1
|
Nisha Aji K, Lalang N, Ramos-Jiménez C, Rahimian R, Mechawar N, Turecki G, Chartrand D, Boileau I, Meyer JH, Rusjan PM, Mizrahi R. Evidence of altered monoamine oxidase B, an astroglia marker, in early psychosis and high-risk state. Mol Psychiatry 2025; 30:2049-2058. [PMID: 39511452 DOI: 10.1038/s41380-024-02816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
A novel radiotracer, [11C]SL25.1188, targets monoamine oxidase-B (MAO-B) enzyme, found primarily in astrocytes, which metabolizes monoamines (including dopamine), particularly in subcortical regions. Altered astrocyte function in schizophrenia is supported by convergent evidence from post-mortem, genetic, transcriptomic, peripheral and preclinical findings. We aimed to test whether levels of MAO-B, an index of astrocyte function are low in the living brains of early psychosis and their high-risk states. Thirty-eight participants including antipsychotic-free/minimally exposed clinical participants with first-episode psychosis (FEP), clinical high-risk (CHR) individuals and healthy volunteers (HVs) underwent a 90-min positron emission tomography (PET) scan with [11C]SL25.1188, to measure MAO-B VT, an index of MAO-B concentration. Participants were excluded if tested positive on urine drug screen (except for cannabis). This study of 14 FEP (mean[SD] age, 25.7[5.7] years; 6 F), 7 CHR (mean[SD] age, 20.9[3.7] years; 4 F) and 17 HV (mean[SD] age, 31.2[13.9] years; 9 F) demonstrated significant group differences in regional MAO-B VT (F(2,37.42) = 4.56, p = 0.02, Cohen's f = 0.49), controlling for tobacco (F (1,37.42) = 5.37, p = 0.03) and cannabis use (F(1,37.42) = 5.11, p = 0.03) with significantly lower MAO-B VT in CHR compared to HV (Cohen's d = 0.99). We report a significant cannabis effect on MAO-B VT (F(1,39.19) = 12.57, p = 0.001, Cohen's f = 0.57), with a significant group-by-cannabis interaction (F(2,37.30) = 3.82, p = 0.03, Cohen's f = 0.45), indicating lower MAO-B VT in cannabis-using clinical groups. Lower MAO-B VT levels were more robust in striatal than cortical regions, in both clinical groups (F(12,46.84) = 2.08, p = 0.04, Cohen's f = 0.73) and in cannabis users (F(6,46.84) = 6.42, p < 0.001, Cohen's f = 0.91). Lower MAO-B concentration supports astrocyte dysfunction in cannabis-using CHR and FEP clinical populations. Lower MAO-B is consistent with replicated striatal dopamine elevation in psychosis, as well as astrocyte dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nittha Lalang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Christian Ramos-Jiménez
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Reza Rahimian
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Daniel Chartrand
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Department of Anesthesia, Montreal Neurological Institute, Montreal, QC, Canada
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeffrey H Meyer
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo M Rusjan
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Romina Mizrahi
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Mitamura K, Norikane T, Yamamoto Y, Miyake K, Nishiyama Y. Increased Uptake of 18F-THK5351 in Glioblastoma But Not in Metastatic Brain Tumor. Clin Nucl Med 2025:00003072-990000000-01655. [PMID: 40247452 DOI: 10.1097/rlu.0000000000005909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025]
Abstract
18F-THK5351 was developed as a tracer with high binding affinity and selectivity for tau protein. However, its off-target binding to monoamine oxidase B (MAO-B), an enzyme highly expressed in astrocytes, has also been demonstrated. In the case of glioblastoma, a strong accumulation of both 11C-methionine (MET) and 18F-THK5351 was observed in the tumor. Conversely, in the case of metastatic brain tumor, while 11C-MET PET showed strong uptake in the lesion, 18F-THK5351 PET revealed no significant accumulation. Differentiating solitary metastatic brain tumors from glioblastoma on MRI can be challenging. This report suggests that 18F-THK5351 PET might serve as a valuable tool for distinguishing glioblastoma from metastatic brain tumors.
Collapse
Affiliation(s)
| | | | | | - Keisuke Miyake
- Neurological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | |
Collapse
|
3
|
Braga J, Kuik EJY, Lepra M, Rusjan PM, Kish SJ, Vieira EL, Nasser Z, Verhoeff N, Vasdev N, Chao T, Bagby M, Boileau I, Kloiber S, Husain MI, Kolla N, Koshimori Y, Faiz K, Wang W, Meyer JH. Astrogliosis Marker [ 11C]SL25.1188 After COVID-19 With Ongoing Depressive and Cognitive Symptoms. Biol Psychiatry 2025; 97:816-824. [PMID: 39395470 DOI: 10.1016/j.biopsych.2024.09.027] [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: 04/30/2024] [Revised: 08/30/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND After acute COVID-19, 5% of people experience persistent depressive symptoms and reduced cognitive function (COVID-DC). Theoretical models propose that astrogliosis is important in long COVID, but measures primarily indicative of astrogliosis have not been studied in the brain of long COVID or COVID-DC. The objective of the current study was to measure [11C]SL25.1188 total distribution volume ([11C]SL25.1188 VT), an index of monoamine oxidase B density and a marker of astrogliosis, with positron emission tomography in participants with COVID-DC and compare with healthy control participants. METHODS In 21 COVID-DC cases and 21 healthy control participants, [11C]SL25.1188 VT was measured in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum. Depressive symptoms were measured with the Beck Depression Inventory-II, and cognitive symptoms were measured with neuropsychological tests. RESULTS [11C]SL25.1188 VT was higher in participants with COVID-DC in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum than in healthy control participants. Depressive symptom severity negatively correlated with [11C]SL25.1188 VT across prioritized brain regions. More recent acute COVID-19 positively correlated with [11C]SL25.1188 VT, reflecting higher values since predominance of the Omicron variant. Exploratory analyses found greater [11C]SL25.1188 VT in the hippocampus, dorsal putamen, and ventral striatum of COVID-DC participants than control participants with a major depressive episode with no history of COVID-19, and there was no relationship to cognitive testing in prioritized regions. CONCLUSIONS Results strongly support the presence of monoamine oxidase B-labeled astrogliosis in COVID-DC throughout the regions assessed, although the association of greater astrogliosis with fewer symptoms raises the possibility of a protective role. The magnitude of astrogliosis in COVID-DC is greater since the emergence of the Omicron variant.
Collapse
Affiliation(s)
- Joeffre Braga
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Emily J Y Kuik
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mariel Lepra
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M Rusjan
- Douglas Research Centre and Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Stephen J Kish
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Erica L Vieira
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Zahra Nasser
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Natasha Verhoeff
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Neil Vasdev
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Thomas Chao
- Institute of Mental Health, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Bagby
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stefan Kloiber
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - M Ishrat Husain
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Kolla
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Waypoint Centre for Mental Health Care, Penetanguishene, Ontario, Canada
| | - Yuko Koshimori
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Khunsa Faiz
- Department of Diagnostic Radiology, Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Wei Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jeffrey H Meyer
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Sun D, Mu M, Jiang Y, Wang B, Kong Z, Tan J, Hu Y. 1, 4-benzodioxan-substituted Thienyl chalcone derivatives as novel reversible inhibitors of human monoamine oxidase B with anti-neuroinflammatory activity. Sci Rep 2025; 15:8690. [PMID: 40082573 PMCID: PMC11906766 DOI: 10.1038/s41598-025-93076-4] [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: 10/10/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
In this study, a series of 1, 4-benzodioxan-substituted thienyl chalcone derivatives were designed, synthesized and evaluated for their inhibitory activities against human MAO-B (hMAO-B). The structure-activity relationship was investigated and summarized. Among the 22 derivatives, compound 12 showed the most potent inhibitory activity, which exhibited an IC50 of 0.11 µM with a selectivity index greater than 333. Kinetics and reversibility studies confirmed that compound 12 acted as a competitive and reversible inhibitor of hMAO-B. Molecular docking studies revealed the enzyme-inhibitor interactions and the rationale was provided. Moreover, compound 12 could effectively inhibit the release of nitric oxide, tumor necrosis factor-alpha and interleukin-1 beta in both lipopolysaccharide and amyloid β-protein 1-42 (Aβ1-42)-stimulated BV2 cells and attenuate the cytotoxicity induced by Aβ1-42 in BV2 cells. As compound 12 exhibited low neurotoxicity, we believe the hit compound which combines the activities of MAO-B inhibiting and anti-neuroinflammation could be further investigated as a novel potential lead for future studies.
Collapse
Affiliation(s)
- Demeng Sun
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Mengxue Mu
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Yanmei Jiang
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Bo Wang
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Zuo Kong
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Jingbo Tan
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Yun Hu
- School of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China.
| |
Collapse
|
5
|
Rullo M, La Spada G, Stefanachi A, Macchia E, Pisani L, Leonetti F. Playing Around the Coumarin Core in the Discovery of Multimodal Compounds Directed at Alzheimer's-Related Targets: A Recent Literature Overview. Molecules 2025; 30:891. [PMID: 40005200 PMCID: PMC11857976 DOI: 10.3390/molecules30040891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) causes a great socioeconomic burden because of its increasing prevalence and the lack of effective therapies. The multifactorial nature of AD prompts researchers to search for new strategies for discovering disease-modifying therapeutics. To this extent, the multitarget approach holds the potential of synergic or cooperative activities arising from compounds that are properly designed to address two or more pathogenetic mechanisms. As a privileged and nature-friendly scaffold, coumarin has successfully been enrolled as the heterocyclic core in the design of multipotent anti-Alzheimer's agents. Herein, we comprehensively summarize the most recent literature (2018-2023), covering the rational design and the discovery of coumarin-containing multitarget directed ligands (MTDLs) whose anti-AD profile encompassed at least two different biological activities relevant for disease onset and progression. To enhance the clarity of presentation, synthetic coumarin-based MTDLs are categorized into four clusters based on their substitution pattern and reported bioactivities: (i) mono-, (ii) di-, and (iii) polysubstituted coumarins directed at protein targets, and (iv) coumarins directed at protein targets with additional metal-chelating features. Before discussing multimodal coumarins, the rationale for addressing each biological target is briefly presented.
Collapse
Affiliation(s)
| | | | | | | | - Leonardo Pisani
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona, 4, 70125 Bari, Italy; (M.R.); (G.L.S.); (A.S.); (E.M.); (F.L.)
| | | |
Collapse
|
6
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
7
|
Naskar R, Ghosh A, Bhattacharya R, Chakraborty S. A critical appraisal of geroprotective activities of flavonoids in terms of their bio-accessibility and polypharmacology. Neurochem Int 2024; 180:105859. [PMID: 39265701 DOI: 10.1016/j.neuint.2024.105859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Flavonoids, a commonly consumed natural product, elicit health-benefits such as antioxidant, anti-inflammatory, antiviral, anti-allergic, hepatoprotective, anti-carcinogenic and neuroprotective activities. Several studies have reported the beneficial role of flavonoids in improving memory, learning, and cognition in clinical settings. Their mechanism of action is mediated through the modulation of multiple signalling cascades. This polypharmacology makes them an attractive natural scaffold for designing and developing new effective therapeutics for complex neurological disorders like Alzheimer's disease and Parkinson's disease. Flavonoids are shown to inhibit crucial targets related to neurodegenerative disorders (NDDs), including acetylcholinesterase, butyrylcholinesterase, β-secretase, γ-secretase, α-synuclein, Aβ protein aggregation and neurofibrillary tangles formation. Conserved neuro-signalling pathways related to neurotransmitter biogenesis and inactivation, ease of genetic manipulation and tractability, cost-effectiveness, and their short lifespan make Caenorhabditis elegans one of the most frequently used models in neuroscience research and high-throughput drug screening for neurodegenerative disorders. Here, we critically appraise the neuroprotective activities of different flavonoids based on clinical trials and epidemiological data. This review provides critical insights into the absorption, metabolism, and tissue distribution of various classes of flavonoids, as well as detailed mechanisms of the observed neuroprotective activities at the molecular level, to rationalize the clinical data. We further extend the review to critically evaluate the scope of flavonoids in the disease management of neurodegenerative disorders and review the suitability of C. elegans as a model organism to study the neuroprotective efficacy of flavonoids and natural products.
