1
|
Saeed U, Piracha ZZ, Tariq MN, Syed S, Rauf M, Razaq L, Iftikhar MK, Maqsood A, Ahsan SM. Decoding the genetic blueprints of neurological disorders: disease mechanisms and breakthrough gene therapies. Front Neurol 2025; 16:1422707. [PMID: 40291849 PMCID: PMC12022314 DOI: 10.3389/fneur.2025.1422707] [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: 04/24/2024] [Accepted: 02/13/2025] [Indexed: 04/30/2025] Open
Abstract
Neurological disorders pose a rapidly growing global health burden, significantly affecting cognitive and motor functions with profound societal repercussions. This comprehensive review probe into the genetic foundations of various neurological conditions while exploring innovative RNA-based therapeutics particularly gene therapies as cutting edge treatment strategies. Through an in-depth analysis of existing literature, the study examines the genetic landscape, disease mechanisms, and gene-based intervention possibilities across a range of neurological disorders, including Cerebellar Ataxias, Autosomal Recessive Ataxia, Mitochondrial Cerebellar Ataxia, Multiple System Atrophy (MSA), Idiopathic Late-Onset Cerebellar Ataxia, Hereditary Spastic Paraplegias, Alzheimer's Disease, Vascular Dementia, Lewy Body Dementia, Frontotemporal Dementias, Inherited Prion Diseases, and Huntington's Disease. It uncovers the intricate network of genetic mutations driving these disorders, shedding light on their mechanisms and uncovering promising therapeutic targets. The review also highlights the remarkable potential of RNA-based therapeutics, with gene therapies standing at the forefront of precision treatment approaches. By offering an up-to-date understanding of the genetic intricacies and emerging therapeutic possibilities in neurological disorders, this study significantly contributes to the advancement of precision medicine in neurology. It also paves the way for future research and clinical applications aimed at improving patient care and outcomes.
Collapse
Affiliation(s)
- Umar Saeed
- Operational Research Center in Healthcare, Near East University, Nicosia, Türkiye
- Foundation University School of Health Sciences (FUSH), Foundation University Islamabad, Islamabad, Pakistan
| | - Zahra Zahid Piracha
- International Center of Medical Sciences Research (ICMSR), Islamabad, Pakistan
- International Center of Medical Sciences Research (ICMSR), Austin, TX, United States
- International Center of Medical Sciences Research (ICMSR), Essex, United Kingdom
| | | | - Shayan Syed
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | - Maria Rauf
- University College of Medicine and Dentistry, Lahore, Pakistan
| | - Laiba Razaq
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | | | - Amna Maqsood
- Akhtar Saeed Medical & Dental College, Lahore, Pakistan
| | | |
Collapse
|
2
|
Shahwan M, Prasad P, Yadav DK, Altwaijry N, Khan MS, Shamsi A. Identification of high-affinity Monoamine oxidase B inhibitors for depression and Parkinson's disease treatment: bioinformatic approach of drug repurposing. Front Pharmacol 2024; 15:1422080. [PMID: 39444620 PMCID: PMC11496130 DOI: 10.3389/fphar.2024.1422080] [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: 04/23/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
Depression and Parkinson's disease (PD) are devastating psychiatric and neurological disorders that require the development of novel therapeutic interventions. Drug repurposing targeting predefined pharmacological targets is a widely use approach in modern drug discovery. Monoamine oxidase B (MAO-B) is a critical protein implicated in Depression and PD. In this study, we undertook a systematic exploration of repurposed drugs as potential inhibitors of MAO-B. Exploring a library of 3,648 commercially available drug molecules, we conducted virtual screening using a molecular docking approach to target the MAO-B binding pocket. Two promising drug molecules, Brexpiprazole and Trifluperidol, were identified based on their exceptional binding potential and drug profiling. Subsequently, all-atom molecular dynamics (MD) simulations were performed on the MAO-B-ligand complexes for a trajectory of 300 nanoseconds (ns). Simulation results demonstrated that the binding of Brexpiprazole and Trifluperidol induced only minor structural alterations in MAO-B and showed significant stabilization throughout the simulation trajectory. Overall, the finding suggests that Brexpiprazole and Trifluperidol exhibit strong potential as repurposed inhibitors of MAO-B that might be explored further in experimental investigations for the development of targeted therapies for depression and PD.
