +351-253-60-49-47
| |
Collapse
|