Collapse
Affiliation(s)
- Roumi Naskar
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, 500046, India
| | - Anirrban Ghosh
- Amity Institute of Biotechnology, Amity University, Kolkata, 700135, India
| | - Raja Bhattacharya
- Amity Institute of Biotechnology, Amity University, Kolkata, 700135, India.
| | - Sandipan Chakraborty
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, 500046, India.
| |
Collapse
|
8
|
Lopresti BJ, Stehouwer J, Reese AC, Mason NS, Royse SK, Narendran R, Laymon CM, Lopez OL, Cohen AD, Mathis CA, Villemagne VL. Kinetic modeling of the monoamine oxidase-B radioligand [ 18F]SMBT-1 in human brain with positron emission tomography. J Cereb Blood Flow Metab 2024; 44:1262-1276. [PMID: 38735059 PMCID: PMC11542143 DOI: 10.1177/0271678x241254679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/29/2024] [Accepted: 04/07/2024] [Indexed: 05/14/2024]
Abstract
This paper describes pharmacokinetic analyses of the monoamine-oxidase-B (MAO-B) radiotracer [18F](S)-(2-methylpyrid-5-yl)-6-[(3-fluoro-2-hydroxy)propoxy]quinoline ([18F]SMBT-1) for positron emission tomography (PET) brain imaging. Brain MAO-B expression is widespread, predominantly within astrocytes. Reactive astrogliosis in response to neurodegenerative disease pathology is associated with MAO-B overexpression. Fourteen elderly subjects (8 control, 5 mild cognitive impairment, 1 Alzheimer's disease) with amyloid ([11C]PiB) and tau ([18F]flortaucipir) imaging assessments underwent dynamic [18F]SMBT-1 PET imaging with arterial input function determination. [18F]SMBT-1 showed high brain uptake and a retention pattern consistent with the known MAO-B distribution. A two-tissue compartment (2TC) model where the K1/k2 ratio was fixed to a whole brain value best described [18F]SMBT-1 kinetics. The 2TC total volume of distribution (VT) was well identified and highly correlated (r2∼0.8) with post-mortem MAO-B indices. Cerebellar grey matter (CGM) showed the lowest mean VT of any region and is considered the optimal pseudo-reference region. Simplified analysis methods including reference tissue models, non-compartmental models, and standard uptake value ratios (SUVR) agreed with 2TC outcomes (r2 > 0.9) but with varying bias. We found the CGM-normalized 70-90 min SUVR to be highly correlated (r2 = 0.93) with the 2TC distribution volume ratio (DVR) with acceptable bias (∼10%), representing a practical alternative for [18F]SMBT-1 analyses.
Collapse
Affiliation(s)
- Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey Stehouwer
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandria C Reese
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neale S Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah K Royse
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Dept. of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Clinical and Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Mitamura K, Norikane T, Yamamoto Y, Miyake K, Nishiyama Y. Increased Uptake of 18 F-THK5351 in Isocitrate Dehydrogenase-Wildtype Glioblastoma But Not in Meningioma. Clin Nucl Med 2024; 49:962-963. [PMID: 38968597 DOI: 10.1097/rlu.0000000000005381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
ABSTRACT 18 F-THK5351 demonstrates a strong binding affinity and selectivity for tau. However, off-target binding with monoamine oxidase-B enzyme, highly expressed in the outer mitochondrial membranes of astrocytes, is possible. In a case with isocitrate dehydrogenase-wildtype glioblastoma, 11 C-MET PET and 18 F-THK5351 PET exhibited increased uptake in the tumor. Conversely, in another case with intracranial meningioma, MET PET revealed increased uptake in the tumor, whereas 18 F-THK5351 PET showed no abnormal uptake in the tumor. However, it is challenging to distinguish meningiomas from glioblastomas on MRI. 18 F-THK5351 PET might help differentiate between isocitrate dehydrogenase-wildtype glioblastoma and meningioma.
Collapse
Affiliation(s)
| | | | | | - Keisuke Miyake
- Neurological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | |
Collapse
|
10
|
Banerjee C, Tripathy D, Kumar D, Chakraborty J. Monoamine oxidase and neurodegeneration: Mechanisms, inhibitors and natural compounds for therapeutic intervention. Neurochem Int 2024; 179:105831. [PMID: 39128624 DOI: 10.1016/j.neuint.2024.105831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Mammalian flavoenzyme Monoamine oxidase (MAO) resides on the outer mitochondrial membrane (OMM) and it is involved in the metabolism of different monoamine neurotransmitters in brain. During MAO mediated oxidative deamination of relevant substrates, H2O2 is released as a catalytic by-product, thus serving as a major source of reactive oxygen species (ROS). Under normal conditions, MAO mediated ROS is reported to propel the functioning of mitochondrial electron transport chain and phasic dopamine release. However, due to its localization onto mitochondria, sudden elevation in its enzymatic activity could directly impact the form and function of the organelle. For instance, in the case of Parkinson's disease (PD) patients who are on l-dopa therapy, the enzyme could be a concurrent source of extensive ROS production in the presence of uncontrolled substrate (dopamine) availability, thus further impacting the health of surviving neurons. It is worth mentioning that the expression of the enzyme in different brain compartments increases with age. Moreover, the involvement of MAO in the progression of neurological disorders such as PD, Alzheimer's disease and depression has been extensively studied in recent times. Although the usage of available synthetic MAO inhibitors has been instrumental in managing these conditions, the associated complications have raised significant concerns lately. Natural products have served as a major source of lead molecules in modern-day drug discovery; however, there is still no FDA-approved MAO inhibitor which is derived from natural sources. In this review, we have provided a comprehensive overview of MAO and how the enzyme system is involved in the pathogenesis of different age-associated neuropathologic conditions. We further discussed the applications and drawbacks of the long-term usage of presently available synthetic MAO inhibitors. Additionally, we have highlighted the prospect and worth of natural product derived molecules in addressing MAO associated complications.
Collapse
Affiliation(s)
- Chayan Banerjee
- Cell Biology and Physiology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debasmita Tripathy
- Department of Zoology, Netaji Nagar College for Women, Kolkata, 700092, India
| | - Deepak Kumar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700032, India.
| | - Joy Chakraborty
- Cell Biology and Physiology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India.
| |
Collapse
|
11
|
Sun D, Wang B, Jiang Y, Kong Z, Mu M, Yang C, Tan J, Hu Y. Benzodioxane Carboxamide Derivatives As Novel Monoamine Oxidase B Inhibitors with Antineuroinflammatory Activity. ACS Med Chem Lett 2024; 15:798-805. [PMID: 38894921 PMCID: PMC11181489 DOI: 10.1021/acsmedchemlett.3c00532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/17/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
In this study, a series of N-phenyl-2,3-dihydrobenzo[b][1,4]dioxine-6-carboxamide derivatives were designed, synthesized, and evaluated for their inhibitory activities against human MAO-B (hMAO-B). The structure-activity relationship (SAR) was investigated and summarized. Compound 1l (N-(3,4-dichlorophenyl)-2,3-dihydrobenzo[b][1,4]dioxine-6-carboxamide) showed the most potent inhibitory activity with an IC50 value of 0.0083 μM and the selectivity index (IC50 (hMAO-A)/IC50 (hMAO-B)) was >4819. Kinetics and reversibility studies confirmed that compound 1l acted as a competitive and reversible inhibitor of hMAO-B. Molecular docking studies revealed the enzyme-inhibitor interactions, and the rationale was provided. Additionally, compound 1l could effectively inhibit the release of NO, TNF-α, and IL-1β in both LPS- and Aβ1-42-stimulated BV2 cells and attenuate the cytotoxicity induced by Aβ1-42. Since compound 1l exhibited low neurotoxicity, we believe that the hit compound with dual activities of inhibiting MAO-B and antineuroinflammation could be further investigated as a novel potential lead for future studies in vivo.
Collapse
Affiliation(s)
| | | | - Yanmei Jiang
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| | - Zuo Kong
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| | - Mengxue Mu
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| | - Changhuan Yang
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| | - Jingbo Tan
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| | - Yun Hu
- School of Bioengineering, Zhuhai Campus, Zunyi Medical University, Zhuhai 519041, China
| |
Collapse
|
12
|
Kang J, Park M, Kim T. Vitamin D Reduces GABA-Positive Astrocytes in the 5xFAD Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 97:1939-1950. [PMID: 38339931 PMCID: PMC10894571 DOI: 10.3233/jad-231033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
Background Vitamin D has neuroprotective and immunomodulating functions that may impact glial cell function in the brain. Previously, we reported molecular and behavioral changes caused by deficiency and supplementation of vitamin D in an Alzheimer's disease (AD) mouse model. Recent studies have highlighted reactive astrocytes as a new therapeutic target for AD treatment. However, the mechanisms underlying the therapeutic effects of vitamin D on the glial cells of AD remain unclear. Objective To investigate the potential association between vitamin D deficiency/supplementation and the pathological progression of AD, including amyloid-β (Aβ) pathology and reactive astrogliosis. Methods Transgenic hemizygous 5XFAD male mice were subjected to different dietary interventions and intraperitoneal vitamin D injections to examine the effects of vitamin D deficiency and supplementation on AD. Brain tissue was then analyzed using immunohistochemistry for Aβ plaques, microglia, and astrocytes, with quantifications performed via ImageJ software. Results Our results demonstrated that vitamin D deficiency exacerbated Aβ plaque formation and increased GABA-positive reactive astrocytes in AD model mice, while vitamin D supplementation ameliorated these effects, leading to a reduction in Aβ plaques and GABA-positive astrocytes. Conclusions Our findings highlight the significant impact of vitamin D status on Aβ pathology and reactive astrogliosis, underscoring its potential role in the prevention and treatment of AD. This study provides the first in vivo evidence of the association between vitamin D and reactive astrogliosis in AD model mice, indicating the potential for targeting vitamin D levels as a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|
13
|
Rodriguez-Vieitez E, Kumar A, Malarte ML, Ioannou K, Rocha FM, Chiotis K. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2024; 2785:195-218. [PMID: 38427196 DOI: 10.1007/978-1-0716-3774-6_13] [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] [Indexed: 03/02/2024]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed and how clinical trials are designed today. Alzheimer's disease (AD) - the most common neurodegenerative disorder - is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells, i.e., astrocytes and microglia, and neuroinflammatory response, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers is available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is a great interest to develop PET tracers to image glial reactivity and neuroinflammation. While most research to date has focused on imaging microgliosis, there is an upsurge of interest in imaging reactive astrocytes in the AD continuum. There is increasing evidence that reactive astrocytes are morphologically and functionally heterogeneous, with different subtypes that express different markers and display various homeostatic or detrimental roles across disease stages. Therefore, multiple biomarkers are desirable to unravel the complex phenomenon of reactive astrocytosis. In the field of in vivo PET imaging in AD, the research concerning reactive astrocytes has predominantly focused on targeting monoamine oxidase B (MAO-B), most often using either 11C-deuterium-L-deprenyl (11C-DED) or 18F-SMBT-1 PET tracers. Additionally, imidazoline2 binding (I2BS) sites have been imaged using 11C-BU99008 PET. Recent studies in our group using 11C-DED PET imaging suggest that astrocytosis may be present from the early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, glucose metabolism, and brain structural changes. It may also contribute to understanding the potential role of novel plasma biomarkers of reactive astrocytes, in particular the glial fibrillary acidic protein (GFAP), at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial response in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial reactivity and neuroinflammation as biomarkers with clinical application and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Ioannou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Filipa M Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Yang C, Wang X, Gao C, Liu Y, Ma Z, Zang J, Wang H, Liu L, Liu Y, Sun H, Wang W. Molecular Mechanism and Structure-activity Relationship of the Inhibition Effect between Monoamine Oxidase and Selegiline Analogues. Curr Comput Aided Drug Des 2024; 20:474-485. [PMID: 37138424 DOI: 10.2174/1573409919666230503143055] [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: 10/08/2022] [Revised: 02/19/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023]