Collapse
Affiliation(s)
- Moyad Shahwan
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Pratibha Prasad
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
- Basic Medical and Dental Sciences Department, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | - Dharmendra Kumar Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Seongnam, Republic of Korea
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
3
|
Chaudhuri KR, Batzu L. Can Continuous Levodopa Delivery Be Achieved in the Absence of Intrajejunal Levodopa Infusion? Implications for India and Underserved Countries. Mov Disord Clin Pract 2024; 11:21-29. [PMID: 38291849 PMCID: PMC10828610 DOI: 10.1002/mdc3.13915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/24/2023] [Accepted: 10/12/2023] [Indexed: 02/01/2024] Open
Affiliation(s)
- K. Ray Chaudhuri
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology, and Neuroscience, King's College LondonLondonUK
- Parkinson's Foundation Centre of Excellence, King's College HospitalLondonUK
| | - Lucia Batzu
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology, and Neuroscience, King's College LondonLondonUK
- Parkinson's Foundation Centre of Excellence, King's College HospitalLondonUK
| |
Collapse
|
4
|
Khan BA, Rehman OU, Alsfouk AA, Ejaz SA, Channar PA, Saeed A, Ghafoor A, Ujan R, Mughal EU, Kumar R, Yousuf S, Hökelek T. Substituted piperidine as a novel lead molecule for the treatment of Parkinson's disease: Synthesis, crystal structure, hirshfeld surface analysis, and molecular modeling. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
5
|
Koschel J, Ray Chaudhuri K, Tönges L, Thiel M, Raeder V, Jost WH. Implications of dopaminergic medication withdrawal in Parkinson's disease. J Neural Transm (Vienna) 2021; 129:1169-1178. [PMID: 34324057 PMCID: PMC8319886 DOI: 10.1007/s00702-021-02389-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022]
Abstract
The trajectory of the use of dopamine replacement therapy (DRT) in Parkinson's disease (PD) is variable and doses may need to be increased, but also tapered. The plan for dose adjustment is usually done as per drug information recommendations from the licensing bodies, but there are no clear guidelines with regards to the best practice regarding the tapering off schedule given sudden dose reductions of drugs such as dopamine agonists may have serious adverse consequences. A systematic literature search was, therefore, performed to derive recommendations and the data show that there are no controlled studies or evidence-based recommendations how to taper or discontinue PD medication in a systematic manner. Most of the data were available on the dopamine agonist withdrawal syndrome (DAWS) and we found only two instructions on how to reduce pramipexole and rotigotine published by the EMA. We suggest that based on the available data, levodopa, dopamine agonists (DA), and amantadine should not be discontinued abruptly. Abrupt or sudden reduction of DA or amantadine in particular can lead to severe life-threatening withdrawal symptoms. Tapering off levodopa, COMT inhibitors, and MAO-B inhibitors may worsen motor and non-motor symptoms. Based on our clinical experience, we have proposed how to reduce PD medication and this work will form the basis of a future Delphi panel to define the recommendations in a consensus.
Collapse
Affiliation(s)
- J Koschel
- Parkinson-Klinik Ortenau, Kreuzbergstr. 12, 77709, Wolfach, Germany
| | - K Ray Chaudhuri
- Biomedical Research Centre, Institute of Psychiatry, Psychology and Neurosciences, King's College and Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, UK
| | - L Tönges
- Neurologische Klinik der Ruhr-Universität Bochum, Bochum, Germany
| | - M Thiel
- Parkinson-Klinik Ortenau, Kreuzbergstr. 12, 77709, Wolfach, Germany
| | - V Raeder
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London, UK.,Department of Neurology, Technical University Dresden, Dresden, Germany
| | - W H Jost
- Parkinson-Klinik Ortenau, Kreuzbergstr. 12, 77709, Wolfach, Germany.