Abstract
INTRODUCTION To investigate the inhibition properties and structure-activity relationship between monoamine oxidase (MAO) and selected monoamine oxidase inhibitors (MAOIs, including selegiline, rasagiline and clorgiline). METHODS The inhibition effect and molecular mechanism between MAO and MAOIs were identified via the half maximal inhibitory concentration (IC50) and molecular docking technology. RESULTS It was indicated that selegiline and rasagiline were MAO B inhibitors, but clorgiline was MAO-A inhibitor based on the selectivity index (SI) of MAOIs (0.000264, 0.0197 and 14607.143 for selegiline, rasagiline and clorgiline, respectively). The high-frequency amino acid residues of the MAOIs and MAO were Ser24, Arg51, Tyr69 and Tyr407 for MAO-A and Arg42 and Tyr435 for MAO B. The MAOIs and MAO A/B pharmacophores included the aromatic core, hydrogen bond acceptor, hydrogen bond donor-acceptor and hydrophobic core. CONCLUSION This study shows the inhibition effect and molecular mechanism between MAO and MAOIs and provides valuable findings on the design and treatment of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Chuanxi Yang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Xiaoning Wang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Chang Gao
- Qingdao Jiaming Measurement and Control Technology Co., Ltd., Qingdao, Shandong, 266000, China
| | - Yunxiang Liu
- Environmental Monitoring Station of Yuncheng County Environmental Protection Bureau, Heze, Shandong, 274700, China
| | - Ziyi Ma
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Jinqiu Zang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Haoce Wang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Lin Liu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Yonglin Liu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Haofen Sun
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| | - Weiliang Wang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao, Shandong, 266520, China
| |
Collapse
|
15
|
Krishna A, Kumar S, Sudevan ST, Singh AK, Pappachen LK, Rangarajan TM, Abdelgawad MA, Mathew B. A Comprehensive Review of the Docking Studies of Chalcone for the Development of Selective MAO-B Inhibitors. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:697-714. [PMID: 37190818 DOI: 10.2174/1871527322666230515155000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
Monoamine oxidase B is a crucial therapeutic target for neurodegenerative disorders like Alzheimer's and Parkinson's since they assist in disintegrating neurotransmitters such as dopamine in the brain. Pursuing efficacious monoamine oxidase B inhibitors is a hot topic, as contemporary therapeutic interventions have many shortcomings. Currently available FDA-approved monoamine oxidase inhibitors like safinamide, selegiline and rasagiline also have a variety of side effects like depression and insomnia. In the quest for a potent monoamine oxidase B inhibitor, sizeable, diverse chemical entities have been uncovered, including chalcones. Chalcone is a renowned structural framework that has been intensively explored for its monoamine oxidase B inhibitory activity.The structural resemblance of chalcone (1,3-diphenyl-2-propen-1-one) based compounds and 1,4-diphenyl- 2-butene, a recognized MAO-B inhibitor, accounts for their MAO-B inhibitory activity. Therefore, multiple revisions to the chalcone scaffold have been attempted by the researchers to scrutinize the implications of substitutions onthe molecule's potency. In this work, we outline the docking investigation results of various chalcone analogues with monoamine oxidase B available in the literature until now to understand the interaction modes and influence of substituents. Here we focused on the interactions between reported chalcone derivatives and the active site of monoamine oxidase B and the influence of substitutions on those interactions. Detailed images illustrating the interactions and impact of the substituents or structural modifications on these interactions were used to support the docking results.
Collapse
Affiliation(s)
- Athulya Krishna
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Sachithra Thazhathuveedu Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Ashutosh Kumar Singh
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Leena K Pappachen
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - T M Rangarajan
- Department of Chemistry, Sri Venketeswara College, University of Delhi, New Delhi-110021, India
| | - Mohamed A Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf 72341, Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| |
Collapse
|
16
|
Mathew B, Oh JM, Parambi DGT, Sudevan ST, Kumar S, Kim H. Enzyme Inhibition Assays for Monoamine Oxidase. Methods Mol Biol 2024; 2761:329-336. [PMID: 38427248 DOI: 10.1007/978-1-0716-3662-6_24] [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] [Indexed: 03/02/2024]
Abstract
Monoamine oxidase (MAO) catalyzes the oxidative deamination of monoamines with two isoforms, namely, MAO-A and MAO-B, in mitochondrial outer membranes. These two types of MAO-A and MAO-B participate in changes in levels of neurotransmitter such as serotonin (5-hydroxytryptamine) and dopamine. Selective MAO-A inhibitors have been targeted for anti-depression treatment, while selective MAO-B inhibitors are targets of therapeutic agents for Alzheimer's disease and Parkinson's disease. For this reason, study on the development of MAO inhibitors has recently become important. Here, we describe methods of MAO activity assay, especially continuous spectrophotometric methods, which give relatively high accuracy. MAO-A and MAO-B can be assayed using kynuramine and benzylamine as substrates, respectively, at 316 nm and 250 nm, respectively, to measure their respective products, 4-hydroxyquinoline and benzaldehyde. Inhibition degree and pattern can be analyzed by using the Lineweaver-Burk and secondary plots in the presence of inhibitor, and reversibility of inhibitor can be determined by using the dialysis method.
Collapse
Affiliation(s)
- Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India.
| | - Jong Min Oh
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | | | - Sachithra Thazhathuveedu Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea.
| |
Collapse
|
17
|
Jayan J, Lee J, Kumar S, Manoharan A, Narayanan AP, Jauhari R, Abdelgawad MA, Ghoneim MM, Ebrahim HA, Mary Zachariah S, Kim H, Mathew B. Development of a New Class of Monoamine Oxidase-B Inhibitors by Fine-Tuning the Halogens on the Acylhydrazones. ACS OMEGA 2023; 8:47606-47615. [PMID: 38144071 PMCID: PMC10733988 DOI: 10.1021/acsomega.3c05719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023]
Abstract
A total of 14 acyl hydrazine derivatives (ACH1-ACH14) were developed and examined for their ability to block monoamine oxidase (MAO). Thirteen analogues showed stronger inhibition potency against MAO-B than MAO-A. With a half-maximum inhibitory concentration of 0.14 μM, ACH10 demonstrated the strongest inhibitory activity against MAO-B, followed by ACH14, ACH13, ACH8, and ACH3 (IC50 = 0.15, 0.18, 0.20, and 0.22 μM, respectively). Structure-activity relationships suggested that the inhibition effect on MAO-B resulted from the combination of halogen substituents of the A- and/or B-rings. This series concluded that when -F was substituted to the B-ring, MAO-B inhibitory activities were high, except for ACH6. In the inhibition kinetics study, the compounds ACH10 and ACH14 were identified as competitive inhibitors, with Ki values of 0.097 ± 0.0021 and 0.10 ± 0.038 μM, respectively. In a reversibility experiment using the dialysis methods, ACH10 and ACH14 showed effective recoveries of MAO-B inhibition as much as lazabemide, a reversible reference. These experiments proposed that ACH10 and ACH14 were efficient, reversible competitive MAO-B inhibitors. In addition, the lead molecules showed good blood-brain barrier permeation with the PAMPA method. The molecular docking and molecular dynamics simulation study confirmed that the hit compound ACH10 can form a stable protein-ligand complex by forming a hydrogen bond with the NH atom in the hydrazide group of the compound.
Collapse
Affiliation(s)
- Jayalakshmi Jayan
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Jiseong Lee
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Sunil Kumar
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Amritha Manoharan
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | | | - Reenoo Jauhari
- School
of Pharmacy, Graphic Era Hill University, Dehradun 248002, Uttarakhand, India
| | - Mohamed A. Abdelgawad
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
- Pharmaceutical
Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62514, Egypt
| | - Mohammed M. Ghoneim
- Department
of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Riyadh 13713, Saudi Arabia
- Pharmacognosy
and Medicinal Plants Department, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Hasnaa Ali Ebrahim
- Department
of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Subin Mary Zachariah
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| | - Hoon Kim
- Department
of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Bijo Mathew
- Department
of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682 041, India
| |
Collapse
|
18
|
Koshimori Y, Cusimano MD, Vieira EL, Rusjan PM, Kish SJ, Vasdev N, Moriguchi S, Boileau I, Chao T, Nasser Z, Ishrat Husain M, Faiz K, Braga J, Meyer JH. Astrogliosis marker 11C-SL25.1188 PET in traumatic brain injury with persistent symptoms. Brain 2023; 146:4469-4475. [PMID: 37602426 PMCID: PMC10629767 DOI: 10.1093/brain/awad279] [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: 10/14/2022] [Revised: 06/22/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Traumatic brain injury (TBI) is common but little is known why up to a third of patients have persisting symptoms. Astrogliosis, a pathophysiological response to brain injury, may be a potential therapeutic target, but demonstration of astrogliosis in the brain of humans with TBI and persistent symptoms is lacking. Astroglial marker monoamine oxidase B (MAO-B) total distribution volume (11C-SL25.1188 VT), an index of MAO-B density, was measured in 29 TBI and 29 similarly aged healthy control cases with 11C-SL25.1188 PET, prioritizing prefrontal cortex (PFC) and cortex proximal to cortical convexity. Correlations of PFC 11C-SL25.1188 VT with psychomotor and processing speed; and serum blood measures implicated in astrogliosis were determined. 11C-SL25.1188 VT was greater in TBI in PFC (P = 0.00064) and cortex (P = 0.00038). PFC 11C-SL25.1188 VT inversely correlated with Comprehensive Trail Making Test psychomotor and processing speed (r = -0.48, P = 0.01). In participants scanned within 2 years of last TBI, PFC 11C-SL25.1188 VT correlated with serum glial fibrillary acid protein (r = 0.51, P = 0.037) and total tau (r = 0.74, P = 0.001). Elevated 11C-SL25.1188 VT argues strongly for astrogliosis and therapeutics modifying astrogliosis towards curative phenotypes should be tested in TBI with persistent symptoms. Given substantive effect size, astrogliosis PET markers should be applied to stratify cases and/or assess target engagement for putative therapeutics targeting astrogliosis.
Collapse
Affiliation(s)
- Yuko Koshimori
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
| | - Michael D Cusimano
- Neurosurgery, St. Michael’s Hospital, University of Toronto, Toronto, M5B 1W8, Canada
| | - Erica L Vieira
- Molecular Neurobiology and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
| | - Pablo M Rusjan
- Douglas Research Centre and Department of Psychiatry, McGill University, Montreal, H3A 1A1, Canada
| | - Stephen J Kish
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Neil Vasdev
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
| | - Sho Moriguchi
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Thomas Chao
- Institute of Mental Health, Department of Psychiatry, University of British Columbia, Vancouver, V6T 2A1, Canada
| | - Zahra Nasser
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
| | - M Ishrat Husain
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Khunsa Faiz
- Department of Diagnostic Radiology, Hamilton Health Sciences, McMaster University, Hamilton, L8S 4K1, Canada
| | - Joeffre Braga
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Jeffrey H Meyer
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
| |
Collapse
|
19
|
Tian J, Du E, Guo L. Mitochondrial Interaction with Serotonin in Neurobiology and Its Implication in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1165-1177. [PMID: 38025801 PMCID: PMC10657725 DOI: 10.3233/adr-230070] [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: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a lethal neurodegenerative disorder characterized by severe brain pathologies and progressive cognitive decline. While the exact cause of this disease remains unknown, emerging evidence suggests that dysregulation of neurotransmitters contributes to the development of AD pathology and symptoms. Serotonin, a critical neurotransmitter in the brain, plays a pivotal role in regulating various brain processes and is implicated in neurological and psychiatric disorders, including AD. Recent studies have shed light on the interplay between mitochondrial function and serotonin regulation in brain physiology. In AD, there is a deficiency of serotonin, along with impairments in mitochondrial function, particularly in serotoninergic neurons. Additionally, altered activity of mitochondrial enzymes, such as monoamine oxidase, may contribute to serotonin dysregulation in AD. Understanding the intricate relationship between mitochondria and serotonin provides valuable insights into the underlying mechanisms of AD and identifies potential therapeutic targets to restore serotonin homeostasis and alleviate AD symptoms. This review summarizes the recent advancements in unraveling the connection between brain mitochondria and serotonin, emphasizing their significance in AD pathogenesis and underscoring the importance of further research in this area. Elucidating the role of mitochondria in serotonin dysfunction will promote the development of therapeutic strategies for the treatment and prevention of this neurodegenerative disorder.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Eric Du
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
- Blue Valley West High School, Overland Park, KS, USA
| | - Lan Guo
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
20
|
Kim KT, Cho DW, Cho JW, Im WJ, Kim DH, Park JH, Park KD, Yang YS, Han SC. Two weeks dose range-finding and four weeks repeated dose oral toxicity study of a novel reversible monoamine oxidase B inhibitor KDS2010 in cynomolgus monkeys. Toxicol Res 2023; 39:693-709. [PMID: 37779583 PMCID: PMC10541392 DOI: 10.1007/s43188-023-00182-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 03/05/2023] [Accepted: 04/04/2023] [Indexed: 10/03/2023] Open
Abstract
A novel reversible monoamine oxidase B inhibitor, KDS2010, has been developed as a therapeutic candidate for neurodegenerative diseases. This study investigated its potential toxicity in non-human primates before human clinical trials. Daily KDS2010 doses (25, 50, or 100 mg/kg) were orally administered to cynomolgus monkeys (1 animal/sex/group, 4 males and 4 females) for 2 weeks to determine the dose range. One male was moribund, and one female was found dead in the 100 mg/kg/day group. One male was also found dead in the 50 mg/kg/day group. The death was considered an adverse effect in both sexes since distal tubules/collecting duct dilation and hypertrophy in the epithelium of the papillary duct were observed in their kidneys. Based on dose range finding results, KDS2010 (10, 20, or 40 mg/kg/day) was administered orally for 4 weeks, and animals were given 2 weeks for recovery. No significant changes were observed during daily clinical observations and macro-and microscopic examinations, including body weight, food consumption, hematology, clinical chemistry, and organ weight. And, the kidney was seen as the primary target organ of KDS2010 in the 2 weeks study, but no adverse effect was observed in the 4 weeks study. Therefore, 40 mg/kg/day is considered the no-observed-adverse-effect level in both sexes of cynomolgus monkeys. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00182-4.
Collapse
Affiliation(s)
- Kyung-Tai Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeonbuk, 56212 Republic of Korea
| | - Doo-Wan Cho
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeonbuk, 56212 Republic of Korea
| | - Jae-woo Cho
- Department of Advanced Toxicology Research, Korea Institute of Toxicology (KIT), 141 Gajeong-Ro, Yuseong-Gu, Daejeon, Republic of Korea
| | - Wan-Jung Im
- Department of Advanced Toxicology Research, Korea Institute of Toxicology (KIT), 141 Gajeong-Ro, Yuseong-Gu, Daejeon, Republic of Korea
| | - Da-Hee Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeonbuk, 56212 Republic of Korea
| | - Jong-Hyun Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Young-Su Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeonbuk, 56212 Republic of Korea
| | - Su-Cheol Han
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeonbuk, 56212 Republic of Korea
| |
Collapse
|
21
|
Singh A, Singh K, Kaur J, Kaur R, Sharma A, Kaur J, Kaur U, Chadha R, Bedi PMS. Pathogenesis of Alzheimer's Disease and Diversity of 1,2,3-Triazole Scaffold in Drug Development: Design Strategies, Structural Insights, and Therapeutic Potential. ACS Chem Neurosci 2023; 14:3291-3317. [PMID: 37683129 DOI: 10.1021/acschemneuro.3c00393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease is a most prevalent form of dementia all around the globe and currently poses a significant challenge to the healthcare system. Currently available drugs only slow the progression of this disease rather than provide proper containment. Identification of multiple targets responsible for this disease in the last three decades established it as a multifactorial neurodegenerative disorder that needs novel multifunctional agents for its management and the possible reason for the failure of currently available single target clinical drugs. 1,2,3-Triazole is a miraculous nucleus in medicinal chemistry and the first choice for development of multifunctional hybrid molecules. Apart from that, it is an integral component of various drugs in clinical trials as well as in clinical practice. This review is focused on the pathogenesis of Alzheimer's disease and 1,2,3-triazole containing derivatives developed in recent decades as potential anti-Alzheimer's agents. The review will provide (A) precise insight of various established targets of Alzheimer's disease including cholinergic, amyloid, tau, monoamine oxidases, glutamate, calcium, and reactive oxygen species hypothesis and (B) design hypothesis, structure-activity relationships, and pharmacological outcomes of 1,2,3-triazole containing multifunctional anti-Alzheimer's agents. This review will provide a baseline for various research groups working on Alzheimer's drug development in designing potent, safer, and effective multifunctional anti-Alzheimer's candidates of the future.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Jashandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Ramanpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Jasleen Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Uttam Kaur
- University School of Business, Chandigarh University, Mohali, Punjab 140413, India
| | - Renu Chadha
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
- Drug and Pollution Testing Laboratory, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| |
Collapse
|
22
|
Ipe RS, Kumar S, Benny F, Jayan J, Manoharan A, Sudevan ST, George G, Gahtori P, Kim H, Mathew B. A Concise Review of the Recent Structural Explorations of Chromones as MAO-B Inhibitors: Update from 2017 to 2023. Pharmaceuticals (Basel) 2023; 16:1310. [PMID: 37765118 PMCID: PMC10534638 DOI: 10.3390/ph16091310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Monoamine oxidases (MAOs) are a family of flavin adenine dinucleotide-dependent enzymes that catalyze the oxidative deamination of a wide range of endogenous and exogenous amines. Multiple neurological conditions, including Parkinson's disease (PD) and Alzheimer's disease (AD), are closely correlated with altered biogenic amine concentrations in the brain caused by MAO. Toxic byproducts of this oxidative breakdown, including hydrogen peroxide, reactive oxygen species, and ammonia, can cause oxidative damage and mitochondrial dysfunction in brain cells. Certain MAO-B blockers have been recognized as effective treatment options for managing neurological conditions, including AD and PD. There is still a pressing need to find potent therapeutic molecules to fight these disorders. However, the focus of neurodegeneration studies has recently increased, and certain compounds are now in clinical trials. Chromones are promising structures for developing therapeutic compounds, especially in neuronal degeneration. This review focuses on the MAO-B inhibitory potential of several synthesized chromones and their structural activity relationships. Concerning the discovery of a novel class of effective chromone-based selective MAO-B-inhibiting agents, this review offers readers a better understanding of the most recent additions to the literature.
Collapse
Affiliation(s)
- Reshma Susan Ipe
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Feba Benny
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Jayalakshmi Jayan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Amritha Manoharan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Sachitra Thazhathuveedu Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Ginson George
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India;
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India; (R.S.I.); (S.K.); (F.B.); (J.J.); (A.M.); (S.T.S.); (G.G.)
| |
Collapse
|
23
|
Rullo M, La Spada G, Miniero DV, Gottinger A, Catto M, Delre P, Mastromarino M, Latronico T, Marchese S, Mangiatordi GF, Binda C, Linusson A, Liuzzi GM, Pisani L. Bioisosteric replacement based on 1,2,4-oxadiazoles in the discovery of 1H-indazole-bearing neuroprotective MAO B inhibitors. Eur J Med Chem 2023; 255:115352. [PMID: 37178666 DOI: 10.1016/j.ejmech.2023.115352] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
Following a hybridization strategy, a series of 5-substituted-1H-indazoles were designed and evaluated in vitro as inhibitors of human monoamine oxidase (hMAO) A and B. Among structural modifications, the bioisostere-based introduction of 1,2,4-oxadiazole ring returned the most potent and selective human MAO B inhibitor (compound 20, IC50 = 52 nM, SI > 192). The most promising inhibitors were studied in cell-based neuroprotection models of SH-SY5Y and astrocytes line against H2O2. Moreover, preliminary drug-like features (aqueous solubility at pH 7.4; hydrolytic stability at acidic and neutral pH) were assessed for selected 1,2,4-oxadiazoles and compared to amide analogues through RP-HPLC methods. Molecular docking simulations highlighted the crucial role of molecular flexibility in providing a better shape complementarity for compound 20 within MAO B enzymatic cleft than rigid analogue 18. Enzymatic kinetics analysis along with thermal stability curves (Tm shift = +2.9 °C) provided clues of a tight-binding mechanism for hMAO B inhibition by 20.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Gabriella La Spada
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Daniela Valeria Miniero
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andrea Gottinger
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Marco Catto
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Pietro Delre
- CNR, Institute of Crystallography, 70126, Bari, Italy
| | - Margherita Mastromarino
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy
| | - Tiziana Latronico
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Sara Marchese
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | | | - Claudia Binda
- Dept. of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Anna Linusson
- Department of Chemistry, Umeå University, 90187, Umeå, Sweden
| | - Grazia Maria Liuzzi
- Dept. of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Leonardo Pisani
- Dept. of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
24
|
Sharma P, Singh M. An ongoing journey of chalcone analogues as single and multi-target ligands in the field of Alzheimer's disease: A review with structural aspects. Life Sci 2023; 320:121568. [PMID: 36925061 DOI: 10.1016/j.lfs.2023.121568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disorder with progressive dementia and cognitive impairment. AD poses severe health challenge in elderly people and become one of the leading causes of death worldwide. It possesses complex pathophysiology with several hypotheses (cholinergic hypothesis, amyloid hypothesis, tau hypothesis, oxidative stress, mitochondrial dysfunction etc.). Several attempts have been made for the management of multifactorial AD. Acetylcholinesterase is the only target has been widely explored in the management of AD to the date. The current review set forth the chalcone based natural, semi-synthetic and synthetic compounds in the search of potential anti-Alzheimer's agents. The main highlights of current review emphasizes on chalcone target different enzymes and pathways like Acetylcholinesterase, β-secretase (BACE1), tau proteins, MAO, free radicals, Advanced glycation end Products (AGEs) etc. and their structure activity relationships contributing in the inhibition of above mentioned various targets of AD.
Collapse
Affiliation(s)
- Pratibha Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
25
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [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: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
26
|
Chatterjee P, Doré V, Pedrini S, Krishnadas N, Thota R, Bourgeat P, Ikonomovic MD, Rainey-Smith SR, Burnham SC, Fowler C, Taddei K, Mulligan R, Ames D, Masters CL, Fripp J, Rowe CC, Martins RN, Villemagne VL. Plasma Glial Fibrillary Acidic Protein Is Associated with 18F-SMBT-1 PET: Two Putative Astrocyte Reactivity Biomarkers for Alzheimer's Disease. J Alzheimers Dis 2023; 92:615-628. [PMID: 36776057 PMCID: PMC10041433 DOI: 10.3233/jad-220908] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Astrocyte reactivity is an early event along the Alzheimer's disease (AD) continuum. Plasma glial fibrillary acidic protein (GFAP), posited to reflect astrocyte reactivity, is elevated across the AD continuum from preclinical to dementia stages. Monoamine oxidase-B (MAO-B) is also elevated in reactive astrocytes observed using 18F-SMBT-1 PET in AD. OBJECTIVE The objective of this study was to evaluate the association between the abovementioned astrocyte reactivity biomarkers. METHODS Plasma GFAP and Aβ were measured using the Simoa ® platform in participants who underwent brain 18F-SMBT-1 and Aβ-PET imaging, comprising 54 healthy control (13 Aβ-PET+ and 41 Aβ-PET-), 11 mild cognitively impaired (3 Aβ-PET+ and 8 Aβ-PET-) and 6 probable AD (5 Aβ-PET+ and 1 Aβ-PET-) individuals. Linear regressions were used to assess associations of interest. RESULTS Plasma GFAP was associated with 18F-SMBT-1 signal in brain regions prone to early Aβ deposition in AD, such as the supramarginal gyrus (SG), posterior cingulate (PC), lateral temporal (LT) and lateral occipital cortex (LO). After adjusting for age, sex, APOE ɛ4 genotype, and soluble Aβ (plasma Aβ 42/40 ratio), plasma GFAP was associated with 18F-SMBT-1 signal in the SG, PC, LT, LO, and superior parietal cortex (SP). On adjusting for age, sex, APOE ɛ4 genotype and insoluble Aβ (Aβ-PET), plasma GFAP was associated with 18F-SMBT-1 signal in the SG. CONCLUSION There is an association between plasma GFAP and regional 18F-SMBT-1 PET, and this association appears to be dependent on brain Aβ load.