| |
Collapse
|
6
|
Safety and Tolerability of Pharmacotherapies for Parkinson’s Disease in Geriatric Patients. Drugs Aging 2019; 36:511-530. [DOI: 10.1007/s40266-019-00654-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Baluchnejadmojarad T, Rabiee N, Zabihnejad S, Roghani M. Ellagic acid exerts protective effect in intrastriatal 6-hydroxydopamine rat model of Parkinson’s disease: Possible involvement of ERβ/Nrf2/HO-1 signaling. Brain Res 2017; 1662:23-30. [DOI: 10.1016/j.brainres.2017.02.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/26/2017] [Accepted: 02/21/2017] [Indexed: 01/14/2023]
|
8
|
Nuebling G, Hensler M, Paul S, Zwergal A, Crispin A, Lorenzl S. PROSPERA: a randomized, controlled trial evaluating rasagiline in progressive supranuclear palsy. J Neurol 2016; 263:1565-74. [PMID: 27230855 DOI: 10.1007/s00415-016-8169-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 12/22/2022]
Abstract
To date, pharmacological treatment options for progressive supranuclear palsy (PSP), a neurodegenerative tauopathy, are limited. The MAO-B inhibitor rasagiline has shown neuroprotective effects in preclinical models of neurodegeneration. To evaluate the safety, tolerability and therapeutic effect of rasagiline on symptom progression in PSP. In this 1-year randomized, double-blind, placebo-controlled trial, 44 patients fulfilling the NINDS-PSP criteria were randomized to 1 mg/d rasagiline or placebo. The combined primary endpoint included symptom progression as measured by the PSP rating scale (PSP-RS) and the requirement of L-dopa rescue medication. Secondary endpoints included Schwab and England Activities of Daily Living (SEADL), Montgomery-Åsberg Depression Rating Scale, Mini Mental State Examination, Frontal Assessment Battery and posturographic measurements. Of the 44 patients randomized, 26 completed the trial per protocol. Rasagiline was well tolerated, with a slight increase of known side effects (hallucinations, ventricular extrasystoles). No effect on the primary endpoint (p = 0.496) was detected. Symptom progression averaged at 11.2 (rasagiline) and 10.8 (placebo) points per year (ΔPSP-RS). No difference was seen in SEADL, depression, cognitive function, frontal executive function and posturographic measurements. Post hoc analyses of PSP-RS subdomains indicate a potential beneficial effect in the "limb motor" subdomain, whereas performance appeared lower in the "mentation" and "history" subdomains in the treatment group. While rasagiline is well tolerated in PSP, a beneficial effect on overall symptom progression was not detected. Post hoc analyses suggest the implementation of more specific endpoints in future studies.
Collapse
Affiliation(s)
- Georg Nuebling
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany. .,Department of Palliative Care, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.
| | - Mira Hensler
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Sabine Paul
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,German Center for Vertigo and Balance Disorders, DSGZ, Ludwig-Maximilians-University, Munich, Germany
| | - Alexander Crispin
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Lorenzl
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Department of Palliative Care, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Endowed Professorship for Interdisciplinary Research in Palliative Care, Institute of Nursing Science and Practice, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
9
|
Burciu RG, Ofori E, Shukla P, Pasternak O, Chung JW, McFarland NR, Okun MS, Vaillancourt DE. Free-water and BOLD imaging changes in Parkinson's disease patients chronically treated with a MAO-B inhibitor. Hum Brain Mapp 2016; 37:2894-903. [PMID: 27089850 DOI: 10.1002/hbm.23213] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/14/2016] [Accepted: 04/04/2016] [Indexed: 12/25/2022] Open
Abstract
Rasagiline is a monoamine oxidase type B inhibitor that possesses no amphetamine-like properties, and provides symptomatic relief in early and late stages of Parkinson's disease (PD). Data in animal models of PD suggest that chronic administration of rasagiline is associated with structural changes in the substantia nigra, and raise the question whether the structure and function of the basal ganglia could be different in PD patients treated chronically with rasagiline as compared with PD patients not treated with rasagiline. Here, we performed a retrospective cross-sectional magnetic resonance imaging (MRI) study at 3 T that investigated nigrostriatal function and structure in PD patients who had taken rasagiline before testing (∼8 months), PD who had not taken rasagiline before testing, and age-matched controls. The two PD groups were selected a priori to not differ significantly in age, sex, disease duration, severity of symptoms, cognitive status, and total levodopa equivalent daily dose of medication. We evaluated percent signal change in the posterior putamen during force production using functional MRI, free-water in the posterior substantia nigra using diffusion MRI, and performance on a bimanual coordination task using a pegboard test. All patients were tested after overnight withdrawal from antiparkinsonian medication. The rasagiline group had greater percent signal change in the posterior putamen, less free-water in the posterior substantia nigra, and better performance on the coordination task than the group not taking rasagiline. These findings point to a possible chronic effect of rasagiline on the structure and function of the basal ganglia in PD. Hum Brain Mapp 37:2894-2903, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roxana G Burciu