Collapse
Affiliation(s)
- Pratishtha Chatterjee
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Vincent Doré
- The Australian eHealth Research Centre, CSIRO, Brisbane, Queensland, Australia.,Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Steve Pedrini
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Natasha Krishnadas
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Rohith Thota
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Queensland, Australia
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pennsylvania, PA, USA.,Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, PA, USA
| | - Stephanie R Rainey-Smith
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Samantha C Burnham
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Queensland, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin Taddei
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Rachel Mulligan
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, Victoria, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jürgen Fripp
- The Australian eHealth Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,Department of Psychiatry, University of Pittsburgh, Pennsylvania, PA, USA
| | | |
Collapse
|
27
|
Discovery of reversible selective monoamine oxidase B inhibitors with anti-acetylcholinesterase activity derived from 4-oxo-N-4-diphenyl butanamides. Future Med Chem 2023; 15:189-210. [PMID: 36799336 DOI: 10.4155/fmc-2022-0169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Aim: Multitargeted drugs are essential for the treatment of various neurodegenerative disorders, because of their complex nature. This study aimed to develop novel small molecules as selective monoamine oxidase B (MAO-B) inhibitors with cholinesterase inhibition. Materials & methods: With the help of fragment-based drug design, some 4-oxo-N-4-diphenyl butanamides were designed and synthesized as MAO-B inhibitors with anti-acetylcholinesterase (AChE) activity. Results: Compound 6m showed the best neuroprotection, with reversible selective MAO-B inhibition activity (IC50 = 11.54 ± 0.64 nM). Compounds 6b, 6h, 6j, 6n and 6p (IC50 = 20.90 ± 0.50, 17.25 ± 0.90, 15.85 ± 0.16, 16.81 ± 0.85 and 25.19 ± 0.17 nM, respectively) also appeared as potent and selective MAO-B inhibitors with anti-AChE activity. Conclusion: The present study suggests potent, neuroprotective and nontoxic lead compounds as selective MAO-B inhibitors with anti-AChE activity.
Collapse
|
28
|
Nisha Aji K, Meyer JH, Rusjan PM, Mizrahi R. Monoamine Oxidase B (MAO-B): A Target for Rational Drug Development in Schizophrenia Using PET Imaging as an Example. ADVANCES IN NEUROBIOLOGY 2023; 30:335-362. [PMID: 36928857 DOI: 10.1007/978-3-031-21054-9_14] [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: 03/18/2023]
Abstract
Monoamine oxidase B (MAO-B) is an important high-density enzyme involved in the generation of oxidative stress and central in the catabolism of dopamine, particularly in brain subcortical regions with putative implications in the pathophysiology of schizophrenia. In this chapter, we review postmortem studies, preclinical models, and peripheral and genetic studies implicating MAO-B in psychosis. A literature search in PubMed was conducted and 64 studies were found to be eligible for systematic review. We found that MAO-B could be identified as a potential target in schizophrenia. Evidence comes mostly from studies of peripheral markers, showing reduced platelet MAO-B activity in schizophrenia, together with preclinical results from MAO-B knock-out mice resulting in a hyperdopaminergic state and behavioral disinhibition. However, whether brain MAO-B is altered in vivo in patients with schizophrenia remains unknown. We therefore review methodological studies involving MAO-B positron emission tomography (PET) radioligands used to quantify MAO-B in vivo in the human brain. Given the limitations of currently available treatments for schizophrenia, elucidating whether MAO-B could be used as a target for risk stratification or clinical staging in schizophrenia could allow for a rational search for newer antipsychotics and the development of new treatments.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, QC, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jeffrey H Meyer
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo M Rusjan
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Romina Mizrahi
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
29
|
Akomolafe SF, Asowata-Ayodele AM. Roasted cashew ( Anacardium occidentale L.) nut-enhanced diet forestalls cisplatin-initiated brain harm in rats. Heliyon 2022; 8:e11066. [PMID: 36276737 PMCID: PMC9578995 DOI: 10.1016/j.heliyon.2022.e11066] [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: 11/26/2021] [Revised: 03/16/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022] Open
Abstract
The incessant dose constraining symptom of the chemotherapeutic agent, cisplatin is neurotoxicity. This examination tried to explore the neuroprotective impact of roasted cashew nut-enhanced diet against brain deficits related with treatment with cisplatin. Rats were separated in to six groups: Control, CIS (cisplatin [7 mg/kg body weight, i.p]), CIS +10% CN (cisplatin plus 10% roasted cashew nut), CIS +20% CN (cisplatin plus 20% roasted cashew nut), 10% CN (10% roasted cashew nut) and 20% CN (20% roasted cashew nut) for 28 days. Key enzymes associated with brain function, including cholinesterases (AChE and BChE), monoaminergic enzyme (MAO), arginase, and adenosine deaminase (ADA), were investigated after the treatment. The following oxidative stress indicators were also measured in the rat brain: glutathione-S-transferase (GST), glutathione peroxidase (GPx), total antioxidant capacity (TAC), total thiol (T-SH), non-protein thiol (NPSH), thiobarbituric acid reactive substances (TBARS), reactive oxygen species (ROS), nitric oxide (NO), superoxide dismutase (SOD). Our outcomes demonstrated that roasted cashew nut enhanced diet showed inhibitory impact on activities of AChE, BChE, ADA, MAO and arginase in cisplatin-induced rats. The roasted cashew nut supplemented diet also boosted redox equilibrium and displayed protection against cispaltin-induced oxidative damage to rats' brains by an increase in SOD, CAT, GST and GPx activities, TAC, T-SH, NPSH and NO levels as well as a considerable drop in ROS and RBARS levels. Roasted cashew nut enhanced diet additionally forestalled neuronal degeneration in rat brain. Thus, roasted cashew nuts could be used as a nutraceutical or functional food to treat cisplatin-induced neurotoxicity. Practical applications The results show that increasing roasted cashew nut consumption can significantly improve antioxidant status, reduce lipid peroxidation, and suppress cholinesterase, adenosine deaminase, monoamine oxidase, and arginase activities in the brain under cisplatin-induced circumstances.
Collapse
Affiliation(s)
- Seun F. Akomolafe
- Department of Biochemistry, Ekiti State University, P.M.B. 5363, Ado Ekiti, Nigeria,Corresponding author.
| | | |
Collapse
|
30
|
Xu Y, Cen P, Ma L, Tian M, Zhang X, Zhang Q, Yu K, Zhang H, Gu W, He Q. Highly efficient radiosynthesis and biological evaluation of [18F]safinamide, a radiolabelled anti-parkinsonian drug for PET imaging. ChemMedChem 2022; 17:e202200472. [PMID: 36068922 DOI: 10.1002/cmdc.202200472] [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: 08/31/2022] [Indexed: 11/06/2022]
Abstract
As an add-on drug approved for Parkinson's disease treatment, safinamide has multiple functions, such as selective and reversible monoamine oxidase-B inhibition, voltage-sensitive sodium/potassium channel blockage, and glutamate release inhibition. Meanwhile, safinamide shows tremendous therapeutic potential in the context of other central nervous system diseases (e.g., ischaemic stroke, amyotrophic lateral sclerosis, depression, etc.). In this work, [18F]safinamide, which is safinamide labelled by the positron-emitting radionuclide [18F]fluorine, was synthesized automatically based on iodonium ylide precursors with high radiochemical yield and high molar activity. Density functional theory was applied to calculate the Gibbs free energy change during iodonium ylide-mediated fluorination and to interpret the effect of tetraethylammonium (TEA+) as the counter cation in these reactions to improve the nucleophilicity of [18F/19F]fluoride. In addition, positron emission tomography studies on Sprague Dawley rats were carried out to determine the imaging characteristics, pharmacokinetics, and metabolism of the [18F]safinamide radiotracer. The results displayed the complete biodistribution of the radiotracer, especially in rat brains, and revealed that [18F]safinamide has moderate brain uptake, rapid and reversible binding kinetics, and good stability.
Collapse
Affiliation(s)
- Yangyang Xu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Peili Cen
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Lijuan Ma
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Mei Tian
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Xue Zhang
- Chinese Academy of Sciences, Shenzhen Institutes of Advanced Technology, CHINA
| | - Qinghua Zhang
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Kaiwu Yu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Hong Zhang
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Wangjun Gu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Qinggang He
- Zhejiang University, Chemical Engineering, 38 Zheda Rd., 310027, Hangzhou, CHINA
| |
Collapse
|
31
|
Diaz-Amarilla P, Arredondo F, Dapueto R, Boix V, Carvalho D, Santi MD, Vasilskis E, Mesquita-Ribeiro R, Dajas-Bailador F, Abin-Carriquiry JA, Engler H, Savio E. Isolation and characterization of neurotoxic astrocytes derived from adult triple transgenic Alzheimer's disease mice. Neurochem Int 2022; 159:105403. [PMID: 35853553 DOI: 10.1016/j.neuint.2022.105403] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/02/2022] [Accepted: 07/09/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease has been considered mostly as a neuronal pathology, although increasing evidence suggests that glial cells might play a key role in the disease onset and progression. In this sense, astrocytes, with their central role in neuronal metabolism and function, are of great interest for increasing our understanding of the disease. Thus, exploring the morphological and functional changes suffered by astrocytes along the course of this disorder has great therapeutic and diagnostic potential. In this work we isolated and cultivated astrocytes from symptomatic 9-10-months-old adult 3xTg-AD mice, with the aim of characterizing their phenotype and exploring their pathogenic potential. These "old" astrocytes occurring in the 3xTg-AD mouse model of Alzheimer's Disease presented high proliferation rate and differential expression of astrocytic markers compared with controls. They were neurotoxic to primary neuronal cultures both, in neuronal-astrocyte co-cultures and when their conditioned media (ACM) was added into neuronal cultures. ACM caused neuronal GSK3β activation, changes in cytochrome c pattern, and increased caspase 3 activity, suggesting intrinsic apoptotic pathway activation. Exposure of neurons to ACM caused different subcellular responses. ACM application to the somato-dendritic domain in compartmentalised microfluidic chambers caused degeneration both locally in soma/dendrites and distally in axons. However, exposure of axons to ACM did not affect somato-dendritic nor axonal integrity. We propose that this newly described old 3xTg-AD neurotoxic astrocytic population can contribute towards the mechanistic understanding of the disease and shed light on new therapeutical opportunities.
Collapse
Affiliation(s)
- Pablo Diaz-Amarilla
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Florencia Arredondo
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay; Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay.
| | - Rosina Dapueto
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Victoria Boix
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Diego Carvalho
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - María Daniela Santi
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Elena Vasilskis
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Raquel Mesquita-Ribeiro
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH, Nottingham, UK
| | - Federico Dajas-Bailador
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH, Nottingham, UK
| | - Juan Andrés Abin-Carriquiry
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Henry Engler
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay.
| | - Eduardo Savio
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay.
| |
Collapse
|
32
|
Phytochemicals as potential inhibitors for COVID-19 revealed by molecular docking, molecular dynamic simulation and DFT studies. Struct Chem 2022; 33:1423-1443. [PMID: 35729939 PMCID: PMC9189813 DOI: 10.1007/s11224-022-01982-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/30/2022] [Indexed: 10/26/2022]
Abstract
The COVID-19 pandemic outbreak demands the designing of potential drugs as there is no specific treatment available. Thanks to their safety and effectiveness, phytochemicals have been used to treat various diseases, including antiviral therapeutics. Molecular docking is a simple, quick, and effective way to screen a variety of molecules for structure-based drug design. Here, we investigate molecular docking experiments on compounds present in plant species, Cocculus hirsutus and Rhodiola rosea and show their potential for the treatment of COVID-19. Almost all the components showed higher binding affinity than the built-in ligand, and those with significantly higher binding affinity were explored further. Molecular mechanics-based generalized born surface area calculations were used to re-rank the top candidates, rhodionidin and cocsoline, and their stability in association with viral protease was confirmed. Density functional theory was used for detailed investigations of the geometries, and electrical properties of rhodionidin and cocsoline. Using the frontier molecular orbitals method, the charge transfer within the molecule was calculated. Chemical reactivity and intermolecular interactions were studied using molecular electrostatic potential maps. These in silico discoveries will simulate the identification of powerful COVID-19 inhibitors, and similar research is likely to make a significant contribution to antiviral drug discovery. Supplementary information The online version contains supplementary material available at 10.1007/s11224-022-01982-4.
Collapse
|
33
|
Wright JP, Goodman JR, Lin YG, Lieberman BP, Clemens J, Gomez LF, Liang Q, Hoye AT, Pontecorvo MJ, Conway KA. Monoamine oxidase binding not expected to significantly affect [ 18F]flortaucipir PET interpretation. Eur J Nucl Med Mol Imaging 2022; 49:3797-3808. [PMID: 35596745 PMCID: PMC9399028 DOI: 10.1007/s00259-022-05822-9] [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: 12/07/2021] [Accepted: 04/25/2022] [Indexed: 11/28/2022]
Abstract
Purpose [18F]-labeled positron emission tomography (PET) radioligands permit in vivo assessment of Alzheimer’s disease biomarkers, including aggregated neurofibrillary tau (NFT) with [18F]flortaucipir. Due to structural similarities of flortaucipir with some monoamine oxidase A (MAO-A) inhibitors, this study aimed to evaluate flortaucipir binding to MAO-A and MAO-B and any potential impact on PET interpretation. Methods [18F]Flortaucipir autoradiography was performed on frozen human brain tissue slices, and PET imaging was conducted in rats. Dissociation constants were determined by saturation binding, association and dissociation rates were measured by kinetic binding experiments, and IC50 values were determined by competition binding. Results Under stringent wash conditions, specific [18F]flortaucipir binding was observed on tau NFT-rich Alzheimer’s disease tissue and not control tissue. In vivo PET experiments in rats revealed no evidence of [18F]flortaucipir binding to MAO-A; pre-treatment with MAO inhibitor pargyline did not impact uptake or wash-out of [18F]flortaucipir. [18F]Flortaucipir bound with low nanomolar affinity to human MAO-A in a microsomal preparation in vitro but with a fast dissociation rate relative to MAO-A ligand fluoroethyl-harmol, consistent with no observed in vivo binding in rats of [18F]flortaucipir to MAO-A. Direct binding of flortaucipir to human MAO-B was not detected in a microsomal preparation. A high concentration of flortaucipir (IC50 of 1.3 μM) was found to block binding of the MAO-B ligand safinamide to MAO-B on microsomes suggesting that, at micromolar concentrations, flortaucipir weakly binds to MAO-B in vitro. Conclusion These data suggest neither MAO-A nor MAO-B binding will contribute significantly to the PET signal in cortical target areas relevant to the interpretation of [18F]flortaucipir. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-022-05822-9.
Collapse
Affiliation(s)
- Justin P Wright
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Jason R Goodman
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Yin-Guo Lin
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Brian P Lieberman
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Jennifer Clemens
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Luis F Gomez
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Qianwa Liang
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | - Adam T Hoye
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA
| | | | - Kelly A Conway
- Avid Radiopharmaceuticals, Eli Lilly & Company, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Sung JS, Bong JH, Yun TG, Han Y, Park Y, Jung J, Lee SJ, Kang MJ, Jose J, Lee M, Pyun JC. Antibody-Mediated Screening of Peptide Inhibitors for Monoamine Oxidase-B (MAO-B) from an Autodisplayed F V Library. Bioconjug Chem 2022; 33:1166-1178. [PMID: 35587267 DOI: 10.1021/acs.bioconjchem.2c00107] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inhibitors for monoamine oxidase-B (MAO-B) were screened from an FV library with a randomized complementarity-determining region 3 (CDR3) region using a monoclonal antibody against dopamine. As the first step, the FV library was expressed on the outer membrane of E. coli by site-directed mutagenesis of the randomized CDR3 region. Among the FV library, variants with a binding affinity to monoclonal antibodies against dopamine were screened and cloned. From the comparison of the binding activity of the screened clones to a control clone with a modified FV antibody (only with CDR1 and CDR2), the CDR3 regions of screened clones were determined to directly interact with the monoclonal antibody against dopamine. These CDR3 sequences were then synthesized as mimotopes (mimicking peptides) of dopamine. The inhibitory activity of two mimotopes against MAO-B was analyzed using HeLa cells overexpressing MAO-B, as well as using activated human astrocytes; their inhibitory activity was compared to that of a commercial inhibitor of MAO-B, selegiline. The inhibition efficiency of the two mimotopes (in comparison with selegiline) was estimated to be 67.2% and 69.4% in the HeLa cells and 64.4% and 58.0% in the human astrocytes. The gene expression pattern in astrocytes after treatment with the two mimotopes was also analyzed and compared with that in the human astrocytes treated with selegiline. Finally, the interaction between two mimotopes and MAO-B was analyzed using docking simulation, and the candidate regions of MAO-B for the interaction with each mimotope were explored through the docking simulation.
Collapse
Affiliation(s)
- Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Tae Gyeong Yun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Yeonju Han
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yusun Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Soo Jeong Lee
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, PharmaCampus, Westphalian Wilhelms-University Münster, Corrensstr. 48, Münster 48149, Germany
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea.,Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea
| |
Collapse
|
35
|
Sharma K. Chromone Scaffolds in the Treatment of Alzheimer's and Parkinson's Disease: An Overview. ChemistrySelect 2022. [DOI: 10.1002/slct.202200540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kamlesh Sharma
- Department of Chemistry Faculty of Science Shree Guru Gobind Singh Tricentenary University Gurugram 122505 Haryana INDIA
| |
Collapse
|
36
|
Meyer JH, Braga J. Development and Clinical Application of Positron Emission Tomography Imaging Agents for Monoamine Oxidase B. Front Neurosci 2022; 15:773404. [PMID: 35280341 PMCID: PMC8914088 DOI: 10.3389/fnins.2021.773404] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Monoamine oxidase B (MAO-B) is a high-density protein in the brain mainly found on outer mitochondrial membranes, primarily in astroglia, but additionally in serotonergic neurons and in the substantia nigra in the midbrain. It is an enzyme that participates in the oxidative metabolism of important monoamines including dopamine, norepinephrine, benzylamine, and phenylethylamine. Elevated MAO-B density may be associated with astrogliosis and inhibiting MAO-B may reduce astrogliosis. MAO-B density is elevated in postmortem sampling of pathology for many neuropsychiatric diseases including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and alcohol use disorder. Initial development of positron emission tomography (PET) imaging agents focused on analogs of [11C]L-deprenyl, with the most commonly applied being the deuterium substituted [11C]L-deprenyl-D2. This latter radiotracer was modeled with an irreversible trapping compartment reflecting its irreversible binding to MAO-B. Subsequently, [11C]SL25.1188, a reversible binding MAO-B radioligand with outstanding properties including high specific binding and excellent reversibility was developed. [11C]SL25.1188 PET was applied to discover a substantive elevation of MAO-B binding in the prefrontal cortex in major depressive disorder (MDD) with an effect size of more than 1.5. Longer duration of MDD was associated with greater MAO-B binding throughout most gray matter regions in the brain, suggesting progressive astrogliosis. Important applications of [11C]L-deprenyl-D2 PET are detecting a 40% loss in radiotracer accumulation in cigarette smokers, and substantial occupancy of novel therapeutics like EVT301 and sembragiline. Given the number of diseases with elevations of MAO-B density and astrogliosis, and the advance of [11C]SL25.1188, clinical applications of MAO-B imaging are still at an early stage.
Collapse
Affiliation(s)
- Jeffrey H. Meyer
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Jeffrey H. Meyer,
| | - Joeffre Braga
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Rullo M, Cipolloni M, Catto M, Colliva C, Miniero DV, Latronico T, de Candia M, Benicchi T, Linusson A, Giacchè N, Altomare CD, Pisani L. Probing Fluorinated Motifs onto Dual AChE-MAO B Inhibitors: Rational Design, Synthesis, Biological Evaluation, and Early-ADME Studies. J Med Chem 2022; 65:3962-3977. [PMID: 35195417 DOI: 10.1021/acs.jmedchem.1c01784] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bioisosteric H/F or CH2OH/CF2H replacement was introduced in coumarin derivatives previously characterized as dual AChE-MAO B inhibitors to probe the effects on both inhibitory potency and drug-likeness. Along with in vitro screening, we investigated early-ADME parameters related to solubility and lipophilicity (Sol7.4, CHI7.4, log D7.4), oral bioavailability and central nervous system (CNS) penetration (PAMPA-HDM and PAMPA-blood-brain barrier (BBB) assays, Caco-2 bidirectional transport study), and metabolic liability (half-lives and clearance in microsomes, inhibition of CYP3A4). Both specific and nonspecific tissue toxicities were determined in SH-SY5Y and HepG2 lines, respectively. Compound 15 bearing a -CF2H motif emerged as a water-soluble, orally bioavailable CNS-permeant potent inhibitor of both human AChE (IC50 = 550 nM) and MAO B (IC50 = 8.2 nM, B/A selectivity > 1200). Moreover, 15 behaved as a safe and metabolically stable neuroprotective agent, devoid of cytochrome liability.
Collapse
Affiliation(s)
- Mariagrazia Rullo
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Marco Catto
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via Orabona, 4, 70125 Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | | | - Anna Linusson
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Nicola Giacchè
- TES Pharma s.r.l., Corso Vannucci 47, 06121 Perugia, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | - Leonardo Pisani
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
38
|
Youdim MBH. Site-activated multi target iron chelators with acetylcholinesterase (AChE) and monoamine oxidase (MAO) inhibitory activities for Alzheimer's disease therapy. J Neural Transm (Vienna) 2022; 129:715-721. [PMID: 35190910 DOI: 10.1007/s00702-022-02462-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/11/2022] [Indexed: 11/29/2022]
Abstract
The first class of site-activated chelators with dual inhibition of acetyl-cholinesterase (AChE) and monoamine oxidase (MAO), rationally designed for simultaneously targeting the multiple pathogenic processes in Alzheimer's disease (AD) without significantly disrupting healthy metal metabolism in the body are discussed. It is demonstrated that the novel prochelator 2 was a selective and potent MAO-A inhibitor in vitro (IC50: 0.0077 ± 0.0007 μM) with moderate inhibition of MAO-B (IC50: 7.90 ± 1.34 μM). In vitro prochelator 2 also selectively inhibited AChE in a time-dependent manner and reach maximum inhibition of AChE after 2 h preincubation (IC50: 0.52 ± 0.07 μM for AChE, versus 44.90 ± 6.10 μM for BuChE). Prochelator 2 showed little affinity for metal (Fe, Cu, and Zn) ions until it bound to and was activated by AChE that is located predominately in the brain, releasing an active iron chelator M30. M30 is an efficient chelator for metal (Fe, Cu, and Zn) ions with the capabilities to suppress oxidative stress, to selectively inhibit MAO-A and B in the brain, and to regulate cerebral biometals dyshomeostasis in vivo; M30 is also a neuroprotective-neurorestorative chelator with a broad spectrum of activities against β-amyloid (Aβ) generation, amyloid plaques and neurofibrillary tangles (NFT) formation, and Aβ aggregation induced by metal (Cu and Zn) ions. Both M30 and prochelator 2 were not toxic to Human SH-SY5Y neuroblastoma cells at low concentrations, but prochelator 2 shows limited cytotoxicity, at high concentrations. Together, these data suggest that prochelator 2 is a promise lead for simultaneously modulating multiple targets in AD.