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Edward Ofori
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Priyank Shukla
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jae Woo Chung
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Nikolaus R McFarland
- Department of Neurology, University of Florida, Gainesville, Florida.,Center for Movement Disorders and Neurorestoration, College of Medicine, University of Florida, University of Florida, Gainesville, Florida
| | - Michael S Okun
- Department of Neurology, University of Florida, Gainesville, Florida.,Center for Movement Disorders and Neurorestoration, College of Medicine, University of Florida, University of Florida, Gainesville, Florida.,Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida.,Department of Neurology, University of Florida, Gainesville, Florida.,Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| |
Collapse
|
10
|
Wu Y, Shamoto-Nagai M, Maruyama W, Osawa T, Naoi M. Rasagiline prevents cyclosporine A-sensitive superoxide flashes induced by PK11195, the initial signal of mitochondrial membrane permeabilization and apoptosis. J Neural Transm (Vienna) 2016; 123:491-4. [PMID: 26931622 DOI: 10.1007/s00702-016-1531-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/20/2016] [Indexed: 12/25/2022]
Abstract
Rasagiline, a neuroprotective inhibitor of type B monoamine oxidase, prevented PK111195-induced apoptosis in SH-SY5Y cells through inhibition of mitochondrial apoptosis signaling (J Neural Transm 120:1539-1551, 2013, J Neural Transm 122:1399-1407, 2015). This paper presents that PK11195 induced superoxide flashes, the transit production burst, mediated by cyclosporine A-sensitive membrane permeability transition. Rasagiline prevented superoxide flashes, calcium efflux, and cell death by PK11195. Regulation of the initial pore formation at the inner mitochondrial membrane was confirmed as the decisive mechanism of neuroprotection by rasagiline.
Collapse
Affiliation(s)
- Yuqiu Wu
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Masayo Shamoto-Nagai
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Wakako Maruyama
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Toshihiko Osawa
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan.
| |
Collapse
|
11
|
Propargylamine as functional moiety in the design of multifunctional drugs for neurodegenerative disorders: MAO inhibition and beyond. Future Med Chem 2016; 7:609-29. [PMID: 25921401 DOI: 10.4155/fmc.15.12] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Much progress has been made in designing analogues that can potentially confer neuroprotection against debilitating neurodegenerative disorders, yet the multifactorial pathogenesis of this cluster of diseases remains a stumbling block for the successful design of an 'ultimate' drug. However, with the growing popularity of the "one drug, multiple targets" paradigm, many researchers have successfully synthesized and evaluated drug-like molecules incorporating a propargylamine function that shows potential to serve as multifunctional drugs or multitarget-directed ligands. It is the aim of this review to highlight the reported activities of these propargylamine derivatives and their prospect to serve as drug candidates for the treatment of neurodegenerative disorders.
Collapse
|
12
|
Bar-Am O, Amit T, Youdim MB, Weinreb O. Neuroprotective and neurorestorative potential of propargylamine derivatives in ageing: focus on mitochondrial targets. J Neural Transm (Vienna) 2015; 123:125-35. [PMID: 25859841 DOI: 10.1007/s00702-015-1395-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/23/2015] [Indexed: 01/14/2023]
Abstract
The mitochondrial theory of ageing proposes that accumulation of damage to mitochondrial function and DNA mutation lead to ageing of humans and animals. It has been suggested that mitochondria play dynamic roles in regulating synaptogenesis and morphological/functional responses of synaptic activity, and thus, deteriorating of mitochondrial function (e.g., deficits of the mitochondrial respiratory enzymes, reduced calcium influx, increased accumulation of mitochondrial DNA defects/apoptotic proteins and impairment of mitochondrial membrane potential) can lead to severe neuronal energy deficit, and in the long run, to modifications in neuronal synapses and neurodegeneration in the ageing brain. Hence, considering the mechanisms by which mitochondrial impairment can lead to neuronal death, the development of neuroprotective molecules that target various mitochondrial pathogenic processes can be effective in the treatment of ageing and age-related neurodegenerative diseases. This review addresses several aspects of the neuroprotective effects of propargylamine derivatives (e.g., the monoamine oxidase-B inhibitors, selegiline and rasagiline and the multifunctional drugs, ladostigil, M30 and VAR10303) in ageing with a special focus on mitochondrial molecular protective mechanisms.