Collapse
Affiliation(s)
- Moussa B H Youdim
- Youdim Pharmaceutical, New Northern Industrial Park, 1 Ha- Tsmika St. Stern Building, Fl-3, P.O. Box 72, 2069207, Yokneam, Israel.
| |
Collapse
|
39
|
Harada R, Furumoto S, Kudo Y, Yanai K, Villemagne VL, Okamura N. Imaging of Reactive Astrogliosis by Positron Emission Tomography. Front Neurosci 2022; 16:807435. [PMID: 35210989 PMCID: PMC8862631 DOI: 10.3389/fnins.2022.807435] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Many neurodegenerative diseases are neuropathologically characterized by neuronal loss, gliosis, and the deposition of misfolded proteins such as β-amyloid (Aβ) plaques and tau tangles in Alzheimer’s disease (AD). In postmortem AD brains, reactive astrocytes and activated microglia are observed surrounding Aβ plaques and tau tangles. These activated glial cells secrete pro-inflammatory cytokines and reactive oxygen species, which may contribute to neurodegeneration. Therefore, in vivo imaging of glial response by positron emission tomography (PET) combined with Aβ and tau PET would provide new insights to better understand the disease process, as well as aid in the differential diagnosis, and monitoring glial response disease-specific therapeutics. There are two promising targets proposed for imaging reactive astrogliosis: monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS), which are predominantly expressed in the mitochondrial membranes of astrocytes and are upregulated in various neurodegenerative conditions. PET tracers targeting these two MAO-B and I2BS have been evaluated in humans. [18F]THK-5351, which was originally designed to target tau aggregates in AD, showed high affinity for MAO-B and clearly visualized reactive astrocytes in progressive supranuclear palsy (PSP). However, the lack of selectivity of [18F]THK-5351 binding to both MAO-B and tau, severely limits its clinical utility as a biomarker. Recently, [18F]SMBT-1 was developed as a selective and reversible MAO-B PET tracer via compound optimization of [18F]THK-5351. In this review, we summarize the strategy underlying molecular imaging of reactive astrogliosis and clinical studies using MAO-B and I2BS PET tracers.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Ryuichi Harada,
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development and Aging, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Nobuyuki Okamura,
| |
Collapse
|
40
|
de Oliveira RL, Voss GT, da C. Rodrigues K, Pinz MP, Biondi JV, Becker NP, Blodorn E, Domingues WB, Larroza A, Campos VF, Alves D, Wilhelm EA, Luchese C. Prospecting for a quinoline containing selenium for comorbidities depression and memory impairment induced by restriction stress in mice. Psychopharmacology (Berl) 2022; 239:59-81. [PMID: 35013761 PMCID: PMC8747877 DOI: 10.1007/s00213-021-06039-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022]
Abstract
RATIONALE Depression is often associated with memory impairment, a clinical feature of Alzheimer's disease (AD), but no effective treatment is available. 7-Chloro-4-(phenylselanyl) quinoline (4-PSQ) has been studied in experimental models of diseases that affect the central nervous system. OBJECTIVES The pharmacological activity of 4-PSQ in depressive-like behavior associated with memory impairment induced by acute restraint stress (ARS) in male Swiss mice was evaluated. METHODS ARS is an unavoidable stress model that was applied for a period of 240 min. Ten minutes after ARS, animals were intragastrically treated with canola oil (10 ml/kg) or 4-PSQ (10 mg/kg) or positive controls (paroxetine or donepezil) (10 mg/kg). Then, after 30 min, mice were submitted to behavioral tests. Corticosterone levels were evaluated in plasma and oxidative stress parameters; monoamine oxidase (MAO)-A and MAO -B isoform activity; mRNA expression levels of kappa nuclear factor B (NF-κB); interleukin (IL)-1β, IL-18, and IL-33; phosphatidylinositol-se-kinase (PI3K); protein kinase B (AKT2), as well as acetylcholinesterase activity were evaluated in the prefrontal cortex and hippocampus. RESULTS 4-PSQ attenuated the depressive-like behavior, self-care, and memory impairment caused by ARS. Based on the evidence, we believe that effects of 4-PSQ may be associated, at least in part, with the attenuation of HPA axis activation, attenuation of alterations in the monoaminergic system, modulation of oxidative stress, reestablishment of AChE activity, modulation of the PI3K/AKT2 pathway, and reduction of neuroinflammation. CONCLUSIONS These results suggested that 4-PSQ exhibited an antidepressant-like effect and attenuated the memory impairment induced by ARS, and it is a promising molecule to treat these comorbidities.
Collapse
Affiliation(s)
- Renata L. de Oliveira
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Guilherme T. Voss
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Karline da C. Rodrigues
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Mikaela P. Pinz
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Julia V. Biondi
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Nicole P. Becker
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Eduardo Blodorn
- grid.411221.50000 0001 2134 6519Laboratório de Genômica Estrutural, Programa de Pós-Graduação Em Biotecnologia, Universidade Federal de Pelotas, Pelotas, RS Brazil
| | - William B. Domingues
- grid.411221.50000 0001 2134 6519Laboratório de Genômica Estrutural, Programa de Pós-Graduação Em Biotecnologia, Universidade Federal de Pelotas, Pelotas, RS Brazil
| | - Allya Larroza
- grid.411221.50000 0001 2134 6519Laboratório de Síntese Orgânica Limpa (LaSOL), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Programa de Pós-Graduação Em Química, Universidade Federal de Pelotas, Pelotas, RS Brazil
| | - Vinícius F. Campos
- grid.411221.50000 0001 2134 6519Laboratório de Genômica Estrutural, Programa de Pós-Graduação Em Biotecnologia, Universidade Federal de Pelotas, Pelotas, RS Brazil
| | - Diego Alves
- grid.411221.50000 0001 2134 6519Laboratório de Síntese Orgânica Limpa (LaSOL), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Programa de Pós-Graduação Em Química, Universidade Federal de Pelotas, Pelotas, RS Brazil
| | - Ethel A. Wilhelm
- grid.411221.50000 0001 2134 6519Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS CEP 96010-900 Brazil
| | - Cristiane Luchese
- Programa de Pós-Graduação Em Bioquímica E Bioprospecção (PPGBBio), Laboratório de Pesquisa Em Farmacologia Bioquímica (LaFarBio), Centro de Ciências Químicas, Farmacêuticas E de Alimentos, Universidade Federal de Pelotas, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
41
|
Segura-Aguilar J, Paris I. Mechanisms of Dopamine Oxidation and Parkinson’s Disease. HANDBOOK OF NEUROTOXICITY 2022:1433-1468. [DOI: 10.1007/978-3-031-15080-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
42
|
Chen K, Palagashvili T, Hsu W, Chen Y, Tabakoff B, Hong F, Shih AT, Shih JC. Brain injury and inflammation genes common to a number of neurological diseases and the genes involved in the genesis of GABAnergic neurons are altered in monoamine oxidase B knockout mice. Brain Res 2022; 1774:147724. [PMID: 34780749 PMCID: PMC8638699 DOI: 10.1016/j.brainres.2021.147724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 01/03/2023]
Abstract
Monoamine oxidase B (MAO B) oxidizes trace amine phenylethylamine (PEA), and neurotransmitters serotonin and dopamine in the brain. We reported previously that PEA levels increased significantly in all brain regions, but serotonin and dopamine levels were unchanged in MAO B knockout (KO) mice. PEA and dopamine are both synthesized from phenylalanine by aromatic L-amino acid decarboxylase in dopaminergic neurons in the striatum. A high concentration of PEA in the striatum may cause dopaminergic neuronal death in the absence of MAO B. We isolated the RNA from brain tissue of MAO B KO mice (2-month old) and age-matched wild type (WT) male mice and analyzed the altered genes by Affymetrix microarray. Differentially expressed genes (DEGs) in MAO B KO compared to WT mice were analyzed by Partek Genomics Suite, followed by Ingenuity Pathway Analysis (IPA) to assess their functional relationships. DEGs in MAO B KO mice are involved in brain inflammation and the genesis of GABAnergic neurons. The significant DEGs include four brain injury or inflammation genes (upregulated: Ido1, TSPO, AVP, Tdo2), five gamma-aminobutyric acid (GABA) receptors (down-regulated: GABRA2, GABRA3, GABRB1, GABRB3, GABRG3), five transcription factors related to adult neurogenesis (upregulated: Wnt7b, Hes5; down-regulated: Pax6, Tcf4, Dtna). Altered brain injury and inflammation genes in MAO B knockout mice are involved in various neurological disorders: attention deficit hyperactive disorder, panic disorder, obsessive compulsive disorder, autism, amyotrophic lateral sclerosis, Parkinson's diseases, Alzheimer's disease, bipolar affective disorder. Many were commonly involved in these disorders, indicating that there are overlapping molecular pathways.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Tamara Palagashvili
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - W Hsu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, CA, USA
| | - Boris Tabakoff
- University of Colorado Health Science Center, Denver, CO, USA
| | - Frank Hong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Abigail T Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 1985 Zonal Ave., Los Angeles, CA, USA; Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles CA, USA.
| |
Collapse
|
43
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
44
|
Mirzaei N, Davis N, Chau TW, Sastre M. Astrocyte Reactivity in Alzheimer's Disease: Therapeutic Opportunities to Promote Repair. Curr Alzheimer Res 2021; 19:1-15. [PMID: 34719372 DOI: 10.2174/1567205018666211029164106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/02/2021] [Accepted: 07/31/2021] [Indexed: 11/22/2022]
Abstract
Astrocytes are fast climbing the ladder of importance in neurodegenerative disorders, particularly in Alzheimer's disease (AD), with the prominent presence of reactive astrocytes sur- rounding amyloid β- plaques, together with activated microglia. Reactive astrogliosis, implying morphological and molecular transformations in astrocytes, seems to precede neurodegeneration, suggesting a role in the development of the disease. Single-cell transcriptomics has recently demon- strated that astrocytes from AD brains are different from "normal" healthy astrocytes, showing dys- regulations in areas such as neurotransmitter recycling, including glutamate and GABA, and im- paired homeostatic functions. However, recent data suggest that the ablation of astrocytes in mouse models of amyloidosis results in an increase in amyloid pathology as well as in the inflammatory profile and reduced synaptic density, indicating that astrocytes mediate neuroprotective effects. The idea that interventions targeting astrocytes may have great potential for AD has therefore emerged, supported by a range of drugs and stem cell transplantation studies that have successfully shown a therapeutic effect in mouse models of AD. In this article, we review the latest reports on the role and profile of astrocytes in AD brains and how manipulation of astrocytes in animal mod- els has paved the way for the use of treatments enhancing astrocytic function as future therapeutic avenues for AD.
Collapse
Affiliation(s)
- Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048. United States
| | - Nicola Davis
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Tsz Wing Chau
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| |
Collapse
|
45
|
Babić Leko M, Nikolac Perković M, Nedić Erjavec G, Klepac N, Švob Štrac DK, Borovečki F, Pivac N, Hof PR, Šimić G. Association of the MAOB rs1799836 Single Nucleotide Polymorphism and APOE ɛ4 Allele in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:585-594. [PMID: 34533445 DOI: 10.2174/1567205018666210917162843] [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: 05/20/2020] [Revised: 07/01/2021] [Accepted: 08/22/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dopaminergic system is functionally compromised in Alzheimer's dis-ease (AD). The activity of monoamine oxidase B (MAOB), the enzyme involved in the degradation of dopamine, is increased during AD. Also, increased expression of MAOB occurs in the post-mortem hippocampus and neocortex of patients with AD. The MAOB rs1799836 polymorphism modulates MAOB transcription, consequently influencing protein translation and MAOB activity. We recently showed that cerebrospinal fluid levels of amyloid β1-42 are decreased in patients carry- ing the A allele in MAOB rs1799836 polymorphism. OBJECTIVE The present study compares MAOB rs1799836 polymorphism and APOE, the only con- firmed genetic risk factor for sporadic AD. METHOD We included 253 participants, 127 of whom had AD, 57 had mild cognitive impairment, 11 were healthy controls, and 58 suffered from other primary causes of dementia. MAOB and APOE polymorphisms were determined using TaqMan SNP Genotyping Assays. RESULTS We observed that the frequency of APOE ɛ4/ɛ4 homozygotes and APOE ɛ4 carriers is sig- nificantly increased among patients carrying the AA MAOB rs1799836 genotype. CONCLUSION These results indicate that the MAOB rs1799836 polymorphism is a potential genetic biomarker of AD and a potential target for the treatment of decreased dopaminergic transmission and cognitive deterioration in AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
46
|
Mitamura K, Norikane T, Yamamoto Y, Miyake K, Nishiyama Y. Increased Uptake of 18F-THK5351 in Glioblastoma But Not in Primary Central Nervous System Lymphoma. Clin Nucl Med 2021; 46:772-773. [PMID: 34028419 DOI: 10.1097/rlu.0000000000003699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT 18F-THK5351 was initially developed to target tau aggregates in neurofibrillary tangles. However, it was also shown to bind to an enzyme, monoamine oxidase B, which is highly expressed in the outer mitochondrial membrane of astrocytes. In a case with glioblastoma, both l-[methyl-11C]-methionine PET and 18F-THK5351 PET showed increased uptake in the tumor. On the other hand, in another case with primary central nervous system lymphoma, l-[methyl-11C]-methionine PET showed increased uptake in the tumor, but 18F-THK5351 PET showed no abnormal uptake in the tumor. 18F-THK5351 PET might be helpful in differentiating between glioblastoma and primary central nervous system lymphoma.
Collapse
Affiliation(s)
| | | | | | - Keisuke Miyake
- Neurological Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | |
Collapse
|
47
|
In vitro and in vivo evaluation of fluorinated indanone derivatives as potential positron emission tomography agents for the imaging of monoamine oxidase B in the brain. Bioorg Med Chem Lett 2021; 48:128254. [PMID: 34256118 DOI: 10.1016/j.bmcl.2021.128254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
Monoamine oxidases (MAOs) play a key role in the metabolism of major monoamine neurotransmitters. In particular, the upregulation of MAO-B in Parkinson's disease, Alzheimer's disease and cancer augmented the development of selective MAO-B inhibitors for diagnostic and therapeutic purposes, such as the anti-parkinsonian MAO-B irreversible binder l-deprenyl (Selegiline®). Herein we report on the synthesis of novel fluorinated indanone derivatives for PET imaging of MAO-B in the brain. Out of our series, the derivatives 6, 8, 9 and 13 are amongst the most affine and selective ligands for MAO-B reported so far. For the derivative 6-((3-fluorobenzyl)oxy)-2,3-dihydro-1H-inden-1-one (6) exhibiting an outstanding affinity (KiMAO-B = 6 nM), an automated copper-mediated radiofluorination starting from the pinacol boronic ester 17 is described. An in vitro screening in different species revealed a MAO-B region-specific accumulation of [18F]6 in rats and piglets in comparison to L-[3H]deprenyl. The pre-clinical in vivo assessment of [18F]6 in mice demonstrated the potential of indanones to readily cross the blood-brain barrier. Nonetheless, parallel in vivo metabolism studies indicated the presence of blood-brain barrier metabolites, thus arguing for further structural modifications. With the matching analytical profiles of the radiometabolite analysis from the in vitro liver microsome studies and the in vivo evaluation, the structure's elucidation of the blood-brain barrier penetrant radiometabolites is possible and will serve as basis for the development of new indanone derivatives suitable for the PET imaging of MAO-B.
Collapse
|
48
|
Ikeda M, Okamoto K, Suzuki K, Takai E, Kasahara H, Furuta N, Furuta M, Tashiro Y, Shimizu C, Takatama S, Naito I, Sato M, Sakai Y, Takahashi M, Amari M, Takatama M, Higuchi T, Tsushima Y, Yokoo H, Kurabayashi M, Ishibashi S, Ishii K, Ikeda Y. Recurrent Lobar Hemorrhages and Multiple Cortical Superficial Siderosis in a Patient of Alzheimer's Disease With Homozygous APOE ε2 Allele Presenting Hypobetalipoproteinemia and Pathological Findings of 18F-THK5351 Positron Emission Tomography: A Case Report. Front Neurol 2021; 12:645625. [PMID: 34305778 PMCID: PMC8294698 DOI: 10.3389/fneur.2021.645625] [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: 01/15/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
In Alzheimer's disease, the apolipoprotein E gene (APOE) ε2 allele is a protective genetic factor, whereas the APOE ε4 allele is a genetic risk factor. However, both the APOE ε2 and the APOE ε4 alleles are genetic risk factors for lobar intracerebral hemorrhage. The reasons for the high prevalence of lobar intracerebral hemorrhage and the low prevalence of Alzheimer's disease with the APOE ε2 allele remains unknown. Here, we describe the case of a 79-year-old Japanese female with Alzheimer's disease, homozygous for the APOE ε2 allele. This patient presented with recurrent lobar hemorrhages and multiple cortical superficial siderosis. The findings on the 11C-labeled Pittsburgh Compound B-positron emission tomography (PET) were characteristic of Alzheimer's disease. 18F-THK5351 PET revealed that the accumulation of 18F-THK 5351 in the right pyramidal tract at the pontine level, the cerebral peduncle of the midbrain, and the internal capsule, reflecting the lesions of the previous lobar intracerebral hemorrhage in the right frontal lobe. Moreover, 18F-THK5351 accumulated in the bilateral globus pallidum, amygdala, caudate nuclei, and the substantia nigra of the midbrain, which were probably off-target reaction, by binding to monoamine oxidase B (MAO-B). 18F-THK5351 were also detected in the periphery of prior lobar hemorrhages and a cortical subarachnoid hemorrhage, as well as in some, but not all, areas affected by cortical siderosis. Besides, 18F-THK5351 retentions were observed in the bilateral medial temporal cortices and several cortical areas without cerebral amyloid angiopathy or prior hemorrhages, possibly where tau might accumulate. This is the first report of a patient with Alzheimer's disease, carrying homozygous APOE ε2 allele and presenting with recurrent lobar hemorrhages, multiple cortical superficial siderosis, and immunohistochemically vascular amyloid β. The 18F-THK5351 PET findings suggested MAO-B concentrated regions, astroglial activation, Waller degeneration of the pyramidal tract, neuroinflammation due to CAA related hemorrhages, and possible tau accumulation.
Collapse
Affiliation(s)
- Masaki Ikeda
- Division of General Education (Neurology), Faculty of Health & Medical Care, Saitama Medical University, Saitama, Japan.,Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan.,Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koichi Okamoto
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Keiji Suzuki
- Department of Pathology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Eriko Takai
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroo Kasahara
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Natsumi Furuta
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Minori Furuta
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yuichi Tashiro
- Department of Neurology, Mito Medical Center, Mito, Japan
| | - Chisato Shimizu
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Shin Takatama
- Department of Neurosurgery, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Isao Naito
- Department of Neurosurgery, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Mie Sato
- Department of Anesthesiology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Yasujiro Sakai
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Manabu Takahashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masakuni Amari
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Masamitsu Takatama
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hideaki Yokoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Masahiko Kurabayashi
- Department of Pathology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Kenji Ishii
- Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
49
|
Behl T, Kaur D, Sehgal A, Singh S, Sharma N, Zengin G, Andronie-Cioara FL, Toma MM, Bungau S, Bumbu AG. Role of Monoamine Oxidase Activity in Alzheimer's Disease: An Insight into the Therapeutic Potential of Inhibitors. Molecules 2021; 26:molecules26123724. [PMID: 34207264 PMCID: PMC8234097 DOI: 10.3390/molecules26123724] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 02/03/2023] Open
Abstract
Despite not being utilized as considerably as other antidepressants in the therapy of depression, the monoamine oxidase inhibitors (MAOIs) proceed to hold a place in neurodegeneration and to have a somewhat broad spectrum in respect of the treatment of neurological and psychiatric conditions. Preclinical and clinical studies on MAOIs have been developing in recent times, especially on account of rousing discoveries manifesting that these drugs possess neuroprotective activities. The altered brain levels of monoamine neurotransmitters due to monoamine oxidase (MAO) are directly associated with various neuropsychiatric conditions like Alzheimer’s disease (AD). Activated MAO induces the amyloid-beta (Aβ) deposition via abnormal cleavage of the amyloid precursor protein (APP). Additionally, activated MAO contributes to the generation of neurofibrillary tangles and cognitive impairment due to neuronal loss. No matter the attention of researchers on the participation of MAOIs in neuroprotection has been on monoamine oxidase-B (MAO-B) inhibitors, there is a developing frame of proof indicating that monoamine oxidase-A (MAO-A) inhibitors may also play a role in neuroprotection. The therapeutic potential of MAOIs alongside the complete understanding of the enzyme’s physiology may lead to the future advancement of these drugs.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (D.K.); (A.S.); (S.S.); (N.S.)
- Correspondence: (T.B.); (S.B.)
| | - Dapinder Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (D.K.); (A.S.); (S.S.); (N.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (D.K.); (A.S.); (S.S.); (N.S.)
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (D.K.); (A.S.); (S.S.); (N.S.)
| | - Neelam Sharma
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; (D.K.); (A.S.); (S.S.); (N.S.)
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, 42130 Konya, Turkey;
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Mirela Marioara Toma
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
- Doctoral School of Biomedical Sciences, University of Oradea, 410073 Oradea, Romania
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
- Doctoral School of Biomedical Sciences, University of Oradea, 410073 Oradea, Romania
- Correspondence: (T.B.); (S.B.)
| | - Adrian Gheorghe Bumbu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| |
Collapse
|
50
|
Rahman MS, Uddin MS, Rahman MA, Samsuzzaman M, Behl T, Hafeez A, Perveen A, Barreto GE, Ashraf GM. Exploring the Role of Monoamine Oxidase Activity in Aging and Alzheimer's Disease. Curr Pharm Des 2021; 27:4017-4029. [PMID: 34126892 DOI: 10.2174/1381612827666210612051713] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Monoamine oxidases (MAOs) are a family of flavin adenine dinucleotide-dependent enzymes that exert a crucial role in the metabolism of neurotransmitters of the central nervous system. The impaired function of MAOs is associated with copious brain diseases. The alteration of monoamine metabolism is a characteristics feature of aging. MAO plays a crucial role in the pathogenesis of Alzheimer's disease (AD) - a progressive neurodegenerative disorder associated with an excessive accumulation of amyloid-beta (Aβ) peptide and neurofibrillary tangles (NFTs). Activated MAO has played a critical role in the development of amyloid plaques from Aβ, as well as the formation of the NFTs. In the brain, MAO mediated metabolism of monoamines is the foremost source of reactive oxygen species formation. The elevated level of MAO-B expression in astroglia has been reported in the AD brains adjacent to amyloid plaques. Increased MAO-B activity in the cortical and hippocampal regions is associated with AD. This review describes the pathogenic mechanism of MAOs in aging as well as the development and propagation of Alzheimer's pathology.
Collapse
Affiliation(s)
- Md Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul. Korea
| | - Md Samsuzzaman
- Department of Food and Life Science, Pukyong National University, Busan 48513. Korea
| | - Tapan Behl
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick. Ireland
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah. Saudi Arabia
| |
Collapse
|