Collapse
Affiliation(s)
- Orit Bar-Am
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Faculty of Medicine, Technion-Israel Institute of Technology, P.O.B. 9697, 31096, Haifa, Israel
| | - Tamar Amit
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Faculty of Medicine, Technion-Israel Institute of Technology, P.O.B. 9697, 31096, Haifa, Israel
| | - Moussa B Youdim
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Faculty of Medicine, Technion-Israel Institute of Technology, P.O.B. 9697, 31096, Haifa, Israel
| | - Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Faculty of Medicine, Technion-Israel Institute of Technology, P.O.B. 9697, 31096, Haifa, Israel.
| |
Collapse
|
13
|
Naoi M, Riederer P, Maruyama W. Modulation of monoamine oxidase (MAO) expression in neuropsychiatric disorders: genetic and environmental factors involved in type A MAO expression. J Neural Transm (Vienna) 2015; 123:91-106. [PMID: 25604428 DOI: 10.1007/s00702-014-1362-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 12/27/2014] [Indexed: 12/18/2022]
Abstract
Monoamine oxidase types A and B (MAO-A, MAO-B) regulate the levels of monoamine neurotransmitters in the brain, and their dysfunction may be involved in the pathogenesis and influence the clinical phenotypes of neuropsychiatric disorders. Reversible MAO-A inhibitors, such as moclobemide and befloxatone, are currently employed in the treatment of emotional disorders by inhibiting the enzymatic degradation of dopamine, serotonin and norepinephrine in the central nervous system (CNS). It has been suggested that the irreversible MAO-B inhibitors selegiline and rasagiline exert a neuroprotective effect in Parkinson's and Alzheimer's diseases. This effect, however, is not related to their inhibition of MAO activity; in animal and cellular models, selegiline and rasagiline protect neuronal cells through their anti-apoptotic activity and induction of pro-survival genes. There is increasing evidence that MAO-A activity, but not that of MAO-B, is implicated in the pathophysiology of neurodegenerative disorders, but also in gene induction by MAO-B inhibitors; on the other hand, selegiline and rasagiline increase MAO-A mRNA, protein, and enzyme activity levels. Taken together, these results suggest that each MAO subtype exerts effects that modulate the expression and activity of the other isoenzyme. The roles of MAO-A and -B in the CNS should therefore be re-evaluated with respect to the "type-specificity" of their inhibitors, which may not be unconditional during chronic treatment. Mao-a expression, in particular, may be implicated in pathogenesis and phenotypes in neuropsychiatric disorders. MAO-A expression is modified by mao polymorphisms affecting its transcriptional efficiency, as well as by mutations and polymorphism of parkin, Sirt1, FOXO, microRNA, presenilin-1, and other regulatory proteins. In addition, childhood maltreatment has been shown to have an impact upon adolescent social behavior in children with mao-a polymorphisms of low transcriptional activity. Low MAO-A activity may increase the levels of serotonin and norepinephrine, resulting in disturbed neurotransmitter system development and behavior. This review discusses genetic and environmental factors involved in the regulation of MAO-A expression, in the contexts of neuropsychiatric function and of the regulation of neuronal survival and death.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan.
| | - Peter Riederer
- Clinical Neurochemistry, National Parkinson's Foundation Centre of Excellence Laboratories, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Wakako Maruyama
- Department of Cognitive Brain Science, National Research Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
14
|
Gershanik OS. Improving L-dopa therapy: the development of enzyme inhibitors. Mov Disord 2014; 30:103-13. [PMID: 25335824 DOI: 10.1002/mds.26050] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/05/2014] [Accepted: 09/11/2014] [Indexed: 11/11/2022] Open
Abstract
The introduction of levodopa produced a monumental change in the treatment of Parkinson's disease (PD). Limitations in its bioavailability and tolerability led to the search for drugs that could improve its pharmacokinetics and safety profile. Dopa-decarboxylase inhibitors were the first such drugs that were developed, and their use in combination with L-dopa has become standard practice. Increasing knowledge on the metabolism of L-dopa allowed the identification of additional targets for intervention in an attempt to improve the symptomatic efficacy of L-dopa. Monoamineoxidase inhibitors, enhancing the central bioavailability of dopamine by blocking its metabolism, were the next step, and despite controversies regarding their efficacy, they have remained as valuable adjuncts to l-dopa in the treatment of PD. More recently, the introduction of potent, selective catechol-O-methyl transferase inhibitors have found their place in the therapeutic armamentarium of PD and are prescribed in combination with l-dopa to prolong the duration of its action.
Collapse
Affiliation(s)
- Oscar S Gershanik
- Institute of Neuroscience, Favaloro Foundation University Hospital, Solis 461, Buenos Aires, Argentina
| |
Collapse
|
15
|
Faulkner MA. Safety overview of FDA-approved medications for the treatment of the motor symptoms of Parkinson's disease. Expert Opin Drug Saf 2014; 13:1055-69. [PMID: 24962891 DOI: 10.1517/14740338.2014.931369] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Parkinson's disease (PD) is among the most common of the neurodegenerative disorders. Treatment is primarily focused on correcting neurotransmitter imbalances. Several classes of medication are available for this purpose. AREAS COVERED A Medline search was performed to gather information about the safety of the medications approved for the treatment of the motor symptoms of PD. This was supplemented with additional articles obtained from online sources and information provided by the FDA and the manufacturers. The focus of this review is the side-effect and safety profiles of carbidopa/levodopa, dopamine agonists, selective monoamine oxidase inhibitors, catechol-o-methyltransferase inhibitors, anticholinergics and amantadine. EXPERT OPINION Though serious side-effects may occur, as a group, the medications used for the treatment of PD motor symptoms tend to produce side-effects that are mild to moderate in nature, and that primarily reflect the focus on dopaminergic therapies. Care plans for Parkinson's patients should be approached based on the needs of the individual as disease presentation, lifestyle, level of disability, concurrent disease states and the presence of non-motor symptoms make each case unique. Patients and caregivers must have realistic expectations about the use of PD medications.
Collapse
Affiliation(s)
- Michele A Faulkner
- Creighton University School of Pharmacy and Health Professions and School of Medicine , 2500 California Plaza, Omaha, NE 68178 , USA +1 402 280 3145 ;
| |
Collapse
|
16
|
Polony G, Humli V, Andó R, Aller M, Horváth T, Harnos A, Tamás L, Vizi ES, Zelles T. Protective effect of rasagiline in aminoglycoside ototoxicity. Neuroscience 2014; 265:263-73. [PMID: 24508748 DOI: 10.1016/j.neuroscience.2014.01.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/13/2014] [Accepted: 01/29/2014] [Indexed: 12/28/2022]
Abstract
Sensorineural hearing losses (SNHLs; e.g., ototoxicant- and noise-induced hearing loss or presbycusis) are among the most frequent sensory deficits, but they lack effective drug therapies. The majority of recent therapeutic approaches focused on the trials of antioxidants and reactive oxygen species (ROS) scavengers in SNHLs. The rationale for these studies was the prominent role of disturbed redox homeostasis and the consequent ROS elevation. Although the antioxidant therapies in several animal studies seemed to be promising, clinical trials have failed to fulfill expectations. We investigated the potential of rasagiline, an FDA-approved monomanine oxidase type B inhibitor (MAO-B) inhibitor type anti-parkinsonian drug, as an otoprotectant. We showed a dose-dependent alleviation of the kanamycin-induced threshold shifts measured by auditory brainstem response (ABR) in an ototoxicant aminoglycoside antibiotic-based hearing loss model in mice. This effect proved to be statistically significant at a 6-mg/kg (s.c.) dose. The most prominent effect appeared at 16kHz, which is the hearing sensitivity optimum for mice. The neuroprotective, antiapoptotic and antioxidant effects of rasagiline in animal models, all targeting a specific mechanism of aminoglycoside injury, may explain this otoprotection. The dopaminergic neurotransmission enhancer effect of rasagiline might also contribute to the protection. Dopamine (DA), released from lateral olivocochlear (LOC) fibers, was shown to exert a protective action against excitotoxicity, a pathological factor in the aminoglycoside-induced SNHL. We have shown that rasagiline enhanced the electric stimulation-evoked release of DA from an acute mouse cochlea preparation in a dose-dependent manner. Using inhibitors of voltage-gated Na(+)-, Ca(2+) channels and DA transporters, we revealed that rasagiline potentiated the action potential-evoked release of DA by inhibiting the reuptake. The complex, multifactorial pathomechanism of SNHLs most likely requires drugs acting on multiple targets for effective therapy. Rasagiline, with its multi-target action and favorable adverse effects profile, might be a good candidate for a clinical trial testing the otoprotective indication.
Collapse
Affiliation(s)
- G Polony
- Department of Otorhinolaryngology, Head and Neck Surgery, Semmelweis University, Budapest, Hungary; Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - V Humli
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - R Andó
- Department of Otorhinolaryngology, Head and Neck Surgery, Semmelweis University, Budapest, Hungary
| | - M Aller
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - T Horváth
- Department of Otorhinolaryngology, Bajcsy-Zsilinszky Hospital, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - A Harnos
- Department of Biomathematics and Informatics, Szent István University, Budapest, Hungary
| | - L Tamás
- Department of Otorhinolaryngology, Head and Neck Surgery, Semmelweis University, Budapest, Hungary
| | - E S Vizi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - T Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|